Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Cell ; 179(6): 1409-1423.e17, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31778655

RESUMEN

The evolution of flight in feathered dinosaurs and early birds over millions of years required flight feathers whose architecture features hierarchical branches. While barb-based feather forms were investigated, feather shafts and vanes are understudied. Here, we take a multi-disciplinary approach to study their molecular control and bio-architectural organizations. In rachidial ridges, epidermal progenitors generate cortex and medullary keratinocytes, guided by Bmp and transforming growth factor ß (TGF-ß) signaling that convert rachides into adaptable bilayer composite beams. In barb ridges, epidermal progenitors generate cylindrical, plate-, or hooklet-shaped barbule cells that form fluffy branches or pennaceous vanes, mediated by asymmetric cell junction and keratin expression. Transcriptome analyses and functional studies show anterior-posterior Wnt2b signaling within the dermal papilla controls barbule cell fates with spatiotemporal collinearity. Quantitative bio-physical analyses of feathers from birds with different flight characteristics and feathers in Burmese amber reveal how multi-dimensional functionality can be achieved and may inspire future composite material designs. VIDEO ABSTRACT.


Asunto(s)
Adaptación Fisiológica , Plumas/anatomía & histología , Plumas/fisiología , Vuelo Animal/fisiología , Animales , Evolución Biológica , Aves/anatomía & histología , Moléculas de Adhesión Celular/metabolismo , Citoesqueleto/metabolismo , Dermis/anatomía & histología , Células Madre/citología , Factores de Tiempo , Transcriptoma/genética , Vía de Señalización Wnt/genética
2.
Cell ; 161(2): 277-90, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25860610

RESUMEN

Coordinated organ behavior is crucial for an effective response to environmental stimuli. By studying regeneration of hair follicles in response to patterned hair plucking, we demonstrate that organ-level quorum sensing allows coordinated responses to skin injury. Plucking hair at different densities leads to a regeneration of up to five times more neighboring, unplucked resting hairs, indicating activation of a collective decision-making process. Through data modeling, the range of the quorum signal was estimated to be on the order of 1 mm, greater than expected for a diffusible molecular cue. Molecular and genetic analysis uncovered a two-step mechanism, where release of CCL2 from injured hairs leads to recruitment of TNF-α-secreting macrophages, which accumulate and signal to both plucked and unplucked follicles. By coupling immune response with regeneration, this mechanism allows skin to respond predictively to distress, disregarding mild injury, while meeting stronger injury with full-scale cooperative activation of stem cells.


Asunto(s)
Folículo Piloso/citología , Células Madre/citología , Animales , Comunicación Celular , Quimiocina CCL2/metabolismo , Folículo Piloso/fisiología , Queratinocitos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Regeneración , Piel/citología , Piel/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
3.
Cell ; 158(5): 1212-1212.e1, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171418

RESUMEN

Ectodermal appendages such as feathers, hair, mammary glands, salivary glands, and sweat glands form branches, allowing much-increased surface for functional differentiation and secretion. Here, the principles of branching morphogenesis are exemplified by the mammary gland and feathers.


Asunto(s)
Plumas/crecimiento & desarrollo , Glándulas Mamarias Humanas/crecimiento & desarrollo , Morfogénesis , Transducción de Señal , Animales , Aves/crecimiento & desarrollo , Aves/metabolismo , Plumas/citología , Femenino , Humanos , Masculino , Mamíferos/crecimiento & desarrollo , Mamíferos/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Humanas/citología
4.
PLoS Biol ; 22(5): e3002636, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38743770

RESUMEN

Periodic patterning requires coordinated cell-cell interactions at the tissue level. Turing showed, using mathematical modeling, how spatial patterns could arise from the reactions of a diffusive activator-inhibitor pair in an initially homogeneous 2D field. Most activators and inhibitors studied in biological systems are proteins, and the roles of cell-cell interaction, ions, bioelectricity, etc. are only now being identified. Gap junctions (GJs) mediate direct exchanges of ions or small molecules between cells, enabling rapid long-distance communications in a cell collective. They are therefore good candidates for propagating nonprotein-based patterning signals that may act according to the Turing principles. Here, we explore the possible roles of GJs in Turing-type patterning using feather pattern formation as a model. We found 7 of the 12 investigated GJ isoforms are highly dynamically expressed in the developing chicken skin. In ovo functional perturbations of the GJ isoform, connexin 30, by siRNA and the dominant-negative mutant applied before placode development led to disrupted primary feather bud formation. Interestingly, inhibition of gap junctional intercellular communication (GJIC) in the ex vivo skin explant culture allowed the sequential emergence of new feather buds at specific spatial locations relative to the existing primary buds. The results suggest that GJIC may facilitate the propagation of long-distance inhibitory signals. Thus, inhibition of GJs may stimulate Turing-type periodic feather pattern formation during chick skin development, and the removal of GJ activity would enable the emergence of new feather buds if the local environment were competent and the threshold to form buds was reached. We further propose Turing-based computational simulations that can predict the sequential appearance of these ectopic buds. Our models demonstrate how a Turing activator-inhibitor system can continue to generate patterns in the competent morphogenetic field when the level of intercellular communication at the tissue scale is modulated.


Asunto(s)
Comunicación Celular , Plumas , Uniones Comunicantes , Animales , Uniones Comunicantes/metabolismo , Plumas/crecimiento & desarrollo , Plumas/metabolismo , Embrión de Pollo , Conexinas/metabolismo , Conexinas/genética , Tipificación del Cuerpo/fisiología , Pollos , Piel/metabolismo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética
5.
Proc Natl Acad Sci U S A ; 120(36): e2221982120, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37643215

RESUMEN

Stem cells in organoids self-organize into tissue patterns with unknown mechanisms. Here, we use skin organoids to analyze this process. Cell behavior videos show that the morphological transformation from multiple spheroidal units with morphogenesis competence (CMU) to planar skin is characterized by two abrupt cell motility-increasing events before calming down. The self-organizing processes are controlled by a morphogenetic module composed of molecular sensors, modulators, and executers. Increasing dermal stiffness provides the initial driving force (driver) which activates Yap1 (sensor) in epidermal cysts. Notch signaling (modulator 1) in epidermal cyst tunes the threshold of Yap1 activation. Activated Yap1 induces Wnts and MMPs (epidermal executers) in basal cells to facilitate cellular flows, allowing epidermal cells to protrude out from the CMU. Dermal cell-expressed Rock (dermal executer) generates a stiff force bridge between two CMU and accelerates tissue mixing via activating Laminin and ß1-integrin. Thus, this self-organizing coalescence process is controlled by a mechano-chemical circuit. Beyond skin, self-organization in organoids may use similar mechano-chemical circuit structures.


Asunto(s)
Epidermis , Piel , Personalidad , Organoides , Emociones , Proteínas Adaptadoras Transductoras de Señales
6.
Development ; 148(18)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34344024

RESUMEN

How dermis maintains tissue homeostasis in cyclic growth and wounding is a fundamental unsolved question. Here, we study how dermal components of feather follicles undergo physiological (molting) and plucking injury-induced regeneration in chickens. Proliferation analyses reveal quiescent, transient-amplifying (TA) and long-term label-retaining dermal cell (LRDC) states. During the growth phase, LRDCs are activated to make new dermal components with distinct cellular flows. Dermal TA cells, enriched in the proximal follicle, generate both peripheral pulp, which extends distally to expand the epithelial-mesenchymal interactive interface for barb patterning, and central pulp, which provides nutrition. Entering the resting phase, LRDCs, accompanying collar bulge epidermal label-retaining cells, descend to the apical dermal papilla. In the next cycle, these apical dermal papilla LRDCs are re-activated to become new pulp progenitor TA cells. In the growth phase, lower dermal sheath can generate dermal papilla and pulp. Transcriptome analyses identify marker genes and highlight molecular signaling associated with dermal specification. We compare the cyclic topological changes with those of the hair follicle, a convergently evolved follicle configuration. This work presents a model for analyzing homeostasis and tissue remodeling of mesenchymal progenitors.


Asunto(s)
Pollos/fisiología , Dermis/fisiología , Células Epidérmicas/fisiología , Plumas/fisiología , Folículo Piloso/fisiología , Regeneración/fisiología , Células Madre/fisiología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Cabello/fisiología , Muda/fisiología , Transducción de Señal/fisiología
7.
Proc Natl Acad Sci U S A ; 116(14): 6884-6890, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30886106

RESUMEN

Animal skin pigment patterns are excellent models to study the mechanism of biological self-organization. Theoretical approaches developed mathematical models of pigment patterning and molecular genetics have brought progress; however, the responsible cellular mechanism is not fully understood. One long unsolved controversy is whether the patterning information is autonomously determined by melanocytes or nonautonomously determined from the environment. Here, we transplanted purified melanocytes and demonstrated that melanocytes could form periodic pigment patterns cell autonomously. Results of heterospecific transplantation among quail strains are consistent with this finding. Further, we observe that developing melanocytes directly connect with each other via filopodia to form a network in culture and in vivo. This melanocyte network is reminiscent of zebrafish pigment cell networks, where connexin is implicated in stripe formation via genetic studies. Indeed, we found connexin40 (cx40) present on developing melanocytes in birds. Stripe patterns can form in quail skin explant cultures. Several calcium channel modulators can enhance or suppress pigmentation globally, but a gap junction inhibitor can change stripe patterning. Most interestingly, in ovo, misexpression of dominant negative cx40 expands the black region, while overexpression of cx40 expands the yellow region. Subsequently, melanocytes instruct adjacent dermal cells to express agouti signaling protein (ASIP), the regulatory factor for pigment switching, which promotes pheomelanin production. Thus, we demonstrate Japanese quail melanocytes have an autonomous periodic patterning role during body pigment stripe formation. We also show dermal agouti stripes and how the coupling of melanocytes with dermal cells may confer stable and distinct pigment stripe patterns.


Asunto(s)
Pollos/metabolismo , Codorniz/metabolismo , Pigmentación de la Piel/fisiología , Piel/metabolismo , Animales , Proteínas Aviares/metabolismo , Embrión de Pollo , Conexinas/metabolismo , Melanocitos/citología , Piel/citología , Proteína alfa-5 de Unión Comunicante
8.
Proc Natl Acad Sci U S A ; 116(22): 10858-10867, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31072931

RESUMEN

Networked structures integrate numerous elements into one functional unit, while providing a balance between efficiency, robustness, and flexibility. Understanding how biological networks self-assemble will provide insights into how these features arise. Here, we demonstrate how nature forms exquisite muscle networks that can repair, regenerate, and adapt to external perturbations using the feather muscle network in chicken embryos as a paradigm. The self-assembled muscle networks arise through the implementation of a few simple rules. Muscle fibers extend outward from feather buds in every direction, but only those muscle fibers able to connect to neighboring buds are eventually stabilized. After forming such a nearest-neighbor configuration, the network can be reconfigured, adapting to perturbed bud arrangement or mechanical cues. Our computational model provides a bioinspired algorithm for network self-assembly, with intrinsic or extrinsic cues necessary and sufficient to guide the formation of these regenerative networks. These robust principles may serve as a useful guide for assembling adaptive networks in other contexts.


Asunto(s)
Aves/crecimiento & desarrollo , Tipificación del Cuerpo/fisiología , Plumas/crecimiento & desarrollo , Modelos Biológicos , Desarrollo de Músculos/fisiología , Algoritmos , Animales , Regeneración/fisiología , Piel/crecimiento & desarrollo
9.
Proc Natl Acad Sci U S A ; 114(34): E7101-E7110, 2017 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-28798065

RESUMEN

Organoids made from dissociated progenitor cells undergo tissue-like organization. This in vitro self-organization process is not identical to embryonic organ formation, but it achieves a similar phenotype in vivo. This implies genetic codes do not specify morphology directly; instead, complex tissue architectures may be achieved through several intermediate layers of cross talk between genetic information and biophysical processes. Here we use newborn and adult skin organoids for analyses. Dissociated cells from newborn mouse skin form hair primordia-bearing organoids that grow hairs robustly in vivo after transplantation to nude mice. Detailed time-lapse imaging of 3D cultures revealed unexpected morphological transitions between six distinct phases: dissociated cells, cell aggregates, polarized cysts, cyst coalescence, planar skin, and hair-bearing skin. Transcriptome profiling reveals the sequential expression of adhesion molecules, growth factors, Wnts, and matrix metalloproteinases (MMPs). Functional perturbations at different times discern their roles in regulating the switch from one phase to another. In contrast, adult cells form small aggregates, but then development stalls in vitro. Comparative transcriptome analyses suggest suppressing epidermal differentiation in adult cells is critical. These results inspire a strategy that can restore morphological transitions and rescue the hair-forming ability of adult organoids: (i) continuous PKC inhibition and (ii) timely supply of growth factors (IGF, VEGF), Wnts, and MMPs. This comprehensive study demonstrates that alternating molecular events and physical processes are in action during organoid morphogenesis and that the self-organizing processes can be restored via environmental reprogramming. This tissue-level phase transition could drive self-organization behavior in organoid morphogenies beyond the skin.


Asunto(s)
Cabello/fisiología , Organoides/fisiología , Animales , Animales Recién Nacidos , Femenino , Cabello/enzimología , Cabello/crecimiento & desarrollo , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Desnudos , Morfogénesis , Organoides/enzimología , Organoides/crecimiento & desarrollo , Regeneración , Piel/enzimología , Piel/crecimiento & desarrollo , Fenómenos Fisiológicos de la Piel , Células Madre/fisiología
10.
Mol Biol Evol ; 35(2): 417-430, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29177513

RESUMEN

The origin of feathers is an important question in Evo-Devo studies, with the eventual evolution of vaned feathers which are aerodynamic, allowing feathered dinosaurs and early birds to fly and venture into new ecological niches. Studying how feathers and scales are developmentally specified provides insight into how a new organ may evolve. We identified feather-associated genes using genomic analyses. The candidate genes were tested by expressing them in chicken and alligator scale forming regions. Ectopic expression of these genes induced intermediate morphotypes between scales and feathers which revealed several major morphogenetic events along this path: Localized growth zone formation, follicle invagination, epithelial branching, feather keratin differentiation, and dermal papilla formation. In addition to molecules known to induce feathers on scales (retinoic acid, ß-catenin), we identified novel scale-feather converters (Sox2, Zic1, Grem1, Spry2, Sox18) which induce one or more regulatory modules guiding these morphogenetic events. Some morphotypes resemble filamentous appendages found in feathered dinosaur fossils, whereas others exhibit characteristics of modern avian feathers. We propose these morpho-regulatory modules were used to diversify archosaur scales and to initiate feather evolution. The regulatory combination and hierarchical integration may have led to the formation of extant feather forms. Our study highlights the importance of integrating discoveries between developmental biology and paleontology.


Asunto(s)
Escamas de Animales , Evolución Biológica , Plumas , Morfogénesis/genética , Caimanes y Cocodrilos , Animales , Embrión de Pollo , Perfilación de la Expresión Génica , Genómica , Fenotipo , Factores de Transcripción , beta-Queratinas/genética , beta-Queratinas/metabolismo
11.
Dev Growth Differ ; 61(1): 124-138, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30569461

RESUMEN

Many animals can change the size, shape, texture and color of their regenerated coats in response to different ages, sexes, or seasonal environmental changes. Here, we propose that the feather core branching morphogenesis module can be regulated by sex hormones or other environmental factors to change feather forms, textures or colors, thus generating a large spectrum of complexity for adaptation. We use sexual dimorphisms of the chicken to explore the role of hormones. A long-standing question is whether the sex-dependent feather morphologies are autonomously controlled by the male or female cell types, or extrinsically controlled and reversible. We have recently identified core feather branching molecular modules which control the anterior-posterior (bone morphogenetic orotein [BMP], Wnt gradient), medio-lateral (Retinoic signaling, Gremlin), and proximo-distal (Sprouty, BMP) patterning of feathers. We hypothesize that morpho-regulation, through quantitative modulation of existing parameters, can act on core branching modules to topologically tune the dimension of each parameter during morphogenesis and regeneration. Here, we explore the involvement of hormones in generating sexual dimorphisms using exogenously delivered hormones. Our strategy is to mimic male androgen levels by applying exogenous dihydrotestosterone and aromatase inhibitors to adult females and to mimic female estradiol levels by injecting exogenous estradiol to adult males. We also examine differentially expressed genes in the feathers of wildtype male and female chickens to identify potential downstream modifiers of feather morphogenesis. The data show male and female feather morphology and their color patterns can be modified extrinsically through molting and resetting the stem cell niche during regeneration.


Asunto(s)
Plumas/crecimiento & desarrollo , Plumas/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Morfogénesis/genética , Animales , Pollos , Femenino , Masculino , Caracteres Sexuales
12.
Exp Dermatol ; 28(4): 480-484, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30916811

RESUMEN

Keloids are wounding-induced fibroproliferative human tumor-like skin scars of complex genetic makeup and poorly defined pathogenesis. To reveal dynamic epigenetic and transcriptome changes of keloid fibroblasts, we performed RNA-seq and ATAC-seq analysis on an early passage keloid fibroblast cell strain and its paired normal control fibroblasts. This keloid strain produced keloid-like scars in a plasma clot-based skin equivalent humanized keloid animal model. RNA-seq analysis reveals gene ontology terms including hepatic fibrosis, Wnt-ß-catenin, TGF-ß, regulation of epithelial-mesenchymal transition (EMT), STAT3 and adherens junction. ATAC-seq analysis suggests STAT3 signalling is the most significantly enriched gene ontology term in keloid fibroblasts, followed by Wnt signalling (Wnt5) and regulation of the EMT pathway. Immunohistochemistry confirms that STAT3 (Tyr705 phospho-STAT3) is activated and ß-catenin is up-regulated in the dermis of keloid clinical specimens and keloid skin equivalent implants from the humanized mouse model. A non-linear dose-response of cucurbitacin I, a selective JAK2/STAT3 inhibitor, in collagen type I expression of keloid-derived plasma clot-based skin equivalents implicates a likely role of STAT3 signalling in keloid pathogenesis. This work also demonstrates the utility of the recently established humanized keloid mouse model in exploring the mechanism of keloid formation.


Asunto(s)
Queloide/etiología , Queloide/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Humanos , Ratones , Transcriptoma , Vía de Señalización Wnt
13.
BMC Genomics ; 19(1): 780, 2018 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-30373532

RESUMEN

BACKGROUND: The molecular mechanism controlling regional specific skin appendage phenotypes is a fundamental question that remains unresolved. We recently identified feather and scale primordium associated genes and with functional studies, proposed five major modules are involved in scale-to-feather conversion and their integration is essential to form today's feathers. Yet, how the molecular networks are wired and integrated at the genomic level is still unknown. RESULTS: Here, we combine classical recombination experiments and systems biology technology to explore the molecular mechanism controlling cell fate specification. In the chimeric explant, dermal fate is more stable, while epidermal fate is reprogrammed to be similar to the original appendage type of the mesenchyme. We analyze transcriptome changes in both scale-to-feather and feather-to-scale transition in the epidermis. We found a highly interconnected regulatory gene network controlling skin appendage types. These gene networks are organized around two molecular hubs, ß-catenin and retinoic acid (RA), which can bind to regulatory elements controlling downstream gene expression, leading to scale or feather fates. ATAC sequencing analyses revealed about 1000 altered widely distributed chromatin open sites. We find that perturbation of a key gene alters the expression of many other co-expressed genes in the same module. CONCLUSIONS: Our findings suggest that these feather / scale fate specification genes form an interconnected network and rewiring of the gene network can lead to changes of appendage phenotypes, acting similarly to endogenous reprogramming at the tissue level. This work shows that key hub molecules, ß-catenin and retinoic acid, regulate scale / feather fate specification gene networks, opening up new possibilities to understand the switches controlling organ phenotypes in a two component (epithelial and mesenchyme) system.


Asunto(s)
Plumas , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Fenotipo , Piel , Transcriptoma , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Especificidad de Órganos/genética , Elementos de Respuesta , Tretinoina/farmacología
14.
Proc Natl Acad Sci U S A ; 112(49): E6770-9, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26598683

RESUMEN

Avian integumentary organs include feathers, scales, claws, and beaks. They cover the body surface and play various functions to help adapt birds to diverse environments. These keratinized structures are mainly composed of corneous materials made of α-keratins, which exist in all vertebrates, and ß-keratins, which only exist in birds and reptiles. Here, members of the keratin gene families were used to study how gene family evolution contributes to novelty and adaptation, focusing on tissue morphogenesis. Using chicken as a model, we applied RNA-seq and in situ hybridization to map α- and ß-keratin genes in various skin appendages at embryonic developmental stages. The data demonstrate that temporal and spatial α- and ß-keratin expression is involved in establishing the diversity of skin appendage phenotypes. Embryonic feathers express a higher proportion of ß-keratin genes than other skin regions. In feather filament morphogenesis, ß-keratins show intricate complexity in diverse substructures of feather branches. To explore functional interactions, we used a retrovirus transgenic system to ectopically express mutant α- or antisense ß-keratin forms. α- and ß-keratins show mutual dependence and mutations in either keratin type results in disrupted keratin networks and failure to form proper feather branches. Our data suggest that combinations of α- and ß-keratin genes contribute to the morphological and structural diversity of different avian skin appendages, with feather-ß-keratins conferring more possible composites in building intrafeather architecture complexity, setting up a platform of morphological evolution of functional forms in feathers.


Asunto(s)
Evolución Biológica , Mapeo Cromosómico , Queratinas/genética , Piel/embriología , beta-Queratinas/genética , Animales , Embrión de Pollo , Hibridación in Situ , Queratina-13/genética , ARN sin Sentido/farmacología , Piel/metabolismo
15.
Proc Natl Acad Sci U S A ; 110(4): 1351-6, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23292934

RESUMEN

Hair follicles facilitate the study of stem cell behavior because stem cells in progressive activation stages, ordered within the follicle architecture, are capable of cyclic regeneration. To study the gene network governing the homeostasis of hair bulge stem cells, we developed a Keratin 15-driven genetic model to directly perturb molecular signaling in the stem cells. We visualize the behavior of these modified stem cells, evaluating their hair-regenerating ability and profile their molecular expression. Bone morphogenetic protein (BMP)-inactivated stem cells exhibit molecular profiles resembling those of hair germs, yet still possess multipotentiality in vivo. These cells also exhibit up-regulation of Wnt7a, Wnt7b, and Wnt16 ligands and Frizzled (Fzd) 10 receptor. We demonstrate direct transcriptional modulation of the Wnt7a promoter. These results highlight a previously unknown intra-stem cell antagonistic competition, between BMP and Wnt signaling, to balance stem cell activity. Reduced BMP signaling and increased Wnt signaling tilts each stem cell toward a hair germ fate and, vice versa, based on a continuous scale dependent on the ratio of BMP/Wnt activity. This work reveals one more hierarchical layer regulating stem cell homeostasis beneath the stem cell-dermal papilla-based epithelial-mesenchymal interaction layer and the hair follicle-intradermal adipocyte-based tissue interaction layer. Although hierarchical layers are all based on BMP/Wnt signaling, the multilayered control ensures that all information is taken into consideration and allows hair stem cells to sum up the total activators/inhibitors involved in making the decision of activation.


Asunto(s)
Células Madre Adultas/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Redes Reguladoras de Genes , Folículo Piloso/citología , Folículo Piloso/metabolismo , Vía de Señalización Wnt , Células Madre Adultas/citología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/deficiencia , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Cabello/crecimiento & desarrollo , Folículo Piloso/efectos de los fármacos , Homeostasis/genética , Queratina-15/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Modelos Genéticos , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Regiones Promotoras Genéticas , Transcriptoma , Proteínas Wnt/administración & dosificación
16.
Proc Natl Acad Sci U S A ; 110(16): E1452-61, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23576731

RESUMEN

How organs are shaped to specific forms is a fundamental issue in developmental biology. To address this question, we used the repetitive, periodic pattern of feather morphogenesis on chicken skin as a model. Avian feathers within a single tract extend from dome-shaped primordia to thin conical structures with a common axis of orientation. From a systems biology perspective, the process is precise and robust. Using tissue transplantation assays, we demonstrate that a "zone of polarizing activity," localized in the posterior feather bud, is necessary and sufficient to mediate the directional elongation. This region contains a spatially well-defined nuclear ß-catenin zone, which is induced by wingless-int (Wnt)7a protein diffusing in from posterior bud epithelium. Misexpressing nuclear ß-catenin randomizes feather polarity. This dermal nuclear ß-catenin zone, surrounded by Notch1 positive dermal cells, induces Jagged1. Inhibition of Notch signaling disrupts the spatial configuration of the nuclear ß-catenin zone and leads to randomized feather polarity. Mathematical modeling predicts that lateral inhibition, mediated by Notch signaling, functions to reduce Wnt7a gradient variations and fluctuations to form the sharp boundary observed for the dermal ß-catenin zone. This zone is also enriched for nonmuscle myosin IIB. Suppressing nonmuscle myosin IIB disrupts directional cell rearrangements and abolishes feather bud elongation. These data suggest that a unique molecular module involving chemical-mechanical coupling converts a pliable chemical gradient to a precise domain, ready for subsequent mechanical action, thus defining the position, boundary, and duration of localized morphogenetic activity that molds the shape of growing organs.


Asunto(s)
Proteínas Aviares/metabolismo , Polaridad Celular/fisiología , Plumas/embriología , Morfogénesis/fisiología , Miosina Tipo IIB no Muscular/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , Animales , Bromodesoxiuridina , Embrión de Pollo , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Electroporación , Hibridación in Situ , Modelos Biológicos , Simulación de Dinámica Molecular , beta Catenina/metabolismo
17.
Proc Natl Acad Sci U S A ; 110(22): E2009-18, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23671090

RESUMEN

Reptiles and fish have robust regenerative powers for tooth renewal. However, extant mammals can either renew their teeth one time (diphyodont dentition) or not at all (monophyodont dentition). Humans replace their milk teeth with permanent teeth and then lose their ability for tooth renewal. Here, we study tooth renewal in a crocodilian model, the American alligator, which has well-organized teeth similar to mammals but can still undergo life-long renewal. Each alligator tooth is a complex family unit composed of the functional tooth, successional tooth, and dental lamina. Using multiple mitotic labeling, we map putative stem cells to the distal enlarged bulge of the dental lamina that contains quiescent odontogenic progenitors that can be activated during physiological exfoliation or artificial extraction. Tooth cycle initiation correlates with ß-catenin activation and soluble frizzled-related protein 1 disappearance in the bulge. The dermal niche adjacent to the dermal lamina dynamically expresses neural cell adhesion molecule, tenascin-C, and other molecules. Furthermore, in development, asymmetric ß-catenin localization leads to the formation of a heterochronous and complex tooth family unit configuration. Understanding how these signaling molecules interact in tooth development in this model may help us to learn how to stimulate growth of adult teeth in mammals.


Asunto(s)
Modelos Animales , Modelos Biológicos , Odontogénesis/fisiología , Regeneración/fisiología , Transducción de Señal/fisiología , Células Madre/fisiología , Diente/fisiología , Caimanes y Cocodrilos , Animales , Bromodesoxiuridina , Proliferación Celular , Glicoproteínas/metabolismo , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Tenascina/metabolismo , Diente/citología , Microtomografía por Rayos X , beta Catenina/metabolismo
18.
Dev Dyn ; 244(8): 905-20, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25858668

RESUMEN

BACKGROUND: How tissue patterns form in development and regeneration is a fundamental issue remaining to be fully understood. The integument often forms repetitive units in space (periodic patterning) and time (cyclic renewal), such as feathers and hairs. Integument patterns are visible and experimentally manipulatable, helping us reveal pattern formative processes. Variability is seen in regional phenotypic specificities and temporal cycling at different physiological stages. RESULTS: Here we show some cellular/molecular bases revealed by analyzing integument patterns. (1) Localized cellular activity (proliferation, rearrangement, apoptosis, differentiation) transforms prototypic organ primordia into specific shapes. Combinatorial positioning of different localized activity zones generates diverse and complex organ forms. (2) Competitive equilibrium between activators and inhibitors regulates stem cells through cyclic quiescence and activation. CONCLUSIONS: Dynamic interactions between stem cells and their adjacent niche regulate regenerative behavior, modulated by multi-layers of macro-environmental factors (dermis, body hormone status, and external environment). Genomics studies may reveal how positional information of localized cellular activity is stored. In vivo skin imaging and lineage tracing unveils new insights into stem cell plasticity. Principles of self-assembly obtained from the integumentary organ model can be applied to help restore damaged patterns during regenerative wound healing and for tissue engineering to rebuild tissues. Developmental Dynamics 244:905-920, 2015. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Morfogénesis/fisiología , Animales , Plumas/citología , Plumas/metabolismo , Genómica , Cabello/citología , Cabello/metabolismo , Piel/citología , Piel/metabolismo , Células Madre/citología , Células Madre/metabolismo , Biología de Sistemas
19.
Dev Biol ; 387(2): 167-78, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24463139

RESUMEN

Avian feathers have robust growth and regeneration capability. To evaluate the contribution of signaling molecules and pathways in these processes, we profiled gene expression in the feather follicle using an absolute quantification approach. We identified hundreds of genes that mark specific components of the feather follicle: the dermal papillae (DP) which controls feather regeneration and axis formation, the pulp mesenchyme (Pp) which is derived from DP cells and nourishes the feather follicle, and the ramogenic zone epithelium (Erz) where a feather starts to branch. The feather DP is enriched in BMP/TGF-ß signaling molecules and inhibitors for Wnt signaling including Dkk2/Frzb. Wnt ligands are mainly expressed in the feather epithelium and pulp. We find that while Wnt signaling is required for the maintenance of DP marker gene expression and feather regeneration, excessive Wnt signaling delays regeneration and reduces pulp formation. Manipulating Dkk2/Frzb expression by lentiviral-mediated overexpression, shRNA-knockdown, or by antibody neutralization resulted in dual feather axes formation. Our results suggest that the Wnt signaling in the proximal feather follicle is fine-tuned to accommodate feather regeneration and axis formation.


Asunto(s)
Plumas/fisiología , Glicoproteínas/metabolismo , Regeneración/genética , Animales , Anticuerpos Neutralizantes/inmunología , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/inmunología , Pollos , Epitelio/metabolismo , Perfilación de la Expresión Génica , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intracelular , Mesodermo/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Piel/embriología , Factor de Crecimiento Transformador beta/metabolismo , Vía de Señalización Wnt , Proteína Wnt3A/metabolismo , Xenopus laevis
20.
Dig Dis Sci ; 60(12): 3669-80, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26173507

RESUMEN

BACKGROUND: Because the molecular mechanisms of morphogenesis of the hepatic cord and sinus are unclear, we investigated the involvement of bone morphogenetic protein (BMP4) in hepatic sinusoid morphogenesis. METHODS: We used embryonic chicken livers, which develop rapidly, as our model, and investigated expression of BMP-related genes. BMP4 activity was manipulated by overexpressing BMP4 and its antagonist, noggin. RESULTS: During hepatic cord morphogenesis, BMP4 and its receptors are expressed in both peri-sinusoidal cells and hepatoblasts as the sinusoids form, whereas noggin is expressed transiently in peri-sinusoidal cells at early stages. Suppression of BMP activity with noggin overexpression disrupted normal hepatic sinusoid structure, leading to liver congestion, failure of fibronectin deposition, and markedly reduced numbers of peri-sinusoidal cells. However, overexpression of BMP did not change sinusoidal morphology but increased endothelial cell number. Noggin overexpression resulted in disrupted cord organization, and dilated sinusoidal space, eventually leading to increased apoptosis and failed hepatocyte differentiation. CONCLUSIONS: Our results show that proper BMP signaling mediates peri-sinusoidal cell-hepatoblast interactions during development; this is essential for hepatic cord organization among hepatoblasts, endothelium, and presumptive hepatic stellate cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/metabolismo , Células Estrelladas Hepáticas/fisiología , Hígado/embriología , Hígado/metabolismo , Células Madre Mesenquimatosas/fisiología , Animales , Proteínas Portadoras/genética , Comunicación Celular , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA