Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
EMBO J ; 41(17): e108780, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35815410

RESUMEN

Schwann cell precursors (SCPs) are nerve-associated progenitors that can generate myelinating and non-myelinating Schwann cells but also are multipotent like the neural crest cells from which they originate. SCPs are omnipresent along outgrowing peripheral nerves throughout the body of vertebrate embryos. By using single-cell transcriptomics to generate a gene expression atlas of the entire neural crest lineage, we show that early SCPs and late migratory crest cells have similar transcriptional profiles characterised by a multipotent "hub" state containing cells biased towards traditional neural crest fates. SCPs keep diverging from the neural crest after being primed towards terminal Schwann cells and other fates, with different subtypes residing in distinct anatomical locations. Functional experiments using CRISPR-Cas9 loss-of-function further show that knockout of the common "hub" gene Sox8 causes defects in neural crest-derived cells along peripheral nerves by facilitating differentiation of SCPs towards sympathoadrenal fates. Finally, specific tumour populations found in melanoma, neurofibroma and neuroblastoma map to different stages of SCP/Schwann cell development. Overall, SCPs resemble migrating neural crest cells that maintain multipotency and become transcriptionally primed towards distinct lineages.


Asunto(s)
Cresta Neural , Células de Schwann , Diferenciación Celular/fisiología , Neurogénesis/fisiología , Nervios Periféricos , Células de Schwann/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(30): 17854-17863, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32647059

RESUMEN

Pacemaker neurons exert control over neuronal circuit function by their intrinsic ability to generate rhythmic bursts of action potential. Recent work has identified rhythmic gut contractions in human, mice, and hydra to be dependent on both neurons and the resident microbiota. However, little is known about the evolutionary origin of these neurons and their interaction with microbes. In this study, we identified and functionally characterized prototypical ANO/SCN/TRPM ion channel-expressing pacemaker cells in the basal metazoan Hydra by using a combination of single-cell transcriptomics, immunochemistry, and functional experiments. Unexpectedly, these prototypical pacemaker neurons express a rich set of immune-related genes mediating their interaction with the microbial environment. Furthermore, functional experiments gave a strong support to a model of the evolutionary emergence of pacemaker cells as neurons using components of innate immunity to interact with the microbial environment and ion channels to generate rhythmic contractions.


Asunto(s)
Relojes Biológicos , Hydra/fisiología , Microbiota , Neuronas/fisiología , Potenciales de Acción , Animales , Evolución Biológica , Análisis por Conglomerados , Biología Computacional/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Ratones
3.
Cell Mol Life Sci ; 78(16): 6033-6049, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34274976

RESUMEN

Melanocytes are pigmented cells residing mostly in the skin and hair follicles of vertebrates, where they contribute to colouration and protection against UV-B radiation. However, the spectrum of their functions reaches far beyond that. For instance, these pigment-producing cells are found inside the inner ear, where they contribute to the hearing function, and in the heart, where they are involved in the electrical conductivity and support the stiffness of cardiac valves. The embryonic origin of such extracutaneous melanocytes is not clear. We took advantage of lineage-tracing experiments combined with 3D visualizations and gene knockout strategies to address this long-standing question. We revealed that Schwann cell precursors are recruited from the local innervation during embryonic development and give rise to extracutaneous melanocytes in the heart, brain meninges, inner ear, and other locations. In embryos with a knockout of the EdnrB receptor, a condition imitating Waardenburg syndrome, we observed only nerve-associated melanoblasts, which failed to detach from the nerves and to enter the inner ear. Finally, we looked into the evolutionary aspects of extracutaneous melanocytes and found that pigment cells are associated mainly with nerves and blood vessels in amphibians and fish. This new knowledge of the nerve-dependent origin of extracutaneous pigment cells might be directly relevant to the formation of extracutaneous melanoma in humans.


Asunto(s)
Encéfalo/fisiología , Oído Interno/fisiología , Corazón/fisiología , Meninges/fisiología , Sistema Nervioso/fisiopatología , Células de Schwann/fisiología , Anfibios/metabolismo , Anfibios/fisiología , Animales , Encéfalo/metabolismo , Linaje de la Célula/fisiología , Oído Interno/metabolismo , Desarrollo Embrionario/fisiología , Femenino , Peces/metabolismo , Peces/fisiología , Melanocitos/metabolismo , Melanocitos/fisiología , Meninges/metabolismo , Ratones , Sistema Nervioso/metabolismo , Embarazo , Receptor de Endotelina B/metabolismo , Células de Schwann/metabolismo
4.
Proc Natl Acad Sci U S A ; 116(30): 15068-15073, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31285319

RESUMEN

Immature multipotent embryonic peripheral glial cells, the Schwann cell precursors (SCPs), differentiate into melanocytes, parasympathetic neurons, chromaffin cells, and dental mesenchymal populations. Here, genetic lineage tracing revealed that, during murine embryonic development, some SCPs detach from nerve fibers to become mesenchymal cells, which differentiate further into chondrocytes and mature osteocytes. This occurred only during embryonic development, producing numerous craniofacial and trunk skeletal elements, without contributing to development of the appendicular skeleton. Formation of chondrocytes from SCPs also occurred in zebrafish, indicating evolutionary conservation. Our findings reveal multipotency of SCPs, providing a developmental link between the nervous system and skeleton.


Asunto(s)
Huesos/citología , Linaje de la Célula/genética , Condrocitos/citología , Células Madre Mesenquimatosas/citología , Tejido Nervioso/citología , Células de Schwann/citología , Animales , Biomarcadores/metabolismo , Huesos/embriología , Huesos/metabolismo , Diferenciación Celular , Condrocitos/metabolismo , Células Cromafines/citología , Células Cromafines/metabolismo , Embrión de Mamíferos , Embrión no Mamífero , Desarrollo Embrionario , Expresión Génica , Melanocitos/citología , Melanocitos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Fibras Nerviosas/metabolismo , Tejido Nervioso/embriología , Tejido Nervioso/metabolismo , Cresta Neural/citología , Cresta Neural/crecimiento & desarrollo , Cresta Neural/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Osteocitos/citología , Osteocitos/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Células de Schwann/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
5.
Dev Dyn ; 250(9): 1300-1317, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33511716

RESUMEN

BACKGROUND: In vertebrates, the skull evolves from a complex network of dermal bones and cartilage-the latter forming the pharyngeal apparatus and the chondrocranium. Squamates are particularly important in this regard as they maintain at least part of the chondrocranium throughout their whole ontogeny until adulthood. Anguid lizards represent a unique group of squamates, which contains limbed and limbless forms and show conspicuous variation of the adult skull. RESULTS: Based on several emboadryonic stages of the limbless lizards Pseudopus apodus and Anguis fragilis, and by comparing with other squamates, we identified and interpreted major differences in chondrocranial anatomy. Among others, the most important differences are in the orbitotemporal region. P. apodus shows a strikingly similar development of this region to other squamates. Unexpectedly, however, A. fragilis differs considerably in the composition of the orbitotemporal region. In addition, A. fragilis retains a paedomorphic state of the nasal region. CONCLUSIONS: Taxonomic comparisons indicate that even closely related species with reduced limbs show significant differences in chondrocranial anatomy. The Pearson correlation coefficient suggests strong correlation between chondrocranial reduction and limb reduction. We pose the hypothesis that limb reduction could be associated with the reduction in chondrocrania by means of genetic mechanisms.


Asunto(s)
Evolución Biológica , Lagartos , Animales , Cartílago , Extremidades , Cráneo/anatomía & histología
6.
Semin Cell Dev Biol ; 91: 2-12, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-29248472

RESUMEN

Chrondrocranium, the cartilaginous skull, is one of the major innovations that underlie evolution of the vertebrate head. Control of the induction and shaping of the cartilage is a key for the formation of the facial bones and largely defines facial shape. The appearance of cartilage in the head enabled many new functions such as protection of central nervous system and sensory structures, support of the feeding apparatus and formation of muscle attachment points ensuring faster and coordinated jaw movements. Here we review the evolution of cartilage in the cranial region and discuss shaping of the chondrocranium in different groups of vertebrates.


Asunto(s)
Evolución Biológica , Cartílago/embriología , Huesos Faciales/embriología , Anfioxos/embriología , Cráneo/embriología , Vertebrados/embriología , Animales , Cartílago/anatomía & histología , Cartílago/crecimiento & desarrollo , Huesos Faciales/anatomía & histología , Huesos Faciales/crecimiento & desarrollo , Humanos , Anfioxos/anatomía & histología , Anfioxos/crecimiento & desarrollo , Modelos Biológicos , Cráneo/anatomía & histología , Cráneo/crecimiento & desarrollo , Vertebrados/anatomía & histología , Vertebrados/crecimiento & desarrollo
7.
Nature ; 513(7519): 551-4, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25079316

RESUMEN

Mesenchymal stem cells occupy niches in stromal tissues where they provide sources of cells for specialized mesenchymal derivatives during growth and repair. The origins of mesenchymal stem cells have been the subject of considerable discussion, and current consensus holds that perivascular cells form mesenchymal stem cells in most tissues. The continuously growing mouse incisor tooth offers an excellent model to address the origin of mesenchymal stem cells. These stem cells dwell in a niche at the tooth apex where they produce a variety of differentiated derivatives. Cells constituting the tooth are mostly derived from two embryonic sources: neural crest ectomesenchyme and ectodermal epithelium. It has been thought for decades that the dental mesenchymal stem cells giving rise to pulp cells and odontoblasts derive from neural crest cells after their migration in the early head and formation of ectomesenchymal tissue. Here we show that a significant population of mesenchymal stem cells during development, self-renewal and repair of a tooth are derived from peripheral nerve-associated glia. Glial cells generate multipotent mesenchymal stem cells that produce pulp cells and odontoblasts. By combining a clonal colour-coding technique with tracing of peripheral glia, we provide new insights into the dynamics of tooth organogenesis and growth.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Incisivo/citología , Células Madre Mesenquimatosas/citología , Neuroglía/citología , Animales , Rastreo Celular , Células Clonales/citología , Pulpa Dental/citología , Femenino , Incisivo/embriología , Masculino , Ratones , Modelos Biológicos , Cresta Neural/citología , Odontoblastos/citología , Regeneración , Células de Schwann/citología
8.
Dev Biol ; 444 Suppl 1: S308-S324, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29807017

RESUMEN

Carotid body glomus cells mediate essential reflex responses to arterial blood hypoxia. They are dopaminergic and secrete growth factors that support dopaminergic neurons, making the carotid body a potential source of patient-specific cells for Parkinson's disease therapy. Like adrenal chromaffin cells, which are also hypoxia-sensitive, glomus cells are neural crest-derived and require the transcription factors Ascl1 and Phox2b; otherwise, their development is little understood at the molecular level. Here, analysis in chicken and mouse reveals further striking molecular parallels, though also some differences, between glomus and adrenal chromaffin cell development. Moreover, histology has long suggested that glomus cell precursors are 'émigrés' from neighbouring ganglia/nerves, while multipotent nerve-associated glial cells are now known to make a significant contribution to the adrenal chromaffin cell population in the mouse. We present conditional genetic lineage-tracing data from mice supporting the hypothesis that progenitors expressing the glial marker proteolipid protein 1, presumably located in adjacent ganglia/nerves, also contribute to glomus cells. Finally, we resolve a paradox for the 'émigré' hypothesis in the chicken - where the nearest ganglion to the carotid body is the nodose, in which the satellite glia are neural crest-derived, but the neurons are almost entirely placode-derived - by fate-mapping putative nodose neuronal 'émigrés' to the neural crest.


Asunto(s)
Cuerpo Carotídeo/embriología , Células Cromafines/metabolismo , Pericitos/metabolismo , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Tipificación del Cuerpo/fisiología , Diferenciación Celular , Hipoxia de la Célula/fisiología , Embrión de Pollo , Pollos/metabolismo , Ratones , Ratones Noqueados , Proteína Proteolipídica de la Mielina/fisiología , Cresta Neural/metabolismo , Neuronas/metabolismo , Pericitos/fisiología , Factores de Transcripción/metabolismo
9.
Cell Commun Signal ; 13: 2, 2015 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-25627785

RESUMEN

BACKGROUND: The WNT/planar-cell-polarity (PCP) pathway is a key regulator of cell polarity and directional cell movements. Core PCP proteins such as Van Gogh-like2 (VANGL2) are evolutionarily highly conserved; however, the mammalian PCP machinery is still poorly understood mainly due to lack of suitable models and quantitative methodology. WNT/PCP has been implicated in many human diseases with the most distinguished positive role in the metastatic process, which accounts for more than 90% of cancer related deaths, and presents therefore an attractive target for pharmacological interventions. However, cellular assays for the assessment of PCP signaling, which would allow a more detailed mechanistic analysis of PCP function and possibly also high throughput screening for chemical compounds targeting mammalian PCP signaling, are still missing. RESULTS: Here we describe a mammalian cell culture model, which correlates B lymphocyte migration of patient-derived MEC1 cells and asymmetric localization of fluorescently-tagged VANGL2. We show by live cell imaging that PCP proteins are polarized in MEC1 cells and that VANGL2 polarization is controlled by the same mechanism as in tissues i.e. it is dependent on casein kinase 1 activity. In addition, destruction of the actin cytoskeleton leads to migratory arrest and cell rounding while VANGL2-EGFP remains polarized suggesting that active PCP signaling visualized by polarized distribution of VANGL2 is a cause for and not a consequence of the asymmetric shape of a migrating cell. CONCLUSIONS: The presented imaging-based methodology allows overcoming limitations of earlier approaches to study the mammalian WNT/PCP pathway, which required in vivo models and analysis of complex tissues. Our system investigating PCP-like signaling on a single-cell level thus opens new possibilities for screening of compounds, which control asymmetric distribution of proteins in the PCP pathway.


Asunto(s)
Linfocitos B/metabolismo , Movimiento Celular/inmunología , Polaridad Celular/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Proteínas de la Membrana/inmunología , Vía de Señalización Wnt/inmunología , Linfocitos B/patología , Línea Celular Tumoral , Movimiento Celular/genética , Polaridad Celular/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Proteínas de la Membrana/genética , Vía de Señalización Wnt/genética
10.
Exp Cell Res ; 321(1): 17-24, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24140263

RESUMEN

The peripheral nervous system (PNS) is complex and omnipresent. The PNS targets all parts of the body starting from early stages of embryonic development, and in large part, is derived from multipotent migratory neural crest stem cells. Current opinion mostly perceives the PNS as a means of communication and information exchange between the central nervous system, the rest of the body and the environment. Additionally, the PNS is largely associated with autonomic control. Being an "alternative brain" it provides local regulation of processes in organs. However, it has become evident in recent years that in addition to these main canonical functions the PNS possesses a number of other important roles in development and homeostasis of targeted tissues, for instance, in nerve-dependent regeneration. The PNS represents a niche that hosts neural crest-derived peripheral glial cells, or, in other words, neural crest-like multipotent cells throughout the entire body. These multipotent nerve-adjacent cells can be reprogrammed in vivo and play a number of roles from creating pigmentation to controlling regeneration of a limb in amphibians or skin in rodents. In the current review we outline newly emerged, non-canonical functions of the PNS and briefly describe cellular and molecular aspects of these alternative functions.


Asunto(s)
Diferenciación Celular , Células Madre Multipotentes/metabolismo , Regeneración Nerviosa/fisiología , Sistema Nervioso Periférico/embriología , Sistema Nervioso Periférico/metabolismo , Animales , Humanos , Células Madre Multipotentes/citología
11.
Sci Rep ; 14(1): 16088, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38997306

RESUMEN

Environmental change is frequent. To adjust and survive, animals need behavioural flexibility. Recently, cognitive flexibility has emerged as a driving force for adjusting to environmental change. Understanding how environmental factors, such as food quality, influence behavioural and/or more costly cognitive flexibility. Here, we investigate the effects of high-quality versus standard food as well as the effects of different housing conditions on both types of flexibility. Our results show that mice that experienced a poorer diet under seminatural conditions showed greater behavioural but not cognitive flexibility. For cage-housed mice, the results were less clear. However, mice fed a poorer diet performed better in innovative problem-solving, thus showing enhanced cognitive flexibility, which was not apparent in the reversal learning paradigm. The observed differences were most likely due to differences in motivation to obtain food rewards. Additionally, animals on poorer diet had lower brain volume, usually related to lower cognitive task performance at the between-species level. Thus, our study emphasises the importance of environmental conditions on behavioural flexibility at the within-species level, highlights that different test paradigms may lead to different conclusions, and finally shows that cage housing of wild animals may lead to patterns that do not necessarily reflect natural conditions.


Asunto(s)
Conducta Animal , Cognición , Calidad de los Alimentos , Animales , Cognición/fisiología , Ratones , Conducta Animal/fisiología , Masculino , Femenino , Dieta , Encéfalo/fisiología , Recompensa , Animales Salvajes/fisiología , Vivienda para Animales
12.
Philos Trans R Soc Lond B Biol Sci ; 378(1880): 20220079, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37183897

RESUMEN

Extensive morphological variation found in mammals reflects the wide spectrum of their ecological adaptations. The highest morphological diversity is present in the craniofacial region, where geometry is mainly dictated by the bony skull. Mammalian craniofacial development represents complex multistep processes governed by numerous conserved genes that require precise spatio-temporal control. A central question in contemporary evolutionary biology is how a defined set of conserved genes can orchestrate formation of fundamentally different structures, and therefore how morphological variability arises. In principle, differential gene expression patterns during development are the source of morphological variation. With the emergence of multicellular organisms, precise regulation of gene expression in time and space is attributed to cis-regulatory elements. These elements contribute to higher-order chromatin structure and together with trans-acting factors control transcriptional landscapes that underlie intricate morphogenetic processes. Consequently, divergence in cis-regulation is believed to rewire existing gene regulatory networks and form the core of morphological evolution. This review outlines the fundamental principles of the genetic code and genomic regulation interplay during development. Recent work that deepened our comprehension of cis-regulatory element origin, divergence and function is presented here to illustrate the state-of-the-art research that uncovered the principles of morphological novelty. This article is part of the theme issue 'The mammalian skull: development, structure and function'.


Asunto(s)
Evolución Molecular , Mamíferos , Animales , Mamíferos/genética , Redes Reguladoras de Genes , Cráneo , Cabeza
13.
Philos Trans R Soc Lond B Biol Sci ; 378(1880): 20220080, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37183900

RESUMEN

Mammals possess impressive craniofacial variation that mirrors their adaptation to diverse ecological niches, feeding behaviour, physiology and overall lifestyle. The spectrum of craniofacial geometries is established mainly during embryonic development. The formation of the head represents a sequence of events regulated on genomic, molecular, cellular and tissue level, with each step taking place under tight spatio-temporal control. Even minor variations in timing, position or concentration of the molecular drivers and the resulting events can affect the final shape, size and position of the skeletal elements and the geometry of the head. Our knowledge of craniofacial development increased substantially in the last decades, mainly due to research using conventional vertebrate model organisms. However, how developmental differences in head formation arise specifically within mammals remains largely unexplored. This review highlights three evolutionary mechanisms acknowledged to modify ontogenesis: heterochrony, heterotopy and heterometry. We present recent research that links changes in developmental timing, spatial organization or gene expression levels to the acquisition of species-specific skull morphologies. We highlight how these evolutionary modifications occur on the level of the genes, molecules and cellular processes, and alter conserved developmental programmes to generate a broad spectrum of skull shapes characteristic of the class Mammalia. This article is part of the theme issue 'The mammalian skull: development, structure and function'.


Asunto(s)
Cabeza , Cráneo , Animales , Evolución Biológica , Mamíferos , Especificidad de la Especie
14.
Nat Commun ; 14(1): 3060, 2023 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-37244931

RESUMEN

Formation of oriented myofibrils is a key event in musculoskeletal development. However, the mechanisms that drive myocyte orientation and fusion to control muscle directionality in adults remain enigmatic. Here, we demonstrate that the developing skeleton instructs the directional outgrowth of skeletal muscle and other soft tissues during limb and facial morphogenesis in zebrafish and mouse. Time-lapse live imaging reveals that during early craniofacial development, myoblasts condense into round clusters corresponding to future muscle groups. These clusters undergo oriented stretch and alignment during embryonic growth. Genetic perturbation of cartilage patterning or size disrupts the directionality and number of myofibrils in vivo. Laser ablation of musculoskeletal attachment points reveals tension imposed by cartilage expansion on the forming myofibers. Application of continuous tension using artificial attachment points, or stretchable membrane substrates, is sufficient to drive polarization of myocyte populations in vitro. Overall, this work outlines a biomechanical guidance mechanism that is potentially useful for engineering functional skeletal muscle.


Asunto(s)
Músculo Esquelético , Pez Cebra , Animales , Ratones , Pez Cebra/genética , Músculo Esquelético/fisiología , Miofibrillas/fisiología , Morfogénesis , Mioblastos/fisiología
15.
Nat Commun ; 14(1): 3092, 2023 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-37248239

RESUMEN

In this study we use comparative genomics to uncover a gene with uncharacterized function (1700011H14Rik/C14orf105/CCDC198), which we hereby name FAME (Factor Associated with Metabolism and Energy). We observe that FAME shows an unusually high evolutionary divergence in birds and mammals. Through the comparison of single nucleotide polymorphisms, we identify gene flow of FAME from Neandertals into modern humans. We conduct knockout experiments on animals and observe altered body weight and decreased energy expenditure in Fame knockout animals, corresponding to genome-wide association studies linking FAME with higher body mass index in humans. Gene expression and subcellular localization analyses reveal that FAME is a membrane-bound protein enriched in the kidneys. Although the gene knockout results in structurally normal kidneys, we detect higher albumin in urine and lowered ferritin in the blood. Through experimental validation, we confirm interactions between FAME and ferritin and show co-localization in vesicular and plasma membranes.


Asunto(s)
Metabolismo Energético , Estudio de Asociación del Genoma Completo , Animales , Humanos , Peso Corporal , Metabolismo Energético/genética , Ferritinas/genética , Riñón , Hombre de Neandertal
16.
Sci Rep ; 12(1): 8728, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35610276

RESUMEN

The complex shape of embryonic cartilage represents a true challenge for phenotyping and basic understanding of skeletal development. X-ray computed microtomography (µCT) enables inspecting relevant tissues in all three dimensions; however, most 3D models are still created by manual segmentation, which is a time-consuming and tedious task. In this work, we utilised a convolutional neural network (CNN) to automatically segment the most complex cartilaginous system represented by the developing nasal capsule. The main challenges of this task stem from the large size of the image data (over a thousand pixels in each dimension) and a relatively small training database, including genetically modified mouse embryos, where the phenotype of the analysed structures differs from the norm. We propose a CNN-based segmentation model optimised for the large image size that we trained using a unique manually annotated database. The segmentation model was able to segment the cartilaginous nasal capsule with a median accuracy of 84.44% (Dice coefficient). The time necessary for segmentation of new samples shortened from approximately 8 h needed for manual segmentation to mere 130 s per sample. This will greatly accelerate the throughput of µCT analysis of cartilaginous skeletal elements in animal models of developmental diseases.


Asunto(s)
Aprendizaje Profundo , Animales , Cartílago/diagnóstico por imagen , Biología Evolutiva , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Redes Neurales de la Computación , Rayos X
17.
Gigascience ; 112022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35380661

RESUMEN

BACKGROUND: Lightless caves can harbour a wide range of living organisms. Cave animals have evolved a set of morphological, physiological, and behavioural adaptations known as troglomorphisms, enabling their survival in the perpetual darkness, narrow temperature and humidity ranges, and nutrient scarcity of the subterranean environment. In this study, we focused on adaptations of skull shape and sensory systems in the blind cave salamander, Proteus anguinus, also known as olm or simply proteus-the largest cave tetrapod and the only European amphibian living exclusively in subterranean environments. This extraordinary amphibian compensates for the loss of sight by enhanced non-visual sensory systems including mechanoreceptors, electroreceptors, and chemoreceptors. We compared developmental stages of P. anguinus with Ambystoma mexicanum, also known as axolotl, to make an exemplary comparison between cave- and surface-dwelling paedomorphic salamanders. FINDINGS: We used contrast-enhanced X-ray computed microtomography for the 3D segmentation of the soft tissues in the head of P. anguinus and A. mexicanum. Sensory organs were visualized to elucidate how the animal is adapted to living in complete darkness. X-ray microCT datasets were provided along with 3D models for larval, juvenile, and adult specimens, showing the cartilage of the chondrocranium and the position, shape, and size of the brain, eyes, and olfactory epithelium. CONCLUSIONS: P. anguinus still keeps some of its secrets. Our high-resolution X-ray microCT scans together with 3D models of the anatomical structures in the head may help to elucidate the nature and origin of the mechanisms behind its adaptations to the subterranean environment, which led to a series of troglomorphisms.


Asunto(s)
Proteidae , Animales , Oscuridad , Urodelos , Rayos X
18.
Nat Commun ; 13(1): 6949, 2022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-36376278

RESUMEN

There are major differences in duration and scale at which limb development and regeneration proceed, raising the question to what extent regeneration is a recapitulation of development. We address this by analyzing skeletal elements using a combination of micro-CT imaging, molecular profiling and clonal cell tracing. We find that, in contrast to development, regenerative skeletal growth is accomplished based entirely on cartilage expansion prior to ossification, not limiting the transversal cartilage expansion and resulting in bulkier skeletal parts. The oriented extension of salamander cartilage and bone appear similar to the development of basicranial synchondroses in mammals, as we found no evidence for cartilage stem cell niches or growth plate-like structures during neither development nor regeneration. Both regenerative and developmental ossification in salamanders start from the cortical bone and proceeds inwards, showing the diversity of schemes for the synchrony of cortical and endochondral ossification among vertebrates.


Asunto(s)
Osteogénesis , Urodelos , Animales , Huesos , Cartílago , División Celular , Mamíferos
19.
Gigascience ; 10(3)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33677535

RESUMEN

BACKGROUND: X-ray microtomography (µCT) has become an invaluable tool for non-destructive analysis of biological samples in the field of developmental biology. Mouse embryos are a typical model for investigation of human developmental diseases. By obtaining 3D high-resolution scans of the mouse embryo heads, we gain valuable morphological information about the structures prominent in the development of future face, brain, and sensory organs. The development of facial skeleton tracked in these µCT data provides a valuable background for further studies of congenital craniofacial diseases and normal development. FINDINGS: In this work, reusable tomographic data from 7 full 3D scans of mouse embryo heads are presented and made publicly available. The ages of these embryos range from E12.5 to E18.5. The samples were stained by phosphotungstic acid prior to scanning, which greatly enhanced the contrast of various tissues in the reconstructed images and enabled precise segmentation. The images were obtained on a laboratory-based µCT system. Furthermore, we provide manually segmented masks of mesenchymal condensations (for E12.5 and E13.5) and cartilage present in the nasal capsule of the scanned embryos. CONCLUSION: We present a comprehensive dataset of X-ray 3D computed tomography images of the developing mouse head with high-quality manual segmentation masks of cartilaginous nasal capsules. The provided µCT images can be used for studying any other major structure within the developing mouse heads. The high quality of the manually segmented models of nasal capsules may be instrumental to understanding the complex process of the development of the face in a mouse model.


Asunto(s)
Imagenología Tridimensional , Cráneo , Animales , Ratones , Cráneo/diagnóstico por imagen , Microtomografía por Rayos X
20.
Front Cell Dev Biol ; 8: 620735, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33392208

RESUMEN

The head represents the most complex part of the body and a distinctive feature of the vertebrate body plan. This intricate structure is assembled during embryonic development in the four-dimensional process of morphogenesis. The head integrates components of the central and peripheral nervous system, sensory organs, muscles, joints, glands, and other specialized tissues in the framework of a complexly shaped skull. The anterior part of the head is referred to as the face, and a broad spectrum of facial shapes across vertebrate species enables different feeding strategies, communication styles, and diverse specialized functions. The face formation starts early during embryonic development and is an enormously complex, multi-step process regulated on a genomic, molecular, and cellular level. In this review, we will discuss recent discoveries that revealed new aspects of facial morphogenesis from the time of the neural crest cell emergence till the formation of the chondrocranium, the primary design of the individual facial shape. We will focus on molecular mechanisms of cell fate specification, the role of individual and collective cell migration, the importance of dynamic and continuous cellular interactions, responses of cells and tissues to generated physical forces, and their morphogenetic outcomes. In the end, we will examine the spatiotemporal activity of signaling centers tightly regulating the release of signals inducing the formation of craniofacial skeletal elements. The existence of these centers and their regulation by enhancers represent one of the core morphogenetic mechanisms and might lay the foundations for intra- and inter-species facial variability.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA