1
|
Yang S, Palmquist KH, Nathan L, Pfeifer CR, Schultheiss PJ, Sharma A, Kam LC, Miller PW, Shyer AE, Rodrigues AR. Morphogens enable interacting supracellular phases that generate organ architecture. Science 2023; 382:eadg5579. [PMID: 37995219 DOI: 10.1126/science.adg5579] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 09/27/2023] [Indexed: 11/25/2023]
Abstract
During vertebrate organogenesis, increases in morphological complexity are tightly coupled to morphogen expression. In this work, we studied how morphogens influence self-organizing processes at the collective or "supra"-cellular scale in avian skin. We made physical measurements across length scales, which revealed morphogen-enabled material property differences that were amplified at supracellular scales in comparison to cellular scales. At the supracellular scale, we found that fibroblast growth factor (FGF) promoted "solidification" of tissues, whereas bone morphogenetic protein (BMP) promoted fluidity and enhanced mechanical activity. Together, these effects created basement membrane-less compartments within mesenchymal tissue that were mechanically primed to drive avian skin tissue budding. Understanding this multiscale process requires the ability to distinguish between proximal effects of morphogens that occur at the cellular scale and their functional effects, which emerge at the supracellular scale.
Collapse
Affiliation(s)
- Sichen Yang
- Laboratory of Morphogenesis, The Rockefeller University, New York, NY 10065, USA
| | - Karl H Palmquist
- Laboratory of Morphogenesis, The Rockefeller University, New York, NY 10065, USA
| | - Levy Nathan
- Laboratory of Morphogenesis, The Rockefeller University, New York, NY 10065, USA
| | - Charlotte R Pfeifer
- Laboratory of Morphogenesis, The Rockefeller University, New York, NY 10065, USA
| | - Paula J Schultheiss
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Anurag Sharma
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Pearson W Miller
- Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA
| | - Amy E Shyer
- Laboratory of Morphogenesis, The Rockefeller University, New York, NY 10065, USA
| | - Alan R Rodrigues
- Laboratory of Morphogenesis, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
2
|
Evanitsky MN, Di Talia S. An active traveling wave of Eda/NF-κB signaling controls the timing and hexagonal pattern of skin appendages in zebrafish. Development 2023; 150:dev201866. [PMID: 37747266 PMCID: PMC10560567 DOI: 10.1242/dev.201866] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/11/2023] [Indexed: 09/26/2023]
Abstract
Periodic patterns drive the formation of a variety of tissues, including skin appendages such as feathers and scales. Skin appendages serve important and diverse functions across vertebrates, yet the mechanisms that regulate their patterning are not fully understood. Here, we have used live imaging to investigate dynamic signals regulating the ontogeny of zebrafish scales. Scales are bony skin appendages that develop sequentially along the anterior-posterior and dorsal-ventral axes to cover the fish in a hexagonal array. We have found that scale development requires cell-cell communication and is coordinated through an active wave mechanism. Using a live transcriptional reporter, we show that a wave of Eda/NF-κB activity precedes scale initiation and is required for scale formation. Experiments decoupling the propagation of the wave from dermal placode formation and osteoblast differentiation demonstrate that the Eda/NF-κB activity wavefront controls the timing of the sequential patterning of scales. Moreover, this decoupling resulted in defects in scale size and significant deviations in the hexagonal patterning of scales. Thus, our results demonstrate that a biochemical traveling wave coordinates scale initiation and proper hexagonal patterning across the fish body.
Collapse
Affiliation(s)
- Maya N. Evanitsky
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
3
|
Ji G, Zhang M, Tu Y, Liu Y, Shan Y, Ju X, Zou J, Shu J, Sheng Z, Li H. Molecular Regulatory Mechanisms in Chicken Feather Follicle Morphogenesis. Genes (Basel) 2023; 14:1646. [PMID: 37628697 PMCID: PMC10454116 DOI: 10.3390/genes14081646] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
In China, the sale of freshly slaughtered chickens is becoming increasingly popular in comparison with that of live chickens, and due to this emerging trend, the skin and feather follicle traits of yellow-feathered broilers have attracted a great deal of research attention. The feather follicle originates from the interaction between the epidermis and dermis in the early embryonic stage. Feather follicle morphogenesis is regulated by the Wnt, ectodysplasin (Eda), epidermal growth factor (EGF), fibroblast growth factor (FGF), bone morphogenetic protein (BMP), sonic hedgehog (Shh), Notch, and other signaling pathways that exist in epithelial and mesenchymal cells. The Wnt pathway is essential for feather follicle and feather morphogenesis. Eda interacts with Wnt to induce FGF expression, which attracts mesenchymal cell movement and aggregates to form feather follicle primordia. BMP acts as an inhibitor of the above signaling pathways to limit the size of the feather tract and distance between neighboring feather primordia in a dose-dependent manner. The Notch/Delta pathway can interact with the FGF pathway to promote feather bud formation. While not a part of the early morphogenesis of feather follicles, Shh and BMP signaling are involved in late feather branching. This review summarizes the roles of miRNAs/lncRNA in the regulation of feather follicle and feather growth and development and suggests topics that need to be solved in a future study. This review focuses on the regulatory mechanisms involved in feather follicle morphogenesis and analyzes the impact of SNP sites on feather follicle traits in poultry. This work may help us to understand the molecular regulatory networks influencing feather follicle growth and provide basic data for poultry carcass quality.
Collapse
Affiliation(s)
- Gaige Ji
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Chinese Academy of Agricultural Science, Institute of Poultry Science, Yangzhou 225125, China
| | - Ming Zhang
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Chinese Academy of Agricultural Science, Institute of Poultry Science, Yangzhou 225125, China
| | - Yunjie Tu
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Chinese Academy of Agricultural Science, Institute of Poultry Science, Yangzhou 225125, China
| | - Yifan Liu
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Chinese Academy of Agricultural Science, Institute of Poultry Science, Yangzhou 225125, China
| | - Yanju Shan
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Chinese Academy of Agricultural Science, Institute of Poultry Science, Yangzhou 225125, China
| | - Xiaojun Ju
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Chinese Academy of Agricultural Science, Institute of Poultry Science, Yangzhou 225125, China
| | - Jianmin Zou
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Chinese Academy of Agricultural Science, Institute of Poultry Science, Yangzhou 225125, China
| | - Jingting Shu
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Chinese Academy of Agricultural Science, Institute of Poultry Science, Yangzhou 225125, China
| | - Zhongwei Sheng
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Chinese Academy of Agricultural Science, Institute of Poultry Science, Yangzhou 225125, China
| | - Hua Li
- School of Life Science and Engineering, Foshan University, Foshan 528231, China
| |
Collapse
|
4
|
Evanitsky MN, Di Talia S. An active traveling wave of Eda/NF-kB signaling controls the timing and hexagonal pattern of skin appendages in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536269. [PMID: 37090617 PMCID: PMC10120683 DOI: 10.1101/2023.04.10.536269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Periodic patterns make up a variety of tissues, including skin appendages such as feathers and scales. Skin appendages serve important and diverse functions across vertebrates, yet the mechanisms that regulate their patterning are not fully understood. Here, we have used live imaging to investigate dynamic signals regulating the ontogeny of zebrafish scales. Scales are bony skin appendages which develop sequentially along the anterior-posterior and dorsal-ventral axes to cover the fish in a hexagonal array. We have found that scale development requires cell-cell communication and is coordinated through an active wave mechanism. Using a live transcriptional reporter, we show that a wave of Eda/NF-κB activity precedes scale initiation and is required for scale formation. Experiments decoupling the propagation of the wave from dermal placode formation and osteoblast differentiation demonstrate that the Eda/NF-kB activity wavefront times the sequential patterning of scales. Moreover, this decoupling resulted in defects in scale size and significant deviations in the hexagonal patterning of scales. Thus, our results demonstrate that a biochemical traveling wave coordinates scale initiation and proper hexagonal patterning across the fish body.
Collapse
Affiliation(s)
- Maya N Evanitsky
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
5
|
Genetic and ecological drivers of molt in a migratory bird. Sci Rep 2023; 13:814. [PMID: 36646769 PMCID: PMC9842746 DOI: 10.1038/s41598-022-26973-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/22/2022] [Indexed: 01/18/2023] Open
Abstract
The ability of animals to sync the timing and location of molting (the replacement of hair, skin, exoskeletons or feathers) with peaks in resource availability has important implications for their ecology and evolution. In migratory birds, the timing and location of pre-migratory feather molting, a period when feathers are shed and replaced with newer, more aerodynamic feathers, can vary within and between species. While hypotheses to explain the evolution of intraspecific variation in the timing and location of molt have been proposed, little is known about the genetic basis of this trait or the specific environmental drivers that may result in natural selection for distinct molting phenotypes. Here we take advantage of intraspecific variation in the timing and location of molt in the iconic songbird, the Painted Bunting (Passerina ciris) to investigate the genetic and ecological drivers of distinct molting phenotypes. Specifically, we use genome-wide genetic sequencing in combination with stable isotope analysis to determine population genetic structure and molting phenotype across thirteen breeding sites. We then use genome-wide association analysis (GWAS) to identify a suite of genes associated with molting and pair this with gene-environment association analysis (GEA) to investigate potential environmental drivers of genetic variation in this trait. Associations between genetic variation in molt-linked genes and the environment are further tested via targeted SNP genotyping in 25 additional breeding populations across the range. Together, our integrative analysis suggests that molting is in part regulated by genes linked to feather development and structure (GLI2 and CSPG4) and that genetic variation in these genes is associated with seasonal variation in precipitation and aridity. Overall, this work provides important insights into the genetic basis and potential selective forces behind phenotypic variation in what is arguably one of the most important fitness-linked traits in a migratory bird.
Collapse
|
6
|
Zimm R, Oberdick D, Gnetneva A, Schneider P, Cebra-Thomas J, Moustakas-Verho JE. Turing's turtles all the way down: A conserved role of EDAR in the carapacial ridge suggests a deep homology of prepatterns across ectodermal appendages. Anat Rec (Hoboken) 2022; 306:1201-1213. [PMID: 36239299 DOI: 10.1002/ar.25096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 11/11/2022]
Abstract
The scutes of the turtle shell are epidermal shields that begin their formation during the early stages of shell development. Like other skin appendages, turtle scutes are hypothesized to be patterned by reaction-diffusion systems. We have previously established ex vivo and in silico systems to study these mechanisms experimentally and have further shown that mathematical models can explain the dynamics of the induction of turtle scute primordia and the generation of final scute architecture. Using these foundations, we expand our current knowledge and test the roles of ectodysplasin and activin signaling in the development of turtle scutes. We find that these molecules play important roles in the prepatterning of scute primordia along the carapacial ridge and show that blocking Edar signaling may lead to a complete loss of marginal scute primordia. We show that it is possible to reproduce these observations using simple mathematical modeling, thereby suggesting a stabilizing role for ectodysplasin within the reaction-diffusion mechanisms. Finally, we argue that our findings further entrench turtle scutes within a class of developmental systems composed of hierarchically nested reaction-diffusion mechanisms, which is conserved across ectodermal organs.
Collapse
Affiliation(s)
- Roland Zimm
- Institute of Functional Genomics, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Danielle Oberdick
- Department of Biology, Millersville University, Millersville, Pennsylvania, USA
| | - Anna Gnetneva
- Zoological Institute of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Judith Cebra-Thomas
- Department of Biology, Millersville University, Millersville, Pennsylvania, USA
| | | |
Collapse
|
7
|
Cui Y, Wang C, Liu L, Liu N, He J. Expression and distribution of EPHA4 and Ephrin A3 in Aohan fine-wool sheep skin. Arch Anim Breed 2022; 65:11-19. [PMID: 35047658 PMCID: PMC8759078 DOI: 10.5194/aab-65-11-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/16/2021] [Indexed: 11/24/2022] Open
Abstract
The objective of this study was to identify the expression and
distribution of EPHA4 and Ephrin A3 genes in the development and morphogenesis of hair
follicles in fine-wool sheep. The results could lay a theoretical basis for
understanding the molecular mechanism that regulates hair follicle
development. The skin of Aohan fine-wool sheep at different developmental
stages (embryonic day 90, E90d, and 120, E120d, and postnatal day 1, B1d,
and 30, B30d) were selected. Real-time quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry were used to
study the levels of mRNA and proteins, respectively. The RT-qPCR results
showed that the mRNA expression level of EPHA4 at B1d was significantly lower
than at E120d (p<0.01). The expression of Ephrin A3 at E120d was
significantly higher than that at E90d and B1d (p<0.01).
Immunohistochemical detection results showed that the level and localisation
of EPHA4 and Ephrin A3 proteins had spatial and temporal specificity. EPHA4 expression in dermal
papilla cells might be important for inducing Aohan fine-hair follicle
regeneration and for controlling the properties of the hair. Ephrin A3 might play an
important role in the redifferentiation of secondary hair follicles and
might also be involved in the inhibition of apoptosis-related gene
expression in hair follicles. The Ephrin A3 signalling pathway might accelerate the
growth of fine-hair follicles and increase the density of hair follicles.
Collapse
Affiliation(s)
- Yu Cui
- College of Animal Science and Technology, Qingdao Agricultural
University, Qingdao, Shandong 266109, China
| | - Chunliang Wang
- Nanchang police dog base of the Ministry of public security,
Nanchang, Jiangxi 330100, China
| | - Lirong Liu
- China Animal Health and Epidemiology Center, Qingdao, Shandong
266032, China
| | - Nan Liu
- College of Animal Science and Technology, Qingdao Agricultural
University, Qingdao, Shandong 266109, China
| | - Jianning He
- College of Animal Science and Technology, Qingdao Agricultural
University, Qingdao, Shandong 266109, China
| |
Collapse
|
8
|
Dushyanth K, Shukla R, Chatterjee RN, Bhattacharya TK. Expression and polymorphism of Follistatin ( FST) gene and its association with growth traits in native and exotic chicken. Anim Biotechnol 2020; 33:824-834. [PMID: 33170076 DOI: 10.1080/10495398.2020.1838917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Follistatin (FST), a member of the transforming growth factor beta super-family regulates body growth by inhibiting the binding of myostatin (an inhibitor of growth) with its receptor in chicken. An experiment was conducted to explore ontogenic expression of the follistatin gene, determine polymorphism at the coding region of the gene and estimate its effect on growth traits in native (Aseel) and exotic broiler (PD-1) and layer (White Leghorn) chicken. The significant differences of FST gene expression were observed among the breeds revealing significantly (p < 0.05) higher expression in PD-1 line followed by White Leghorn and Aseel breeds during both embryonic and post-hatch period. The polymorphism at the functional domain of the FST gene was identified with the presence of 4 haplotypes. The follistatin haplogroups had the significant effect on body weights (p < 0.05) at 42 days of age in the White Leghorn, PD-1 and Aseel breeds (h1h1 in PD-1, h1h4 in White Leghorn and h1h2 haplogroups in Aseel breeds had the highest body weights of 770.04 ± 12.96, 246.28 ± 7.60 and 270.00 ± 10.68 g, respectively). It is concluded that the follistatin gene expressed differently during the embryonic and post-embryonic period across the breeds and the coding region of the gene was polymorphic having significant effects on growth traits in chicken.
Collapse
Affiliation(s)
- K Dushyanth
- ICAR-Directorate of Poultry Research, Hyderabad, India
| | - R Shukla
- ICAR-Directorate of Poultry Research, Hyderabad, India
| | | | | |
Collapse
|
9
|
Ho WKW, Freem L, Zhao D, Painter KJ, Woolley TE, Gaffney EA, McGrew MJ, Tzika A, Milinkovitch MC, Schneider P, Drusko A, Matthäus F, Glover JD, Wells KL, Johansson JA, Davey MG, Sang HM, Clinton M, Headon DJ. Feather arrays are patterned by interacting signalling and cell density waves. PLoS Biol 2019; 17:e3000132. [PMID: 30789897 PMCID: PMC6383868 DOI: 10.1371/journal.pbio.3000132] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/17/2019] [Indexed: 12/30/2022] Open
Abstract
Feathers are arranged in a precise pattern in avian skin. They first arise during development in a row along the dorsal midline, with rows of new feather buds added sequentially in a spreading wave. We show that the patterning of feathers relies on coupled fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) signalling together with mesenchymal cell movement, acting in a coordinated reaction-diffusion-taxis system. This periodic patterning system is partly mechanochemical, with mechanical-chemical integration occurring through a positive feedback loop centred on FGF20, which induces cell aggregation, mechanically compressing the epidermis to rapidly intensify FGF20 expression. The travelling wave of feather formation is imposed by expanding expression of Ectodysplasin A (EDA), which initiates the expression of FGF20. The EDA wave spreads across a mesenchymal cell density gradient, triggering pattern formation by lowering the threshold of mesenchymal cells required to begin to form a feather bud. These waves, and the precise arrangement of feather primordia, are lost in the flightless emu and ostrich, though via different developmental routes. The ostrich retains the tract arrangement characteristic of birds in general but lays down feather primordia without a wave, akin to the process of hair follicle formation in mammalian embryos. The embryonic emu skin lacks sufficient cells to enact feather formation, causing failure of tract formation, and instead the entire skin gains feather primordia through a later process. This work shows that a reaction-diffusion-taxis system, integrated with mechanical processes, generates the feather array. In flighted birds, the key role of the EDA/Ectodysplasin A receptor (EDAR) pathway in vertebrate skin patterning has been recast to activate this process in a quasi-1-dimensional manner, imposing highly ordered pattern formation.
Collapse
Affiliation(s)
- William K. W. Ho
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Lucy Freem
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Debiao Zhao
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Kevin J. Painter
- School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Thomas E. Woolley
- School of Mathematics, Cardiff University, Cathays, Cardiff, United Kingdom
| | - Eamonn A. Gaffney
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Michael J. McGrew
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Athanasia Tzika
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | | | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Armin Drusko
- FIAS and Faculty of Biological Sciences, University of Frankfurt, Frankfurt, Germany
| | - Franziska Matthäus
- FIAS and Faculty of Biological Sciences, University of Frankfurt, Frankfurt, Germany
| | - James D. Glover
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Kirsty L. Wells
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Jeanette A. Johansson
- Cancer Research UK Edinburgh Centre and MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Megan G. Davey
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Helen M. Sang
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael Clinton
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Denis J. Headon
- Roslin Institute Chicken Embryology, Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
10
|
Toomey MB, Marques CI, Andrade P, Araújo PM, Sabatino S, Gazda MA, Afonso S, Lopes RJ, Corbo JC, Carneiro M. A non-coding region near Follistatin controls head colour polymorphism in the Gouldian finch. Proc Biol Sci 2018; 285:20181788. [PMID: 30282656 PMCID: PMC6191701 DOI: 10.1098/rspb.2018.1788] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/05/2018] [Indexed: 12/17/2022] Open
Abstract
Discrete colour morphs coexisting within a single population are common in nature. In a broad range of organisms, sympatric colour morphs often display major differences in other traits, including morphology, physiology or behaviour. Despite the repeated occurrence of this phenomenon, our understanding of the genetics that underlie multi-trait differences and the factors that promote the long-term maintenance of phenotypic variability within a freely interbreeding population are incomplete. Here, we investigated the genetic basis of red and black head colour in the Gouldian finch (Erythrura gouldiae), a classic polymorphic system in which naturally occurring colour morphs also display differences in aggressivity and reproductive success. We show that the candidate locus is a small (approx. 70 kb) non-coding region mapping to the Z chromosome near the Follistatin (FST) gene. Unlike recent findings in other systems where phenotypic morphs are explained by large inversions containing hundreds of genes (so-called supergenes), we did not identify any structural rearrangements between the two haplotypes using linked-read sequencing technology. Nucleotide divergence between the red and black alleles was high when compared to the remainder of the Z chromosome, consistent with their maintenance as balanced polymorphisms over several million years. Our results illustrate how pleiotropic phenotypes can arise from simple genetic variation, probably regulatory in nature.
Collapse
Affiliation(s)
- Matthew B Toomey
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Cristiana I Marques
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, 4485-661 Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Pedro Andrade
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, 4485-661 Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Pedro M Araújo
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, 4485-661 Vairão, Portugal
- Centro de Ciências do Mar e do Ambiente, Departamento de Ciências da Vida, Universidade de Coimbra, 3004-517 Coimbra, Portugal
| | - Stephen Sabatino
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, 4485-661 Vairão, Portugal
| | - Małgorzata A Gazda
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, 4485-661 Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Sandra Afonso
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, 4485-661 Vairão, Portugal
| | - Ricardo J Lopes
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, 4485-661 Vairão, Portugal
| | - Joseph C Corbo
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Miguel Carneiro
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, 4485-661 Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| |
Collapse
|
11
|
Schneider RA. Neural crest and the origin of species-specific pattern. Genesis 2018; 56:e23219. [PMID: 30134069 PMCID: PMC6108449 DOI: 10.1002/dvg.23219] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/20/2022]
Abstract
For well over half of the 150 years since the discovery of the neural crest, the special ability of these cells to function as a source of species-specific pattern has been clearly recognized. Initially, this observation arose in association with chimeric transplant experiments among differentially pigmented amphibians, where the neural crest origin for melanocytes had been duly noted. Shortly thereafter, the role of cranial neural crest cells in transmitting species-specific information on size and shape to the pharyngeal arch skeleton as well as in regulating the timing of its differentiation became readily apparent. Since then, what has emerged is a deeper understanding of how the neural crest accomplishes such a presumably difficult mission, and this includes a more complete picture of the molecular and cellular programs whereby neural crest shapes the face of each species. This review covers studies on a broad range of vertebrates and describes neural-crest-mediated mechanisms that endow the craniofacial complex with species-specific pattern. A major focus is on experiments in quail and duck embryos that reveal a hierarchy of cell-autonomous and non-autonomous signaling interactions through which neural crest generates species-specific pattern in the craniofacial integument, skeleton, and musculature. By controlling size and shape throughout the development of these systems, the neural crest underlies the structural and functional integration of the craniofacial complex during evolution.
Collapse
Affiliation(s)
- Richard A. Schneider
- Department of Orthopedic SurgeryUniversity of California at San Francisco, 513 Parnassus AvenueS‐1161San Francisco, California
| |
Collapse
|
12
|
Kahata K, Dadras MS, Moustakas A. TGF-β Family Signaling in Epithelial Differentiation and Epithelial-Mesenchymal Transition. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a022194. [PMID: 28246184 DOI: 10.1101/cshperspect.a022194] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epithelia exist in the animal body since the onset of embryonic development; they generate tissue barriers and specify organs and glands. Through epithelial-mesenchymal transitions (EMTs), epithelia generate mesenchymal cells that form new tissues and promote healing or disease manifestation when epithelial homeostasis is challenged physiologically or pathologically. Transforming growth factor-βs (TGF-βs), activins, bone morphogenetic proteins (BMPs), and growth and differentiation factors (GDFs) have been implicated in the regulation of epithelial differentiation. These TGF-β family ligands are expressed and secreted at sites where the epithelium interacts with the mesenchyme and provide paracrine queues from the mesenchyme to the neighboring epithelium, helping the specification of differentiated epithelial cell types within an organ. TGF-β ligands signal via Smads and cooperating kinase pathways and control the expression or activities of key transcription factors that promote either epithelial differentiation or mesenchymal transitions. In this review, we discuss evidence that illustrates how TGF-β family ligands contribute to epithelial differentiation and induce mesenchymal transitions, by focusing on the embryonic ectoderm and tissues that form the external mammalian body lining.
Collapse
Affiliation(s)
- Kaoru Kahata
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Mahsa Shahidi Dadras
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Aristidis Moustakas
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
13
|
A chemotaxis model of feather primordia pattern formation during avian development. J Theor Biol 2018; 437:225-238. [DOI: 10.1016/j.jtbi.2017.10.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/17/2017] [Accepted: 10/25/2017] [Indexed: 11/21/2022]
|
14
|
Saad K, Theis S, Otto A, Luke G, Patel K. Detailed expression profile of the six Glypicans and their modifying enzyme, Notum during chick limb and feather development. Gene 2017; 610:71-79. [PMID: 28192166 DOI: 10.1016/j.gene.2017.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/30/2017] [Accepted: 02/06/2017] [Indexed: 11/29/2022]
Abstract
The development of vertebrate appendages, especially the limb and feather buds are orchestrated by numerous secreted signalling molecules including Sonic Hedgehog, Bone Morphogenetic Proteins, Fibroblast Growth Factors and Wnts. These proteins coordinate the growth and patterning of ectodermal and mesenchymal cells. The influence of signalling molecules is affected over large distances by their concentration (morphogen activity) but also at local levels by the presence of proteins that either attenuate or promote their activity. Glypicans are cell surface molecules that regulate the activity of the major secreted signalling molecules expressed in the limb and feather bud. Here we investigated the expression of all Glypicans during chick limb and feather development. In addition we profiled the expression of Notum, an enzyme that regulates Glypican activity. We show that five of the six Glypicans and Notum are expressed in a dynamic manner during the development of limbs and feathers. We also investigated the expression of key Glypicans and show that they are controlled by signalling molecules highlighting the presence of feedback loops. Lastly we show that Glypicans and Notum are expressed in a tissue specific manner in adult chicken tissues. Our results strongly suggest that the Glypicans and Notum have many as yet undiscovered roles to play during the development of vertebrate appendages.
Collapse
Affiliation(s)
- Kawakeb Saad
- School of Biological Sciences, University of Reading, Reading, UK
| | - Susanne Theis
- School of Biological Sciences, University of Reading, Reading, UK
| | - Anthony Otto
- School of Biological Sciences, University of Reading, Reading, UK
| | - Graham Luke
- School of Biological Sciences, University of Reading, Reading, UK
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK; Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
15
|
Functional roles of Aves class-specific cis-regulatory elements on macroevolution of bird-specific features. Nat Commun 2017; 8:14229. [PMID: 28165450 PMCID: PMC5473641 DOI: 10.1038/ncomms14229] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 12/12/2016] [Indexed: 01/01/2023] Open
Abstract
Unlike microevolutionary processes, little is known about the genetic basis of macroevolutionary processes. One of these magnificent examples is the transition from non-avian dinosaurs to birds that has created numerous evolutionary innovations such as self-powered flight and its associated wings with flight feathers. By analysing 48 bird genomes, we identified millions of avian-specific highly conserved elements (ASHCEs) that predominantly (>99%) reside in non-coding regions. Many ASHCEs show differential histone modifications that may participate in regulation of limb development. Comparative embryonic gene expression analyses across tetrapod species suggest ASHCE-associated genes have unique roles in developing avian limbs. In particular, we demonstrate how the ASHCE driven avian-specific expression of gene Sim1 driven by ASHCE may be associated with the evolution and development of flight feathers. Together, these findings demonstrate regulatory roles of ASHCEs in the creation of avian-specific traits, and further highlight the importance of cis-regulatory rewiring during macroevolutionary changes.
Collapse
|
16
|
Toews D, Taylor S, Vallender R, Brelsford A, Butcher B, Messer P, Lovette I. Plumage Genes and Little Else Distinguish the Genomes of Hybridizing Warblers. Curr Biol 2016; 26:2313-8. [DOI: 10.1016/j.cub.2016.06.034] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/13/2016] [Accepted: 06/15/2016] [Indexed: 01/04/2023]
|
17
|
Chen CC, Murray PJ, Jiang TX, Plikus MV, Chang YT, Lee OK, Widelitz RB, Chuong CM. Regenerative hair waves in aging mice and extra-follicular modulators follistatin, dkk1, and sfrp4. J Invest Dermatol 2014; 134:2086-2096. [PMID: 24618599 PMCID: PMC4102635 DOI: 10.1038/jid.2014.139] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/12/2014] [Accepted: 02/20/2014] [Indexed: 01/03/2023]
Abstract
Hair cycling is modulated by factors both intrinsic and extrinsic to hair follicles. Cycling defects lead to conditions such as aging-associated alopecia. Recently, we demonstrated that mouse skin exhibits regenerative hair waves, reflecting a coordinated regenerative behavior in follicle populations. Here, we use this model to explore the regenerative behavior of aging mouse skin. Old mice (>18 months) tracked over several months show that with progressing age, hair waves slow down, wave propagation becomes restricted, and hair cycle domains fragment into smaller domains. Transplanting aged donor mouse skin to a young host can restore donor cycling within a 3 mm range of the interface, suggesting that changes are due to extracellular factors. Therefore, hair stem cells in aged skin can be reactivated. Molecular studies show that extra-follicular modulators Bmp2, Dkk1, and Sfrp4 increase in early anagen. Further, we identify follistatin as an extra-follicular modulator, which is highly expressed in late telogen and early anagen. Indeed, follistatin induces hair wave propagation and its level decreases in aging mice. We present an excitable medium model to simulate the cycling behavior in aging mice and illustrate how the interorgan macroenvironment can regulate the aging process by integrating both "activator" and "inhibitor" signals.
Collapse
Affiliation(s)
- Chih-Chiang Chen
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Institute of Clinical Medicine and Department of Dermatology, National Yang-Ming University, Taipei, Taiwan; Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | - Ting Xin Jiang
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Yun-Ting Chang
- Institute of Clinical Medicine and Department of Dermatology, National Yang-Ming University, Taipei, Taiwan
| | - Oscar K Lee
- Institute of Clinical Medicine and Department of Dermatology, National Yang-Ming University, Taipei, Taiwan; Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei and Center for Stem Cell Research, National Yang-Ming University and Veterans General Hospital, Taipei, Taiwan
| | - Randall B Widelitz
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Institute of Clinical Medicine and Department of Dermatology, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
18
|
Abstract
Tooth replacement is a common trait to most vertebrates, including mammals. Mammals, however, have lost the capacity for continuous tooth renewal seen in most other vertebrates, and typically have only 1–2 generations of teeth. Here, we review the mechanisms of tooth replacement in reptiles and mammals, and discuss in detail the current and historical theories on control of timing and pattern of tooth replacement and development.
Collapse
|
19
|
Origin and Development of Hair Cell Orientation in the Inner Ear. INSIGHTS FROM COMPARATIVE HEARING RESEARCH 2013. [DOI: 10.1007/2506_2013_28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Painter KJ, Hunt GS, Wells KL, Johansson JA, Headon DJ. Towards an integrated experimental-theoretical approach for assessing the mechanistic basis of hair and feather morphogenesis. Interface Focus 2012; 2:433-50. [PMID: 23919127 DOI: 10.1098/rsfs.2011.0122] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 01/23/2012] [Indexed: 01/21/2023] Open
Abstract
In his seminal 1952 paper, 'The Chemical Basis of Morphogenesis', Alan Turing lays down a milestone in the application of theoretical approaches to understand complex biological processes. His deceptively simple demonstration that a system of reacting and diffusing chemicals could, under certain conditions, generate spatial patterning out of homogeneity provided an elegant solution to the problem of how one of nature's most intricate events occurs: the emergence of structure and form in the developing embryo. The molecular revolution that has taken place during the six decades following this landmark publication has now placed this generation of theoreticians and biologists in an excellent position to rigorously test the theory and, encouragingly, a number of systems have emerged that appear to conform to some of Turing's fundamental ideas. In this paper, we describe the history and more recent integration between experiment and theory in one of the key models for understanding pattern formation: the emergence of feathers and hair in the skins of birds and mammals.
Collapse
Affiliation(s)
- K J Painter
- Department of Mathematics/Maxwell Institute for Mathematical Sciences , Heriot-Watt University , Edinburgh EH14 4AS , UK
| | | | | | | | | |
Collapse
|
21
|
Lehtonen PK, Laaksonen T, Artemyev AV, Belskii E, Berg PR, Both C, Buggiotti L, Bureš S, Burgess MD, Bushuev AV, Krams I, Moreno J, Mägi M, Nord A, Potti J, Ravussin PA, Sirkiä PM, Sætre GP, Winkel W, Primmer CR. Candidate genes for colour and vision exhibit signals of selection across the pied flycatcher (Ficedula hypoleuca) breeding range. Heredity (Edinb) 2011; 108:431-40. [PMID: 22027894 DOI: 10.1038/hdy.2011.93] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The role of natural selection in shaping adaptive trait differentiation in natural populations has long been recognized. Determining its molecular basis, however, remains a challenge. Here, we search for signals of selection in candidate genes for colour and its perception in a passerine bird. Pied flycatcher plumage varies geographically in both its structural and pigment-based properties. Both characteristics appear to be shaped by selection. A single-locus outlier test revealed 2 of 14 loci to show significantly elevated signals of divergence. The first of these, the follistatin gene, is expressed in the developing feather bud and is found in pathways with genes that determine the structure of feathers and may thus be important in generating variation in structural colouration. The second is a gene potentially underlying the ability to detect this variation: SWS1 opsin. These two loci were most differentiated in two Spanish pied flycatcher populations, which are also among the populations that have the highest UV reflectance. The follistatin and SWS1 opsin genes thus provide strong candidates for future investigations on the molecular basis of adaptively significant traits and their co-evolution.
Collapse
Affiliation(s)
- P K Lehtonen
- Department of Biology, University of Turku, Turku, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Mou C, Pitel F, Gourichon D, Vignoles F, Tzika A, Tato P, Yu L, Burt DW, Bed'hom B, Tixier-Boichard M, Painter KJ, Headon DJ. Cryptic patterning of avian skin confers a developmental facility for loss of neck feathering. PLoS Biol 2011; 9:e1001028. [PMID: 21423653 PMCID: PMC3057954 DOI: 10.1371/journal.pbio.1001028] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 02/01/2011] [Indexed: 12/04/2022] Open
Abstract
Vertebrate skin is characterized by its patterned array of appendages, whether feathers, hairs, or scales. In avian skin the distribution of feathers occurs on two distinct spatial levels. Grouping of feathers within discrete tracts, with bare skin lying between the tracts, is termed the macropattern, while the smaller scale periodic spacing between individual feathers is referred to as the micropattern. The degree of integration between the patterning mechanisms that operate on these two scales during development and the mechanisms underlying the remarkable evolvability of skin macropatterns are unknown. A striking example of macropattern variation is the convergent loss of neck feathering in multiple species, a trait associated with heat tolerance in both wild and domestic birds. In chicken, a mutation called Naked neck is characterized by a reduction of body feathering and completely bare neck. Here we perform genetic fine mapping of the causative region and identify a large insertion associated with the Naked neck trait. A strong candidate gene in the critical interval, BMP12/GDF7, displays markedly elevated expression in Naked neck embryonic skin due to a cis-regulatory effect of the causative mutation. BMP family members inhibit embryonic feather formation by acting in a reaction-diffusion mechanism, and we find that selective production of retinoic acid by neck skin potentiates BMP signaling, making neck skin more sensitive than body skin to suppression of feather development. This selective production of retinoic acid by neck skin constitutes a cryptic pattern as its effects on feathering are not revealed until gross BMP levels are altered. This developmental modularity of neck and body skin allows simple quantitative changes in BMP levels to produce a sparsely feathered or bare neck while maintaining robust feather patterning on the body.
Collapse
Affiliation(s)
- Chunyan Mou
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Frederique Pitel
- UMR INRA/ENVT Laboratoire de Génétique Cellulaire, INRA, Castanet-Tolosan, France
| | | | - Florence Vignoles
- UMR INRA/ENVT Laboratoire de Génétique Cellulaire, INRA, Castanet-Tolosan, France
| | - Athanasia Tzika
- Laboratory of Natural and Artificial Evolution, Department of Zoology and Animal Biology, Sciences III, Geneva, Switzerland
| | - Patricia Tato
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Le Yu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Dave W. Burt
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Kevin J. Painter
- Department of Mathematics and Maxwell Institute for Mathematical Sciences, School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Denis J. Headon
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
23
|
Lin CM, Jiang TX, Baker RE, Maini PK, Widelitz RB, Chuong CM. Spots and stripes: pleomorphic patterning of stem cells via p-ERK-dependent cell chemotaxis shown by feather morphogenesis and mathematical simulation. Dev Biol 2009; 334:369-82. [PMID: 19647731 PMCID: PMC2811698 DOI: 10.1016/j.ydbio.2009.07.036] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 07/22/2009] [Accepted: 07/27/2009] [Indexed: 02/01/2023]
Abstract
A key issue in stem cell biology is the differentiation of homogeneous stem cells towards different fates which are also organized into desired configurations. Little is known about the mechanisms underlying the process of periodic patterning. Feather explants offer a fundamental and testable model in which multi-potential cells are organized into hexagonally arranged primordia and the spacing between primordia. Previous work explored roles of a Turing reaction-diffusion mechanism in establishing chemical patterns. Here we show that a continuum of feather patterns, ranging from stripes to spots, can be obtained when the level of p-ERK activity is adjusted with chemical inhibitors. The patterns are dose-dependent, tissue stage-dependent, and irreversible. Analyses show that ERK activity-dependent mesenchymal cell chemotaxis is essential for converting micro-signaling centers into stable feather primordia. A mathematical model based on short-range activation, long-range inhibition, and cell chemotaxis is developed and shown to simulate observed experimental results. This generic cell behavior model can be applied to model stem cell patterning behavior at large.
Collapse
Affiliation(s)
- Chih-Min Lin
- Department of Pathology, University of Southern California, Los Angeles, California 90033, USA
| | - Ting Xin Jiang
- Department of Pathology, University of Southern California, Los Angeles, California 90033, USA
| | - Ruth E. Baker
- Centre for Mathematical Biology, Mathematical Institute, University of Oxford, 24-29 St Giles', Oxford OX1 3LB, UK
| | - Philip K. Maini
- Centre for Mathematical Biology, Mathematical Institute, University of Oxford, 24-29 St Giles', Oxford OX1 3LB, UK
- Oxford Centre for Integrative Systems Biology, Department for Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Randall B. Widelitz
- Department of Pathology, University of Southern California, Los Angeles, California 90033, USA
| | - Cheng-Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California 90033, USA
| |
Collapse
|
24
|
Michon F, Forest L, Collomb E, Demongeot J, Dhouailly D. BMP2 and BMP7 play antagonistic roles in feather induction. Development 2008; 135:2797-805. [PMID: 18635609 DOI: 10.1242/dev.018341] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Feathers, like hairs, first appear as primordia consisting of an epidermal placode associated with a dermal condensation that is necessary for the continuation of their differentiation. Previously, the BMPs have been proposed to inhibit skin appendage formation. We show that the function of specific BMPs during feather development is more complex. BMP2 and BMP7, which are expressed in both the epidermis and the dermis, are involved in an antagonistic fashion in regulating the formation of dermal condensations, and thus are both necessary for subsequent feather morphogenesis. BMP7 is expressed earlier and functions as a chemoattractant that recruits cells into the condensation, whereas BMP2 is expressed later, and leads to an arrest of cell migration, likely via its modulation of the EIIIA fibronectin domain and alpha4 integrin expression. Based on the observed cell proliferation, chemotaxis and the timing of BMP2 and BMP7 expression, we propose a mathematical model, a reaction-diffusion system, which not only simulates feather patterning, but which also can account for the negative effects of excess BMP2 or BMP7 on feather formation.
Collapse
Affiliation(s)
- Frederic Michon
- Equipe Ontogenèse et Cellules Souches du Tégument, Centre de Recherche INSERM UJF - U823, Institut Albert Bonniot, Site Santé, La Tronche, BP170, 38042 Grenoble Cedex 9, France
| | | | | | | | | |
Collapse
|
25
|
Eichberger T, Kaser A, Pixner C, Schmid C, Klingler S, Winklmayr M, Hauser-Kronberger C, Aberger F, Frischauf AM. GLI2-specific transcriptional activation of the bone morphogenetic protein/activin antagonist follistatin in human epidermal cells. J Biol Chem 2008; 283:12426-37. [PMID: 18319260 DOI: 10.1074/jbc.m707117200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hedgehog (HH) signaling in the epidermis is primarily mediated by the zinc finger transcription factors GLI1 and GLI2. Exquisite regulation of HH/GLI signaling is crucial for proper specification of the epidermal lineage and development of its derivatives, whereas dysregulation of HH/GLI signaling disrupts tissue homeostasis and causes basal cell carcinoma (BCC). Similarly, bone morphogenetic proteins (BMPs) and activins have been described as key signaling factors in the complex regulation of epidermal fate decisions, although their precise interplay with HH/GLI is largely elusive. Here we show that, in human epidermal cells, expression of the activin/BMP antagonist follistatin (FST) is predominantly up-regulated by the HH effector GLI2. Consistently, we found strong FST expression in the outer root sheath of human hair follicles and BCC. Detailed promoter analysis showed that two sequences with homology to the GLI consensus binding site are required for GLI2-mediated activation. Interestingly, activation of the FST promoter is highly GLI2-specific, because neither GLI1 nor GLI3 can significantly increase FST transcription. GLI2 specificity requires the presence of a 518-bp fragment in the proximal FST promoter region. On the protein level, sequences C-terminal to the zinc finger are responsible for GLI2-specific activation of FST transcription, pointing to the existence of GLI-interacting cofactors that modulate GLI target specificity. Our results reveal a key role of GLI2 in activation of the activin/BMP antagonist FST in response to HH signaling and provide new evidence for a regulatory interaction between HH and activin/BMP signaling in hair follicle development and BCC.
Collapse
Affiliation(s)
- Thomas Eichberger
- Department of Molecular Biology, University of Salzburg, Hellbrunnerstrasse 34, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wu W, Xu R, Xiao L, Xu H, Gao G. Expression of the β-Catenin Gene in the Skin of Embryonic Geese During Feather Bud Development. Poult Sci 2008; 87:204-11. [DOI: 10.3382/ps.2007-00197] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
27
|
Chen W, Fu X, Ge S, Sun T, Zhou G, Han B, Li H, Sheng Z. Profiling of genes differentially expressed in a rat of early and later gestational ages with high-density oligonucleotide DNA array. Wound Repair Regen 2007; 15:147-55. [PMID: 17244330 DOI: 10.1111/j.1524-475x.2006.00195.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The early gestational fetus heals dermal wounds rapidly and scarlessly. This phenomenon appears to be intrinsic to fetal skin and is probably modulated by interplay of many genes. We ventured to study differences in gene expression between earlier gestational skin (EGS) and later gestational skin (LGS) with the aid of high-density oligonucleotide DNA array to explore the molecular mechanism underlying scarless healing. Total RNA was isolated from fetal Wistar rat skin of the scarless (E15) and scar-forming (E18) periods of gestation (term=21.5 days), and purified to mRNAs. Both the mRNAs from EGS and LGS were reversely transcribed to cDNAs, and were labeled with the incorporation of fluorescent dCTP for preparing the hybridization probes through single primer amplification reaction and Klenow labeling methods. The mixed probes were then hybridized to the oligonucleotide DNA arrays that contained 5,705 DNA fragments representing 5,705 rat genes. After highly stringent washing, the microarray was scanned for fluorescent signals to display the differentially expressed genes between two groups of tissues. Among 5,705 rat genes, there were 53 genes (0.93%) with differentially expressed levels between EGS and LGS; 27 genes, including fibroblast growth factor 8 and follistatin, were up-regulated (0.47%); and 26 genes, containing lymphoid enhancer binding factor-1 and beta-catenin, were down-regulated (0.46%) in fetal skin of scarless period vs. scar-forming period. Analyses of genes related to ion channels, growth factors, extracellular matrix and cellular skeleton, and movement confirmed that our molecular data obtained by oligonucleotide DNA array were consistent with the published biochemical and clinical findings of fetal scarless healing. Stronger expression of fibroblast growth factor 8, follistatin, and weaker expression of lymphoid enhancer binding factor-1 and beta-catenin in EGS vs. LGS were also testified with reverse transcription-polymerase chain reaction and Western blotting methods. Oligonucleotide DNA array was a powerful tool for investigating different gene expression between scarless and scar-forming periods of gestation in the rat fetal skin. Many genes were involved in the phenotypic transition from scarless to scar-forming wound repair during gestation. Further analysis of the obtained genes will help to understand the molecular mechanism of fetal scarless healing.
Collapse
Affiliation(s)
- Wei Chen
- Wound Healing and Cell Biology Laboratory, Burns Institute, The First Affiliated Hospital (304 Hospital) of the General Hospital of PLA, Trauma Center of Postgraduate Medical College, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Pummila M, Fliniaux I, Jaatinen R, James MJ, Laurikkala J, Schneider P, Thesleff I, Mikkola ML. Ectodysplasin has a dual role in ectodermal organogenesis: inhibition of Bmp activity and induction of Shh expression. Development 2007; 134:117-25. [PMID: 17164417 DOI: 10.1242/dev.02708] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Ectodermal organogenesis is regulated by inductive and reciprocal signalling cascades that involve multiple signal molecules in several conserved families. Ectodysplasin-A (Eda), a tumour necrosis factor-like signalling molecule, and its receptor Edar are required for the development of a number of ectodermal organs in vertebrates. In mice, lack of Edaleads to failure in primary hair placode formation and missing or abnormally shaped teeth, whereas mice overexpressing Eda are characterized by enlarged hair placodes and supernumerary teeth and mammary glands. Here, we report two signalling outcomes of the Eda pathway: suppression of bone morphogenetic protein (Bmp) activity and upregulation of sonic hedgehog (Shh)signalling. Recombinant Eda counteracted Bmp4 activity in developing teeth and, importantly, inhibition of BMP activity by exogenous noggin partially restored primary hair placode formation in Eda-deficient skin in vitro, indicating that suppression of Bmp activity was compromised in the absence of Eda. The downstream effects of the Eda pathway are likely to be mediated by transcription factor nuclear factor-κB (NF-κB), but the transcriptional targets of Edar have remained unknown. Using a quantitative approach, we show in cultured embryonic skin that Eda induced the expression of two Bmp inhibitors, Ccn2/Ctgf (CCN family protein 2/connective tissue growth factor) and follistatin. Moreover, our data indicate that Shh is a likely transcriptional target of Edar, but, unlike noggin, recombinant Shh was unable to rescue primary hair placode formation in Eda-deficient skin explants.
Collapse
Affiliation(s)
- Marja Pummila
- Institute of Biotechnology, Developmental Biology Program, University of Helsinki, 00014 Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Lin CM, Jiang TX, Widelitz RB, Chuong CM. Molecular signaling in feather morphogenesis. Curr Opin Cell Biol 2006; 18:730-41. [PMID: 17049829 PMCID: PMC4406286 DOI: 10.1016/j.ceb.2006.10.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Accepted: 10/05/2006] [Indexed: 12/17/2022]
Abstract
The development and regeneration of feathers have gained much attention recently because of progress in the following areas. First, pattern formation. The exquisite spatial arrangement provides a simple model for decoding the rules of morphogenesis. Second, stem cell biology. In every molting, a few stem cells have to rebuild the entire epithelial organ, providing much to learn on how to regenerate an organ physiologically. Third, evolution and development ('Evo-Devo'). The discovery of feathered dinosaur fossils in China prompted enthusiastic inquiries about the origin and evolution of feathers. Progress has been made in elucidating feather morphogenesis in five successive phases: macro-patterning, micro-patterning, intra-bud morphogenesis, follicle morphogenesis and regenerative cycling.
Collapse
Affiliation(s)
- Chih-Min Lin
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, Los Angeles, CA 90033, USA
| | | | | | | |
Collapse
|
30
|
Geetha-Loganathan P, Nimmagadda S, Huang R, Scaal M, Christ B. Expression pattern of BMPs during chick limb development. ACTA ACUST UNITED AC 2006; 211 Suppl 1:87-93. [PMID: 17024298 DOI: 10.1007/s00429-006-0129-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2006] [Indexed: 10/24/2022]
Abstract
In vertebrates, BMPs (bone morphogenic proteins) play critical roles in establishing the basic embryonic body plan and are involved in the development of a large variety of organs and tissues. Here, we analyzed the expression pattern of various BMPs (2, 4, 5 and 7) by whole mount in situ hybridization during chick limb development. In limb, expression of BMPs suggests evolutionary conserved mechanisms of BMP-dependent differentiation between lower and higher vertebrates. During the early developmental stages, BMP-2 and BMP-7 are expressed in the posterior distal mesenchyme leaving a less prominent expression anteriorly. BMP-4 is initially expressed in the anterior mesenchyme and spreads later to the whole mesenchyme leaving a stronger expression at the anterior side. From HH-stage 25, expression of BMP-4 is observed in the anterior-posterior margins of the limb bud. The BMPs 2, 4 and 7 are expressed strongly in the AER, whereas BMP-5 is expressed as a weak signal in the distal mesoderm during the early stages of limb development. Later from HH-stage 25 onwards, BMP-5 is expressed in the dorsal and ventral muscular mass of the developing limb. As digits become identifiable, expression of BMPs are observed in the interdigital mesenchyme and can also be detected along the contours of the developing phalanges and at the distal tips of the digits. All these BMPs are found to be expressed in the developing feather buds from day 8 onwards.
Collapse
Affiliation(s)
- P Geetha-Loganathan
- Institute of Anatomy and Cell Biology, University of Freiburg, Albertstrasse 17, 79104, Freiburg, Germany
| | | | | | | | | |
Collapse
|
31
|
Zhou Y, Liu HX, Mistretta CM. Bone morphogenetic proteins and noggin: inhibiting and inducing fungiform taste papilla development. Dev Biol 2006; 297:198-213. [PMID: 16828469 DOI: 10.1016/j.ydbio.2006.05.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2006] [Revised: 05/13/2006] [Accepted: 05/17/2006] [Indexed: 11/18/2022]
Abstract
Fungiform papillae are epithelial specializations that develop in a linear pattern on the anterior mammalian tongue and differentiate to eventually contain taste buds. Little is known about morphogenetic and pattern regulation of these crucial taste organs. We used embryonic rat tongue, organ cultures to test roles for bone morphogenetic proteins, BMP2, 4 and 7, and antagonists noggin and follistatin, in development of papillae from a stage before morphological initiation (E13) or from a stage after the pre-papilla placodes have formed (E14). BMPs and noggin proteins become progressively restricted to papilla locations during tongue development. In E13 cultures, exogenous BMPs or noggin induce increased numbers of fungiform papillae, in a concentration-dependent manner, compared to standard tongue cultures; BMPs, but not noggin, lead to a decreased tongue size at this stage. In E14 cultures, however, exogenous BMP2, 4 or 7 each inhibits papilla formation so that there is a decrease in papilla number. Noggin substantially increases number of papillae in E14 cultures. Using beads for a highly localized protein delivery, papillae are inhibited in the surround of BMP-soaked beads and induced in large clusters around noggin-soaked beads. Follistatin, presented in culture medium or by bead, does not alter papilla formation or number. In all fungiform papillae that form under various culture conditions, the molecular marker, sonic hedgehog, is within each papilla. However, the BMP inhibitory effect on papillae is not prevented by disrupting sonic hedgehog signaling through addition of cyclopamine to cultures. BMPs and noggin alter cell proliferation in tongue epithelium in opposite ways, demonstrated with Ki67 immunostaining. We propose that the BMPs and noggin, colocalized within papilla placodes and the fungiform papillae per se, have opposing inhibitory and activating or inducing roles in papilla development in linear patterns. We present a model for these effects.
Collapse
Affiliation(s)
- Yanqiu Zhou
- Department of Biologic and Materials Sciences, Room 6217, School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
32
|
Fraser GJ, Graham A, Smith MM. Developmental and evolutionary origins of the vertebrate dentition: molecular controls for spatio-temporal organisation of tooth sites in osteichthyans. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2006; 306:183-203. [PMID: 16496402 DOI: 10.1002/jez.b.21097] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The rainbow trout (Oncorhynchus mykiss) as a developmental model surpasses both zebrafish and mouse for a more widespread distribution of teeth in the oro-pharynx as the basis for general vertebrate odontogenesis, one in which replacement is an essential requirement. Studies on the rainbow trout have led to the identification of the initial sequential appearance of teeth, through differential gene expression as a changing spatio-temporal pattern, to set in place the primary teeth of the first generation, and also to regulate the continuous production of replacement tooth families. Here we reveal gene expression data that address both the field and clone theories for patterning a polyphyodont osteichthyan dentition. These data inform how the initial pattern may be established through up-regulation at tooth loci from a broad odontogenic band. It appears that control and regulation of replacement pattern resides in the already primed dental epithelium at the sides of the predecessor tooth. A case is presented for the developmental changes that might have occurred during vertebrate evolution, for the origin of a separate successional dental lamina, by comparison with an osteichthyan tetrapod dentition (Ambystoma mexicanum). The evolutionary origins of such a permanent dental lamina are proposed to have occurred from the transient one demonstrated here in the trout. This has implications for phylogenies based on the homology of teeth as only those developed from a dental lamina. Utilising the data generated from the rainbow trout model, we propose this as a standard for comparative development and evolutionary theories of the vertebrate dentition.
Collapse
Affiliation(s)
- Gareth J Fraser
- MRC Centre for Developmental Neurobiology, King's College London, London, UK.
| | | | | |
Collapse
|
33
|
Abstract
Beaks and feathers epitomize inimitable avian traits. Within individuals and across species there exists astounding diversity in the size, shape, arrangement, and colour of beaks and feathers in association with various functional adaptations. What has enabled the concomitantly divergent evolution of beaks and feathers? The common denominator may lie in their developmental programmes. As revealed through recent transplant experiments using quail and duck embryos, the developmental programme for each structure utilizes mesenchyme as a dominant source of species-specific patterning information, acts as a module of closely coupled molecular and histogenic events, and operates with a high degree of spatial and temporal plasticity. By synergizing these three features, the developmental programmes underlying beaks and feathers likely have the essential potential to react spontaneously to novel conditions and new gene functions, and as a consequence are well equipped to generate and accommodate innovative phenotypes during the course of evolution.
Collapse
Affiliation(s)
- Richard A Schneider
- Department of Orthopaedic Surgery, University of California at San Francisco, CA 94143-0514, USA.
| |
Collapse
|
34
|
Eames BF, Schneider RA. Quail-duck chimeras reveal spatiotemporal plasticity in molecular and histogenic programs of cranial feather development. Development 2005; 132:1499-509. [PMID: 15728671 PMCID: PMC2835538 DOI: 10.1242/dev.01719] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The avian feather complex represents a vivid example of how a developmental module composed of highly integrated molecular and histogenic programs can become rapidly elaborated during the course of evolution. Mechanisms that facilitate this evolutionary diversification may involve the maintenance of plasticity in developmental processes that underlie feather morphogenesis. Feathers arise as discrete buds of mesenchyme and epithelium, which are two embryonic tissues that respectively form dermis and epidermis of the integument. Epithelial-mesenchymal signaling interactions generate feather buds that are neatly arrayed in space and time. The dermis provides spatiotemporal patterning information to the epidermis but precise cellular and molecular mechanisms for generating species-specific differences in feather pattern remain obscure. In the present study, we exploit the quail-duck chimeric system to test the extent to which the dermis regulates the expression of genes required for feather development. Quail and duck have distinct feather patterns and divergent growth rates, and we exchange pre-migratory neural crest cells destined to form the craniofacial dermis between them. We find that donor dermis induces host epidermis to form feather buds according to the spatial pattern and timetable of the donor species by altering the expression of members and targets of the Bone Morphogenetic Protein, Sonic Hedgehog and Delta/Notch pathways. Overall, we demonstrate that there is a great deal of spatiotemporal plasticity inherent in the molecular and histogenic programs of feather development, a property that may have played a generative and regulatory role throughout the evolution of birds.
Collapse
Affiliation(s)
- B. Frank Eames
- Department of Orthopaedic Surgery, University of California at San Francisco, 533 Parnassus Avenue, U-453, San Francisco, CA 94143-0514, USA
| | - Richard A. Schneider
- Department of Orthopaedic Surgery, University of California at San Francisco, 533 Parnassus Avenue, U-453, San Francisco, CA 94143-0514, USA
| |
Collapse
|
35
|
Hornik C, Krishan K, Yusuf F, Scaal M, Brand-Saberi B. cDermo-1 misexpression induces dense dermis, feathers, and scales. Dev Biol 2005; 277:42-50. [PMID: 15572138 DOI: 10.1016/j.ydbio.2004.08.050] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2003] [Revised: 08/26/2004] [Accepted: 08/26/2004] [Indexed: 11/24/2022]
Abstract
Reciprocal epithelio-mesenchymal interactions between the prospective epidermis and the underlying dermis are the major driving forces in the development of skin appendages. Feather development is initiated by a still unknown signal from the dermis in feather-forming skin. The morphological response of the ectoderm to this signal is the formation of an epidermal placode, which signals back to the mesenchyme to induce dermal condensations. Together, epidermal and dermal components constitute the outgrowing feather bud. The bHLH transcription factor cDermo-1 is expressed in developing dermis and is the earliest known marker of prospective feather tracts. To test its function during feather development, we forced cDermo-1 expression in embryonic chicken dermis using a retroviral expression vector. In featherless (apteric) regions, cDermo-1 misexpression induced dense, thickened dermis normally observed in feathered skin (pterylae), and leads to the development of regularly spaced and normally shaped ectopic feather buds. In pterylae, cDermo-1 misexpression enhanced feather growth. In hindlimb skin, according to the local skin identity, misexpression of cDermo-1 induced ectopic scale formation. Thus, we show that forced cDermo-1 expression in developing dermis is sufficient to launch the developmental program leading to skin appendage formation. We propose a role of cDermo-1 at the initial stages of feather induction upstream of FGF10.
Collapse
Affiliation(s)
- Christoph Hornik
- Institute of Anatomy and Cell Biology II, Albert-Ludwigs-Universität, D-79104 Freiburg, Germany
| | | | | | | | | |
Collapse
|
36
|
Wang XP, Suomalainen M, Jorgez CJ, Matzuk MM, Werner S, Thesleff I. Follistatin regulates enamel patterning in mouse incisors by asymmetrically inhibiting BMP signaling and ameloblast differentiation. Dev Cell 2004; 7:719-30. [PMID: 15525533 DOI: 10.1016/j.devcel.2004.09.012] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2004] [Revised: 09/01/2004] [Accepted: 09/03/2004] [Indexed: 01/01/2023]
Abstract
Rodent incisors are covered by enamel only on their labial side. This asymmetric distribution of enamel is instrumental to making the cutting edge sharp. Enamel matrix is secreted by ameloblasts derived from dental epithelium. Here we show that overexpression of follistatin in the dental epithelium inhibits ameloblast differentiation in transgenic mouse incisors, whereas in follistatin knockout mice, ameloblasts differentiate ectopically on the lingual enamel-free surface. Consistent with this, in wild-type mice, follistatin was continuously expressed in the lingual dental epithelium but downregulated in the labial epithelium. Experiments on cultured tooth explants indicated that follistatin inhibits the ameloblast-inducing activity of BMP4 from the underlying mesenchymal odontoblasts and that follistatin expression is induced by activin from the surrounding dental follicle. Hence, ameloblast differentiation is regulated by antagonistic actions of BMP4 and activin A from two mesenchymal cell layers flanking the dental epithelium, and asymmetrically expressed follistatin regulates the labial-lingual patterning of enamel formation.
Collapse
Affiliation(s)
- Xiu-Ping Wang
- Developmental Biology Programme, Institute of Biotechnology, Viikki Biocenter, PO Box 56, University of Helsinki, FIN-00014 Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
37
|
McKinnell IW, Turmaine M, Patel K. Sonic Hedgehog functions by localizing the region of proliferation in early developing feather buds. Dev Biol 2004; 272:76-88. [PMID: 15242792 DOI: 10.1016/j.ydbio.2004.04.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2003] [Revised: 04/07/2004] [Accepted: 04/14/2004] [Indexed: 11/28/2022]
Abstract
Feathers are formed following a series of reciprocal signals between the epithelium and the mesenchyme. Initially, the formation of a dense dermis leads to the induction of a placode in the overlying ectoderm. The ectoderm subsequently signals back to the dermis to promote cell division. Sonic Hedgehog (Shh) is a secreted protein expressed in the ectoderm that has previously been implicated in mitogenic and morphogenetic processes throughout feather bud development. We therefore interfered with Shh signaling during early feather bud development and observed a dramatic change in feather form and prominence. Surprisingly, outgrowth did occur and was manifest as irregular, fused, and ectopic feather domains at both molecular and morphological levels. Experiments with Di-I and BrdU indicated that this effect was at least in part caused by the dispersal of previously aggregated proliferating dermal cells. We propose that Shh maintains bud development by localizing the dermal feather progenitors.
Collapse
Affiliation(s)
- Iain W McKinnell
- Department of Veterinary Basic Science, Royal Veterinary College, London NW1 0TU, UK
| | | | | |
Collapse
|
38
|
Fliniaux I, Viallet JP, Dhouailly D. Signaling dynamics of feather tract formation from the chick somatopleure. Development 2004; 131:3955-66. [PMID: 15269169 DOI: 10.1242/dev.01263] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the chick, most feathers are restricted to specific areas of the skin, the feather tracts or pterylae, while other areas, such as the apteria, remain bare. In the embryo, the expansion and closure of the somatopleure leads to the juxtaposition of the ventral pteryla, midventral apterium and amnion. The embryonic proximal somatopleural mesoderm is determined to form a feather-forming dermis at 2 days of incubation (E2), while the embryonic distal and the extra-embryonic somatopleure remain open to determination. We found a progressive, lateral expression of Noggin in the embryonic area, and downregulation of Msx1, a BMP4 target gene, with Msx1 expression being ultimately restricted to the most distal embryonic and extra-embryonic somatopleural mesoderm. Msx1 downregulation thus correlates with the formation of the pterylae, and its maintenance to that of the apterium. Suspecting that the inhibition of BMP4 signaling might be linked to the determination of a feather-forming dermis, we grafted Noggin-expressing cells in the distal somatopleure at E2. This elicited the formation of a supplementary pteryla in the midventral apterium. Endogenous Noggin, which is secreted by the intermediate mesoderm at E2, then by the proximal somatopleure at E4, could be sufficient to suppress BMP4 signaling in the proximal somatopleural mesoderm and then in part of the distal somatopleure, thus in turn allowing the formation of the dense dermis of the future pterylae. The same result was obtained with the graft of Shh-producing cells, but Noggin and Shh are both required in order to change the future amnion into a feather-bearing skin. A possible synergistic role of endogenous Shh from the embryonic endoderm remains to be confirmed.
Collapse
Affiliation(s)
- Ingrid Fliniaux
- Equipe Biologie de la Différenciation Epithéliale, UMR CNRS 5538, LEDAC, Institut Albert Bonniot, Université Joseph Fourier, BP 53-38041 Grenoble Cedex 9, France
| | | | | |
Collapse
|
39
|
Mandler M, Neubüser A. FGF signaling is required for initiation of feather placode development. Development 2004; 131:3333-43. [PMID: 15201222 DOI: 10.1242/dev.01203] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Morphogenesis of hairs and feathers is initiated by an as yet unknown dermal signal that induces placode formation in the overlying ectoderm. To determine whether FGF signals are required for this process we over-expressed soluble versions of FGFR1 or FGFR2 in the skin of chicken embryos. This produced a complete failure of feather formation prior to any morphological or molecular signs of placode development. We further show that Fgf10 is expressed in the dermis of nascent feather primordia, and that anti-FGF10 antibodies block feather placode development in skin explants. In addition we show that FGF10 can induce expression of positive and negative regulators of feather development and can induce its own expression under conditions of low BMP signaling. Together these results demonstrate that FGF signaling is required for the initiation of feather placode development and implicate FGF10 as an early dermal signal involved in this process.
Collapse
Affiliation(s)
- Markus Mandler
- Research Institute of Molecular Pathology, Dr Bohr-Gasse 7, A-1030 Vienna, Austria
| | | |
Collapse
|
40
|
McKinnell IW, Makarenkova H, de Curtis I, Turmaine M, Patel K. EphA4, RhoB and the molecular development of feather buds are maintained by the integrity of the actin cytoskeleton. Dev Biol 2004; 270:94-105. [PMID: 15136143 DOI: 10.1016/j.ydbio.2004.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2003] [Revised: 01/29/2004] [Accepted: 02/06/2004] [Indexed: 01/17/2023]
Abstract
The development of feather buds is a highly ordered process involving epithelial-mesenchymal signalling. Cellular morphology is determined by the actin cytoskeleton, which is controlled by networks of regulators such as the GTPases. EphA4 belongs to a receptor tyrosine kinase family that has been consistently shown to regulate the cytoskeleton via Rho family GTPases in neural development and is expressed in early stages of feather bud development though its role has not been defined. We therefore used an in vitro skin culture system to interfere with EphA4 levels in feather buds using anti-sense oligonucleotides, demonstrating a severe effect on both their number and morphological form. Analysis of the Rho family of GTPases revealed that this effect was mediated by the GTPase RhoB, the expression of which was altered in response to altered levels of EphA4. In addition, the inhibition of RhoB mimicked the effects of reduced EphA4 levels on feather development. Significantly, manipulation of cytoskeletal dynamics revealed that those cells undergoing morphogenetic change regulate the patterning signals responsible for initiating feather development. We propose that this molecular maintenance mechanism between EphA4-RhoB and the actin cytoskeleton converges or coordinates with other morphogenic signalling systems to control feather bud development.
Collapse
Affiliation(s)
- Iain W McKinnell
- Department of Veterinary Basic Science, Royal Veterinary College, London NW1 0TU, UK
| | | | | | | | | |
Collapse
|
41
|
Boughner JC, Dean MC. Does space in the jaw influence the timing of molar crown initiation? A model using baboons (Papio anubis) and great apes (Pan troglodytes, Pan paniscus). J Hum Evol 2004; 46:255-77. [PMID: 14984783 DOI: 10.1016/j.jhevol.2003.11.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2003] [Revised: 11/20/2003] [Accepted: 11/26/2003] [Indexed: 11/18/2022]
Abstract
Radiographic and histological studies of baboon (Papio hamadryas, P. anubis) and chimpanzee (Pan troglodytes) permanent tooth development have found that periods of molar crown mineralization overlap markedly in chimpanzees but are staggered in baboons. Here we test the hypothesis that these intertaxon differences in molar initiation are primarily due to the space available in the mandibles of each species for these teeth. This study includes radiographic, linear measurement, and three-dimensional (3D) coordinate landmark data taken from baboon (Papio anubis n=51) and great ape (Pan paniscus n=43, P. troglodytes n=60) mandibles and permanent molars across a broad developmental range for each taxon. Unexpectedly, 3D multivariate statistical shape analysis of the molar crypt, crown, and root data shows that all three species trajectories of molar row shape change are indistinguishable from each other. Qualitative analysis of these 3D data reveals subtle and inconclusive intergeneric differences in the space maintained between adjacent molars during growth. The space distal to each newly initiated molar is slightly greater in the baboon. Bivariate analyses comparing molar row and mandibular corpus proportions in Papio and Pan fail to show clear or consistent taxonomic differences in the ratio of space afforded developing molars in the alveolar bone. Thus, there is a poor correlation between mandibular proportion and both intermolar spacing and 3D molar development pattern. Contrary to earlier studies, these results suggest that pattern of molar crown initiation and temporal overlap of adjacent mineralizing crowns is not significantly different between Papio and Pan. This may be due in part to the inclusion here of not only 3D molar crown data but also 3D molar crypt data. This study strongly refutes the hypothesis that space available in the mandible directly underlies different times of permanent molar crown initiation between Papio and Pan.
Collapse
Affiliation(s)
- Julia C Boughner
- Department of Oral Health Sciences, Faculty of Dentistry, University of British Columbia, 2199 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada.
| | | |
Collapse
|
42
|
Wang XP, Suomalainen M, Jorgez CJ, Matzuk MM, Wankell M, Werner S, Thesleff I. Modulation of activin/bone morphogenetic protein signaling by follistatin is required for the morphogenesis of mouse molar teeth. Dev Dyn 2004; 231:98-108. [PMID: 15305290 DOI: 10.1002/dvdy.20118] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Teeth form as ectodermal appendages, and their morphogenesis is regulated by conserved signaling pathways. The shape of the tooth crown results from growth and folding of inner dental epithelium, and the cusp patterning is regulated by transient signaling centers, the enamel knots. Several signal proteins in the transforming growth factor-beta (TGF beta) superfamily are required for tooth development. Follistatin is an extracellular inhibitor of TGF beta signaling. To investigate the roles of follistatin during tooth development, we analyzed in detail the expression patterns of follistatin, activin beta A, as well as Bmp2, Bmp4, and Bmp7 during tooth morphogenesis. We also examined the tooth phenotypes of follistatin knockout mice and of transgenic mice overexpressing follistatin in the epithelium under the keratin 14 (K14) promoter. The folding of the dental epithelium was aberrant in the molars of follistatin knockout mice, and the cusps were shallow with reduced cell proliferation and lack of anteroposterior polarization. The functions of both primary and secondary enamel knots were apparently disturbed. In K14-follistatin transgenic mice, the molar cusp pattern was also seriously affected (although different from the follistatin knockouts) and the occlusal surfaces of the molars were whorled. Their enamel was prematurely worn. In addition, all of the third molars were missing. Our results indicate that follistatin regulates morphogenesis and shaping of the tooth crown. We propose that finely tuned antagonistic effects between follistatin and TGF beta superfamily signals are critical for enamel knot formation and function, as well as for patterning of tooth cusps.
Collapse
Affiliation(s)
- Xiu-Ping Wang
- Developmental Biology Programme, Institute of Biotechnology, Viikki Biocenter, University of Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
43
|
Harris MP, Linkhart BL, Fallon JF. Bmp7 mediates early signaling events during induction of chick epidermal organs. Dev Dyn 2004; 231:22-32. [PMID: 15305284 DOI: 10.1002/dvdy.20096] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The induction and specification of a large number of vertebrate organs require reciprocal signaling between an epithelium and subjacent mesenchyme. In the formation of integumentary organs, the initial inductive signaling events leading to the formation of the organ primordia stem from the mesenchyme. However, the epithelium must have the capacity to respond to these signals. We demonstrate that bone morphogenetic protein 7 (Bmp7) is an early molecular marker for epidermal organ development during development of feathers and scales of the chick. Bmp7 is expressed broadly in the preplacode epidermis and subsequently becomes localized to the forming placodes of feathers and scales. An examination of Bmp7 expression in the scaleless mutant chicken integument indicates that Bmp7 expression in the epidermis is associated with the ability to form epidermal organs. We show that BMP7 function is necessary for the formation of epidermal placodes in both feather and scale forming epidermis. In addition, precocious expression of Bmp7 in the metatarsal epidermis of the Silkie mutant or treatment of the metatarsus with ectopic BMP7 protein results in feather development from scale forming integument. From these data, we propose that Bmp7 is necessary and sufficient, in a developmental context, to mediate the competence of an epithelium to respond to inductive signals from the underlying mesenchyme to form epidermal organs in the chick. We propose that regulation of Bmp7 in localized areas of the embryonic epidermis facilitates the development of regional formation of integumentary organs.
Collapse
Affiliation(s)
- Matthew P Harris
- Anatomy Department, University of Wisconsin-Madison, 1300 University Avenue, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
44
|
Jiang TX, Widelitz RB, Shen WM, Will P, Wu DY, Lin CM, Jung HS, Chuong CM. Integument pattern formation involves genetic and epigenetic controls: feather arrays simulated by digital hormone models. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2004; 48:117-35. [PMID: 15272377 PMCID: PMC4386648 DOI: 10.1387/ijdb.041788tj] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Pattern formation is a fundamental morphogenetic process. Models based on genetic and epigenetic control have been proposed but remain controversial. Here we use feather morphogenesis for further evaluation. Adhesion molecules and/or signaling molecules were first expressed homogenously in feather tracts (restrictive mode, appear earlier) or directly in bud or inter-bud regions ( de novo mode, appear later). They either activate or inhibit bud formation, but paradoxically colocalize in the bud. Using feather bud reconstitution, we showed that completely dissociated cells can reform periodic patterns without reference to previous positional codes. The patterning process has the characteristics of being self-organizing, dynamic and plastic. The final pattern is an equilibrium state reached by competition, and the number and size of buds can be altered based on cell number and activator/inhibitor ratio, respectively. We developed a Digital Hormone Model which consists of (1) competent cells without identity that move randomly in a space, (2) extracellular signaling hormones which diffuse by a reaction-diffusion mechanism and activate or inhibit cell adhesion, and (3) cells which respond with topological stochastic actions manifested as changes in cell adhesion. Based on probability, the results are cell clusters arranged in dots or stripes. Thus genetic control provides combinational molecular information which defines the properties of the cells but not the final pattern. Epigenetic control governs interactions among cells and their environment based on physical-chemical rules (such as those described in the Digital Hormone Model). Complex integument patterning is the sum of these two components of control and that is why integument patterns are usually similar but non-identical. These principles may be shared by other pattern formation processes such as barb ridge formation, fingerprints, pigmentation patterning, etc. The Digital Hormone Model can also be applied to swarming robot navigation, reaching intelligent automata and representing a self-re-configurable type of control rather than a follow-the-instruction type of control.
Collapse
Affiliation(s)
- Ting-Xin Jiang
- Department of Pathology, University of Southern California, Los Angeles, California, USA
| | - Randall B. Widelitz
- Department of Pathology, University of Southern California, Los Angeles, California, USA
| | - Wei-Min Shen
- Computer Science Information Sciences Institute, University of Southern California, Los Angeles, California, USA
| | - Peter Will
- Computer Science Information Sciences Institute, University of Southern California, Los Angeles, California, USA
| | - Da-Yu Wu
- Department of Cellular and Neurobiology, University of Southern California, Los Angeles, California, USA
| | - Chih-Min Lin
- Department of Pathology, University of Southern California, Los Angeles, California, USA
| | - Han-Sung Jung
- Dept. of Oral Biology, Division in Histology, College of Dentistry, Yonsei University, Seoul, Korea
| | - Cheng-Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
45
|
WIDELITZ RANDALLB, JIANG TINGXIN, YU MINGKE, SHEN TED, SHEN JENYEE, WU PING, YU ZHICAO, CHUONG CHENGMING. Molecular biology of feather morphogenesis: a testable model for evo-devo research. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2003; 298:109-22. [PMID: 12949772 PMCID: PMC4382008 DOI: 10.1002/jez.b.29] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Darwin's theory describes the principles that are responsible for evolutionary change of organisms and their attributes. The actual mechanisms, however, need to be studied for each species and each organ separately. Here we have investigated the mechanisms underlying these principles in the avian feather. Feathers comprise one of the most complex and diverse epidermal organs as demonstrated by their shape, size, patterned arrangement and pigmentation. Variations can occur at several steps along each level of organization, leading to highly diverse forms and functions. Feathers develop gradually during ontogeny through a series of steps that may correspond to the evolutionary steps that were taken during the phylogeny from a reptilian ancestor to birds. These developmental steps include 1) the formation of feather tract fields on the skin surfaces; 2) periodic patterning of the individual feather primordia within the feather tract fields; 3) feather bud morphogenesis establishing anterio-posterior (along the cranio-caudal axis) and proximo-distal axes; 4) branching morphogenesis to create the rachis, barbs and barbules within a feather bud; and 5) gradual modulations of these basic morphological parameters within a single feather or across a feather tract. Thus, possibilities for variation in form and function of feathers occur at every developmental step. In this paper, principles guiding feather tract formation, distributions of individual feathers within the tracts and variations in feather forms are discussed at a cellular and molecular level.
Collapse
Affiliation(s)
- RANDALL B. WIDELITZ
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - TING XIN JIANG
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - MINGKE YU
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - TED SHEN
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - JEN-YEE SHEN
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - PING WU
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - ZHICAO YU
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - CHENG-MING CHUONG
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| |
Collapse
|
46
|
Salingcarnboriboon R, Yoshitake H, Tsuji K, Obinata M, Amagasa T, Nifuji A, Noda M. Establishment of tendon-derived cell lines exhibiting pluripotent mesenchymal stem cell-like property. Exp Cell Res 2003; 287:289-300. [PMID: 12837285 DOI: 10.1016/s0014-4827(03)00107-1] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Development of the musculoskeletal system requires coordinated formation of distinct types of tissues, including bone, cartilage, muscle, and tendon. Compared to muscle, cartilage, and bone, cellular and molecular bases of tendon development have not been well understood due to the lack of tendon cell lines. The purpose of this study was to establish and characterize tendon cell lines. Three clonal tendon cell lines (TT-E4, TT-G11, and TT-D6) were established using transgenic mice harboring a temperature-sensitive mutant of SV40 large T antigen. Proliferation of these cells was significantly enhanced by treatment with bFGF and TGF-beta but not BMP2. Tendon phenotype-related genes such as those encoding scleraxis, Six1, EphA4, COMP, and type I collagen were expressed in these tendon cell clones. In addition to tendon phenotype-related genes, expression of osteopontin and Cbfal was observed. These clonal cell lines formed hard fibrous connective tissue when implanted onto chorioallantoic membrane in ovo. Furthermore, these cells also formed tendon-like tissues when they were implanted into defects made in patella tendon in mice. As these tendon cell lines also produced fibrocartilaginous tissues in tendon defect implantation experiments, mesenchymal stem cell properties were examined. Interestingly, these cells expressed genes related to osteogenic, chondrogenic, and adipogenic lineages at low levels when examined by RT-PCR. TT-G11 and TT-E4 cells differentiated into either osteoblasts or adipocytes, respectively, when they were cultured in cognate differentiation medium. These observations indicated that the established tendon cell line possesses mesenchymal stem cell-like properties, suggesting the existence of mesenchymal stem cell in tendon tissue.
Collapse
Affiliation(s)
- R Salingcarnboriboon
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
The evolutionarily conserved Hox gene family of transcriptional regulators has originally been known for specifying positional identities along the longitudinal body axis of bilateral metazoans, including mouse and man. It is believed that subsequent to this archaic role, subsets of Hox genes have been co-opted for patterning functions in phylogenetically more recent structures, such as limbs and epithelial appendages. Among these, the hair follicle is of particular interest, as it is the only organ undergoing cyclical phases of regression and regeneration during the entire life span of an organism. Furthermore, the hair follicle is increasingly capturing the attention of developmental geneticists, as this abundantly available miniature organ mimics key aspects of embryonic patterning and, in addition, presents a model for studying organ renewal. The first Hox gene shown to play a universal role in hair follicle development is Hoxc13, as both Hoxc13-deficient and overexpressing mice exhibit severe hair growth and patterning defects. Differential gene expression analyses in the skin of these mutants, as well as in vitro DNA binding studies performed with potential targets for HOXC13 transcriptional regulation in human hair, identified genes encoding hair-specific keratins and keratin-associated proteins (KAPs) as major groups of presumptive Hoxc13 downstream effectors in the control of hair growth. The Hoxc13 mutant might thus serve as a paradigm for studying hair-specific roles of Hoxc13 and other members of this gene family, whose distinct spatio-temporally restricted expression patterns during hair development and cycling suggest discrete functions in follicular patterning and hair cycle control. The main conclusion from a discussion of these potential roles vis-à-vis current expression data in mouse and man, and from the perspective of the results obtained with the Hoxc13 transgenic models, is that members of the Hox family are likely to fulfill essential roles of great functional diversity in hair that require complex transcriptional control mechanisms to ensure proper spatio-temporal patterns of Hox gene expression at homeostatic levels.
Collapse
Affiliation(s)
- Alexander Awgulewitsch
- Departments of Medicine and Dermatology, and Hollings Cancer Center, Medical University of South Carolina, 96 Jonathan Lucas St., CSB 912, Charleston, SC 29425, USA.
| |
Collapse
|
48
|
Hall JMH, Bell ML, Finger TE. Disruption of sonic hedgehog signaling alters growth and patterning of lingual taste papillae. Dev Biol 2003; 255:263-77. [PMID: 12648489 DOI: 10.1016/s0012-1606(02)00048-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Taste buds on the anterior part of the tongue develop in conjunction with epithelial-mesenchymal specializations in the form of gustatory (taste) papillae. Sonic hedgehog (Shh) and Bone Morphogenetic Protein 4 (BMP4) are expressed in developing taste papillae, but the roles of these signaling molecules in specification of taste bud progenitors and in papillary morphogenesis are unclear. We show here that BMP4 is not expressed in the early tongue, but is precisely coexpressed with Shh in papillary placodes, which serve as a signaling center for both gustatory and papillary development. To elucidate the role of Shh, we used an in vitro model of mouse fungiform papillary development to determine the effects of two functional inhibitors of Shh signaling: anti-Shh (5E1) antibody and cyclopamine. Cultured E11.5 tongue explants express Shh and BMP4(LacZ) in a pattern similar to that of intact embryos, localizing to developing papillary placodes after 2 days in culture. Tongues cultured with 5E1 antibody continue to express these genes in papillary patterns but develop more papillae that are larger and closer together than in controls. Tongues cultured with cyclopamine have a dose-dependent expansion of Shh and BMP4(LacZ) expression domains. Both antibody-treated and cyclopamine-treated tongue explants also are smaller than controls. Taken together, these results suggest that, although Shh is not involved in the initial specification of papillary placodes, Shh does play two key roles during pmcry development: (1) as a morphogen that directs cells toward a nonpapillary fate, and (2) as a mitogen, causing expansion of the interplacodal epithelium and underlying mesenchyme.
Collapse
Affiliation(s)
- Joshua M H Hall
- Department of Cellular and Structural Biology, University of Colorado Health Sciences Center, Denver, CO 80262, USA
| | | | | |
Collapse
|
49
|
Nakamura M, Matzuk MM, Gerstmayer B, Bosio A, Lauster R, Miyachi Y, Werner S, Paus R. Control of pelage hair follicle development and cycling by complex interactions between follistatin and activin. FASEB J 2003; 17:497-9. [PMID: 12514121 DOI: 10.1096/fj.02-0247fje] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Members of the transforming growth factor beta/bone morphogenetic protein (TGF-beta/BMP) family are involved in the control of hair follicle (HF) morphogenesis and cycling. The activities of several members of this family activins and BMP-2, -4, -7, and -11) are controlled by antagonists such as follistatin. Because follistatin-deficient mice show abnormalities in vibrissae development, we explored the role of follistatin and activin in pelage HF development and cycling. We show here that during HF development follistatin mRNA was prominently expressed by hair matrix and outer root sheath keratinocytes as well as by interfollicular epidermal cells, whereas activin betaA mRNA was mainly expressed in dermal papilla cells. Compared with age-matched wild-type controls, both follistatin knockout mice and activin betaA transgenic mice showed a significant retardation of HF morphogenesis. Treatment of wild-type embryonic skin explants with follistatin protein stimulated HF development. This effect was inhibited by addition of recombinant activin A protein. Activin betaA transgenic mice demonstrated retardation of catagen entry, down-regulation of BMP-2, and up-regulation of expression of its antagonist matrix GLA protein. These observations suggest that follistatin and activin interaction plays an important role in both HF development and cycling, possibly in part by regulating expression of BMP-2 and its antagonist.
Collapse
Affiliation(s)
- Motonobu Nakamura
- Department of Dermatology, University Hospital Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Tao H, Yoshimoto Y, Yoshioka H, Nohno T, Noji S, Ohuchi H. FGF10 is a mesenchymally derived stimulator for epidermal development in the chick embryonic skin. Mech Dev 2002; 116:39-49. [PMID: 12128204 DOI: 10.1016/s0925-4773(02)00131-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The development of avian cutaneous appendages, feathers and scales, is known to arise from the epithelial-mesenchymal interaction. Here we show that FGF10 is associated with this developmental process as an early signal from mesenchymal cells underlying nascent cutaneous placodes. Expression of Fgf10 was detected in the mesenchymal cells underneath the developing placodes. Forced expression of Fgf10 in the femoral skin suppressed expression of Shh and a zinc finger gene snail-related (cSnR), while induced expression of Bmp2 in the interbud region, resulting in thickening of the epidermal layer. Furthermore, forced expression of Fgf10 in the foot skin caused marked ingrowings of the epidermis. The cells in the epidermal ingrowings expressed beta-catenin, proliferating cell nuclear antigen, and an epidermal stem cell marker p63. These results support the idea that FGF10 is a mesenchymally derived stimulator of epidermal development through crosstalk with bone morphogenetic protein (BMP), beta-catenin, and other signaling pathways.
Collapse
Affiliation(s)
- Hirotaka Tao
- Department of Biological Science and Technology, Faculty of Engineering, The University of Tokushima, 2-1 Minami-Jyosanjima-cho, Japan
| | | | | | | | | | | |
Collapse
|