Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 731
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 633(8031): 887-894, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39261736

ABSTRACT

Early amniote development is highly self-organized, capable of adapting to interference through local and long-range cell-cell interactions. This process, called embryonic regulation1, has been well illustrated in experiments on avian embryos, in which subdividing the epiblast disk into different parts not only redirects cell fates to eventually form a complete and well-proportioned embryo at its original location, but also leads to the self-organization of additional, fully formed embryos2,3 in the other separated parts. The cellular interactions underlying embryonic self-organization are widely believed to be mediated by molecular signals, yet the identity of such signals is unclear. Here, by analysing intact and mechanically perturbed quail embryos, we show that the mechanical forces that drive embryogenesis self-organize, with contractility locally self-activating and the ensuing tension acting as a long-range inhibitor. This mechanical feedback governs the persistent pattern of tissue flows that shape the embryo4-6 and also steers the concomitant emergence of embryonic territories by modulating gene expression, ensuring the formation of a single embryo under normal conditions, yet allowing the emergence of multiple, well-proportioned embryos after perturbations. Thus, mechanical forces act at the core of embryonic self-organization, shaping both tissues and gene expression to robustly yet plastically canalize early development.


Subject(s)
Biomechanical Phenomena , Body Patterning , Embryo, Nonmammalian , Embryonic Development , Quail , Animals , Biomechanical Phenomena/genetics , Biomechanical Phenomena/physiology , Body Patterning/genetics , Body Patterning/physiology , Cell Communication , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Embryonic Development/physiology , Feedback, Physiological , Gene Expression Regulation, Developmental , Germ Layers/cytology , Germ Layers/embryology , Germ Layers/metabolism , Quail/embryology , Quail/genetics
2.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38856082

ABSTRACT

A major challenge in biology is to understand how mechanical interactions and cellular behavior affect the shapes of tissues and embryo morphology. The extension of the neural tube and paraxial mesoderm, which form the spinal cord and musculoskeletal system, respectively, results in the elongated shape of the vertebrate embryonic body. Despite our understanding of how each of these tissues elongates independently of the others, the morphogenetic consequences of their simultaneous growth and mechanical interactions are still unclear. Our study investigates how differential growth, tissue biophysical properties and mechanical interactions affect embryonic morphogenesis during axial extension using a 2D multi-tissue continuum-based mathematical model. Our model captures the dynamics observed in vivo by time-lapse imaging of bird embryos, and reveals the underestimated influence of differential tissue proliferation rates. We confirmed this prediction in quail embryos by showing that decreasing the rate of cell proliferation in the paraxial mesoderm affects long-term tissue dynamics, and shaping of both the paraxial mesoderm and the neighboring neural tube. Overall, our work provides a new theoretical platform upon which to consider the long-term consequences of tissue differential growth and mechanical interactions on morphogenesis.


Subject(s)
Cell Proliferation , Mesoderm , Models, Biological , Morphogenesis , Neural Tube , Animals , Mesoderm/embryology , Mesoderm/cytology , Neural Tube/embryology , Neural Tube/cytology , Quail/embryology , Embryo, Nonmammalian/cytology , Embryonic Development/physiology , Viscosity
3.
Development ; 151(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39250350

ABSTRACT

Dorsal neural tube-derived retinoic acid promotes the end of neural crest production and transition into a definitive roof plate. Here, we analyze how this impacts the segregation of central and peripheral lineages, a process essential for tissue patterning and function. Localized in ovo inhibition in quail embryos of retinoic acid activity followed by single-cell transcriptomics unraveled a comprehensive list of differentially expressed genes relevant to these processes. Importantly, progenitors co-expressed neural crest, roof plate and dI1 interneuron markers, indicating a failure in proper lineage segregation. Furthermore, separation between roof plate and dI1 interneurons is mediated by Notch activity downstream of retinoic acid, highlighting their crucial role in establishing the roof plate-dI1 boundary. Within the peripheral branch, where absence of retinoic acid resulted in neural crest production and emigration extending into the roof plate stage, sensory progenitors failed to separate from melanocytes, leading to formation of a common glia-melanocyte cell with aberrant migratory patterns. In summary, the implementation of single-cell RNA sequencing facilitated the discovery and characterization of a molecular mechanism responsible for the segregation of dorsal neural fates during development.


Subject(s)
Neural Crest , Tretinoin , Animals , Tretinoin/metabolism , Tretinoin/pharmacology , Neural Crest/metabolism , Neural Crest/cytology , Gene Expression Regulation, Developmental , Quail/embryology , Cell Movement , Receptors, Notch/metabolism , Cell Lineage , Body Patterning/genetics , Body Patterning/drug effects , Neural Plate/metabolism , Neural Plate/embryology , Interneurons/metabolism , Interneurons/cytology , Single-Cell Analysis , Neural Tube/embryology , Neural Tube/metabolism , Cell Differentiation , Melanocytes/metabolism , Melanocytes/cytology
4.
Development ; 147(10)2020 05 26.
Article in English | MEDLINE | ID: mdl-32345743

ABSTRACT

Sonic hedgehog (Shh), produced in the notochord and floor plate, is necessary for both neural and mesodermal development. To reach the myotome, Shh has to traverse the sclerotome and a reduction of sclerotomal Shh affects myotome differentiation. By investigating loss and gain of Shh function, and floor-plate deletions, we report that sclerotomal Shh is also necessary for neural tube development. Reducing the amount of Shh in the sclerotome using a membrane-tethered hedgehog-interacting protein or Patched1, but not dominant active Patched, decreased the number of Olig2+ motoneuron progenitors and Hb9+ motoneurons without a significant effect on cell survival or proliferation. These effects were a specific and direct consequence of Shh reduction in the mesoderm. In addition, grafting notochords in a basal but not apical location, vis-à-vis the tube, profoundly affected motoneuron development, suggesting that initial ligand presentation occurs at the basal side of epithelia corresponding to the sclerotome-neural tube interface. Collectively, our results reveal that the sclerotome is a potential site of a Shh gradient that coordinates the development of mesodermal and neural progenitors.


Subject(s)
Hedgehog Proteins/metabolism , Neural Tube/embryology , Neurulation/genetics , Notochord/metabolism , Quail/embryology , Animals , Body Patterning/genetics , Cell Differentiation/genetics , Chick Embryo , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Mesoderm/metabolism , Motor Neurons/metabolism , Neural Plate/metabolism , Neural Tube/metabolism , Neurogenesis/genetics , Patched-1 Receptor/metabolism , Signal Transduction/genetics , Transfection
5.
Mol Hum Reprod ; 27(2)2021 02 05.
Article in English | MEDLINE | ID: mdl-33313783

ABSTRACT

Experimental autoimmune orchitis (EAO) is a useful model to study organ-specific autoimmunity and chronic testicular inflammation. This model reflects testicular pathological changes reported in immunological infertility in men. Progression of EAO in rodents is associated with a significantly increased percentage of testicular endothelial cells and interstitial testicular blood vessels, indicating an ongoing angiogenic process. Vascular endothelial growth factor A (VEGFA), the main regulator of physiological and pathological angiogenesis, can stimulate endothelial cell proliferation, chemotaxis and vascular permeability. The aim of this study was to explore the role of VEGFA in the pathogenesis of testicular inflammation. Our results found VEGFA expression in Leydig cells, endothelial cells and macrophages in testis of rats with autoimmune orchitis. VEGFA level was significantly higher in testicular fluid and serum of rats at the end of the immunization period, preceding testicular damage. VEGF receptor (VEGFR) 1 is expressed mainly in testicular endothelial cells, whereas VEGFR2 was detected in germ cells and vascular smooth muscle cells. Both receptors were expressed in testicular interstitial cells. VEGFR2 increased after the immunization period in the testicular interstitium and VEGFR1 was downregulated in EAO testis. In-vivo-specific VEGFA inhibition by Bevacizumab prevented the increase in blood vessel number and reduced EAO incidence and severity. Our results unveil relevance of VEGFA-VEGFR axis during orchitis development, suggesting that VEGFA might be an early marker of testicular inflammation and Bevacizumab a therapeutic tool for treatment of testicular inflammation associated with subfertility and infertility.


Subject(s)
Autoimmune Diseases/pathology , Neovascularization, Pathologic , Testis/blood supply , Testis/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/prevention & control , Bevacizumab/pharmacology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Leydig Cells/metabolism , Leydig Cells/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Orchitis/immunology , Orchitis/metabolism , Orchitis/prevention & control , Quail/embryology , Rats, Wistar , Signal Transduction , Testis/drug effects , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
PLoS Comput Biol ; 16(8): e1007874, 2020 08.
Article in English | MEDLINE | ID: mdl-32822340

ABSTRACT

Shear stress induces directed endothelial cell (EC) migration in blood vessels leading to vessel diameter increase and induction of vascular maturation. Other factors, such as EC elongation and interaction between ECs and non-vascular areas are also important. Computational models have previously been used to study collective cell migration. These models can be used to predict EC migration and its effect on vascular remodelling during embryogenesis. We combined live time-lapse imaging of the remodelling vasculature of the quail embryo yolk sac with flow quantification using a combination of micro-Particle Image Velocimetry and computational fluid dynamics. We then used the flow and remodelling data to inform a model of EC migration during remodelling. To obtain the relation between shear stress and velocity in vitro for EC cells, we developed a flow chamber to assess how confluent sheets of ECs migrate in response to shear stress. Using these data as an input, we developed a multiphase, self-propelled particles (SPP) model where individual agents are driven to migrate based on the level of shear stress while maintaining appropriate spatial relationship to nearby agents. These agents elongate, interact with each other, and with avascular agents at each time-step of the model. We compared predicted vascular shape to real vascular shape after 4 hours from our time-lapse movies and performed sensitivity analysis on the various model parameters. Our model shows that shear stress has the largest effect on the remodelling process. Importantly, however, elongation played an especially important part in remodelling. This model provides a powerful tool to study the input of different biological processes on remodelling.


Subject(s)
Hydrodynamics , Vascular Remodeling , Animals , Blood Circulation , Cell Movement/physiology , Cell Shape , Computational Biology , Endothelial Cells/physiology , Quail/anatomy & histology , Quail/embryology , Stress, Mechanical
7.
J Mol Cell Cardiol ; 147: 62-73, 2020 10.
Article in English | MEDLINE | ID: mdl-32777295

ABSTRACT

BACKGROUND: During the formation of the coronary artery stem, endothelial strands from the endothelial progenitor pool surrounding the conotruncus penetrate into the aortic wall. Vascular endothelial growth factors (VEGFs) as well as CXCL12/CXCR4 signaling are thought to play a role in the formation of the coronary stem. However, the mechanisms regulating how endothelial strands exclusively invade into the aorta remain unknown. METHODS AND RESULTS: Immunohistochemistry showed that before the formation of endothelial strands, Sema3a was highly expressed in endothelial progenitors surrounding the great arteries. At the onset of/during invasion of endothelial strands into the aorta, Sema3a was downregulated and CXCR4 was upregulated in the endothelial strands. In situ hybridization showed that Cxcl12 was highly expressed in the aortic wall compared with in the pulmonary artery. Using avian embryonic hearts, we established two types of endothelial penetration assay, in which coronary endothelial strands preferentially invaded into the aorta in culture. Sema3a blocking peptide induced an excess number of endothelial strands penetrating into the pulmonary artery, whereas recombinant Sema3a inhibited the formation of endothelial strands. In cultured coronary endothelial progenitors, recombinant VEGF protein induced CXCR4-positive endothelial strands, which were capable of being attracted by CXCL12-impregnated beads. Monoazo rhodamine detected that hypoxia was predominant in aortic/subaortic region in ovo and hypoxic condition downregulated the expression of Sema3a in culture. CONCLUSION: Results suggested that hypoxia in the aortic region downregulates the expression of Sema3a, thereby enhancing VEGF activity to induce the formation of CXCR4-positive endothelial strands, which are subsequently attracted into the Cxcl12-positive aortic wall to connect the aortic lumen.


Subject(s)
Chemokine CXCL12/metabolism , Coronary Vessels/metabolism , Down-Regulation/genetics , Hypoxia/genetics , Receptors, CXCR4/metabolism , Animals , Aorta/embryology , Aorta/metabolism , Cells, Cultured , Chickens , Coronary Vessels/embryology , Endothelial Cells/metabolism , Quail/embryology , Semaphorin-3A/metabolism , Up-Regulation
8.
Development ; 144(2): 281-291, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28096216

ABSTRACT

Cells may exchange information with other cells and tissues by exerting forces on the extracellular matrix (ECM). Fibronectin (FN) is an important ECM component that forms fibrils through cell contacts and creates directionally biased geometry. Here, we demonstrate that FN is deposited as pillars between widely separated germ layers, namely the somitic mesoderm and the endoderm, in quail embryos. Alongside the FN pillars, long filopodia protrude from the basal surfaces of somite epithelial cells. Loss-of-function of Ena/VASP, α5ß1-integrins or talin in the somitic cells abolished the FN pillars, indicating that FN pillar formation is dependent on the basal filopodia through these molecules. The basal filopodia and FN pillars are also necessary for proper somite morphogenesis. We identified a new mechanism contributing to FN pillar formation by focusing on cyclic expansion of adjacent dorsal aorta. Maintenance of the directional alignment of the FN pillars depends on pulsatile blood flow through the dorsal aortae. These results suggest that the FN pillars are specifically established through filopodia-mediated and pulsating force-related mechanisms.


Subject(s)
Blood Vessels/physiology , Endoderm/metabolism , Mesoderm/metabolism , Pseudopodia/physiology , Quail/embryology , Stress, Mechanical , Animals , Animals, Genetically Modified , Cell Movement , Embryo, Nonmammalian , Extracellular Matrix/metabolism , Fibronectins/metabolism , Morphogenesis
9.
Development ; 144(18): 3315-3324, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28807901

ABSTRACT

We have proposed that independent origins of the tympanic membrane (TM), consisting of the external auditory meatus (EAM) and first pharyngeal pouch, are linked with distinctive middle ear structures in terms of dorsal-ventral patterning of the pharyngeal arches during amniote evolution. However, previous studies have suggested that the first pharyngeal arch (PA1) is crucial for TM formation in both mouse and chick. In this study, we compare TM formation along the anterior-posterior axis in these animals using Hoxa2 expression as a marker of the second pharyngeal arch (PA2). In chick, the EAM begins to invaginate at the surface ectoderm of PA2, not at the first pharyngeal cleft, and the entire TM forms in PA2. Chick-quail chimera that have lost PA2 and duplicated PA1 suggest that TM formation is achieved by developmental interaction between a portion of the EAM and the columella auris in PA2, and that PA1 also contributes to formation of the remaining part of the EAM. By contrast, in mouse, TM formation is highly associated with an interdependent relationship between the EAM and tympanic ring in PA1.


Subject(s)
Branchial Region/embryology , Tympanic Membrane/embryology , Animals , Branchial Region/metabolism , Chick Embryo , Chickens , Ear Canal/embryology , Ear, Middle/embryology , Embryo, Mammalian/metabolism , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Models, Biological , Phenotype , Quail/embryology , Tympanic Membrane/metabolism
10.
Cell Mol Neurobiol ; 40(3): 383-393, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31555941

ABSTRACT

Neural crest cells (NCCs) comprise a population of multipotent progenitors and stem cells at the origin of the peripheral nervous system (PNS) and melanocytes of skin, which are profoundly influenced by microenvironmental factors, among which is basic fibroblast growth factor 2 (FGF2). In this work, we further investigated the role of this growth factor in quail trunk NC morphogenesis and demonstrated its huge effect in NCC growth mainly by stimulating cell proliferation but also reducing cell death, despite that NCC migration from the neural tube explant was not affected. Moreover, following FGF2 treatment, reduced expression of the early NC markers Sox10 and FoxD3 and improved proliferation of HNK1-positive NCC were observed. Since these markers are involved in the regulation of glial and melanocytic fate of NC, the effect of FGF2 on NCC differentiation was investigated. Therefore, in the presence of FGF2, increased proportions of NCCs positives to the melanoblast marker Mitf as well as NCCs double stained to Mitf and BrdU were recorded. In addition, treatment with FGF2, followed by differentiation medium, resulted in increased expression of melanin and improved proportion of melanin-pigmented melanocytes without alteration in the glial marker Schwann myelin protein (SMP). Taken together, these data further reveal the important role of FGF2 in NCC proliferation, survival, and differentiation, particularly in melanocyte development. This is the first demonstration of FGF2 effects in melanocyte commitment of NC and in the proliferation of Mitf-positive melanoblasts. Elucidating the differentiation process of embryonic NCCs brings us a step closer to understanding the development of the PNS and then undertaking the search for advanced technologies to prevent, or treat, injuries caused by NC-related disorders, also known as neurocristopathies.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Embryonic Stem Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , Melanocytes/drug effects , Neural Crest/drug effects , Animals , Cell Movement/drug effects , Cells, Cultured , Chick Embryo , Embryonic Stem Cells/physiology , Melanins/metabolism , Melanocytes/physiology , Neural Crest/cytology , Neural Tube/cytology , Neural Tube/drug effects , Peripheral Nerves/cytology , Peripheral Nerves/drug effects , Peripheral Nerves/physiology , Quail/embryology , Torso
11.
Dev Biol ; 444 Suppl 1: S287-S296, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29391165

ABSTRACT

We quantified cell population increase in the quail embryo enteric nervous system (ENS) from E2.5 (about 1500 cells) to E12 (about 8 million cells). We then probed ENS proliferative capacity by grafting to the chorio-allantoic membrane large (600 cells) and small (40 cells) populations of enteric neural crest (ENC) cells with aneural gut. This demonstrated that ENC cells show an extremely high capacity to regulate their proliferation while forming the ENS. Previous mathematical models and clonal label experiments revealed that a few dominant ENS "superstar" cell clones emerge but most clones are small. The model implied that "superstars" arise stochastically, but the same outcome could arise if "superstars" were pre-determined. We investigated these two modes mathematically and by grafting experiments with large and small numbers of ENCs, each including one EGFP-labelled ENC cell. The stochastic model predicts that the frequency of "superstar" detection increases as the ENC population decreases, the pre-determined model does not. Experimentally, as predicted by the stochastic model, the frequency of "superstar" detection increased with small ENC cell number. We conclude that ENS "superstar" clones achieve this status stochastically. Clonal dominance implies that clonal diversity is greatly reduced and in this case, somatic mutations may affect the phenotype. We suggest that somatic mutations coupled with loss of clonal diversity may contribute to variable penetrance and expressivity in individuals with genetically identical ENS pathologies.


Subject(s)
Enteric Nervous System/embryology , Enteric Nervous System/metabolism , Neural Crest/metabolism , Animals , Cell Movement/physiology , Cells, Cultured , Chick Embryo , Clone Cells , Enteric Nervous System/physiology , Models, Biological , Models, Theoretical , Neural Crest/physiology , Neurons/metabolism , Quail/embryology , Stochastic Processes
12.
Dev Biol ; 444 Suppl 1: S219-S236, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29753626

ABSTRACT

How does form arise during development and change during evolution? How does form relate to function, and what enables embryonic structures to presage their later use in adults? To address these questions, we leverage the distinct functional morphology of the jaw in duck, chick, and quail. In connection with their specialized mode of feeding, duck develop a secondary cartilage at the tendon insertion of their jaw adductor muscle on the mandible. An equivalent cartilage is absent in chick and quail. We hypothesize that species-specific jaw architecture and mechanical forces promote secondary cartilage in duck through the differential regulation of FGF and TGFß signaling. First, we perform transplants between chick and duck embryos and demonstrate that the ability of neural crest mesenchyme (NCM) to direct the species-specific insertion of muscle and the formation of secondary cartilage depends upon the amount and spatial distribution of NCM-derived connective tissues. Second, we quantify motility and build finite element models of the jaw complex in duck and quail, which reveals a link between species-specific jaw architecture and the predicted mechanical force environment. Third, we investigate the extent to which mechanical load mediates FGF and TGFß signaling in the duck jaw adductor insertion, and discover that both pathways are mechano-responsive and required for secondary cartilage formation. Additionally, we find that FGF and TGFß signaling can also induce secondary cartilage in the absence of mechanical force or in the adductor insertion of quail embryos. Thus, our results provide novel insights on molecular, cellular, and biomechanical mechanisms that couple musculoskeletal form and function during development and evolution.


Subject(s)
Fibroblast Growth Factors/metabolism , Jaw/embryology , Transforming Growth Factor beta/metabolism , Animals , Biological Evolution , Cartilage/metabolism , Cell Movement , Chick Embryo , Chondrogenesis , Ducks/embryology , Embryo, Nonmammalian/metabolism , Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental/genetics , Jaw/physiology , Mandible/embryology , Mesoderm/embryology , Neural Crest/embryology , Neural Crest/physiology , Quail/embryology , Signal Transduction/physiology , Species Specificity , Transforming Growth Factor beta/physiology
13.
Bull Math Biol ; 81(7): 2220-2238, 2019 07.
Article in English | MEDLINE | ID: mdl-30945102

ABSTRACT

Growth in biological systems occurs as a consequence of cell proliferation fueled by a nutrient supply. In general, the nutrient gradient of the system will be nonconstant, resulting in biased cell proliferation. We develop a uniaxial discrete cellular automaton with biased cell proliferation using a probability distribution which reflects the nutrient gradient of the system. An explicit probability mass function for the displacement of any tracked cell under the cellular automaton model is derived and verified against averaged simulation results; this displacement distribution has applications in predicting cell trajectories and evolution of expected site occupancies.


Subject(s)
Cell Proliferation/physiology , Models, Biological , Algorithms , Animals , Body Patterning/physiology , Cell Movement/physiology , Computer Simulation , Digestive System/embryology , Linear Models , Markov Chains , Mathematical Concepts , Probability , Quail/embryology , Spatio-Temporal Analysis , Systems Analysis
14.
Genesis ; 56(6-7): e23219, 2018 06.
Article in English | MEDLINE | ID: mdl-30134069

ABSTRACT

For well over half of the 150 years since the discovery of the neural crest, the special ability of these cells to function as a source of species-specific pattern has been clearly recognized. Initially, this observation arose in association with chimeric transplant experiments among differentially pigmented amphibians, where the neural crest origin for melanocytes had been duly noted. Shortly thereafter, the role of cranial neural crest cells in transmitting species-specific information on size and shape to the pharyngeal arch skeleton as well as in regulating the timing of its differentiation became readily apparent. Since then, what has emerged is a deeper understanding of how the neural crest accomplishes such a presumably difficult mission, and this includes a more complete picture of the molecular and cellular programs whereby neural crest shapes the face of each species. This review covers studies on a broad range of vertebrates and describes neural-crest-mediated mechanisms that endow the craniofacial complex with species-specific pattern. A major focus is on experiments in quail and duck embryos that reveal a hierarchy of cell-autonomous and non-autonomous signaling interactions through which neural crest generates species-specific pattern in the craniofacial integument, skeleton, and musculature. By controlling size and shape throughout the development of these systems, the neural crest underlies the structural and functional integration of the craniofacial complex during evolution.


Subject(s)
Body Patterning/physiology , Neural Crest/cytology , Neural Crest/physiology , Animals , Beak/embryology , Bone and Bones/embryology , Branchial Region , Cell Differentiation/physiology , Chimera/embryology , Ducks/embryology , Face/embryology , Gene Expression Regulation, Developmental/physiology , Humans , Neural Crest/embryology , Quail/embryology , Skeleton/embryology , Skull/embryology , Species Specificity
15.
Development ; 142(23): 4151-7, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26552886

ABSTRACT

Angiogenesis is tightly controlled by a number of signalling pathways. Although our understanding of the molecular mechanisms involved in angiogenesis has rapidly increased, the role that biomechanical signals play in this process is understudied. We recently developed a technique to simultaneously analyse flow dynamics and vascular remodelling by time-lapse microscopy in the capillary plexus of avian embryos and used this to study the hemodynamic environment present during angiogenic sprouting. We found that sprouts always form from a vessel at lower pressure towards a vessel at higher pressure, and that sprouts form at the location of a shear stress minimum, but avoid locations where two blood streams merge even if this point is at a lower level of shear stress than the sprouting location. Using these parameters, we were able to successfully predict sprout location in quail embryos. We also found that the pressure difference between two vessels is permissive to elongation, and that sprouts will either change direction or regress if the pressure difference becomes negative. Furthermore, the sprout elongation rate is proportional to the pressure difference between the two vessels. Our results show that flow dynamics are predictive of the location of sprout formation in perfused vascular networks and that pressure differences across the interstitium can guide sprout elongation.


Subject(s)
Neovascularization, Physiologic/physiology , Quail/embryology , Animals , Biomechanical Phenomena , Endothelial Cells/cytology , Hemodynamics , Hydrodynamics , Image Processing, Computer-Assisted , Microscopy, Fluorescence , Morphogenesis , Pressure , Shear Strength , Stress, Mechanical , Time-Lapse Imaging
16.
Dev Growth Differ ; 60(2): 97-111, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29392712

ABSTRACT

The origin of coronary endothelial cells (ECs) has been investigated in avian species, and the results showed that the coronary ECs originate from the proepicardial organ (PEO) and developing epicardium. Genetic approaches in mouse models showed that the major source of coronary ECs is the sinus venosus endothelium or ventricular endocardium. To clarify and reconcile the differences between avian and mouse species, we examined the source of coronary ECs in avian embryonic hearts. Using an enhanced green fluorescent protein-Tol2 system and fluorescent dye labeling, four types of quail-chick chimeras were made and quail-specific endothelial marker (QH1) immunohistochemistry was performed. The developing PEO consisted of at least two cellular populations in origin, one was sinus venosus endothelium-derived inner cells and the other was surface mesothelium-derived cells. The majority of ECs in the coronary stems, ventricular free wall, and dorsal ventricular septum originated from the sinus venosus endothelium. The ventricular endocardium contributed mainly to the septal artery and a few cells to the coronary stems. Surface mesothelial cells of the PEO differentiated mainly into a smooth muscle phenotype, but a few differentiated into ECs. In avian species, the coronary endothelium had a heterogeneous origin in a region-specific manner, and the sources of ECs were basically the same as those observed in mice.


Subject(s)
Coronary Vessels/embryology , Endothelial Cells/cytology , Endothelium, Vascular/embryology , Epithelial Cells/cytology , Heart/embryology , Animals , Cell Differentiation , Chick Embryo , Chickens , Chimera/embryology , Endothelium, Vascular/cytology , Epithelial Cells/physiology , Epithelium/physiology , Green Fluorescent Proteins/genetics , Immunohistochemistry , Myocardium/cytology , Organ Culture Techniques , Pericardium/cytology , Pericardium/embryology , Quail/embryology
17.
BMC Dev Biol ; 17(1): 13, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29017464

ABSTRACT

BACKGROUND: Vertebrate head development depends on a series of interactions between many cell populations of distinct embryological origins. Cranial mesenchymal tissues have a dual embryonic source: - the neural crest (NC), which generates most of craniofacial skeleton, dermis, pericytes, fat cells, and tenocytes; and - the mesoderm, which yields muscles, blood vessel endothelia and some posterior cranial bones. The molecular players that orchestrate co-development of cephalic NC and mesodermal cells to properly construct the head of vertebrates remain poorly understood. In this regard, Six1 gene, a vertebrate homolog of Drosophila Sine Oculis, is known to be required for development of ear, nose, tongue and cranial skeleton. However, the embryonic origin and fate of Six1-expressing cells have remained unclear. In this work, we addressed these issues in the avian embryo model by using quail-chick chimeras, cephalic NC cultures and immunostaining for SIX1. RESULTS: Our data show that, at early NC migration stages, SIX1 is expressed by mesodermal cells but excluded from the NC cells (NCC). Then, SIX1 becomes widely expressed in NCC that colonize the pre-otic mesenchyme. In contrast, in the branchial arches (BAs), SIX1 is present only in mesodermal cells that give rise to jaw muscles. At later developmental stages, the distribution of SIX1-expressing cells in mesoderm-derived tissues is consistent with a possible role of this factor in the myogenic program of all types of head muscles, including pharyngeal, extraocular and tongue muscles. In NC derivatives, SIX1 is notably expressed in perichondrium and chondrocytes of the nasal septum and in the sclera, although other facial cartilages such as Meckel's were negative at the stages considered. Moreover, in cephalic NC cultures, chondrocytes and myofibroblasts, not the neural and melanocytic cells express SIX1. CONCLUSION: The present results point to a dynamic tissue-specific expression of SIX1 in a variety of cephalic NC- and mesoderm-derived cell types and tissues, opening the way for further analysis of Six1 function in the coordinated development of these two cellular populations during vertebrate head formation.


Subject(s)
Embryo, Nonmammalian/embryology , Neural Crest/embryology , Animals , Mesoderm/embryology , Quail/embryology
18.
Genet Sel Evol ; 49(1): 14, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28125975

ABSTRACT

BACKGROUND: Environmental exposures, for instance to chemicals, are known to impact plant and animal phenotypes on the long term, sometimes across several generations. Such transgenerational phenotypes were shown to be promoted by epigenetic alterations such as DNA methylation, an epigenetic mark involved in the regulation of gene expression. However, it is yet unknown whether transgenerational epigenetic inheritance of altered phenotypes exists in birds. The purpose of this study was to develop an avian model to investigate whether changes to the embryonic environment had a transgenerational effect that could alter the phenotypes of third-generation offspring. Given its impact on the mammalian epigenome and the reproductive system in birds, genistein was used as an environment stressor. RESULTS: We compared several third-generation phenotypes of two quail "epilines", which were obtained from genistein-injected eggs (Epi+) or from untreated eggs (Epi-) from the same founders. A "mirrored" crossing strategy was used to minimize between-line genetic variability by maintaining similar ancestor contributions across generations in each line. Three generations after genistein treatment, a significant difference in the sexual maturity of the females, which, after three generations, could not be attributed to direct maternal effects, was observed between the lines, with Epi+ females starting to lay eggs later. Adult body weight was significantly affected by genistein treatment applied in a previous generation, and a significant interaction between line and sex was observed for body weight at 3 weeks. Behavioral traits, such as evaluating the birds' reaction to social isolation, were also significantly affected by genistein treatment. Yet, global methylation analyses revealed no significant difference between the epilines. CONCLUSIONS: These findings demonstrate that embryonic environment affects the phenotype of offspring three generations later in quail. While one cannot rule out the existence of some initial genetic variability between the lines, the mirrored animal design should have minimized its effects, and thus, the observed differences in animals of the third generation may be attributed, at least partly, to transgenerational epigenetic phenomena.


Subject(s)
Embryonic Development/genetics , Environment , Gene-Environment Interaction , Quail/embryology , Quail/genetics , Animals , Behavior, Animal , Body Weight/genetics , DNA Methylation , Epigenesis, Genetic , Female , Genetic Association Studies , Male , Phenotype , Quantitative Trait, Heritable , Reproduction/genetics , Temperature
19.
Nature ; 476(7358): 57-62, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21814276

ABSTRACT

The developing vertebrate gut tube forms a reproducible looped pattern as it grows into the body cavity. Here we use developmental experiments to eliminate alternative models and show that gut looping morphogenesis is driven by the homogeneous and isotropic forces that arise from the relative growth between the gut tube and the anchoring dorsal mesenteric sheet, tissues that grow at different rates. A simple physical mimic, using a differentially strained composite of a pliable rubber tube and a soft latex sheet is consistent with this mechanism and produces similar patterns. We devise a mathematical theory and a computational model for the number, size and shape of intestinal loops based solely on the measurable geometry, elasticity and relative growth of the tissues. The predictions of our theory are quantitatively consistent with observations of intestinal loops at different stages of development in the chick embryo. Our model also accounts for the qualitative and quantitative variation in the distinct gut looping patterns seen in a variety of species including quail, finch and mouse, illuminating how the simple macroscopic mechanics of differential growth drives the morphology of the developing gut.


Subject(s)
Intestines/anatomy & histology , Intestines/embryology , Models, Anatomic , Models, Biological , Animals , Biomechanical Phenomena , Chick Embryo , Computer Simulation , Elasticity , Female , Finches/embryology , Mesentery/anatomy & histology , Mesentery/embryology , Mice , Quail/embryology , Rotation , Rubber
20.
Dev Biol ; 391(2): 241-50, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24809797

ABSTRACT

Corneal avascularity is important for optical clarity and normal vision. However, the molecular mechanisms that prevent angioblast migration and vascularization of the developing cornea are not clear. Previously we showed that periocular angioblasts and forming ocular blood vessels avoid the presumptive cornea despite dynamic ingression of neural crest cells. In the current study, we investigate the role of Semaphorin3A (Sema3A), a cell guidance chemorepellent, on angioblast migration and corneal avascularity during development. We show that Sema3A, Vegf, and Nrp1 are expressed in the anterior eye during cornea development. Sema3A mRNA transcripts are expressed at significantly higher levels than Vegf in the lens that is positioned adjacent to the presumptive cornea. Blockade of Sema3A signaling via lens removal or injection of a synthetic Sema3A inhibitor causes ectopic migration of angioblasts into the cornea and results in its subsequent vascularization. In addition, using bead implantation, we demonstrate that exogenous Sema3A protein inhibits Vegf-induced vascularization of the cornea. In agreement with these findings, loss of Sema/Nrp1 signaling in Nrp1(Sema-) mutant mice results in ectopic angioblasts and vascularization of the embryonic mouse corneas. Altogether, our results reveal Sema3A signaling as an important cue during the establishment of corneal avascularity in both chick and mouse embryos. Our study introduces cornea development as a new model for studying the mechanisms involved in vascular patterning during embryogenesis and it also provides new insights into therapeutic potential for Sema3A in neovascular diseases.


Subject(s)
Cornea/blood supply , Lens, Crystalline/blood supply , Neuropilin-1/genetics , Semaphorin-3A/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Animals, Genetically Modified , Cell Movement , Cells, Cultured , Chick Embryo , Cornea/embryology , Endothelial Cells , Lens, Crystalline/embryology , Mice , Neovascularization, Physiologic , Neuropilin-1/biosynthesis , Quail/embryology , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/genetics , Semaphorin-3A/antagonists & inhibitors , Semaphorin-3A/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL