Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
STAR Protoc ; 2(3): 100661, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34278338

ABSTRACT

During morphogenesis, cellular sheets undergo dynamic folding to build functional forms. Here, we develop an image-based quantitative morphology field (QMorF) protocol that quantifies the morphological features of cellular structures and associated distributions. Using feather shafts with different rigidities as examples, QMorF performs coarse-graining statistical measurements of the fitted cellular objects over a micro-image stack, revealing underlying mechanical coupling and developmental clues. These images give intuitive representations of mechanical forces and should be useful for analyzing tissue images showing clear cellular features. For complete details on the use and execution of this protocol, please refer to Chang et al. (2019).


Subject(s)
Feathers/cytology , Image Processing, Computer-Assisted/methods , Animals , Chickens , Feathers/growth & development , Morphogenesis , Paraffin Embedding
2.
Genes (Basel) ; 12(2)2021 02 10.
Article in English | MEDLINE | ID: mdl-33578693

ABSTRACT

Scaffoldin, an S100 fused-type protein (SFTP) with high amino acid sequence similarity to the mammalian hair follicle protein trichohyalin, has been identified in reptiles and birds, but its functions are not yet fully understood. Here, we investigated the expression pattern of scaffoldin and cornulin, a related SFTP, in the developing beaks of birds. We determined the mRNA levels of both SFTPs by reverse transcription polymerase chain reaction (RT-PCR) in the beak and other ectodermal tissues of chicken (Gallus gallus) and quail (Coturnix japonica) embryos. Immunohistochemical staining was performed to localize scaffoldin in tissues. Scaffoldin and cornulin were expressed in the beak and, at lower levels, in other embryonic tissues of both chickens and quails. Immunohistochemistry revealed scaffoldin in the peridermal compartment of the egg tooth, a transitory cornified protuberance (caruncle) on the upper beak which breaks the eggshell during hatching. Furthermore, scaffoldin marked a multilayered peridermal structure on the lower beak. The results of this study suggest that scaffoldin plays an evolutionarily conserved role in the development of the avian beak with a particular function in the morphogenesis of the egg tooth.


Subject(s)
Avian Proteins/genetics , Beak/metabolism , Chickens/genetics , Coturnix/genetics , Feathers/metabolism , Hoof and Claw/metabolism , Animals , Avian Proteins/metabolism , Beak/cytology , Beak/embryology , Biological Evolution , Chick Embryo , Chickens/growth & development , Chickens/metabolism , Conserved Sequence , Coturnix/embryology , Coturnix/metabolism , Embryo, Nonmammalian , Epidermis/embryology , Epidermis/metabolism , Feathers/cytology , Feathers/embryology , Gene Expression Regulation, Developmental , Hoof and Claw/cytology , Hoof and Claw/embryology , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Mammals , Morphogenesis/genetics , Zygote/growth & development , Zygote/metabolism
3.
Dev Cell ; 53(5): 561-576.e9, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32516596

ABSTRACT

Regional specification is critical for skin development, regeneration, and evolution. The contribution of epigenetics in this process remains unknown. Here, using avian epidermis, we find two major strategies regulate ß-keratin gene clusters. (1) Over the body, macro-regional specificities (scales, feathers, claws, etc.) established by typical enhancers control five subclusters located within the epidermal differentiation complex on chromosome 25; (2) within a feather, micro-regional specificities are orchestrated by temporospatial chromatin looping of the feather ß-keratin gene cluster on chromosome 27. Analyses suggest a three-factor model for regional specification: competence factors (e.g., AP1) make chromatin accessible, regional specifiers (e.g., Zic1) target specific genome regions, and chromatin regulators (e.g., CTCF and SATBs) establish looping configurations. Gene perturbations disrupt morphogenesis and histo-differentiation. This chicken skin paradigm advances our understanding of how regulation of big gene clusters can set up a two-dimensional body surface map.


Subject(s)
Avian Proteins/metabolism , CCCTC-Binding Factor/metabolism , Chromatin Assembly and Disassembly , Epithelial Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Morphogenesis , beta-Keratins/genetics , Animals , Avian Proteins/genetics , CCCTC-Binding Factor/genetics , Cell Differentiation , Chick Embryo , Chromosomes/genetics , Epithelial Cells/cytology , Feathers/cytology , Feathers/embryology , Feathers/metabolism , Gene Expression Regulation, Developmental , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Multigene Family
4.
Article in English | MEDLINE | ID: mdl-31712186

ABSTRACT

Some organisms can modulate gene expression to trigger physiological responses that help adapt to environmental stress. The synthesis of the pigment pheomelanin in melanocytes seems to be one of these responses, as it may contribute to cellular homeostasis. We experimentally induced environmental oxidative stress in male zebra finches Taeniopygia guttata by the administration of the herbicide diquat dibromide during feather growth to test if the expression of genes involved in pheomelanin synthesis shows epigenetic lability. As pheomelanin synthesis implies decreasing the availability of the main cellular antioxidant (glutathione), it is expected to cause oxidative stress unless a protective mechanism limits pheomelanin synthesis and thus favors the antioxidant capacity. However, diquat exposure did not only improve the antioxidant capacity of birds, but also upregulated the expression of a gene (AGRP) that promotes pheomelanin synthesis in feather melanocytes, leading to the development of darker plumage coloration. No changes in the expression of other genes involved in pheomelanin synthesis (Slc7a11, Slc45a2, MC1R, ASIP and CTNS) were detected. DNA methylation levels only changed in MC1R, suggesting that epigenetic modifications other than changes in methylation may regulate AGRP expression lability. Our results suggest that exogenous oxidative stress induced a hormetic response that enhanced the oxidative status of birds and, consequently, promoted pheomelanin-based pigmentation, supporting the idea that birds adjust pheomelanin synthesis to their oxidative stress conditions.


Subject(s)
Diquat/toxicity , Feathers/physiology , Finches/physiology , Herbicides/toxicity , Melanins/biosynthesis , Oxidative Stress , Pigmentation/drug effects , Agouti-Related Protein/metabolism , Animals , Antioxidants/metabolism , Epigenesis, Genetic , Feathers/cytology , Finches/genetics , Glutathione/metabolism , Male , Melanocytes/cytology , Melanocytes/metabolism
5.
PLoS Biol ; 17(10): e3000448, 2019 10.
Article in English | MEDLINE | ID: mdl-31577791

ABSTRACT

The development of an organism involves the formation of patterns from initially homogeneous surfaces in a reproducible manner. Simulations of various theoretical models recapitulate final states of natural patterns, yet drawing testable hypotheses from those often remains difficult. Consequently, little is known about pattern-forming events. Here, we surveyed plumage patterns and their emergence in Galliformes, ratites, passerines, and penguins, together representing the three major taxa of the avian phylogeny, and built a unified model that not only reproduces final patterns but also intrinsically generates shared and varying directionality, sequence, and duration of patterning. We used in vivo and ex vivo experiments to test its parameter-based predictions. We showed that directional and sequential pattern progression depends on a species-specific prepattern: an initial break in surface symmetry launches a travelling front of sharply defined, oriented domains with self-organising capacity. This front propagates through the timely transfer of increased cell density mediated by cell proliferation, which controls overall patterning duration. These results show that universal mechanisms combining prepatterning and self-organisation govern the timely emergence of the plumage pattern in birds.


Subject(s)
Galliformes/genetics , Models, Statistical , Palaeognathae/genetics , Passeriformes/genetics , Pigmentation/genetics , Spheniscidae/genetics , Animals , Color , Embryo, Nonmammalian , Feathers/cytology , Feathers/growth & development , Feathers/metabolism , Galliformes/anatomy & histology , Galliformes/classification , Galliformes/growth & development , Inheritance Patterns , Morphogenesis/genetics , Palaeognathae/anatomy & histology , Palaeognathae/classification , Palaeognathae/growth & development , Passeriformes/anatomy & histology , Passeriformes/classification , Passeriformes/growth & development , Phylogeny , Skin/cytology , Skin/growth & development , Skin/metabolism , Spheniscidae/anatomy & histology , Spheniscidae/classification , Spheniscidae/growth & development
6.
PLoS Biol ; 17(2): e3000132, 2019 02.
Article in English | MEDLINE | ID: mdl-30789897

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.


Subject(s)
Body Patterning , Feathers/cytology , Feathers/embryology , Signal Transduction , Animals , Biomechanical Phenomena , Birds/embryology , Cell Aggregation , Cell Count , Cell Movement , Cell Shape , Ectodysplasins/metabolism , Edar Receptor/metabolism , Fibroblast Growth Factors/metabolism , Flight, Animal/physiology , Mesoderm/cytology , Mesoderm/embryology , Skin/cytology , Skin/embryology , beta Catenin/metabolism
7.
Dev Growth Differ ; 61(2): 141-149, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30675906

ABSTRACT

Selective cell death by apoptosis plays important roles in organogenesis. Apoptotic cells are observed in the developmental and homeostatic processes of several ectodermal organs, such as hairs, feathers, and mammary glands. In chick feather development, apoptotic events have been observed during feather morphogenesis, but have not been investigated during early feather bud formation. Previously, we have reported a method for generating feather buds on a bioengineered skin from dissociated skin epithelial and mesenchymal cells in three-dimensional culture. During the development of the bioengineered skin, epithelial cavity formation by apoptosis was observed in the epithelial tissue. In this study, we examined the selective epithelial cell death during the bioengineered skin development. Histological analyses suggest that the selective epithelial cell death in the bioengineered skin was induced by caspase-3-related apoptosis. The formation of feather buds of the bioengineered skin was disturbed by the treatment with a pan-caspase inhibitor. The pan-caspase inhibitor treatment suppressed the rearrangement of the epithelial layer and the formation of dermal condensation, which are thought to be essential step to form feather buds. The suppression of the formation of feather buds on the pan-caspase inhibitor-treated skin was partially compensated by the addition of a GSK-3ß inhibitor, which activates Wnt/ß-catenin signaling. These results suggest that the epithelial cell death is involved in the formation of feather buds of the bioengineered skin. These observations also suggest that caspase activities and Wnt/ß-catenin signaling may contribute to the formation of epithelial and mesenchymal components in the bioengineered skin.


Subject(s)
Cell Death , Epithelial Cells/cytology , Feathers/cytology , Feathers/growth & development , Skin/cytology , Tissue Engineering , Animals , Cells, Cultured , Chickens , Skin/growth & development
8.
Nat Commun ; 9(1): 5377, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30560870

ABSTRACT

Collective cell migration mediates multiple tissue morphogenesis processes. Yet how multi-dimensional mesenchymal cell movements are coordinated remains mostly unknown. Here we report that coordinated mesenchymal cell migration during chicken feather elongation is accompanied by dynamic changes of bioelectric currents. Transcriptome profiling and functional assays implicate contributions from functional voltage-gated Ca2+ channels (VGCCs), Connexin-43 based gap junctions, and Ca2+ release activated Ca2+ (CRAC) channels. 4-Dimensional Ca2+ imaging reveals that the Sonic hedgehog-responsive mesenchymal cells display synchronized Ca2+ oscillations, which expand progressively in area during feather elongation. Inhibiting VGCCs, gap junctions, or Sonic hedgehog signaling alters the mesenchymal Ca2+ landscape, cell movement patterns and feather bud elongation. Ca2+ oscillations induced by cyclic activation of opto-cCRAC channels enhance feather bud elongation. Functional disruption experiments and promoter analysis implicate synergistic Hedgehog and WNT/ß-Catenin signaling in activating Connexin-43 expression, establishing gap junction networks synchronizing the Ca2+ profile among cells, thereby coordinating cell movement patterns.


Subject(s)
Calcium Signaling/physiology , Cell Movement/physiology , Connexin 43/metabolism , Feathers/growth & development , Hedgehog Proteins/metabolism , Animals , Cells, Cultured , Chickens , Connexin 43/genetics , Embryo, Nonmammalian , Feathers/cytology , Gap Junctions/metabolism , Mesoderm/cytology , Mesoderm/physiology , Morphogenesis/physiology , Promoter Regions, Genetic , Skin/cytology , Wnt Signaling Pathway/physiology
9.
Sci Rep ; 8(1): 16766, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30425309

ABSTRACT

Amniote skin appendages such as feathers, hairs and scales, provide thermoregulation, physical protection and display different color patterns to attract a mate or frighten an adversary. A long-standing question is whether "reptile scale" and "avian leg scales" are of the same origin. Understanding the relation between avian feathers, avian scales and reptilian scales will enhance our understanding of skin appendage evolution. We compared the molecular and cellular profiles in chicken feather, chicken scales and alligator scales and found that chicken scutate scales are similar to chicken feathers in morphogenesis at the early placode stage. When we compared the expression of the recently identified feather-specific genes and scale-specific genes in these skin appendages, we found that at the molecular level alligator scales are significantly different from both chicken feathers and chicken scales. Furthermore, we identified a similarly diffuse putative stem cell niche in morphologically similar chicken and alligator scales. These putative stem cells participate in alligator scale regeneration. In contrast, avian feathers have a more condensed stem cell niche, which may be responsible for cycling. Thus, our results suggest that chicken and alligator scales formed independently through convergent evolution.


Subject(s)
Alligators and Crocodiles , Biological Evolution , Chickens , Feathers/cytology , Feathers/metabolism , Skin/cytology , Skin/metabolism , Animals , Biomarkers/metabolism , Chick Embryo , Diffusion , Gene Expression Regulation , Sequence Analysis, RNA , Species Specificity , Stem Cells/metabolism
10.
Anat Sci Int ; 93(4): 548-558, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29931653

ABSTRACT

The molting cycle of feathers includes an anagen (growth) stage, a likely catagen stage where the feather follicles degenerate, and a resting stage where fully grown feathers remain in their follicles and are functional before molting. However, the cytological changes involved in the resting and molting stages are poorly known, so the results of an ultrastructural analysis of these processes in adult chick feathers are presented here. The study showed that the dermal papilla shrinks, and numerous cells present increased heterochromatin and free collagen fibrils in the extracellular matrix. Degeneration of the germinal epithelium of the follicle-the papillary collar-occurs with an initial substantial contraction of cells followed by an increase in heterochromatin, vesicle and lipid accumulation, and membrane and organelle degeneration. Desmosomes are still present between degenerating epithelial cells, but ribosomes and tonofilaments disappear. This suggests that cell necrosis initially proceeds as a major contraction resembling apoptosis-a process termed necroptosis, which was previously also shown to occur during the formation of barbs and barbules in mature down and pennaceous feathers. This study suggests that, aside from apoptosis, the collar epithelium degenerates due to external factors, in particular the retraction of blood vessels supplying the dermal papilla. In contrast, revascularization of the dermal papilla triggers a new phase of feather growth (anagen).


Subject(s)
Chickens/anatomy & histology , Chickens/physiology , Feathers/cytology , Feathers/growth & development , Hair Follicle/pathology , Hair Follicle/ultrastructure , Immunohistochemistry , Microscopy, Electron, Transmission , Molting/physiology , Animals , Apoptosis , Collagen/metabolism , Dermis/blood supply , Desmosomes , Epithelium/pathology , Extracellular Matrix/metabolism , Feathers/physiology , Hair Follicle/cytology , Hair Follicle/metabolism , Heterochromatin/metabolism , Lipid Metabolism , Necrosis
11.
BMC Genomics ; 19(1): 399, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29801437

ABSTRACT

BACKGROUND: Early feathering and late feathering in chickens are sex-linked phenotypes, which have commercial application in the poultry industry for sexing chicks at hatch and have important impacts on performance traits. However, the genetic mechanism controlling feather development and feathering patterns is unclear. Here, miRNA and mRNA expression profiles in chicken wing skin tissues were analysed through high-throughput transcriptomic sequencing, aiming to understand the biological process of follicle development and the formation of different feathering phenotypes. RESULTS: Compared to the N1 group with no primary feathers extending out, 2893 genes and 31 miRNAs displayed significantly different expression in the F1 group with primary feathers longer than primary-covert feathers, and 1802 genes and 11 miRNAs in the L2 group displayed primary feathers shorter than primary-covert feathers. Only 201 altered genes and 3 altered miRNAs were identified between the N1 and L2 groups (fold change > 2, q value < 0.01). Both sequencing and qPCR tests revealed that PRLR was significantly decreased in the F1 and L2 groups compared to the N1 group, whereas SPEF2 was significantly decreased in the F1 group compared to the N1 or L2 group. Functional analysis revealed that the altered genes or targets of altered miRNAs were involved in multiple biological processes and pathways related to feather growth and development, such as the Wnt signalling pathway, the TGF-beta signalling pathway, the MAPK signalling pathway, epithelial cell differentiation, and limb development. Integrated analysis of miRNA and mRNA showed that 14 pairs of miRNA-mRNA negatively interacted in the process of feather formation. CONCLUSIONS: Transcriptomic sequencing of wing skin tissues revealed large changes in F1 vs. N1 and L2 vs. N1, but few changes in F1 vs. L2 for both miRNA and mRNA expression. PRLR might only contribute to follicle development, while SPEF2 was highly related to the growth rate of primary feathers or primary-covert feathers and could be responsible for early and late feather formation. Interactions between miR-1574-5p/NR2F, miR-365-5p/JAK3 and miR-365-5p/CDK6 played important roles in hair or feather formation. In all, our results provide novel evidence to understand the molecular regulation of follicle development and feathering phenotype.


Subject(s)
Chickens/growth & development , Chickens/genetics , Feathers/growth & development , Gene Expression Profiling , MicroRNAs/genetics , Skin/metabolism , Animals , Chickens/anatomy & histology , Feathers/cytology , RNA, Messenger/genetics , Sequence Analysis, RNA , Signal Transduction/genetics , Time Factors
12.
Nat Commun ; 9(1): 1345, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29632339

ABSTRACT

Branching morphogenesis is a general mechanism that increases the surface area of an organ. In chicken feathers, the flat epithelial sheath at the base of the follicle is transformed into periodic branches. How exactly the keratinocytes are organized into this pattern remains unclear. Here we show that in the feather follicle, the pre-branch basal keratinocytes have extensive filopodia, which contract and smooth out after branching. Manipulating the filopodia via small GTPases RhoA/Cdc42 also regulates branch formation. These basal filopodia help interpret the proximal-distal FGF gradient in the follicle. Furthermore, the topological arrangement of cell adhesion via E-Cadherin re-distribution controls the branching process. Periodic activation of Notch signaling drives the differential cell adhesion and contraction of basal filopodia, which occurs only below an FGF signaling threshold. Our results suggest a coordinated adjustment of cell shape and adhesion orchestrates feather branching, which is regulated by Notch and FGF signaling.


Subject(s)
Avian Proteins/metabolism , Feathers/growth & development , Feathers/metabolism , Fibroblast Growth Factors/metabolism , Receptors, Notch/metabolism , Animals , Cadherins/metabolism , Cell Adhesion , Cell Shape , Cells, Cultured , Chickens , Feathers/cytology , Humans , Keratinocytes/metabolism , Male , Models, Biological , Morphogenesis/physiology , Pseudopodia/metabolism , Signal Transduction
13.
PLoS Biol ; 15(12): e2004412, 2017 12.
Article in English | MEDLINE | ID: mdl-29261650

ABSTRACT

Sixty-five years after Turing first revealed the potential of systems with local activation and long-range inhibition to generate pattern, we have only recently begun to identify the biological elements that operate at many scales to generate periodic patterns in nature. In this Primer, we first review the theoretical framework provided by Turing, Meinhardt, and others that suggests how periodic patterns could self-organize in developing animals. This Primer was developed to provide context for recent studies that reveal how diverse molecular, cellular, and physical mechanisms contribute to the establishment of the periodic pattern of hair or feather buds in the developing skin. From an initial emphasis on trying to disambiguate which specific mechanism plays a primary role in hair or feather bud development, we are beginning to discover that multiple mechanisms may, in at least some contexts, operate together. While the emergence of the diverse mechanisms underlying pattern formation in specific biological contexts probably reflects the contingencies of evolutionary history, an intriguing possibility is that these mechanisms interact and reinforce each other, producing emergent systems that are more robust.


Subject(s)
Body Patterning/physiology , Feathers/cytology , Hair/cytology , Models, Biological , Animals , Feathers/anatomy & histology , Feathers/growth & development , Hair/anatomy & histology , Hair/growth & development , Signal Transduction , Skin/anatomy & histology , Skin/cytology , Skin/growth & development
14.
J Anim Sci ; 95(8): 3479-3486, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28805906

ABSTRACT

Pluripotent stem cells including embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) are regarded as representative tools for conservation of animal genetic resources. Although ESC have been established from chicken, it is very difficult to obtain enough embryos for isolation of stem cells for avian conservation in most wild birds. Therefore, the high feasibility of obtaining the pluripotent cell is most important in avian conservation studies. In this study, we generated induced pluripotent stem cell-like cells (iPSLC) from avian Feather Follicular cells (FFC). Avian FFC are one of the most easily accessible cell sources in most avian species, and their reprogramming into pluripotent stem cells can be an alternative system for preservation of avian species. Intriguingly, FFC had mesenchymal stromal cells (MSC)-like characteristics with regard to gene expression, protein expression, and adipocyte differentiation. Subsequently, we attempted to generate iPSLC from FFC using retroviral vectors. The FFC-iPSLC can proliferate with the stem pluripotent property and differentiate into several types of cells in vitro. Our results suggest that chicken FFC are an alternative cell source for avian cell reprogramming into pluripotent stem cells. This experimental strategy should be useful for conservation and restoration of endangered or high-value avian species without sacrificing embryos.


Subject(s)
Chickens/physiology , Feathers/cytology , Pluripotent Stem Cells/physiology , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Pluripotent Stem Cells/cytology
15.
Science ; 357(6353): 811-815, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28705989

ABSTRACT

The spacing of hair in mammals and feathers in birds is one of the most apparent morphological features of the skin. This pattern arises when uniform fields of progenitor cells diversify their molecular fate while adopting higher-order structure. Using the nascent skin of the developing chicken embryo as a model system, we find that morphological and molecular symmetries are simultaneously broken by an emergent process of cellular self-organization. The key initiators of heterogeneity are dermal progenitors, which spontaneously aggregate through contractility-driven cellular pulling. Concurrently, this dermal cell aggregation triggers the mechanosensitive activation of ß-catenin in adjacent epidermal cells, initiating the follicle gene expression program. Taken together, this mechanism provides a means of integrating mechanical and molecular perspectives of organ formation.


Subject(s)
Epidermal Cells , Epidermis/embryology , Feathers/cytology , Feathers/embryology , Mechanotransduction, Cellular , Organogenesis/physiology , Animals , Chick Embryo , Gene Expression Regulation, Developmental , Organogenesis/genetics , Stem Cells/cytology , Stem Cells/physiology , beta Catenin/metabolism
16.
Integr Biol (Camb) ; 9(9): 751-761, 2017 09 18.
Article in English | MEDLINE | ID: mdl-28726920

ABSTRACT

Vibrations in covalent bonds affect electron delocalization within molecules, as reported in polymers. If synthesized by living cells, the electron delocalization of polymers affects the stabilization of cellular free radicals, but biomolecular vibration has never been considered a potential source of cytotoxicity. Here we show that the vibrational characteristics of natural pheomelanin and eumelanin contribute to feather color expression in four poultry breeds with different melanin-based pigmentation patterns, but only the vibrational characteristics of pheomelanin are related to the production of reactive oxygen species (ROS) in the mitochondria of melanocytes and to systemic levels of cellular oxidative stress and damage. This association may be explained by the close physical contact existing between mitochondria and melanosomes, and reveals an unprecedented factor affecting the viability of organisms through their pigmentation. These findings open a new avenue for understanding the mechanism linking pheomelanin synthesis to human melanoma risk.


Subject(s)
Melanins/chemistry , Melanins/metabolism , Melanocytes/metabolism , Animals , Feathers/cytology , Feathers/metabolism , Female , Humans , Male , Mitochondria/metabolism , Oxidative Stress , Phenotype , Pigmentation , Poultry , Reactive Oxygen Species/metabolism , Spectrum Analysis, Raman , Vibration
17.
J Mol Biol ; 428(7): 1423-40, 2016 Apr 10.
Article in English | MEDLINE | ID: mdl-26196442

ABSTRACT

Hair and feathers are unique because (1) their stem cells are contained within a follicle structure, (2) they undergo cyclic regeneration repetitively throughout life, (3) regeneration occurs physiologically in healthy individuals and (4) regeneration is also induced in response to injury. Precise control of this cyclic regeneration process is essential for maintaining the homeostasis of living organisms. While stem cells are regulated by the intra-follicle-adjacent micro-environmental niche, this niche is also modulated dynamically by extra-follicular macro-environmental signals, allowing stem cells to adapt to a larger changing environment and physiological needs. Here we review several examples of macro-environments that communicate with the follicles: intradermal adipose tissue, innate immune system, sex hormones, aging, circadian rhythm and seasonal rhythms. Related diseases are also discussed. Unveiling the mechanisms of how stem cell niches are modulated provides clues for regenerative medicine. Given that stem cells are hard to manipulate, focusing translational therapeutic applications at the environments appears to be a more practical approach.


Subject(s)
Feathers/physiology , Hair Follicle/physiology , Regeneration/physiology , Stem Cell Niche/physiology , Animals , Feathers/cytology , Hair Follicle/cytology , Humans , Tissue Scaffolds
18.
PLoS One ; 10(9): e0137622, 2015.
Article in English | MEDLINE | ID: mdl-26352664

ABSTRACT

Feathers have been widely used to assess mercury contamination in birds as they reflect metal concentrations accumulated between successive moult periods: they are also easy to sample and have minimum impact on the study birds. Moult is considered the major pathway for mercury excretion in seabirds. Penguins are widely believed to undergo a complete, annual moult during which they do not feed. As penguins lose all their feathers, they are expected to have a low individual-variability in feather mercury concentration as all feathers are formed simultaneously from the same somatic reserves. This assumption is central to penguin studies that use feathers to examine the annual or among-individual variation in mercury concentrations in penguins. To test this assumption, we measured the mercury concentrations in 3-5 body feathers of 52 gentoo penguins (Pygoscelis papua) breeding at Bird Island, South Georgia (54°S 38°W). Twenty-five percent of the penguins studied showed substantial within-individual variation in the amount of mercury in their feathers (Coefficient of Variation: 34.7-96.7%). This variation may be caused by differences in moult patterns among individuals within the population leading to different interpretations in the overall population. Further investigation is now needed to fully understand individual variation in penguins' moult.


Subject(s)
Environmental Monitoring , Feathers/cytology , Mercury/isolation & purification , Animals , Feathers/chemistry , Mercury/toxicity , Spheniscidae
19.
J Invest Dermatol ; 135(7): 1719-1721, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26066892

ABSTRACT

Through cyclic regeneration, feather stem cells are molded into different shapes under different physiological states. With its distinct morphology, context-dependent growth, and experimental manipulability, the feather provides a rich model to study growth control, regeneration, and morphogenesis in vivo. Recent examples include novel insights revealed by transient perturbation with chemotherapeutic reagents and irradiation during feather growth.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Feathers/cytology , Hedgehog Proteins/metabolism , Signal Transduction/drug effects , Animals , Male
20.
Evol Dev ; 17(3): 185-94, 2015.
Article in English | MEDLINE | ID: mdl-25963196

ABSTRACT

Feathers are an evolutionary novelty found in all extant birds. Despite recent progress investigating feather development and a revolution in dinosaur paleontology, the relationship of feathers to other amniote skin appendages, particularly reptile scales, remains unclear. Disagreement arises primarily from the observation that feathers and avian scutate scales exhibit an anatomical placode-defined as an epidermal thickening-in early development, whereas alligator and other avian scales do not. To investigate the homology of feathers and archosaur scales we examined patterns of nuclear ß-catenin localization during early development of feathers and different bird and alligator scales. In birds, nuclear ß-catenin is first localized to the feather placode, and then exhibits a dynamic pattern of localization in both epidermis and dermis of the feather bud. We found that asymmetric avian scutate scales and alligator scales share similar patterns of nuclear ß-catenin localization with feathers. This supports the hypothesis that feathers, scutate scales, and alligator scales are homologous during early developmental stages, and are derived from early developmental stages of an asymmetric scale present in the archosaur ancestor. Furthermore, given that the earliest stage of ß-catenin localization in feathers and archosaur scales is also found in placodes of several mammalian skin appendages, including hair and mammary glands, we hypothesize that a common skin appendage placode originated in the common ancestor of all amniotes. We suggest a skin placode should not be defined by anatomical features, but as a local, organized molecular signaling center from which an epidermal appendage develops.


Subject(s)
Biological Evolution , Birds/genetics , Feathers/embryology , beta Catenin/analysis , Alligators and Crocodiles/anatomy & histology , Animal Structures/chemistry , Animal Structures/cytology , Animals , Birds/embryology , Feathers/chemistry , Feathers/cytology
SELECTION OF CITATIONS
SEARCH DETAIL