Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.942
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Dev Biol ; 515: 60-66, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38964706

RESUMO

Terminal differentiation of epithelial cells is critical for the barrier function of the skin, the growth of skin appendages, such as hair and nails, and the development of the skin of amniotes. Here, we present the hypothesis that the differentiation of cells in the embryonic periderm shares characteristic features with the differentiation of epithelial cells that support the morphogenesis of cornified skin appendages during postnatal life. The periderm prevents aberrant fusion of adjacent epithelial sites during early skin development. It is shed off when keratinocytes of the epidermis form the cornified layer, the stratum corneum. A similar role is played by epithelia that ensheath cornifying skin appendages until they disintegrate to allow the separation of the mature part of the skin appendage from the adjacent tissue. These epithelia, exemplified by the inner root sheath of hair follicles and the epithelia close to the free edge of nails or claws, are referred to as scaffolding epithelia. The periderm and scaffolding epithelia are similar with regard to their transient functions in separating tissues and the conserved expression of trichohyalin and trichohyalin-like genes in mammals and birds. Thus, we propose that parts of the peridermal differentiation program were coopted to a new postnatal function during the evolution of cornified skin appendages in amniotes.


Assuntos
Diferenciação Celular , Diferenciação Celular/fisiologia , Animais , Pele/embriologia , Pele/citologia , Pele/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Epitélio/embriologia , Epitélio/metabolismo , Epiderme/embriologia , Epiderme/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Folículo Piloso/embriologia , Folículo Piloso/citologia , Humanos , Morfogênese
2.
Cell ; 136(6): 992-4, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19303840

RESUMO

Polycomb group proteins control a hierarchy of gene expression during the differentiation of stem cells. In this issue, Ezhkova et al. (2009) show that repression mediated by Polycomb proteins controls the timing of differentiation of precursor cells of the epidermal lineage.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas Repressoras/metabolismo , Pele/citologia , Animais , Diferenciação Celular , Modelos Biológicos , Proteínas do Grupo Polycomb , Pele/embriologia , Pele/metabolismo
3.
Genes Dev ; 30(20): 2325-2338, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27807033

RESUMO

Growth and regeneration of one tissue within an organ compels accommodative changes in the surrounding tissues. However, the molecular nature and operating logic governing these concurrent changes remain poorly defined. The dermal adipose layer expands concomitantly with hair follicle downgrowth, providing a paradigm for studying coordinated changes of surrounding lineages with a regenerating tissue. Here, we discover that hair follicle transit-amplifying cells (HF-TACs) play an essential role in orchestrating dermal adipogenesis through secreting Sonic Hedgehog (SHH). Depletion of Shh from HF-TACs abrogates both dermal adipogenesis and hair follicle growth. Using cell type-specific deletion of Smo, a gene required in SHH-receiving cells, we found that SHH does not act on hair follicles, adipocytes, endothelial cells, and hematopoietic cells for adipogenesis. Instead, SHH acts directly on adipocyte precursors, promoting their proliferation and their expression of a key adipogenic gene, peroxisome proliferator-activated receptor γ (Pparg), to induce dermal adipogenesis. Our study therefore uncovers a critical role for TACs in orchestrating the generation of both their own progeny and a neighboring lineage to achieve concomitant tissue production across lineages.


Assuntos
Adipogenia/fisiologia , Folículo Piloso/citologia , Folículo Piloso/metabolismo , Proteínas Hedgehog/metabolismo , Pele/metabolismo , Adipogenia/genética , Animais , Proliferação de Células/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Folículo Piloso/embriologia , Folículo Piloso/crescimento & desenvolvimento , Masculino , Camundongos , Transdução de Sinais , Pele/embriologia , Pele/crescimento & desenvolvimento
4.
Cell ; 132(2): 299-310, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18243104

RESUMO

Quiescent adult stem cells reside in specialized niches where they become activated to proliferate and differentiate during tissue homeostasis and injury. How stem cell quiescence is governed is poorly understood. We report here that NFATc1 is preferentially expressed by hair follicle stem cells in their niche, where its expression is activated by BMP signaling upstream and it acts downstream to transcriptionally repress CDK4 and maintain stem cell quiescence. As stem cells become activated during hair growth, NFATc1 is downregulated, relieving CDK4 repression and activating proliferation. When calcineurin/NFATc1 signaling is suppressed, pharmacologically or via complete or conditional NFATc1 gene ablation, stem cells are activated prematurely, resulting in precocious follicular growth. Our findings may explain why patients receiving cyclosporine A for immunosuppressive therapy display excessive hair growth, and unveil a functional role for calcium-NFATc1-CDK4 circuitry in governing stem cell quiescence.


Assuntos
Proliferação de Células , Fatores de Transcrição NFATC/metabolismo , Pele/citologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Animais , Antígenos CD34/metabolismo , Biomarcadores , Núcleo Celular/metabolismo , Células Cultivadas , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Ciclosporina/farmacologia , Regulação para Baixo , Embrião de Mamíferos , Deleção de Genes , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Folículo Piloso/citologia , Folículo Piloso/fisiologia , Imuno-Histoquímica , Imunossupressores/farmacologia , Camundongos , Camundongos Knockout , Camundongos Nus , Morfogênese , Fatores de Transcrição NFATC/genética , RNA Mensageiro/metabolismo , Retroviridae/genética , Pele/embriologia , Transplante de Pele , Células-Tronco/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Transgenes , Transplante Homólogo
5.
PLoS Biol ; 17(2): e3000132, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30789897

RESUMO

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.


Assuntos
Padronização Corporal , Plumas/citologia , Plumas/embriologia , Transdução de Sinais , Animais , Fenômenos Biomecânicos , Aves/embriologia , Agregação Celular , Contagem de Células , Movimento Celular , Forma Celular , Ectodisplasinas/metabolismo , Receptor Edar/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Voo Animal/fisiologia , Mesoderma/citologia , Mesoderma/embriologia , Pele/citologia , Pele/embriologia , beta Catenina/metabolismo
6.
PLoS Biol ; 17(5): e3000287, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31141500

RESUMO

Atypical chemokine receptor 2 (ACKR2) is a chemokine-scavenging receptor. ACKR2-/-embryos display a reduction in size of a novel, to our knowledge, embryonic skin macrophage population referred to as 'intermediate' cells. CC chemokine receptor 2 (CCR2)-/-embryos display an identical phenotype, indicating that these cells require CCR2 to enable them to populate embryonic skin. Further analysis revealed that ACKR2-/-embryos have higher circulating concentrations of the CCR2 ligand, CC ligand 2 (CCL2); thus, ACKR2 regulates intraembryonic CCL2 levels. We show that ACKR2 is strongly expressed by trophoblasts and that it blocks movement of inflammatory chemokines, such as CCL2, from the maternal decidua into the embryonic circulation. We propose that trophoblastic ACKR2 is responsible for ensuring chemokine compartmentalisation on the maternal decidua, without which chemokines enter the embryonic circulation, disrupting gradients essential for directed intraembryonic cell migration. Overall, therefore, we describe a novel, to our knowledge, molecular mechanism whereby maternal decidual chemokines can function in a compartmentalised fashion without interfering with intraembryonic leukocyte migration. These data suggest similar functions for other atypical chemokine receptors in the placenta and indicate that defects in such receptors may have unanticipated developmental consequences.


Assuntos
Quimiocinas/metabolismo , Mamíferos/metabolismo , Placenta/metabolismo , Animais , Movimento Celular , Decídua/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Gravidez , Receptores de Quimiocinas/deficiência , Receptores de Quimiocinas/metabolismo , Pele/embriologia , Pele/metabolismo , Transcrição Gênica , Saco Vitelino/metabolismo
7.
Nature ; 539(7630): 518-523, 2016 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-27806375

RESUMO

Mammalian colour patterns are among the most recognizable characteristics found in nature and can have a profound impact on fitness. However, little is known about the mechanisms underlying the formation and subsequent evolution of these patterns. Here we show that, in the African striped mouse (Rhabdomys pumilio), periodic dorsal stripes result from underlying differences in melanocyte maturation, which give rise to spatial variation in hair colour. We identify the transcription factor ALX3 as a regulator of this process. In embryonic dorsal skin, patterned expression of Alx3 precedes pigment stripes and acts to directly repress Mitf, a master regulator of melanocyte differentiation, thereby giving rise to light-coloured hair. Moreover, Alx3 is upregulated in the light stripes of chipmunks, which have independently evolved a similar dorsal pattern. Our results show a previously undescribed mechanism for modulating spatial variation in hair colour and provide insights into how phenotypic novelty evolves.


Assuntos
Padronização Corporal , Regulação da Expressão Gênica no Desenvolvimento , Cor de Cabelo , Murinae/embriologia , Murinae/genética , Animais , Evolução Biológica , Padronização Corporal/genética , Diferenciação Celular , Cor de Cabelo/genética , Proteínas de Homeodomínio/metabolismo , Melaninas/biossíntese , Melanócitos/citologia , Melanócitos/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia/antagonistas & inibidores , Fator de Transcrição Associado à Microftalmia/metabolismo , Murinae/fisiologia , Fenótipo , Regiões Promotoras Genéticas/genética , Sciuridae/genética , Pele/embriologia
8.
Dev Dyn ; 250(4): 584-600, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33354814

RESUMO

BACKGROUND: Cases of parallel evolution offer the possibility to identify adaptive traits and to uncover developmental constraints on the evolutionary trajectories of these traits. The independent evolution of direct development from the ancestral biphasic life history in frogs is such a case of parallel evolution. In frogs, aquatic larvae (tadpoles) differ profoundly from their adult forms and exhibit a stunning diversity regarding their habitats, morphology and feeding behaviors. The transition from the tadpole to the adult is a climactic, thyroid hormone (TH)-dependent process of profound and fast morphological rearrangement called metamorphosis. One of the organ systems that experiences the most comprehensive metamorphic rearrangements is the skin. Direct-developing frogs lack a free-swimming tadpole and hatch from terrestrial eggs as fully formed froglets. In the few species examined, development is characterized by the condensed and transient formation of some tadpole-specific features and the early formation of adult-specific features during a "cryptic" metamorphosis. RESULTS: We show that skin in direct-developing African squeaker frogs (Arthroleptis) is also repatterned from a tadpole-like to an adult-like histology during a cryptic metamorphosis. This repatterning correlates with histological thyroid gland maturation. A comparison with data from the Puerto Rican coqui (Eleutherodactylus coqui) reveals that the evolution of direct development in these frogs is associated with a comparable heterochronic shift of thyroid gland maturation. CONCLUSION: This suggests that the development of many adult features is still dependent on, and possibly constrained by, the ancestral dependency on thyroid hormone signaling.


Assuntos
Anuros/embriologia , Evolução Biológica , Metamorfose Biológica , Pele/embriologia , Glândula Tireoide/embriologia , Animais , Epiderme/crescimento & desenvolvimento , Epiderme/metabolismo , Larva/anatomia & histologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Pigmentação da Pele
9.
Development ; 145(12)2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29769218

RESUMO

To distribute and establish the melanocyte lineage throughout the skin and other developing organs, melanoblasts undergo several rounds of proliferation, accompanied by migration through complex environments and differentiation. Melanoblast migration requires interaction with extracellular matrix of the epidermal basement membrane and with surrounding keratinocytes in the developing skin. Migration has been characterized by measuring speed, trajectory and directionality of movement, but there are many unanswered questions about what motivates and defines melanoblast migration. Here, we have established a general mathematical model to simulate the movement of melanoblasts in the epidermis based on biological data, assumptions and hypotheses. Comparisons between experimental data and computer simulations reinforce some biological assumptions, and suggest new ideas for how melanoblasts and keratinocytes might influence each other during development. For example, it appears that melanoblasts instruct each other to allow a homogeneous distribution in the tissue and that keratinocytes may attract melanoblasts until one is stably attached to them. Our model reveals new features of how melanoblasts move and, in particular, suggest that melanoblasts leave a repulsive trail behind them as they move through the skin.


Assuntos
Movimento Celular/fisiologia , Simulação por Computador , Queratinócitos/metabolismo , Melanócitos/citologia , Pele/embriologia , Animais , Membrana Basal/metabolismo , Adesão Celular/fisiologia , Matriz Extracelular/metabolismo , Melanócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Teóricos
10.
Pediatr Dev Pathol ; 24(2): 96-102, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33470919

RESUMO

BACKGROUND: The immature skin of preterm infants is uniquely vulnerable to pressure and chemical injury. We sought to qualitatively and quantitatively describe the histopathologic patterns of skin development in preterm infants. METHODS: Autopsy skin samples were examined for 48 liveborn preterm infants born at 18+ to 36 weeks, and control groups of term neonates and older infants/children. Quantitative variables included thickness of the stratum corneum, epidermis, and dermis. Qualitative features included stratum corneum, rete ridges, and hair follicles. RESULTS: Patterns of maturation were reproducible. Compact keratin appeared beginning at 21-22 weeks. Basketweave keratin appeared first around hair follicles, and then became more generalized from ∼28 weeks corrected gestational age (CGA) onward. Rete ridges began to appear at ∼30 weeks. Epidemal and dermal thickness increased with age. Infants who survived ≤7 days had thicker dermis than those who survived longer, even adjusted for CGA. CONCLUSIONS: Skin development in preterm infants has reproducible milestones. Significant structural changes occurring around 28-30 weeks may improve barrier function, with implications for use of topical compounds such as chlorhexidine. The findings also highlight challenges in evaluating pressure injuries in preterm infants, and postnatal changes in skin parameters.


Assuntos
Recém-Nascido Prematuro , Pele/patologia , Estudos de Casos e Controles , Desenvolvimento Infantil , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Pele/embriologia , Pele/crescimento & desenvolvimento
11.
Proc Natl Acad Sci U S A ; 115(32): 8173-8178, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30037996

RESUMO

Ectodysplasin A (Eda) signaling activates NF-κB during skin appendage formation, but how Eda controls specific gene transcription remains unclear. Here, we find that Eda triggers the formation of an NF-κB-associated SWI/SNF (BAF) complex in which p50/RelB recruits a linker protein, Tfg, that interacts with BAF45d in the BAF complex. We further reveal that Tfg is initially induced by Eda-mediated RelB activation and then bridges RelB and BAF for subsequent gene regulation. The BAF component BAF250a is particularly up-regulated in skin appendages, and epidermal knockout of BAF250a impairs skin appendage development, resulting in phenotypes similar to those of Eda-deficient mouse models. Transcription profiling identifies several target genes regulated by Eda, RelB, and BAF. Notably, RelB and the BAF complex are indispensable for transcription of Eda target genes, and both BAF complex and Eda signaling are required to open chromatin of Eda targets. Our studies thus suggest that Eda initiates a signaling cascade and recruits a BAF complex to specific gene loci to facilitate transcription during organogenesis.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Ectodisplasinas/metabolismo , Organogênese/genética , Pele/embriologia , Fator de Transcrição RelB/genética , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Animais , Cromatina/metabolismo , Ectodisplasinas/genética , Receptor Edar/genética , Receptor Edar/metabolismo , Feminino , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição RelB/metabolismo , Ativação Transcricional/fisiologia , Regulação para Cima
12.
Int J Mol Sci ; 22(15)2021 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-34361001

RESUMO

Epidermal progenitor cells divide symmetrically and asymmetrically to form stratified epidermis and hair follicles during late embryonic development. Flightless I (Flii), an actin remodelling protein, is implicated in Wnt/ß-cat and integrin signalling pathways that govern cell division. This study investigated the effect of altering Flii on the divisional orientation of epidermal progenitor cells (EpSCs) in the basal layer during late murine embryonic development and early adolescence. The effect of altering Flii expression on asymmetric vs. symmetric division was assessed in vitro in adult human primary keratinocytes and in vivo at late embryonic development stages (E16, E17 and E19) as well as adolescence (P21 day-old) in mice with altered Flii expression (Flii knockdown: Flii+/-, wild type: WT, transgenic Flii overexpressing: FliiTg/Tg) using Western blot and immunohistochemistry. Flii+/- embryonic skin showed increased asymmetrical cell division of EpSCs with an increase in epidermal stratification and elevated talin, activated-Itgb1 and Par3 expression. FliiTg/Tg led to increased symmetrical cell division of EpSCs with increased cell proliferation rate, an elevated epidermal SOX9, Flap1 and ß-cat expression, a thinner epidermis, but increased hair follicle number and depth. Flii promotes symmetric division of epidermal progenitor cells during murine embryonic development.


Assuntos
Divisão Celular , Proteínas dos Microfilamentos/genética , Células-Tronco Embrionárias Murinas/metabolismo , Pele/metabolismo , Transativadores/genética , Animais , Células Cultivadas , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Fatores de Transcrição SOX9/metabolismo , Pele/embriologia , Transativadores/metabolismo , beta Catenina/metabolismo
13.
Dev Biol ; 455(1): 60-72, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31279726

RESUMO

Salamanders are capable of full-thickness skin regeneration where removal of epidermis, dermis and hypodermis results in scar-free repair. What remains unclear is whether regeneration of these tissues recapitulates the cellular events of skin development or occurs through a process unique to regenerative healing. Unfortunately, information on the post-embryonic development of salamander skin is severely lacking, having focused on compartments or cell types, but never on the skin as a complete organ. By examining coordinated development of the epidermis and dermis in axolotls we establish six distinct stages of skin development (I-VI): I-V for normally paedomorphic adults and a sixth stage following metamorphosis. Raising animals either in isolation (zero density pressure) or in groups (density pressure) we find that skin development progresses as a function of animal size and that density directly effects developmental rate. Using keratins, p63, and proliferative markers, we show that when the dermis transforms into the stratum spongiosum and stratum compactum, keratinocytes differentiate into at least three distinct phenotypes that reveal a cryptic stratification program uncoupled from metamorphosis. Lastly, comparing skin regeneration to skin development, we find that dermal regeneration occurs through a unique process, relying heavily on remodeling of the wound extracellular matrix, rather than proceeding through direct development of a dermal lamella produced by the epidermis. By preventing fibroblast influx into the wound bed using beryllium nitrate, we show that in the absence of fibroblast generated ECM production skin regeneration occurs through an alternate route that recapitulates development.


Assuntos
Desenvolvimento Embrionário/fisiologia , Fibroblastos/fisiologia , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Pele/fisiopatologia , Cicatrização/fisiologia , Ambystoma mexicanum/embriologia , Ambystoma mexicanum/fisiologia , Animais , Derme/embriologia , Derme/metabolismo , Derme/fisiologia , Epiderme/embriologia , Epiderme/metabolismo , Epiderme/fisiologia , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiologia , Queratinócitos/citologia , Queratinócitos/metabolismo , Queratinócitos/fisiologia , Queratinas/metabolismo , Masculino , Pele/embriologia , Pele/lesões , Fatores de Tempo
14.
Immunology ; 160(2): 149-156, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32173857

RESUMO

Establishment of a well-functioning immune network in skin is crucial for its barrier function. This begins in utero alongside the structural differentiation and maturation of skin, and continues to expand and diversify across the human lifespan. The microenvironment of the developing human skin supports immune cell differentiation and has an overall anti-inflammatory profile. Immunologically inert and skewed immune populations found in developing human skin promote wound healing, and as such may play a crucial role in the structural changes occurring during skin development.


Assuntos
Desenvolvimento Embrionário/imunologia , Imunidade Celular , Pele/embriologia , Cicatrização/imunologia , Células Apresentadoras de Antígenos/imunologia , Diferenciação Celular/imunologia , Eritrócitos/imunologia , Humanos , Tolerância Imunológica , Linfócitos/imunologia , Mastócitos/imunologia , Pele/citologia , Pele/imunologia
15.
J Exp Zool B Mol Dev Evol ; 334(5): 294-310, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32410344

RESUMO

Studying reptilian embryonic development provides answers to many questions related to the development of tetrapods. Reptilian skin has been recently considered in studies at the evo-devo level. The lizard epidermis has to be shed periodically. At the embryonic level, contention exists regarding the first layers to appear, whether the oberhautchen or the clear layer, and whether the shedding complex develops before hatching. Geckos exhibit diverse morphologies independently evolved multiple times within the clade, such as subdigital pad lamellae. Here we investigate the embryonic development of Tarentola mauritanica and establishing its embryonic table. Primarily we follow the development of the integument. This is a closely related species to Tarentola annularis and it is crucial to investigate whether it has the same derived digital condition of claw regression. Eleven embryonic stages are described according to the external morphological characteristics of the embryos. Interestingly, the oviposition stage appears earlier than its close relative T. annularis, and the total incubation time is less. We also describe skin development, adding clear evidence to the debate on the development of the shedding complex, which we found is developed before hatching. We describe one layer of periderm and the clear layer as the first embryonic epidermal layers. Generally, our results show the genus Tarentola to have the advantage of being a unique taxon, easily breed at the laboratory, with multiple clutches per year, and with an earlier stage at oviposition. That could be a model animal for embryonic development and experimental embryology studies.


Assuntos
Embrião não Mamífero , Lagartos/embriologia , Pele/embriologia , Animais , Desenvolvimento Embrionário , Morfogênese
16.
PLoS Biol ; 15(7): e2002117, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28700594

RESUMO

Two theories address the origin of repeating patterns, such as hair follicles, limb digits, and intestinal villi, during development. The Turing reaction-diffusion system posits that interacting diffusible signals produced by static cells first define a prepattern that then induces cell rearrangements to produce an anatomical structure. The second theory, that of mesenchymal self-organisation, proposes that mobile cells can form periodic patterns of cell aggregates directly, without reference to any prepattern. Early hair follicle development is characterised by the rapid appearance of periodic arrangements of altered gene expression in the epidermis and prominent clustering of the adjacent dermal mesenchymal cells. We assess the contributions and interplay between reaction-diffusion and mesenchymal self-organisation processes in hair follicle patterning, identifying a network of fibroblast growth factor (FGF), wingless-related integration site (WNT), and bone morphogenetic protein (BMP) signalling interactions capable of spontaneously producing a periodic pattern. Using time-lapse imaging, we find that mesenchymal cell condensation at hair follicles is locally directed by an epidermal prepattern. However, imposing this prepattern's condition of high FGF and low BMP activity across the entire skin reveals a latent dermal capacity to undergo spatially patterned self-organisation in the absence of epithelial direction. This mesenchymal self-organisation relies on restricted transforming growth factor (TGF) ß signalling, which serves to drive chemotactic mesenchymal patterning when reaction-diffusion patterning is suppressed, but, in normal conditions, facilitates cell movement to locally prepatterned sources of FGF. This work illustrates a hierarchy of periodic patterning modes operating in organogenesis.


Assuntos
Folículo Piloso/embriologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Padronização Corporal , Diferenciação Celular , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos , Transdução de Sinais , Pele/citologia , Pele/embriologia , Pele/metabolismo , Fator de Crescimento Transformador beta/metabolismo
17.
Genome ; 63(12): 615-628, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32956594

RESUMO

Skin and feather follicle morphogenesis are important processes for duck development; however, the mechanisms underlying morphogenesis at the embryonic stage remain unclear. To improve the understanding of these processes, we used transcriptome and weighted gene co-expression network analyses to identify the critical genes and pathways involved in duck skin development. Five modules were found to be the most related to five key stages in skin development that span from embryonic day 8 (E8) to postnatal day 7 (D7). Using STEM software, 6519 genes from five modules were clustered into 10 profiles to reveal key genes. Above all, we obtained several key module genes including WNT3A, NOTCH1, SHH, BMP2, NOG, SMAD3, and TGFß2. Furthermore, we revealed that several pathways play critical roles throughout the skin development process, including the Wnt pathway and cytoskeletal rearrangement-related pathways, whereas others are involved in specific stages of skin development, such as the Notch, Hedgehog, and TGF-beta signaling pathways. Overall, this study identified the pathways and genes that play critical roles in skin development, which may provide a basis for high-quality down-type meat duck breeding.


Assuntos
Patos/embriologia , Patos/genética , Desenvolvimento Embrionário/genética , Pele/embriologia , Animais , Patos/crescimento & desenvolvimento , Plumas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Anotação de Sequência Molecular , Morfogênese/genética , Organogênese , Pele/metabolismo , Transcriptoma
18.
Cell Mol Life Sci ; 76(10): 1919-1934, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30830237

RESUMO

Here, we review melanocyte development and how the embryonic melanoblast, although specified to become a melanocyte, is prone to cellular plasticity and is not fully committed to the melanocyte lineage. Even fully differentiated and pigment-producing melanocytes do not always have a stable phenotype. The gradual lineage restriction of neural crest cells toward the melanocyte lineage is determined by both cell-intrinsic and extracellular signals in which differentiation and pathfinding ability reciprocally influence each other. These signals are leveraged by subtle differences in timing and axial positioning. The most extensively studied migration route is the dorsolateral path between the dermomyotome and the prospective epidermis, restricted to melanoblasts. In addition, the embryonic origin of the skin dermis through which neural crest derivatives migrate may also affect the segregation between melanogenic and neurogenic cells in embryos. It is widely accepted that, irrespective of the model organism studied, the immediate precursor of both melanoblast and neurogenic populations is a glial-melanogenic bipotent progenitor. Upon exposure to different conditions, melanoblasts may differentiate into other neural crest-derived lineages such as neuronal cells and vice versa. Key factors that regulate melanoblast migration and patterning will regulate melanocyte homeostasis during different stages of hair cycling in postnatal hair follicles.


Assuntos
Diferenciação Celular , Plasticidade Celular , Melanócitos/citologia , Crista Neural/citologia , Animais , Movimento Celular , Humanos , Modelos Biológicos , Crista Neural/embriologia , Pele/citologia , Pele/embriologia , Células-Tronco/citologia
19.
Biol Res ; 53(1): 4, 2020 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-32014065

RESUMO

BACKGROUND: Pigmentation development, is a complex process regulated by many transcription factors during development. With the development of the RNA sequencing (RNA-seq), non-coding RNAs, such as miRNAs, lncRNAs, and circRNAs, are found to play an important role in the function detection of related regulation factors. In this study, we provided the expression profiles and development of ncRNAs related to melanocyte and skin development in mice with black coat color skin and mice with white coat color skin during embryonic day 15 (E15) and postnatal day 7 (P7). The expression profiles of different ncRNAs were detected via RNA-seq and also confirmed by the quantitative real-time PCR (qRT-PCR) method. GO and KEGG used to analyze the function the related target genes. RESULTS: We identified an extensive catalogue of 206 and 183 differently expressed miRNAs, 600 and 800 differently expressed lncRNAs, and 50 and 54 differently expressed circRNAs, respectively. GO terms and pathway analysis showed the target genes of differentially expressed miRNA and lncRNA. The host genes of circRNA were mainly enriched in cellular process, single organism process. The target genes of miRNAs were mainly enriched in chromatin binding and calcium ion binding in the nucleus. The function of genes related to lncRNAs are post translation modification. The competing endogenous RNA (ceRNA) network of lncRNAs and circRNAs displays a complex interaction between ncRNA and mRNA related to skin development, such as Tcf4, Gnas, and Gpnms related to melanocyte development. CONCLUSIONS: The ceRNA network of lncRNA and circRNA displays a complex interaction between ncRNA and mRNA related to skin development and melanocyte development. The embryonic and postnatal development of skin provide a reference for further studies on the development mechanisms of ncRNA during pigmentation.


Assuntos
Perfilação da Expressão Gênica , Melanócitos , MicroRNAs/genética , RNA Longo não Codificante/genética , Pigmentação da Pele/genética , Pele/embriologia , Animais , Diferenciação Celular , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
20.
Dev Biol ; 433(2): 144-154, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29179946

RESUMO

The skin is the largest organ in the body and plays multiple essential roles ranging from regulating temperature, preventing infection and ultimately defining who we are physically. It is a highly dynamic organ that constantly replaces the outermost cells throughout life. However, when faced with a major injury, human skin cannot restore a significant lesion to its original functionality, instead a reparative scar is formed. In contrast to this, many other species have the unique ability to regenerate full thickness skin without formation of scar tissue. Here we review recent advances in the field that shed light on how the skin cells in regenerative species react to injury to prevent scar formation versus scar forming humans.


Assuntos
Cicatriz/prevenção & controle , Modelos Animais , Cicatrização/fisiologia , Anfíbios/fisiologia , Animais , Cicatriz/fisiopatologia , Matriz Extracelular/metabolismo , Fibroblastos/fisiologia , Regulação da Expressão Gênica , Humanos , Invertebrados/fisiologia , Queratinócitos/fisiologia , Mamíferos/fisiologia , Lesões Pré-Natais/fisiopatologia , Regeneração/fisiologia , Pele/citologia , Pele/embriologia , Especificidade da Espécie , Cicatrização/imunologia , Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA