1
|
Myllymäki SM, Lan Q, Mikkola ML. Embryonic Mammary Gland Morphogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:9-27. [PMID: 39821018 DOI: 10.1007/978-3-031-70875-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Embryonic mammary gland development unfolds with the specification of bilateral mammary lines, thereafter progressing through placode, bud, and sprout stages before branching morphogenesis. Extensive epithelial-mesenchymal interactions guide morphogenesis from embryogenesis to adulthood. Two distinct mesenchymal tissues are involved, the primary mammary mesenchyme that harbors mammary inductive capacity, and the secondary mesenchyme, the precursor of the adult stroma. Placode and bud stages are morphologically similar with other ectodermal appendages like the hair follicle, reflecting the mammary gland's assumed evolutionary origin from an ancestral hair follicle-associated glandular unit. The shared features extend to signalling cascades such as the Wnt/β-catenin, fibroblast growth factor (Fgf), and ectodysplasin (Eda) pathways, while pathways unique to mammary gland include parathyroid hormone-like hormone (Pthlh) signalling and Hedgehog activity suppression. Mammary gland branching is highly non-stereotypic, achieved by the dynamic use of two distinct modes of branching: tip bifurcation and side branching and stochastic branch point formation. The cellular mechanisms driving the initial morphogenetic steps are slowly beginning to be unravelled. During placode and bud stages, mammary primordium predominantly grows through cell influx, while sprouting correlates with heightened proliferation. Branch elongation is driven by directional cell migration combined with differential cell motility and proliferation supplying the reservoir of migratory cells, whereas a bifurcating tip is associated with localized repression of the cell cycle and cell motility. Numerous similarities exist between embryonic programs and breast tumorigenesis, spanning cellular plasticity, epithelial-stromal interactions, and molecular regulators. Understanding embryonic mammogenesis may provide insights into how normal developmental processes can go awry, leading to malignancy, or how they can be reversed to prevent cancer progression.
Collapse
Affiliation(s)
- Satu-Marja Myllymäki
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
- Faculty of Medicine, Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Qiang Lan
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Marja L Mikkola
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
2
|
Zheng X, Huang H, Zhou Z, Guo W, Yang G, Chen Z, Chen D, Chen Y, Yuan G. Axin1 regulates tooth root development by inhibiting AKT1-mTORC1 activation and Shh translation in Hertwig's epithelial root sheath. Development 2024; 151:dev202899. [PMID: 39344774 DOI: 10.1242/dev.202899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Hertwig's epithelial root sheath (HERS) interacts with dental apical mesenchyme and guides development of the tooth root, which is integral to the function of the whole tooth. However, the key genes in HERS essential for root development are understudied. Here, we show that Axin1, a scaffold protein that negatively regulates canonical Wnt signaling, is strongly expressed in the HERS. Axin1 ablation in the HERS of mice leads to defective root development, but in a manner independent of canonical Wnt signaling. Further studies reveal that Axin1 in the HERS negatively regulates the AKT1-mTORC1 pathway through binding to AKT1, leading to inhibition of ribosomal biogenesis and mRNA translation. Sonic hedgehog (Shh) protein, a morphogen essential for root development, is over-synthesized by upregulated mTORC1 activity upon Axin1 inactivation. Importantly, either haploinsufficiency of the mTORC1 subunit Rptor or pharmacological inhibition of Shh signaling can rescue the root defects in Axin1 mutant mice. Collectively, our data suggest that, independently of canonical Wnt signaling, Axin1 controls ribosomal biogenesis and selective mRNA translation programs via AKT1-mTORC1 signaling during tooth root development.
Collapse
Affiliation(s)
- Xiaoyu Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Hongcan Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Zhipeng Zhou
- National Key Laboratory of Agricultural Microbiology, College of Life Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Weihua Guo
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan 610041, China
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan 610041, China
| | - Guobin Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institutes of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Guohua Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| |
Collapse
|
3
|
Kalyanakrishnan K, Beaudin A, Jetté A, Ghezelbash S, Hotea DI, Chen J, Lefrançois P, Laurin M. ARHGEF3 Regulates Hair Follicle Morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612256. [PMID: 39314354 PMCID: PMC11419159 DOI: 10.1101/2024.09.13.612256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
During embryogenesis, cells arrange into precise patterns that enable tissues and organs to develop specialized functions. Despite its critical importance, the molecular choreography behind these collective cellular behaviors remains elusive, posing a major challenge in developmental biology and limiting advances in regenerative medicine. By using the mouse hair follicle as a mini-organ system to study the formation of bud-like structures during embryonic development, our work uncovers a crucial role for the Rho GTPase regulator ARHGEF3 in hair follicle morphogenesis. We demonstrate that Arhgef3 expression is upregulated at the onset of hair follicle placode formation. In Arhgef3 knockout animals, we observed defects in placode compaction, leading to impaired hair follicle downgrowth. Through cell culture models, we show that ARHGEF3 promotes F-actin accumulation at the cell cortex and P-cadherin enrichment at cell-cell junctions. Collectively, our study identifies ARHGEF3 as a new regulator of cell shape rearrangements during hair placode morphogenesis, warranting further exploration of its role in other epithelial appendages that arise from similar developmental processes.
Collapse
Affiliation(s)
- Krithika Kalyanakrishnan
- Centre de recherche du CHU de Québec - Université Laval, axe Oncologie, Québec, Canada
- Programme de biologie moléculaire et cellulaire, Université Laval
- Département de biologie moléculaire, biochimie médicale et pathologie, Université Laval
- Faculté de médecine, Université Laval
- Centre de recherche sur le cancer de l'Université Laval (CRC)
- Centre de recherche en organogénèse expérimentale (LOEX)
| | - Amy Beaudin
- Centre de recherche du CHU de Québec - Université Laval, axe Oncologie, Québec, Canada
- Programme de biologie moléculaire et cellulaire, Université Laval
- Département de biologie moléculaire, biochimie médicale et pathologie, Université Laval
- Faculté de médecine, Université Laval
- Centre de recherche sur le cancer de l'Université Laval (CRC)
- Centre de recherche en organogénèse expérimentale (LOEX)
| | - Alexandra Jetté
- Centre de recherche du CHU de Québec - Université Laval, axe Oncologie, Québec, Canada
- Centre de recherche sur le cancer de l'Université Laval (CRC)
- Centre de recherche en organogénèse expérimentale (LOEX)
| | - Sarah Ghezelbash
- Cancer Axis, Lady Davis Institute for Medical Research, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Québec, Canada
| | - Diana Ioana Hotea
- Centre de recherche du CHU de Québec - Université Laval, axe Oncologie, Québec, Canada
- Programme de biologie moléculaire et cellulaire, Université Laval
- Département de biologie moléculaire, biochimie médicale et pathologie, Université Laval
- Faculté de médecine, Université Laval
- Centre de recherche sur le cancer de l'Université Laval (CRC)
- Centre de recherche en organogénèse expérimentale (LOEX)
| | - Jie Chen
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, Urbana, IL 61801
| | - Philippe Lefrançois
- Cancer Axis, Lady Davis Institute for Medical Research, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Québec, Canada
- Division of Dermatology, Department of Medicine, McGill University, Montréal, QC, Canada
| | - Mélanie Laurin
- Centre de recherche du CHU de Québec - Université Laval, axe Oncologie, Québec, Canada
- Programme de biologie moléculaire et cellulaire, Université Laval
- Département de biologie moléculaire, biochimie médicale et pathologie, Université Laval
- Faculté de médecine, Université Laval
- Centre de recherche sur le cancer de l'Université Laval (CRC)
- Centre de recherche en organogénèse expérimentale (LOEX)
| |
Collapse
|
4
|
Satta JP, Lan Q, Taketo MM, Mikkola ML. Stabilization of Epithelial β-Catenin Compromises Mammary Cell Fate Acquisition and Branching Morphogenesis. J Invest Dermatol 2024; 144:1223-1237.e10. [PMID: 38159590 DOI: 10.1016/j.jid.2023.11.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 01/03/2024]
Abstract
The Wnt/β-catenin pathway plays a critical role in cell fate specification, morphogenesis, and stem cell activation across diverse tissues, including the skin. In mammals, the embryonic surface epithelium gives rise to the epidermis as well as the associated appendages including hair follicles and mammary glands, both of which depend on epithelial Wnt/β-catenin activity for initiation of their development. Later on, Wnts are thought to enhance mammary gland growth and branching, whereas in hair follicles, they are essential for hair shaft formation. In this study, we report a strong downregulation of epithelial Wnt/β-catenin activity as the mammary bud progresses to branching. We show that forced activation of epithelial β-catenin severely compromises embryonic mammary gland branching. However, the phenotype of conditional Lef1-deficient embryos implies that a low level of Wnt/β-catenin activity is necessary for mammary cell survival. Transcriptomic profiling suggests that sustained high β-catenin activity leads to maintenance of mammary bud gene signature at the expense of outgrowth/branching gene signature. In addition, it leads to upregulation of epidermal differentiation genes. Strikingly, we find a partial switch to hair follicle fate early on upon stabilization of β-catenin, suggesting that the level of epithelial Wnt/β-catenin signaling activity may contribute to the choice between skin appendage identities.
Collapse
Affiliation(s)
- Jyoti Prabha Satta
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (HILIFE), University of Helsinki, Helsinki, Finland
| | - Qiang Lan
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (HILIFE), University of Helsinki, Helsinki, Finland
| | - Makoto Mark Taketo
- Colon Cancer Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Marja L Mikkola
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (HILIFE), University of Helsinki, Helsinki, Finland.
| |
Collapse
|
5
|
Lee O, Bazzi LA, Xu Y, Pearson E, Wang M, Hosseini O, Akasha AM, Choi JN, Karlan S, Pilewskie M, Kocherginsky M, Benante K, Helland T, Mellgren G, Dimond E, Perloff M, Heckman-Stoddard BM, Khan SA. A randomized Phase I pre-operative window trial of transdermal endoxifen in women planning mastectomy: Evaluation of dermal safety, intra-mammary drug distribution, and biologic effects. Biomed Pharmacother 2024; 171:116105. [PMID: 38171245 DOI: 10.1016/j.biopha.2023.116105] [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: 09/19/2023] [Revised: 12/17/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024] Open
Abstract
Breast cancer prevention only requires local exposure of the breast to active drug. However, oral preventive agents entail systemic exposure, causing adverse effects that limit acceptance by high-risk women. Drug-delivery through the breast skin is an attractive option, but requires demonstration of dermal safety and drug distribution throughout the breast. We formulated the tamoxifen metabolite (E/Z)-endoxifen for transdermal delivery and tested it in a placebo-controlled, double-blinded Phase I trial with dose escalation from 10 to 20 mg daily. The primary endpoint was dermal toxicity. Thirty-two women planning mastectomy were randomized (2:1) to endoxifen-gel or placebo-gel applied to both breasts for 3-5 weeks. Both doses of endoxifen-gel incurred no dermal or systemic toxicity compared to placebo. All endoxifen-treated breasts contained the drug at each of five sampling locations; the median per-person tissue concentration in the treated participants was 0.6 ng/g (IQR 0.4-1.6), significantly higher (p < 0.001) than the median plasma concentration (0.2 ng/mL, IQR 0.2-0.2). The median ratio of the more potent (Z)-isomer to (E)-isomer at each breast location was 1.50 (IQR 0.96-2.54, p < 0.05). No discernible effects of breast size or adiposity on tissue concentrations were observed. At the endoxifen doses and duration used, and the tissue concentration achieved, we observed a non-significant overall reduction of tumor proliferation (Ki67 LI) and significant downregulation of gene signatures known to promote cancer invasion (FN1, SERPINH1, PLOD2, PDGFA, ITGAV) (p = 0.03). Transdermal endoxifen is an important potential breast cancer prevention agent but formulations with better dermal penetration are needed.
Collapse
Affiliation(s)
- Oukseub Lee
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Latifa A Bazzi
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yanfei Xu
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Erik Pearson
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Minhua Wang
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Omid Hosseini
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Azza M Akasha
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jennifer Nam Choi
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Scott Karlan
- Saul and Joyce Brandman Breast Center, Cedars-Sinai Medical Center, West Hollywood, CA, USA
| | | | - Masha Kocherginsky
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kelly Benante
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Thomas Helland
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Eileen Dimond
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Marjorie Perloff
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | | | - Seema A Khan
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
6
|
Sahu S, Sahoo S, Sullivan T, O'Sullivan TN, Turan S, Albaugh ME, Burkett S, Tran B, Salomon DS, Kozlov SV, Koehler KR, Jolly MK, Sharan SK. Spatiotemporal modulation of growth factors directs the generation of multilineage mouse embryonic stem cell-derived mammary organoids. Dev Cell 2024; 59:175-186.e8. [PMID: 38159568 PMCID: PMC10872289 DOI: 10.1016/j.devcel.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 09/20/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
Ectodermal appendages, such as the mammary gland (MG), are thought to have evolved from hair-associated apocrine glands to serve the function of milk secretion. Through the directed differentiation of mouse embryonic stem cells (mESCs), here, we report the generation of multilineage ESC-derived mammary organoids (MEMOs). We adapted the skin organoid model, inducing the dermal mesenchyme to transform into mammary-specific mesenchyme via the sequential activation of Bone Morphogenetic Protein 4 (BMP4) and Parathyroid Hormone-related Protein (PTHrP) and inhibition of hedgehog (HH) signaling. Using single-cell RNA sequencing, we identified gene expression profiles that demonstrate the presence of mammary-specific epithelial cells, fibroblasts, and adipocytes. MEMOs undergo ductal morphogenesis in Matrigel and can reconstitute the MG in vivo. Further, we demonstrate that the loss of function in placode regulators LEF1 and TBX3 in mESCs results in impaired skin and MEMO generation. In summary, our MEMO model is a robust tool for studying the development of ectodermal appendages, and it provides a foundation for regenerative medicine and disease modeling.
Collapse
Affiliation(s)
- Sounak Sahu
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Sarthak Sahoo
- Department of Bioengineering, Indian Institute of Science, Bengaluru 560012, India
| | - Teresa Sullivan
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - T Norene O'Sullivan
- Centre for Advanced Preclinical Research (CAPR), National Cancer Institute, Frederick, MD 21702, USA
| | - Sevilay Turan
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Mary E Albaugh
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Sandra Burkett
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Bao Tran
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - David S Salomon
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Serguei V Kozlov
- Centre for Advanced Preclinical Research (CAPR), National Cancer Institute, Frederick, MD 21702, USA; Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Karl R Koehler
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA; Department of Otolaryngology, Department of Plastic & Oral Surgery, and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bengaluru 560012, India
| | - Shyam K Sharan
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; Centre for Advanced Preclinical Research (CAPR), National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
7
|
Dingwall HL, Tomizawa RR, Aharoni A, Hu P, Qiu Q, Kokalari B, Martinez SM, Donahue JC, Aldea D, Mendoza M, Glass IA, Wu H, Kamberov YG. Sweat gland development requires an eccrine dermal niche and couples two epidermal programs. Dev Cell 2024; 59:20-32.e6. [PMID: 38096824 PMCID: PMC10872420 DOI: 10.1016/j.devcel.2023.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/02/2023] [Accepted: 11/14/2023] [Indexed: 12/22/2023]
Abstract
Eccrine sweat glands are indispensable for human thermoregulation and, similar to other mammalian skin appendages, form from multipotent epidermal progenitors. Limited understanding of how epidermal progenitors specialize to form these vital organs has precluded therapeutic efforts toward their regeneration. Herein, we applied single-nucleus transcriptomics to compare the expression content of wild-type, eccrine-forming mouse skin to that of mice harboring a skin-specific disruption of Engrailed 1 (En1), a transcription factor that promotes eccrine gland formation in humans and mice. We identify two concurrent but disproportionate epidermal transcriptomes in the early eccrine anlagen: one that is shared with hair follicles and one that is En1 dependent and eccrine specific. We demonstrate that eccrine development requires the induction of a dermal niche proximal to each developing gland in humans and mice. Our study defines the signatures of eccrine identity and uncovers the eccrine dermal niche, setting the stage for targeted regeneration and comprehensive skin repair.
Collapse
Affiliation(s)
- Heather L Dingwall
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Reiko R Tomizawa
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Adam Aharoni
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Peng Hu
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Qi Qiu
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Blerina Kokalari
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Joan C Donahue
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Aldea
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Meryl Mendoza
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ian A Glass
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Hao Wu
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Yana G Kamberov
- Department of Genetics, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Box C, Pennington C, Hare S, Porter S, Edwards D, Eccles S, Crompton M, Harvey A. Brk/PTK6 and Involucrin Expression May Predict Breast Cancer Cell Responses to Vitamin D3. Int J Mol Sci 2023; 24:10757. [PMID: 37445934 DOI: 10.3390/ijms241310757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
The process of human embryonic mammary development gives rise to the structures in which mammary cells share a developmental lineage with skin epithelial cells such as keratinocytes. As some breast carcinomas have previously been shown to express high levels of involucrin, a marker of keratinocyte differentiation, we hypothesised that some breast tumours may de-differentiate to a keratinocyte-derived 'evolutionary history'. To confirm our hypothesis, we investigated the frequency of involucrin expression along with that of Brk, a tyrosine kinase expressed in up to 86% of breast carcinomas whose normal expression patterns are restricted to differentiating epithelial cells, most notably those in the skin (keratinocytes) and the gastrointestinal tract. We found that involucrin, a keratinocyte differentiation marker, was expressed in a high proportion (78%) of breast carcinoma samples and cell lines. Interestingly, tumour samples found to express high levels of involucrin were also shown to express Brk. 1,25-dihydroxyvitamin D3, a known differentiation agent and potential anti-cancer agent, decreased proliferation in the breast cancer cell lines that expressed both involucrin and Brk, whereas the Brk/involucrin negative cell lines tested were less susceptible. In addition, responses to 1,25-dihydroxyvitamin D3 were not correlated with vitamin D receptor expression. These data contribute to the growing body of evidence suggesting that cellular responses to 1,25-dihydroxyvitamin D3 are potentially independent of vitamin D receptor status and provide an insight into potential markers, such as Brk and/or involucrin that could predict therapeutic responses to 1,25-dihydroxyvitamin D3.
Collapse
Affiliation(s)
- Carol Box
- The Cancer Research UK Cancer Therapeutics Unit, McElwain Laboratories, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Caroline Pennington
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Stephen Hare
- Centre for Genome Engineering and Maintenance, Institute for Health Medicine and Environment, Brunel University London, Uxbridge UB8 3PH, UK
| | - Sarah Porter
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Dylan Edwards
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Suzanne Eccles
- The Cancer Research UK Cancer Therapeutics Unit, McElwain Laboratories, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Mark Crompton
- School of Biological Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Amanda Harvey
- Centre for Genome Engineering and Maintenance, Institute for Health Medicine and Environment, Brunel University London, Uxbridge UB8 3PH, UK
| |
Collapse
|
9
|
Sulic AM, Das Roy R, Papagno V, Lan Q, Saikkonen R, Jernvall J, Thesleff I, Mikkola ML. Transcriptomic landscape of early hair follicle and epidermal development. Cell Rep 2023; 42:112643. [PMID: 37318953 DOI: 10.1016/j.celrep.2023.112643] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 04/04/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023] Open
Abstract
Morphogenesis of ectodermal organs, such as hair, tooth, and mammary gland, starts with the formation of local epithelial thickenings, or placodes, but it remains to be determined how distinct cell types and differentiation programs are established during ontogeny. Here, we use bulk and single-cell transcriptomics and pseudotime modeling to address these questions in developing hair follicles and epidermis and produce a comprehensive transcriptomic profile of cellular populations in the hair placode and interplacodal epithelium. We report previously unknown cell populations and marker genes, including early suprabasal and genuine interfollicular basal markers, and propose the identity of suprabasal progenitors. By uncovering four different hair placode cell populations organized in three spatially distinct areas, with fine gene expression gradients between them, we posit early biases in cell fate establishment. This work is accompanied by a readily accessible online tool to stimulate further research on skin appendages and their progenitors.
Collapse
Affiliation(s)
- Ana-Marija Sulic
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Rishi Das Roy
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Verdiana Papagno
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Qiang Lan
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Riikka Saikkonen
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Jukka Jernvall
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland; Department of Geosciences and Geography, University of Helsinki, 00014 Helsinki, Finland
| | - Irma Thesleff
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Marja L Mikkola
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland.
| |
Collapse
|
10
|
Wu C, Li J, Xu X, Xu Q, Qin C, Liu G, Wei C, Zhang G, Tian K, Fu X. Effect of the FA2H Gene on cashmere fineness of Jiangnan cashmere goats based on transcriptome sequencing. BMC Genomics 2022; 23:527. [PMID: 35864447 PMCID: PMC9306159 DOI: 10.1186/s12864-022-08763-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Background Cashmere goats are a heterogeneous hairy mammal. The fineness of cashmere can affect its economic value. Therefore, in this study, we used transcriptome sequencing techniques to analyze the gene expression profiles of the skin tissues of cashmere goats with different cashmere fineness. The selected candidate genes were functionally verified with the secondary hair follicle hair papillary cells of cashmere goats. Results We identified 479 DEGs, of which 238 mRNAs were up-regulated in the fine velvet group and 241 mRNA were down-regulated. Based on functional annotation and protein interaction network analysis, we found some genes that may affect the fineness of cashmere, including SOX18, SOX4, WNT5A, IGFBP4, KAP8, KRT36, and FA2H. Using qRT-PCR, Western blot, CCK-8 cell viability detection, EDU cell proliferation detection, and flow cytometry, we found that overexpression of the FA2H gene could promote the proliferation of secondary hair follicle DPCs in cashmere goats. At the same time, we proved that FA2H could regulate the expression levels of the FGF5 and BMP2 genes in DPCs. Conclusion The results of this study provide a useful reference for the genetics and breeding of Jiangnan cashmere goats and goat genome annotation, and provide an experimental basis for improving cashmere quality of the cashmere goat. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08763-7.
Collapse
Affiliation(s)
- Cuiling Wu
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China.,College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China.,Key Laboratory of Genetics Breeding and Reproduction of Xinjiang Wool Sheep and Cashmere-Goat, Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, 830011, China
| | - Jianying Li
- Key Laboratory of Special Environmental Medicine, Xinjiang Military General Hospital, Urumqi, 830000, China
| | - Xinming Xu
- Key Laboratory of Genetics Breeding and Reproduction of Xinjiang Wool Sheep and Cashmere-Goat, Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, 830011, China
| | - Qi Xu
- Key Laboratory of Special Environmental Medicine, Xinjiang Military General Hospital, Urumqi, 830000, China
| | - Chongkai Qin
- Xinjiang Aksu Prefecture Animal Husbandry Technology Extension Center, Aksu, 843000, China
| | - Guifen Liu
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Chen Wei
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Guoping Zhang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Kechuan Tian
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China.
| | - Xuefeng Fu
- Key Laboratory of Genetics Breeding and Reproduction of Xinjiang Wool Sheep and Cashmere-Goat, Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, 830011, China.
| |
Collapse
|
11
|
Thermos G, Piperi E, Tosios KI, Nikitakis NG. Expression of BMP4 and FOXN1 in orthokeratinized odontogenic cyst compared to odontogenic keratocyst suggests an epidermal phenotype. Biotech Histochem 2022; 97:584-592. [PMID: 35527675 DOI: 10.1080/10520295.2022.2048073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Odontogenic keratocysts (OKC) and orthokeratinized odontogenic cysts (OOC) are odontogenic cysts that share histological and immunohistochemical similarity with epidermal appendages and cutaneous cystic lesions despite exhibiting contrasting biological behavior. In epidermal appendages, BMP4 induces expression of FOXN1, which participates in terminal differentiation of keratinocytes and control of proliferation. We compared BMP4 and FOXN1 expression in OOC and OKC to investigate their role in the epithelial differentiation of these cysts. BMP4 and FOXN1 expression was assessed using immunohistochemistry in 20 primary sporadic OKC and compared to 16 OOC. BMP4 epithelial expression was detected in 81.25% OOC compared to 35% in OKC, while its expression in connective tissue was observed in 65% OKC and 75% OOC. FOXN1 was detected in 75% OOC vs. 30% OKC. The "triple positive" phenotype, i.e., BMP4 epithelial and connective tissue positivity and FOXN1 epithelial positivity, was seen in 56.25% OOC compared to 10% OKC. The greater expression of BMP4 and FOXN1 in OOC suggests greater activation of this pathway in OOC, which suggests a role in its more mature epithelium; it also resembles an epidermal phenotype.
Collapse
Affiliation(s)
- Grigorios Thermos
- Department of Oral Medicine and Pathology and Hospital Dentistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Piperi
- Department of Oral Medicine and Pathology and Hospital Dentistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos I Tosios
- Department of Oral Medicine and Pathology and Hospital Dentistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos G Nikitakis
- Department of Oral Medicine and Pathology and Hospital Dentistry, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Pincha N, Marangoni P, Haque A, Klein OD. Parallels in signaling between development and regeneration in ectodermal organs. Curr Top Dev Biol 2022; 149:373-419. [PMID: 35606061 PMCID: PMC10049776 DOI: 10.1016/bs.ctdb.2022.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Ectodermal organs originate from the outermost germ layer of the developing embryo and include the skin, hair, tooth, nails, and exocrine glands. These organs develop through tightly regulated, sequential and reciprocal epithelial-mesenchymal crosstalk, and they eventually assume various morphologies and functions while retaining the ability to regenerate. As with many other tissues in the body, the development and morphogenesis of these organs are regulated by a set of common signaling pathways, such as Shh, Wnt, Bmp, Notch, Tgf-β, and Eda. However, subtle differences in the temporal activation, the multiple possible combinations of ligand-receptor activation, the various cofactors, as well as the underlying epigenetic modulation determine how each organ develops into its adult form. Although each organ has been studied separately in considerable detail, the mechanisms underlying the parallels and differences in signaling that regulate their development have rarely been investigated. First, we will use the tooth, the hair follicle, and the mammary gland as representative ectodermal organs to explore how the development of signaling centers and establishment of stem cell populations influence overall growth and morphogenesis. Then we will compare how some of the major signaling pathways (Shh, Wnt, Notch and Yap/Taz) differentially regulate developmental events. Finally, we will discuss how signaling regulates regenerative processes in all three.
Collapse
Affiliation(s)
- Neha Pincha
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, United States
| | - Pauline Marangoni
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, United States
| | - Ameera Haque
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, United States
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, United States; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA, United States.
| |
Collapse
|
13
|
Lan Q, Satta J, Myllymäki SM, Trela E, Lindström R, Kaczyńska B, Englund J, Mikkola ML. Protocol for Studying Embryonic Mammary Gland Branching Morphogenesis Ex Vivo. Methods Mol Biol 2022; 2471:1-18. [PMID: 35175589 DOI: 10.1007/978-1-0716-2193-6_1] [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] [Indexed: 06/14/2023]
Abstract
Mammary gland development starts during embryogenesis, and the process continues after birth. During development, the mammary gland undergoes massive morphological and physiological alterations including growth, invasion, and branching morphogenesis providing an ideal model for stem cell and cancer biology studies. Great efforts have been made in understanding mammary gland development during puberty and adulthood; however, the process during embryogenesis is still elusive. One reason is that the tools to study tissue dynamics during development are limited, which is partially due to the lack of an ex vivo culture method. Here we describe an updated organ culture protocol of the murine embryonic mammary gland. This powerful tool allows monitoring of growth and branching morphogenesis of mammary gland ex vivo by live imaging. In addition, we introduce a novel method for culturing intact, stroma-free mammary rudiments from late gestation mouse embryos in 3D in Matrigel. This approach can be used to identify the direct stromal cues for branching morphogenesis.
Collapse
Affiliation(s)
- Qiang Lan
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Jyoti Satta
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Satu-Marja Myllymäki
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Ewelina Trela
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Riitta Lindström
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Beata Kaczyńska
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Johanna Englund
- Centre of Excellence in Stem Cell Metabolism, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Marja L Mikkola
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
| |
Collapse
|
14
|
Lee O, Pilewskie M, Karlan S, Tull MB, Benante K, Xu Y, Blanco L, Helenowski I, Kocherginsky M, Yadav S, Hosseini O, Hansen N, Bethke K, Muzzio M, Troester MA, Dimond E, Perloff M, Heckman-Stoddard B, Khan SA. Local Transdermal Delivery of Telapristone Acetate Through Breast Skin, Compared With Oral Treatment: A Randomized Double-Blind, Placebo-Controlled Phase II Trial. Clin Pharmacol Ther 2021; 109:728-738. [PMID: 32996592 PMCID: PMC8388824 DOI: 10.1002/cpt.2041] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
Oral breast cancer prevention medications entail systemic exposure, limiting acceptance by high-risk women. Delivery through the breast skin, although an attractive alternative, requires demonstration of drug distribution throughout the breast. We conducted a randomized double-blind, placebo-controlled phase II clinical trial comparing telapristone acetate, a progesterone receptor antagonist, administered orally (12 mg/day) or transdermally (12 mg/breast) for 4 ± 1 weeks to women planning mastectomy. Plasma and tissue concentrations, measured at five locations in the mastectomy specimen using liquid chromatography tandem mass spectrometry were compared. In 60 evaluable subjects, median drug concentration (ng/g tissue) was 103 (interquartile range (IQR): 46.3-336) in the oral vs. 2.82 (IQR: 1.4-5.5) in the transdermal group. Despite poor dermal permeation, within-breast drug distribution pattern was identical in both groups (R2 = 0.88, P = 0.006), demonstrating that transdermally and orally delivered drug is distributed similarly through the breast, and is strongly influenced by tissue adiposity (P < 0.0001). Other skin-penetrant drugs should be tested for breast cancer prevention.
Collapse
Affiliation(s)
- Oukseub Lee
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Melissa Pilewskie
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Scott Karlan
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mary B Tull
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kelly Benante
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yanfei Xu
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Luis Blanco
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Irene Helenowski
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Masha Kocherginsky
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Shivangi Yadav
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Omid Hosseini
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nora Hansen
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kevin Bethke
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Miguel Muzzio
- Analytical Chemistry Division, Illinois Institute of Technology Research Institute, Chicago, Illinois, USA
| | - Melissa A Troester
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Eileen Dimond
- National Cancer Institute Division of Cancer Prevention, Bethesda, Maryland, USA
| | - Marjorie Perloff
- National Cancer Institute Division of Cancer Prevention, Bethesda, Maryland, USA
| | | | - Seema A Khan
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
15
|
Wu Z, Hai E, Di Z, Ma R, Shang F, Wang Y, Wang M, Liang L, Rong Y, Pan J, Wu W, Su R, Wang Z, Wang R, Zhang Y, Li J. Using WGCNA (weighted gene co-expression network analysis) to identify the hub genes of skin hair follicle development in fetus stage of Inner Mongolia cashmere goat. PLoS One 2020; 15:e0243507. [PMID: 33351808 PMCID: PMC7755285 DOI: 10.1371/journal.pone.0243507] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Mature hair follicles represent an important stage of hair follicle development, which determines the stability of hair follicle structure and its ability to enter the hair cycle. Here, we used weighted gene co-expression network analysis (WGCNA) to identify hub genes of mature skin and hair follicles in Inner Mongolian cashmere goats. METHODS We used transcriptome sequencing data for the skin of Inner Mongolian cashmere goats from fetal days 45-135 days, and divided the co expressed genes into different modules by WGCNA. Characteristic values were used to screen out modules that were highly expressed in mature skin follicles. Module hub genes were then selected based on the correlation coefficients between the gene and module eigenvalue, gene connectivity, and Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. The results were confirmed by quantitative polymerase chain reaction (qPCR). RESULTS Ten modules were successfully defined, of which one, with a total of 3166 genes, was selected as a specific module through sample and gene expression pattern analyses. A total of 584 candidate hub genes in the module were screened by the correlation coefficients between the genes and module eigenvalue and gene connectivity. Finally, GO/KEGG functional enrichment analyses detected WNT10A as a key gene in the development and maturation of skin hair follicles in fetal Inner Mongolian cashmere goats. qPCR showed that the expression trends of 13 genes from seven fetal skin samples were consistent with the sequencing results, indicating that the sequencing results were reliable.n.
Collapse
Affiliation(s)
- Zhihong Wu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Erhan Hai
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Zhengyang Di
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Rong Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Fangzheng Shang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Yu Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Min Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Lili Liang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Youjun Rong
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Jianfeng Pan
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Wenbin Wu
- Zhenlai Hehe Animal Husbandry Development Co., Ltd, Baicheng, China
| | - Rui Su
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Zhiying Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Ruijun Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Yanjun Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Engineering Research Center for Goat Genetics and Breeding, Hohhot, Inner Mongolia Autonomous Region, China
- * E-mail: (JL); , (YZ)
| | - Jinquan Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Hohhot, Inner Mongolia Autonomous Region, China
- Key Laboratory of Mutton Sheep Genetics and Breeding, Ministry of Agriculture, Hohhot, China
- Engineering Research Center for Goat Genetics and Breeding, Hohhot, Inner Mongolia Autonomous Region, China
- * E-mail: (JL); , (YZ)
| |
Collapse
|
16
|
Lan Q, Mikkola ML. Protocol: Adeno-Associated Virus-Mediated Gene Transfer in Ex Vivo Cultured Embryonic Mammary Gland. J Mammary Gland Biol Neoplasia 2020; 25:409-416. [PMID: 33009602 PMCID: PMC7960627 DOI: 10.1007/s10911-020-09461-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/24/2020] [Indexed: 12/31/2022] Open
Abstract
Branching morphogenesis of the murine mammary gland starts during late embryogenesis. It is regulated by the signals emanating both from the epithelium and the mesenchyme, yet the molecular mechanisms regulating this process remain poorly understood. We have previously developed a unique whole organ culture technique for embryonic mammary glands, which provides a powerful tool to monitor and manipulate branching morphogenesis ex vivo. Nowadays, RNA sequencing and other transcriptional profiling techniques provide robust methods to identify components of gene regulatory networks driving branching morphogenesis. However, validation of the candidate genes still mainly depends on the use of the transgenic mouse models, especially in mammary gland studies. By comparing different serotypes of recombinant adeno-associated virus (rAAVs), we found out that rAAVs provide sufficient efficiency for gene transfer with different tissue preferences depending on the serotypes of the virus. AAV-2 and AAV-8 preferentially target epithelial and mesenchymal compartments, respectively, while AAV-9 infects both tissues. Here, we describe a protocol for AAV-mediated gene transfer in ex vivo cultured murine embryonic mammary gland facilitating gene function studies on mammary gland branching morphogenesis.
Collapse
Affiliation(s)
- Qiang Lan
- Cell and tissue dynamics research program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
| | - Marja L Mikkola
- Cell and tissue dynamics research program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
| |
Collapse
|
17
|
Facilitation of Bone Healing Processes Based on the Developmental Function of Meox2 in Tooth Loss Lesion. Int J Mol Sci 2020; 21:ijms21228701. [PMID: 33218046 PMCID: PMC7698889 DOI: 10.3390/ijms21228701] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/10/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
In the present study, we examined the bone healing capacity of Meox2, a homeobox gene that plays essential roles in the differentiation of a range of developing tissues, and identified its putative function in palatogenesis. We applied the knocking down of Meox2 in human periodontal ligament fibroblasts to examine the osteogenic potential of Meox2. Additionally, we applied in vivo periodontitis induced experiment to reveal the possible application of Meox2 knockdown for 1 and 2 weeks in bone healing processes. We examined the detailed histomorphological changes using Masson’s trichrome staining and micro-computed tomography evaluation. Moreover, we observed the localization patterns of various signaling molecules, including α-SMA, CK14, IL-1β, and MPO to examine the altered bone healing processes. Furthermore, we investigated the process of bone formation using immunohistochemistry of Osteocalcin and Runx2. On the basis of the results, we suggest that the knocking down of Meox2 via the activation of osteoblast and modulation of inflammation would be a plausible answer for bone regeneration as a gene therapy. Additionally, we propose that the purpose-dependent selection and application of developmental regulation genes are important for the functional regeneration of specific tissues and organs, where the pathological condition of tooth loss lesion would be.
Collapse
|
18
|
Kashgari G, Meinecke L, Gordon W, Ruiz B, Yang J, Ma AL, Xie Y, Ho H, Plikus MV, Nie Q, Jester JV, Andersen B. Epithelial Migration and Non-adhesive Periderm Are Required for Digit Separation during Mammalian Development. Dev Cell 2020; 52:764-778.e4. [PMID: 32109382 DOI: 10.1016/j.devcel.2020.01.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/26/2019] [Accepted: 01/28/2020] [Indexed: 01/04/2023]
Abstract
The fusion of digits or toes, syndactyly, can be part of complex syndromes, including van der Woude syndrome. A subset of van der Woude cases is caused by dominant-negative mutations in the epithelial transcription factor Grainyhead like-3 (GRHL3), and Grhl3-/-mice have soft-tissue syndactyly. Although impaired interdigital cell death of mesenchymal cells causes syndactyly in multiple genetic mutants, Grhl3-/- embryos had normal interdigital cell death, suggesting alternative mechanisms for syndactyly. We found that in digit separation, the overlying epidermis forms a migrating interdigital epithelial tongue (IET) when the epithelium invaginates to separate the digits. Normally, the non-adhesive surface periderm allows the IET to bifurcate as the digits separate. In contrast, in Grhl3-/- embryos, the IET moves normally between the digits but fails to bifurcate because of abnormal adhesion of the periderm. Our study identifies epidermal developmental processes required for digit separation.
Collapse
Affiliation(s)
- Ghaidaa Kashgari
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Lina Meinecke
- Department of Mathematics, School of Physical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Developmental & Cell Biology, School of the Biological Sciences, University of California, Irvine, Irvine, CA, USA
| | - William Gordon
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Bryan Ruiz
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Jady Yang
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Amy Lan Ma
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Yilu Xie
- The Gavin Herbert Eye Institute, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Hsiang Ho
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Maksim V Plikus
- Department of Developmental & Cell Biology, School of the Biological Sciences, University of California, Irvine, Irvine, CA, USA
| | - Qing Nie
- Department of Mathematics, School of Physical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Developmental & Cell Biology, School of the Biological Sciences, University of California, Irvine, Irvine, CA, USA
| | - James V Jester
- The Gavin Herbert Eye Institute, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Bogi Andersen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA; Department of Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
19
|
Yamada S, Lav R, Li J, Tucker AS, Green JBA. Molar Bud-to-Cap Transition Is Proliferation Independent. J Dent Res 2019; 98:1253-1261. [PMID: 31393749 PMCID: PMC6761786 DOI: 10.1177/0022034519869307] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Tooth germs undergo a series of dynamic morphologic changes through bud, cap, and bell stages, in which odontogenic epithelium continuously extends into the underlying mesenchyme. During the transition from the bud stage to the cap stage, the base of the bud flattens and then bends into a cap shape whose edges are referred to as “cervical loops.” Although genetic mechanisms for cap formation have been well described, little is understood about the morphogenetic mechanisms. Computer modeling and cell trajectory tracking have suggested that the epithelial bending is driven purely by differential cell proliferation and adhesion in different parts of the tooth germ. Here, we show that, unexpectedly, inhibition of cell proliferation did not prevent bud-to-cap morphogenesis. We quantified cell shapes and actin and myosin distributions in different parts of the tooth epithelium at the critical stages and found that these are consistent with basal relaxation in the forming cervical loops and basal constriction around enamel knot at the center of the cap. Inhibition of focal adhesion kinase, which is required for basal constriction in other systems, arrested the molar explant morphogenesis at the bud stage. Together, these results show that the bud-to-cap transition is largely proliferation independent, and we propose that it is driven by classic actomyosin-driven cell shape–dependent mechanisms. We discuss how these results can be reconciled with the previous models and data.
Collapse
Affiliation(s)
- S Yamada
- Centre for Craniofacial Biology and Regeneration, King's College London, London, UK
| | - R Lav
- Centre for Craniofacial Biology and Regeneration, King's College London, London, UK
| | - J Li
- Centre for Craniofacial Biology and Regeneration, King's College London, London, UK
| | - A S Tucker
- Centre for Craniofacial Biology and Regeneration, King's College London, London, UK
| | - J B A Green
- Centre for Craniofacial Biology and Regeneration, King's College London, London, UK
| |
Collapse
|
20
|
Ueyama T, Sakuma M, Nakatsuji M, Uebi T, Hamada T, Aiba A, Saito N. Rac-Dependent Signaling from Keratinocytes Promotes Differentiation of Intradermal White Adipocytes. J Invest Dermatol 2019; 140:75-84.e6. [PMID: 31351086 DOI: 10.1016/j.jid.2019.06.140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/23/2019] [Accepted: 06/28/2019] [Indexed: 01/10/2023]
Abstract
Rac signaling affects numerous downstream targets in vitro; however, few studies have established in vivo levels. We generated mice with a single knockout (KO) of Rac1 (Keratin5(K5)-Cre;Rac1flox/flox, Rac1-KO) and double KO of Rac1 and Rac3 (K5-Cre;Rac1flox/flox;Rac3-/-, Rac1/Rac3-DKO) in keratinocytes. The hairless phenotype in Rac1-KO mice was markedly exacerbated in Rac1/Rac3-DKO mice. Strikingly, Rac1-KO mice exhibited thinner dermal white adipose tissue, which was considerably further reduced in Rac1/Rac3-DKO mice. DNA microarray using primary keratinocytes from Rac1/Rac3-DKO mice exhibited decreased mRNA levels of Bmp2, Bmp5, Fgf20, Fgf21, Fgfbp1, and Pdgfα. Combinational treatment with bone morphogenetic protein (BMP) 2 and fibroblast growth factor (FGF) 21 in culture medium, but not individual purified recombinant proteins, could differentiate 3T3-L1 fibroblasts into adipocytes, as could culture media from primary keratinocytes. Conversely, addition of anti-BMP2 or anti-FGF21 antibodies into the culture medium inhibited fibroblast differentiation. In addition, BMP2 and FGF21 treatment promoted adipocyte differentiation only of rat primary white adipocyte precursors but not rat primary brown adipocyte precursors. Furthermore, BMP2 and FGF21 treatment enhanced adipogenesis of normal human dermal fibroblasts. Notably, brown adipogenesis promoted by FGF21 was inhibited by BMP2. Thus, we propose a complex paracrine pathway from keratinocytes to intradermal pre-adipocytes, which functions as a Rac-dependent modulator of both white and brown adipogenesis.
Collapse
Affiliation(s)
- Takehiko Ueyama
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan.
| | - Megumi Sakuma
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Mio Nakatsuji
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Tatsuya Uebi
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Takeshi Hamada
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Atsu Aiba
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoaki Saito
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| |
Collapse
|
21
|
Lu P, Zhou T, Xu C, Lu Y. Mammary stem cells, where art thou? WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e357. [PMID: 31322329 DOI: 10.1002/wdev.357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022]
Abstract
Tremendous progress has been made in the field of stem cell biology. This is in part due to the emergence of various vertebrate organs, including the mammary gland, as an amenable model system for adult stem cell studies and remarkable technical advances in single cell technology and modern genetic lineage tracing. In the current review, we summarize the recent progress in mammary gland stem cell biology at both the adult and embryonic stages. We discuss current challenges and controversies, and potentially new and exciting directions for future research. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration.
Collapse
Affiliation(s)
- Pengfei Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Tao Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chongshen Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yunzhe Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
22
|
Shpichka A, Butnaru D, Bezrukov EA, Sukhanov RB, Atala A, Burdukovskii V, Zhang Y, Timashev P. Skin tissue regeneration for burn injury. Stem Cell Res Ther 2019; 10:94. [PMID: 30876456 PMCID: PMC6419807 DOI: 10.1186/s13287-019-1203-3] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The skin is the largest organ of the body, which meets the environment most directly. Thus, the skin is vulnerable to various damages, particularly burn injury. Skin wound healing is a serious interaction between cell types, cytokines, mediators, the neurovascular system, and matrix remodeling. Tissue regeneration technology remarkably enhances skin repair via re-epidermalization, epidermal-stromal cell interactions, angiogenesis, and inhabitation of hypertrophic scars and keloids. The success rates of skin healing for burn injuries have significantly increased with the use of various skin substitutes. In this review, we discuss skin replacement with cells, growth factors, scaffolds, or cell-seeded scaffolds for skin tissue reconstruction and also compare the high efficacy and cost-effectiveness of each therapy. We describe the essentials, achievements, and challenges of cell-based therapy in reducing scar formation and improving burn injury treatment.
Collapse
Affiliation(s)
- Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Denis Butnaru
- Sechenov Biomedical Science and Technology Park, Sechenov University, Moscow, Russia
| | | | | | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC USA
| | - Vitaliy Burdukovskii
- Baikal Institute of Nature Management, Siberian Branch of the Russian Academy of Sciences, Ulan-Ude, Russia
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC USA
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Research Center “Crystallography and Photonics” RAS, Institute of Photonic Technologies, Troitsk, Moscow, Russia
- Departments of Polymers and Composites, N.N. Semenov Institute of Chemical Physics, Moscow, Russia
| |
Collapse
|
23
|
Balic A. Concise Review: Cellular and Molecular Mechanisms Regulation of Tooth Initiation. Stem Cells 2018; 37:26-32. [DOI: 10.1002/stem.2917] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Anamaria Balic
- Research Program in Developmental Biology; Institute of Biotechnology, University of Helsinki; Helsinki Finland
| |
Collapse
|
24
|
Perrot-Applanat M, Kolf-Clauw M, Michel C, Beausoleil C. Alteration of mammary gland development by bisphenol a and evidence of a mode of action mediated through endocrine disruption. Mol Cell Endocrinol 2018; 475:29-53. [PMID: 30048677 DOI: 10.1016/j.mce.2018.06.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 01/08/2023]
Abstract
The development and function of the mammary gland are endocrine-dependent processes, depending on the stage of development. Foetal and/or postnatal exposure to low doses of BPA alters tissue organisation through epithelial proliferation and stroma-epithelial interactions. BPA also alters the expression of E2-dependent epithelial and stroma transcriptomes. Several signalling pathways are consistent with the observed phenotype: proliferation and apoptosis, a focal adhesion pathway indicating changes in biomechanical properties of the extracellular matrix, and immune function. Some of BPA's effects are reversed by oestrogen and/or GPER inhibitors. BPA also alters the expression of epigenetic marks (EZH2, HOTAIR), which would explain the delayed effect of foetal BPA exposure. In conclusion, experimental evidence shows that pre- or postnatal BPA exposure consistently causes endocrine modifications in the mammary tissue of different animal species, disrupting stromal-epithelial interactions and ultimately increasing its susceptibility to carcinogens. An interspecies comparison highlights why and how these effects apply to humans.
Collapse
Affiliation(s)
| | - Martine Kolf-Clauw
- CREFRE, Toulouse University, INSERM, Toulouse Veterinary School, 23 chemin des Capelles, BP 87614, F 310176, Toulouse Cedex 3, France
| | - Cécile Michel
- ANSES, Risk Assessment Department, Maisons-Alfort, France.
| | | |
Collapse
|
25
|
Wahlbuhl M, Schuepbach-Mallepell S, Kowalczyk-Quintas C, Dick A, Fahlbusch FB, Schneider P, Schneider H. Attenuation of Mammary Gland Dysplasia and Feeding Difficulties in Tabby Mice by Fetal Therapy. J Mammary Gland Biol Neoplasia 2018; 23:125-138. [PMID: 29855766 DOI: 10.1007/s10911-018-9399-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Hypohidrotic ectodermal dysplasias (HED) are hereditary differentiation disorders of multiple ectodermal structures including the mammary gland. The X-linked form of HED (XLHED) is caused by a lack of the secreted signaling molecule ectodysplasin A1 (EDA1) which is encoded by the gene EDA and belongs to the tumor necrosis factor (TNF) superfamily. Although male patients (hemizygous) are usually more severely affected by XLHED, heterozygous female carriers of an EDA mutation may also suffer from a variety of symptoms, in particular from abnormal development of their breasts. In Tabby mice, a well-studied animal model of XLHED, EDA1 is absent. We investigated the effects of prenatal administration of Fc-EDA, a recombinant EDA1 replacement protein, on mammary gland development in female Tabby mice. Intra-amniotic delivery of Fc-EDA to fetal animals resulted later in improved breastfeeding and thus promoted the growth of their offspring. In detail, such treatment led to a normalization of the nipple shape (protrusion, tapering) that facilitated sucking. Mammary glands of treated female Tabby mice also showed internal changes, including enhanced branching morphogenesis and ductal elongation. Our findings indicate that EDA receptor stimulation during development has a stable impact on later stages of mammary gland differentiation, including lactation, but also show that intra-amniotic administration of an EDA1 replacement protein to fetal Tabby mice partially corrects the mammary gland phenotype in female adult animals.
Collapse
Affiliation(s)
- Mandy Wahlbuhl
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestr. 15, 91054, Erlangen, Germany.
| | | | | | - Angela Dick
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestr. 15, 91054, Erlangen, Germany
| | - Fabian B Fahlbusch
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestr. 15, 91054, Erlangen, Germany
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Holm Schneider
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestr. 15, 91054, Erlangen, Germany
| |
Collapse
|
26
|
Duteil D, Tourrette Y, Eberlin A, Willmann D, Patel D, Friedrichs N, Müller JM, Schüle R. The histone acetyltransferase inhibitor Nir regulates epidermis development. Development 2018; 145:dev.158543. [PMID: 29490983 DOI: 10.1242/dev.158543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/11/2018] [Indexed: 11/20/2022]
Abstract
In addition to its function as an inhibitor of histone acetyltransferases, Nir (Noc2l) binds to p53 and TAp63 to regulate their activity. Here, we show that epidermis-specific ablation of Nir impairs epidermal stratification and barrier function, resulting in perinatal lethality. Nir-deficient epidermis lacks appendages and remains single layered during embryogenesis. Cell proliferation is inhibited, whereas apoptosis and p53 acetylation are increased, indicating that Nir is controlling cell proliferation by limiting p53 acetylation. Transcriptome analysis revealed that Nir regulates the expression of essential factors in epidermis development, such as keratins, integrins and laminins. Furthermore, Nir binds to and controls the expression of p63 and limits H3K18ac at the p63 promoter. Corroborating the stratification defects, asymmetric cell divisions were virtually absent in Nir-deficient mice, suggesting that Nir is required for correct mitotic spindle orientation. In summary, our data define Nir as a key regulator of skin development.
Collapse
Affiliation(s)
- Delphine Duteil
- Urologische Klinik und Zentrale Klinische Forschung, Klinikum der Universität Freiburg, Breisacherstrasse 66, 79106 Freiburg, Germany
| | - Yves Tourrette
- Urologische Klinik und Zentrale Klinische Forschung, Klinikum der Universität Freiburg, Breisacherstrasse 66, 79106 Freiburg, Germany
| | - Adrien Eberlin
- Urologische Klinik und Zentrale Klinische Forschung, Klinikum der Universität Freiburg, Breisacherstrasse 66, 79106 Freiburg, Germany
| | - Dominica Willmann
- Urologische Klinik und Zentrale Klinische Forschung, Klinikum der Universität Freiburg, Breisacherstrasse 66, 79106 Freiburg, Germany
| | - Dharmeshkumar Patel
- Pediatric Blood and Marrow Transplant, University of Minnesota, 2-191 Moos Tower, 515 Delaware St. SE, Minneapolis, MN 55455, USA
| | - Nicolaus Friedrichs
- Institute of Pathology, University of Cologne Medical School, 50937 Cologne, Germany
| | - Judith M Müller
- Urologische Klinik und Zentrale Klinische Forschung, Klinikum der Universität Freiburg, Breisacherstrasse 66, 79106 Freiburg, Germany
| | - Roland Schüle
- Urologische Klinik und Zentrale Klinische Forschung, Klinikum der Universität Freiburg, Breisacherstrasse 66, 79106 Freiburg, Germany .,BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University, 79106 Freiburg, Germany.,Deutsche Konsortium für Translationale Krebsforschung (DKTK), Standort, 79106 Freiburg, Germany
| |
Collapse
|
27
|
Bach K, Pensa S, Grzelak M, Hadfield J, Adams DJ, Marioni JC, Khaled WT. Differentiation dynamics of mammary epithelial cells revealed by single-cell RNA sequencing. Nat Commun 2017; 8:2128. [PMID: 29225342 PMCID: PMC5723634 DOI: 10.1038/s41467-017-02001-5] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 11/01/2017] [Indexed: 11/21/2022] Open
Abstract
Characterising the hierarchy of mammary epithelial cells (MECs) and how they are regulated during adult development is important for understanding how breast cancer arises. Here we report the use of single-cell RNA sequencing to determine the gene expression profile of MECs across four developmental stages; nulliparous, mid gestation, lactation and post involution. Our analysis of 23,184 cells identifies 15 clusters, few of which could be fully characterised by a single marker gene. We argue instead that the epithelial cells-especially in the luminal compartment-should rather be conceptualised as being part of a continuous spectrum of differentiation. Furthermore, our data support the existence of a common luminal progenitor cell giving rise to intermediate, restricted alveolar and hormone-sensing progenitors. This luminal progenitor compartment undergoes transcriptional changes in response to a full pregnancy, lactation and involution. In summary, our results provide a global, unbiased view of adult mammary gland development.
Collapse
Affiliation(s)
- Karsten Bach
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Cancer Research UK Cambridge Cancer Centre, Cambridge, CB2 0RE, UK
| | - Sara Pensa
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
- Cancer Research UK Cambridge Cancer Centre, Cambridge, CB2 0RE, UK
| | - Marta Grzelak
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Cancer Research UK Cambridge Cancer Centre, Cambridge, CB2 0RE, UK
| | - James Hadfield
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Cancer Research UK Cambridge Cancer Centre, Cambridge, CB2 0RE, UK
| | - David J Adams
- Cancer Research UK Cambridge Cancer Centre, Cambridge, CB2 0RE, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1HH, UK
| | - John C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK.
- Cancer Research UK Cambridge Cancer Centre, Cambridge, CB2 0RE, UK.
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1HH, UK.
- European Bioinformatics Institute, European Molecular Biology Laboratory, Hinxton, CB10 1 SD, UK.
| | - Walid T Khaled
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK.
- Cancer Research UK Cambridge Cancer Centre, Cambridge, CB2 0RE, UK.
| |
Collapse
|
28
|
Roles of the Hedgehog Signaling Pathway in Epidermal and Hair Follicle Development, Homeostasis, and Cancer. J Dev Biol 2017; 5:jdb5040012. [PMID: 29615568 PMCID: PMC5831796 DOI: 10.3390/jdb5040012] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/15/2017] [Accepted: 11/18/2017] [Indexed: 12/15/2022] Open
Abstract
The epidermis is the outermost layer of the skin and provides a protective barrier against environmental insults. It is a rapidly-renewing tissue undergoing constant regeneration, maintained by several types of stem cells. The Hedgehog (HH) signaling pathway is one of the fundamental signaling pathways that contributes to epidermal development, homeostasis, and repair, as well as to hair follicle development and follicle bulge stem cell maintenance. The HH pathway interacts with other signal transduction pathways, including those activated by Wnt, bone morphogenetic protein, platelet-derived growth factor, Notch, and ectodysplasin. Furthermore, aberrant activation of HH signaling is associated with various tumors, including basal cell carcinoma. Therefore, an understanding of the regulatory mechanisms of the HH signaling pathway is important for elucidating fundamental mechanisms underlying both organogenesis and carcinogenesis. In this review, we discuss the role of the HH signaling pathway in the development and homeostasis epidermis and hair follicles, and in basal cell carcinoma formation, providing an update of current knowledge in this field.
Collapse
|
29
|
WNT10A mutation causes ectodermal dysplasia by impairing progenitor cell proliferation and KLF4-mediated differentiation. Nat Commun 2017; 8:15397. [PMID: 28589954 PMCID: PMC5467248 DOI: 10.1038/ncomms15397] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 03/27/2017] [Indexed: 02/06/2023] Open
Abstract
Human WNT10A mutations are associated with developmental tooth abnormalities and adolescent onset of a broad range of ectodermal defects. Here we show that β-catenin pathway activity and adult epithelial progenitor proliferation are reduced in the absence of WNT10A, and identify Wnt-active self-renewing stem cells in affected tissues including hair follicles, sebaceous glands, taste buds, nails and sweat ducts. Human and mouse WNT10A mutant palmoplantar and tongue epithelia also display specific differentiation defects that are mimicked by loss of the transcription factor KLF4. We find that β-catenin interacts directly with region-specific LEF/TCF factors, and with KLF4 in differentiating, but not proliferating, cells to promote expression of specialized keratins required for normal tissue structure and integrity. Our data identify WNT10A as a critical ligand controlling adult epithelial proliferation and region-specific differentiation, and suggest downstream β-catenin pathway activation as a potential approach to ameliorate regenerative defects in WNT10A patients. Human WNT10A mutations are associated with dental defects and adult onset ectodermal dysplasia. Xu et al. show that WNT10A-activated ß-catenin plays dual roles in adult epithelial progenitor proliferation and differentiation by complexing with KLF4 in differentiating, but not proliferating, cells.
Collapse
|
30
|
Nanashima N, Horie K, Yamada T, Shimizu T, Tsuchida S. Hair keratin KRT81 is expressed in normal and breast cancer cells and contributes to their invasiveness. Oncol Rep 2017; 37:2964-2970. [DOI: 10.3892/or.2017.5564] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/05/2016] [Indexed: 11/05/2022] Open
|
31
|
Veraitch O, Mabuchi Y, Matsuzaki Y, Sasaki T, Okuno H, Tsukashima A, Amagai M, Okano H, Ohyama M. Induction of hair follicle dermal papilla cell properties in human induced pluripotent stem cell-derived multipotent LNGFR(+)THY-1(+) mesenchymal cells. Sci Rep 2017; 7:42777. [PMID: 28220862 PMCID: PMC5318903 DOI: 10.1038/srep42777] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 01/16/2017] [Indexed: 12/19/2022] Open
Abstract
The dermal papilla (DP) is a specialised mesenchymal component of the hair follicle (HF) that plays key roles in HF morphogenesis and regeneration. Current technical difficulties in preparing trichogenic human DP cells could be overcome by the use of highly proliferative and plastic human induced pluripotent stem cells (hiPSCs). In this study, hiPSCs were differentiated into induced mesenchymal cells (iMCs) with a bone marrow stromal cell phenotype. A highly proliferative and plastic LNGFR(+)THY-1(+) subset of iMCs was subsequently programmed using retinoic acid and DP cell activating culture medium to acquire DP properties. The resultant cells (induced DP-substituting cells [iDPSCs]) exhibited up-regulated DP markers, interacted with human keratinocytes to up-regulate HF related genes, and when co-grafted with human keratinocytes in vivo gave rise to fibre structures with a hair cuticle-like coat resembling the hair shaft, as confirmed by scanning electron microscope analysis. Furthermore, iDPSCs responded to the clinically used hair growth reagent, minoxidil sulfate, to up-regulate DP genes, further supporting that they were capable of, at least in part, reproducing DP properties. Thus, LNGFR(+)THY-1(+) iMCs may provide material for HF bioengineering and drug screening for hair diseases.
Collapse
Affiliation(s)
- Ophelia Veraitch
- Department of Dermatology, Keio University School of Medicine 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yo Mabuchi
- Department of Physiology, Keio University School of Medicine 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yumi Matsuzaki
- Department of Physiology, Keio University School of Medicine 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Laboratory of Tumor Biology, Department of Life Sciences, Faculty of Medicine, Shimane University, Shiojicho 89-1, Izumo-shi, Shimane, 693-8501, Japan
| | - Takashi Sasaki
- KOSÉ Endowed Program for Skin Care and Allergy Prevention, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hironobu Okuno
- Department of Physiology, Keio University School of Medicine 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Aki Tsukashima
- Department of Dermatology, Keio University School of Medicine 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Dermatology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Manabu Ohyama
- Department of Dermatology, Keio University School of Medicine 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Dermatology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo, Japan
| |
Collapse
|
32
|
Control of Hoxd gene transcription in the mammary bud by hijacking a preexisting regulatory landscape. Proc Natl Acad Sci U S A 2016; 113:E7720-E7729. [PMID: 27856734 DOI: 10.1073/pnas.1617141113] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Vertebrate Hox genes encode transcription factors operating during the development of multiple organs and structures. However, the evolutionary mechanism underlying this remarkable pleiotropy remains to be fully understood. Here, we show that Hoxd8 and Hoxd9, two genes of the HoxD complex, are transcribed during mammary bud (MB) development. However, unlike in other developmental contexts, their coexpression does not rely on the same regulatory mechanism. Hoxd8 is regulated by the combined activity of closely located sequences and the most distant telomeric gene desert. On the other hand, Hoxd9 is controlled by an enhancer-rich region that is also located within the telomeric gene desert but has no impact on Hoxd8 transcription, thus constituting an exception to the global regulatory logic systematically observed at this locus. The latter DNA region is also involved in Hoxd gene regulation in other contexts and strongly interacts with Hoxd9 in all tissues analyzed thus far, indicating that its regulatory activity was already operational before the appearance of mammary glands. Within this DNA region and neighboring a strong limb enhancer, we identified a short sequence conserved in therian mammals and capable of enhancer activity in the MBs. We propose that Hoxd gene regulation in embryonic MBs evolved by hijacking a preexisting regulatory landscape that was already at work before the emergence of mammals in structures such as the limbs or the intestinal tract.
Collapse
|
33
|
Li J, Qu H, Jiang H, Zhao Z, Zhang Q. Transcriptome-Wide Comparative Analysis of microRNA Profiles in the Telogen Skins of Liaoning Cashmere Goats (Capra hircus) and Fine-Wool Sheep (Ovis aries) by Solexa Deep Sequencing. DNA Cell Biol 2016; 35:696-705. [PMID: 27754706 DOI: 10.1089/dna.2015.3161] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Compare the microRNA (miRNA) trancriptomes of goat and sheep skin using Solexa sequencing to understand the development of skin and hair follicles (HFs). miRNA expression patterns vary in the two small RNA libraries from goat (G library) and sheep (S library) telogen skin samples. Analysis of the size distribution of 25.32 million clean reads revealed that most are 21-23 nucleotides. A total of 1910 known miRNAs and 2261 novel mature miRNAs were identified in this study. Among them, 107 novel miRNAs and 1246 known miRNAs were differentially expressed in the two libraries; 10 of the known miRNAs were identified using stem-loop quantitative real-time PCR. Furthermore, GO and KEGG pathway analysis of predicted miRNA targets illustrated the roles of these differentially expressed miRNAs in telogen HF development and growth. This study provides important information about the role of miRNAs in the regulation of HF development and their function in the telogen phase. This observation may help future investigations of the regulation of miRNAs during wool quality improvement.
Collapse
Affiliation(s)
- Jianping Li
- 1 College of Veterinary Medicine, Jilin University, Changchun, China .,2 Henan University of Animal Husbandry and Economy, Henan, Zhengzhou, China
| | - HaiE Qu
- 1 College of Veterinary Medicine, Jilin University, Changchun, China
| | - Huaizhi Jiang
- 3 College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Zhihui Zhao
- 4 College of Animal Science and Technology, Jilin University, Changchun, China
| | - Qiaoling Zhang
- 1 College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
34
|
Functional differences between neonatal and adult fibroblasts and keratinocytes: Donor age affects epithelial-mesenchymal crosstalk in vitro. Int J Mol Med 2016; 38:1063-74. [PMID: 27513730 PMCID: PMC5029973 DOI: 10.3892/ijmm.2016.2706] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/25/2016] [Indexed: 12/17/2022] Open
Abstract
Clinical evidence suggests that healing is faster and almost scarless at an early neonatal age in comparison with that in adults. In this study, the phenotypes of neonatal and adult dermal fibroblasts and keratinocytes (nestin, smooth muscle actin, keratin types 8, 14 and 19, and fibronectin) were compared. Furthermore, functional assays (proliferation, migration, scratch wound closure) including mutual epithelial-mesenchymal interactions were also performed to complete the series of experiments. Positivity for nestin and α smooth muscle actin was higher in neonatal fibroblasts (NFs) when compared with their adult counterparts (adult fibroblasts; AFs). Although the proliferation of NFs and AFs was similar, they significantly differed in their migration potential. The keratinocyte experiments revealed small, poorly differentiated cells (positive for keratins 8, 14 and 19) in primary cultures isolated from neonatal tissues. Moreover, the neonatal keratinocytes exhibited significantly faster rates of healing the experimentally induced in vitro defects in comparison with adult cells. Notably, the epithelial/mesenchymal interaction studies showed that NFs in co-culture with adult keratinocytes significantly stimulated the adult epithelial cells to acquire the phenotype of small, non-confluent cells expressing markers of poor differentiation. These results indicate the important differences between neonatal and adult cells that may be associated with improved wound healing during the early neonatal period.
Collapse
|
35
|
Li J, Chatzeli L, Panousopoulou E, Tucker AS, Green JBA. Epithelial stratification and placode invagination are separable functions in early morphogenesis of the molar tooth. Development 2016; 143:670-81. [PMID: 26755699 PMCID: PMC4760321 DOI: 10.1242/dev.130187] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/30/2015] [Indexed: 01/06/2023]
Abstract
Ectodermal organs, which include teeth, hair follicles, mammary ducts, and glands such as sweat, mucous and sebaceous glands, are initiated in development as placodes, which are epithelial thickenings that invaginate and bud into the underlying mesenchyme. These placodes are stratified into a basal and several suprabasal layers of cells. The mechanisms driving stratification and invagination are poorly understood. Using the mouse molar tooth as a model for ectodermal organ morphogenesis, we show here that vertical, stratifying cell divisions are enriched in the forming placode and that stratification is cell division dependent. Using inhibitor and gain-of-function experiments, we show that FGF signalling is necessary and sufficient for stratification but not invagination as such. We show that, instead, Shh signalling is necessary for, and promotes, invagination once suprabasal tissue is generated. Shh-dependent suprabasal cell shape suggests convergent migration and intercalation, potentially accounting for post-stratification placode invagination to bud stage. We present a model in which FGF generates suprabasal tissue by asymmetric cell division, while Shh triggers cell rearrangement in this tissue to drive invagination all the way to bud formation. Summary: During tooth development in mice, FGF-dependent vertical cell divisions thicken the tooth placode while Shh drives cell rearrangements that cause invagination.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Craniofacial Development & Stem Cell Biology, King's College London, London SE1 9RT, UK
| | - Lemonia Chatzeli
- Department of Craniofacial Development & Stem Cell Biology, King's College London, London SE1 9RT, UK
| | - Eleni Panousopoulou
- Department of Craniofacial Development & Stem Cell Biology, King's College London, London SE1 9RT, UK
| | - Abigail S Tucker
- Department of Craniofacial Development & Stem Cell Biology, King's College London, London SE1 9RT, UK
| | - Jeremy B A Green
- Department of Craniofacial Development & Stem Cell Biology, King's College London, London SE1 9RT, UK
| |
Collapse
|
36
|
Bloomquist RF, Parnell NF, Phillips KA, Fowler TE, Yu TY, Sharpe PT, Streelman JT. Coevolutionary patterning of teeth and taste buds. Proc Natl Acad Sci U S A 2015; 112:E5954-62. [PMID: 26483492 PMCID: PMC4640805 DOI: 10.1073/pnas.1514298112] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Teeth and taste buds are iteratively patterned structures that line the oro-pharynx of vertebrates. Biologists do not fully understand how teeth and taste buds develop from undifferentiated epithelium or how variation in organ density is regulated. These organs are typically studied independently because of their separate anatomical location in mammals: teeth on the jaw margin and taste buds on the tongue. However, in many aquatic animals like bony fishes, teeth and taste buds are colocalized one next to the other. Using genetic mapping in cichlid fishes, we identified shared loci controlling a positive correlation between tooth and taste bud densities. Genome intervals contained candidate genes expressed in tooth and taste bud fields. sfrp5 and bmper, notable for roles in Wingless (Wnt) and bone morphogenetic protein (BMP) signaling, were differentially expressed across cichlid species with divergent tooth and taste bud density, and were expressed in the development of both organs in mice. Synexpression analysis and chemical manipulation of Wnt, BMP, and Hedgehog (Hh) pathways suggest that a common cichlid oral lamina is competent to form teeth or taste buds. Wnt signaling couples tooth and taste bud density and BMP and Hh mediate distinct organ identity. Synthesizing data from fish and mouse, we suggest that the Wnt-BMP-Hh regulatory hierarchy that configures teeth and taste buds on mammalian jaws and tongues may be an evolutionary remnant inherited from ancestors wherein these organs were copatterned from common epithelium.
Collapse
Affiliation(s)
- Ryan F Bloomquist
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332; College of Dental Medicine, Georgia Regents University, Augusta, GA 30912;
| | - Nicholas F Parnell
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Kristine A Phillips
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Teresa E Fowler
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Tian Y Yu
- Department of Craniofacial Development and Stem Cell Biology, Dental Institute, King's College London, London WC2R 2LS, United Kingdom
| | - Paul T Sharpe
- Department of Craniofacial Development and Stem Cell Biology, Dental Institute, King's College London, London WC2R 2LS, United Kingdom
| | - J Todd Streelman
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332;
| |
Collapse
|
37
|
Figueroa F, Singer SS, LeClair EE. Making maxillary barbels with a proximal-distal gradient of Wnt signals in matrix-bound mesenchymal cells. Evol Dev 2015; 17:367-79. [PMID: 26492827 PMCID: PMC4620582 DOI: 10.1111/ede.12167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The evolution of specific appendages is made possible by the ontogenetic deployment of general cell signaling pathways. Many fishes, amphibians and reptiles have unique skin appendages known as barbels, which are poorly understood at the cellular and molecular level. In this study, we examine the cell arrangements, cell division patterns, and gene expression profiles associated with the zebrafish maxillary barbel, or ZMB. The earliest cellular organization of the ZMB is an internal whorl of mesenchymal cells in the dermis of the maxilla; there is no epithelial placode, nor any axially-elongated epithelial cells as expected of an apical ectodermal ridge (AER). As the ZMB develops, cells in S-phase are at first distributed randomly throughout the appendage, gradually transitioning to a proliferative population concentrated at the distal end. By observing ZMB ontogenetic stages in a Wnt-responsive transgenic reporter line, TCFsiam, we identified a strongly fluorescent mesenchymal cell layer within these developing appendages. Using an in vitro explant culture technique on developing barbel tissues, we co-localized the fluorescent label in these cells with the mitotic marker EdU. Surprisingly, the labeled cells showed little proliferation, indicating a slow-cycling subpopulation. Transmission electron microscopy of the ZMB located these cells in a single, circumferential layer within the barbel's matrix core. Morphologically, these cells resemble fibroblasts or osteoblasts; in addition to their matrix-bound location, they are identified by their pancake-shaped nuclei, abundant rough endoplasmic reticulum, and cytoplasmic extensions into the surrounding extracellular matrix. Taken together, these features define a novel mesenchymal cell population in zebrafish, the "TCF(+) core cells." A working model of barbel development is proposed, in which these minimally mitotic mesodermal cells produce collagenous matrix in response to ectodermally-derived Wnt signals deployed in a proximal-distal gradient along the appendage. This documents a novel mechanism of vertebrate appendage outgrowth. Similar genetic signals and cell behaviors may be responsible for the independent and repeated evolution of barbel structures in other fish species.
Collapse
Affiliation(s)
- Francisco Figueroa
- DePaul University Department of Biological Sciences, Chicago, Il 60614 USA
| | - Susan S. Singer
- DePaul University Department of Biological Sciences, Chicago, Il 60614 USA
| | | |
Collapse
|
38
|
Carroll LS, Capecchi MR. Hoxc8 initiates an ectopic mammary program by regulating Fgf10 and Tbx3 expression and Wnt/β-catenin signaling. Development 2015; 142:4056-67. [PMID: 26459221 DOI: 10.1242/dev.128298] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/01/2015] [Indexed: 01/22/2023]
Abstract
The role of Hox genes in the formation of cutaneous accessory organs such as hair follicles and mammary glands has proved elusive, a likely consequence of overlapping function and expression among various homeobox factors. Lineage and immunohistochemical analysis of Hoxc8 in mice revealed that this midthoracic Hox gene has transient but strong regional expression in ventrolateral surface ectoderm at E10.5, much earlier than previously reported. Targeted mice were generated to conditionally misexpress Hoxc8 from the Rosa locus using select Cre drivers, which significantly expanded the domain of thoracic identity in mutant embryos. Accompanying this expansion was the induction of paired zones of ectopic mammary development in the cervical region, which generated between three and five pairs of mammary placodes anterior to the first wild-type mammary rudiment. These rudiments expressed the mammary placode markers Wnt10b and Tbx3 and were labeled by antibodies to the mammary mesenchyme markers ERα and androgen receptor. Somitic Fgf10 expression, which is required for normal mammary line formation, was upregulated in mutant cervical somites, and conditional ablation of ectodermal Tbx3 expression eliminated all normally positioned and ectopic mammary placodes. We present evidence that Hoxc8 participates in regulating the initiation stages of mammary placode morphogenesis, and suggest that this and other Hox genes are likely to have important roles during regional specification and initiation of these and other cutaneous accessory organs.
Collapse
Affiliation(s)
- Lara S Carroll
- Moran Eye Center, University of Utah, Salt Lake City, UT 84132, USA
| | - Mario R Capecchi
- Department of Human Genetics and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
39
|
Cui CY, Schlessinger D. Eccrine sweat gland development and sweat secretion. Exp Dermatol 2015; 24:644-50. [PMID: 26014472 DOI: 10.1111/exd.12773] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2015] [Indexed: 12/21/2022]
Abstract
Eccrine sweat glands help to maintain homoeostasis, primarily by stabilizing body temperature. Derived from embryonic ectoderm, millions of eccrine glands are distributed across human skin and secrete litres of sweat per day. Their easy accessibility has facilitated the start of analyses of their development and function. Mouse genetic models find sweat gland development regulated sequentially by Wnt, Eda and Shh pathways, although precise subpathways and additional regulators require further elucidation. Mature glands have two secretory cell types, clear and dark cells, whose comparative development and functional interactions remain largely unknown. Clear cells have long been known as the major secretory cells, but recent studies suggest that dark cells are also indispensable for sweat secretion. Dark cell-specific Foxa1 expression was shown to regulate a Ca(2+) -dependent Best2 anion channel that is the candidate driver for the required ion currents. Overall, it was shown that cholinergic impulses trigger sweat secretion in mature glands through second messengers - for example InsP3 and Ca(2+) - and downstream ion channels/transporters in the framework of a Na(+) -K(+) -Cl(-) cotransporter model. Notably, the microenvironment surrounding secretory cells, including acid-base balance, was implicated to be important for proper sweat secretion, which requires further clarification. Furthermore, multiple ion channels have been shown to be expressed in clear and dark cells, but the degree to which various ion channels function redundantly or indispensably also remains to be determined.
Collapse
Affiliation(s)
- Chang-Yi Cui
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
40
|
Kimura R, Watanabe C, Kawaguchi A, Kim YI, Park SB, Maki K, Ishida H, Yamaguchi T. Common polymorphisms in WNT10A affect tooth morphology as well as hair shape. Hum Mol Genet 2015; 24:2673-80. [DOI: 10.1093/hmg/ddv014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 01/16/2015] [Indexed: 11/14/2022] Open
|
41
|
|
42
|
Cui CY, Yin M, Sima J, Childress V, Michel M, Piao Y, Schlessinger D. Involvement of Wnt, Eda and Shh at defined stages of sweat gland development. Development 2014; 141:3752-60. [PMID: 25249463 DOI: 10.1242/dev.109231] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
To maintain body temperature, sweat glands develop from embryonic ectoderm by a poorly defined mechanism. We demonstrate a temporal cascade of regulation during mouse sweat gland formation. Sweat gland induction failed completely when canonical Wnt signaling was blocked in skin epithelium, and was accompanied by sharp downregulation of downstream Wnt, Eda and Shh pathway genes. The Wnt antagonist Dkk4 appeared to inhibit this induction: Dkk4 was sharply downregulated in β-catenin-ablated mice, indicating that it is induced by Wnt/β-catenin; however, its overexpression repressed Wnt target genes and significantly reduced gland numbers. Eda signaling succeeded Wnt. Wnt signaling was still active and nascent sweat gland pre-germs were still seen in Eda-null mice, but the pre-germs failed to develop further and the downstream Shh pathway was not activated. When Wnt and Eda were intact but Shh was ablated, germ induction and subsequent duct formation occurred normally, but the final stage of secretory coil formation failed. Thus, sweat gland development shows a relay of regulatory steps initiated by Wnt/β-catenin - itself modulated by Dkk4 - with subsequent participation of Eda and Shh pathways.
Collapse
Affiliation(s)
- Chang-Yi Cui
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Mingzhu Yin
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jian Sima
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Victoria Childress
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Marc Michel
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Yulan Piao
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
43
|
Paulose T, Speroni L, Sonnenschein C, Soto AM. Estrogens in the wrong place at the wrong time: Fetal BPA exposure and mammary cancer. Reprod Toxicol 2014; 54:58-65. [PMID: 25277313 DOI: 10.1016/j.reprotox.2014.09.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/19/2014] [Accepted: 09/09/2014] [Indexed: 01/27/2023]
Abstract
Iatrogenic gestational exposure to diethylstilbestrol (DES) induced alterations of the genital tract and predisposed individuals to develop clear cell carcinoma of the vagina as well as breast cancer later in life. Gestational exposure of rodents to a related compound, the xenoestrogen bisphenol-A (BPA) increases the propensity to develop mammary cancer during adulthood, long after cessation of exposure. Exposure to BPA during gestation induces morphological alterations in both the stroma and the epithelium of the fetal mammary gland at 18 days of age. We postulate that the primary target of BPA is the fetal stroma, the only mammary tissue expressing estrogen receptors during fetal life. BPA would then alter the reciprocal stroma-epithelial interactions that mediate mammogenesis. In addition to this direct effect on the mammary gland, BPA is postulated to affect the hypothalamus and thus in turn affect the regulation of mammotropic hormones at puberty and beyond.
Collapse
MESH Headings
- Age Factors
- Animals
- Benzhydryl Compounds/toxicity
- Breast Neoplasms/chemically induced
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Communication/drug effects
- Cell Transformation, Neoplastic/chemically induced
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Dose-Response Relationship, Drug
- Endocrine Disruptors/toxicity
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Estrogens/toxicity
- Female
- Gestational Age
- Humans
- Hypothalamus/drug effects
- Hypothalamus/metabolism
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Glands, Human/drug effects
- Mammary Glands, Human/growth & development
- Mammary Glands, Human/metabolism
- Mammary Glands, Human/pathology
- Maternal Exposure/adverse effects
- Phenols/toxicity
- Pregnancy
- Prenatal Exposure Delayed Effects
- Risk Assessment
- Risk Factors
- Stromal Cells/drug effects
- Stromal Cells/metabolism
- Stromal Cells/pathology
Collapse
Affiliation(s)
- Tessie Paulose
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, United States.
| | - Lucia Speroni
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, United States.
| | - Carlos Sonnenschein
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, United States.
| | - Ana M Soto
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, United States.
| |
Collapse
|
44
|
Zhang X, Martinez D, Koledova Z, Qiao G, Streuli CH, Lu P. FGF ligands of the postnatal mammary stroma regulate distinct aspects of epithelial morphogenesis. Development 2014; 141:3352-62. [PMID: 25078648 DOI: 10.1242/dev.106732] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
FGF signaling is essential for mammary gland development, yet the mechanisms by which different members of the FGF family control stem cell function and epithelial morphogenesis in this tissue are not well understood. Here, we have examined the requirement of Fgfr2 in mouse mammary gland morphogenesis using a postnatal organ regeneration model. We found that tissue regeneration from basal stem cells is a multistep event, including luminal differentiation and subsequent epithelial branching morphogenesis. Basal cells lacking Fgfr2 did not generate an epithelial network owing to a failure in luminal differentiation. Moreover, Fgfr2 null epithelium was unable to undergo ductal branch initiation and elongation due to a deficiency in directional migration. We identified FGF10 and FGF2 as stromal ligands that control distinct aspects of mammary ductal branching. FGF10 regulates branch initiation, which depends on directional epithelial migration. By contrast, FGF2 controls ductal elongation, requiring cell proliferation and epithelial expansion. Together, our data highlight a pleiotropic role of Fgfr2 in stem cell differentiation and branch initiation, and reveal that different FGF ligands regulate distinct aspects of epithelial behavior.
Collapse
Affiliation(s)
- Xiaohong Zhang
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Denisse Martinez
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Zuzana Koledova
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Guijuan Qiao
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Charles H Streuli
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Pengfei Lu
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
45
|
Chalkias H, Ekman E, Lundeheim N, Rydhmer L, Jacobson M. Inverted teats (Mammillae invertitae) in gilts – Effect on piglet survival and growth rate. J Anim Sci 2014; 92:2587-94. [DOI: 10.2527/jas.2013-7387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- H. Chalkias
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Box 7023, SE-750 07 Uppsala, Sweden
| | - E. Ekman
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Box 7028, SE-750 07 Uppsala, Sweden
| | - N. Lundeheim
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Box 7023, SE-750 07 Uppsala, Sweden
| | - L. Rydhmer
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Box 7023, SE-750 07 Uppsala, Sweden
| | - M. Jacobson
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, SE-750 07 Uppsala, Sweden
| |
Collapse
|
46
|
Ahtiainen L, Lefebvre S, Lindfors PH, Renvoisé E, Shirokova V, Vartiainen MK, Thesleff I, Mikkola ML. Directional cell migration, but not proliferation, drives hair placode morphogenesis. Dev Cell 2014; 28:588-602. [PMID: 24636260 DOI: 10.1016/j.devcel.2014.02.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 01/08/2014] [Accepted: 02/04/2014] [Indexed: 12/27/2022]
Abstract
Epithelial reorganization involves coordinated changes in cell shapes and movements. This restructuring occurs during formation of placodes, ectodermal thickenings that initiate the morphogenesis of epithelial organs including hair, mammary gland, and tooth. Signaling pathways in ectodermal placode formation are well known, but the cellular mechanisms have remained ill defined. We established imaging methodology for live visualization of embryonic skin explants during the first wave of hair placode formation. We found that the vast majority of placodal cells were nonproliferative throughout morphogenesis. We show that cell compaction and centripetal migration are the main cellular mechanisms associated with hair placode morphogenesis and that inhibition of actin remodeling suppresses placode formation. Stimulation of both ectodysplasin/NF-κB and Wnt/β-catenin signaling increased cell motility and the number of cells committed to placodal fate. Thus, cell fate choices and morphogenetic events are controlled by the same molecular pathways, providing the framework for coordination of these two processes.
Collapse
Affiliation(s)
- Laura Ahtiainen
- Developmental Biology Program, Institute of Biotechnology, 00014 University of Helsinki, Helsinki, Finland.
| | - Sylvie Lefebvre
- Developmental Biology Program, Institute of Biotechnology, 00014 University of Helsinki, Helsinki, Finland
| | - Päivi H Lindfors
- Developmental Biology Program, Institute of Biotechnology, 00014 University of Helsinki, Helsinki, Finland
| | - Elodie Renvoisé
- Developmental Biology Program, Institute of Biotechnology, 00014 University of Helsinki, Helsinki, Finland
| | - Vera Shirokova
- Developmental Biology Program, Institute of Biotechnology, 00014 University of Helsinki, Helsinki, Finland
| | - Maria K Vartiainen
- Cell and Molecular Biology Program, Institute of Biotechnology, 00014 University of Helsinki, Helsinki, Finland
| | - Irma Thesleff
- Developmental Biology Program, Institute of Biotechnology, 00014 University of Helsinki, Helsinki, Finland
| | - Marja L Mikkola
- Developmental Biology Program, Institute of Biotechnology, 00014 University of Helsinki, Helsinki, Finland.
| |
Collapse
|
47
|
Burger K, Schneider AT, Wohlfart S, Kiesewetter F, Huttner K, Johnson R, Schneider H. Genotype-phenotype correlation in boys with X-linked hypohidrotic ectodermal dysplasia. Am J Med Genet A 2014; 164A:2424-32. [PMID: 24715423 DOI: 10.1002/ajmg.a.36541] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/16/2014] [Indexed: 11/06/2022]
Abstract
X-linked hypohidrotic ectodermal dysplasia (XLHED), the most frequent form of ectodermal dysplasia, is a genetic disorder of ectoderm development characterized by malformation of multiple ectodermal structures such as skin, hair, sweat and sebaceous glands, and teeth. The disease is caused by a broad spectrum of mutations in the gene EDA. Although XLHED symptoms show inter-familial and intra-familial variability, genotype-phenotype correlation has been demonstrated with respect to sweat gland function. In this study, we investigated to which extent the EDA genotype correlates with the severity of XLHED-related skin and hair signs. Nineteen male children with XLHED (age range 3-14 years) and seven controls (aged 6-14 years) were examined by confocal microscopy of the skin, quantification of pilocarpine-induced sweating, semi-quantitative evaluation of full facial photographs with respect to XLHED-related skin issues, and phototrichogram analysis. All eight boys with known hypomorphic EDA mutations were able to produce at least some sweat and showed less severe cutaneous signs of XLHED than the anhidrotic XLHED patients (e.g., perioral and periorbital eczema or hyperpigmentation, regional hyperkeratosis, characteristic wrinkles under the eyes). As expected, individuals with XLHED had significantly less and thinner hair than healthy controls. However, there were also significant differences in hair number, diameter, and other hair characteristics between the group with hypomorphic EDA mutations and the anhidrotic patients. In summary, this study indicated a remarkable genotype-phenotype correlation of skin and hair findings in prepubescent males with XLHED.
Collapse
Affiliation(s)
- Kristin Burger
- Department of Pediatrics, German Competence Center for Children with Ectodermal Dysplasias, University Hospital Erlangen, University of Erlangen-Nürnberg, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Iida Y, Hibiya K, Inohaya K, Kudo A. Eda/Edar signaling guides fin ray formation with preceding osteoblast differentiation, as revealed by analyses of the medaka all-fin less mutantafl. Dev Dyn 2014; 243:765-77. [DOI: 10.1002/dvdy.24120] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 12/20/2022] Open
Affiliation(s)
- Yuuki Iida
- Department of Biological Information; Tokyo Institute of Technology; Yokohama Japan
| | - Kenta Hibiya
- Department of Biological Information; Tokyo Institute of Technology; Yokohama Japan
| | - Keiji Inohaya
- Department of Biological Information; Tokyo Institute of Technology; Yokohama Japan
| | - Akira Kudo
- Department of Biological Information; Tokyo Institute of Technology; Yokohama Japan
| |
Collapse
|
49
|
Lu C, Fuchs E. Sweat gland progenitors in development, homeostasis, and wound repair. Cold Spring Harb Perspect Med 2014; 4:4/2/a015222. [PMID: 24492848 DOI: 10.1101/cshperspect.a015222] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The human body is covered with several million sweat glands. These tiny coiled tubular skin appendages produce the sweat that is our primary source of cooling and hydration of the skin. Numerous studies have been published on their morphology and physiology. Until recently, however, little was known about how glandular skin maintains homeostasis and repairs itself after tissue injury. Here, we provide a brief overview of sweat gland biology, including newly identified reservoirs of stem cells in glandular skin and their activation in response to different types of injuries. Finally, we discuss how the genetics and biology of glandular skin has advanced our knowledge of human disorders associated with altered sweat gland activity.
Collapse
Affiliation(s)
- Catherine Lu
- Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065
| | | |
Collapse
|
50
|
Howard BA, Lu P. Stromal regulation of embryonic and postnatal mammary epithelial development and differentiation. Semin Cell Dev Biol 2014; 25-26:43-51. [DOI: 10.1016/j.semcdb.2014.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/19/2013] [Accepted: 01/09/2014] [Indexed: 01/06/2023]
|