Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Elife ; 122024 May 10.
Article in English | MEDLINE | ID: mdl-38727576

ABSTRACT

Large-scale cell flow characterizes gastrulation in animal development. In amniote gastrulation, particularly in avian gastrula, a bilateral vortex-like counter-rotating cell flow, called 'polonaise movements', appears along the midline. Here, through experimental manipulations, we addressed relationships between the polonaise movements and morphogenesis of the primitive streak, the earliest midline structure in amniotes. Suppression of the Wnt/planar cell polarity (PCP) signaling pathway maintains the polonaise movements along a deformed primitive streak. Mitotic arrest leads to diminished extension and development of the primitive streak and maintains the early phase of the polonaise movements. Ectopically induced Vg1, an axis-inducing morphogen, generates the polonaise movements, aligned to the induced midline, but disturbs the stereotypical cell flow pattern at the authentic midline. Despite the altered cell flow, induction and extension of the primitive streak are preserved along both authentic and induced midlines. Finally, we show that ectopic axis-inducing morphogen, Vg1, is capable of initiating the polonaise movements without concomitant PS extension under mitotic arrest conditions. These results are consistent with a model wherein primitive streak morphogenesis is required for the maintenance of the polonaise movements, but the polonaise movements are not necessarily responsible for primitive streak morphogenesis. Our data describe a previously undefined relationship between the large-scale cell flow and midline morphogenesis in gastrulation.


Subject(s)
Gastrulation , Morphogenesis , Animals , Cell Movement , Primitive Streak/embryology , Cell Polarity , Gastrula/embryology , Chick Embryo
2.
bioRxiv ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38712212

ABSTRACT

A bilateral body plan is predominant throughout the animal kingdom. Bilaterality of amniote embryos becomes recognizable as midline morphogenesis begins at gastrulation, bisecting an embryonic field into the left and right sides. Soon after, left-right asymmetry also starts. While a series of laterality genes expressed after the left-right compartmentalization has been extensively studied, the laterality patterning prior to and during midline morphogenesis has remained unclear. Here, through a biophysical quantification in a high spatial and temporal resolution, applied to a chick model system, we show that a large-scale bilateral counter-rotating cell flow, termed as 'polonaise movements', display left-right asymmetries in early gastrulation. This cell movement starts prior to the formation of the primitive streak, which is the earliest midline structure, and earlier than expression of laterality genes. The cell flow speed and vorticity unravel the location and timing of the left-right asymmetries. The bilateral cell flow exhibited a Left side asymmetry at the beginning, but a transition towards Right dominance. Mitotic arrest that diminishes primitive streak formation resulted in changes in the bilateral flow pattern, but the Right dominance persisted. Our data indicate that the left-right asymmetry in amniote gastrula becomes detectable prior to the point when the asymmetric regulation of the laterality signals at the node leads to the left-right patterning. More broadly, our results suggest that physical processes can play an unexpected but significant role in influencing left-right laterality during embryonic development.

3.
bioRxiv ; 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37293063

ABSTRACT

Large-scale cell flow characterizes gastrulation in animal development. In amniote gastrulation, particularly in avian gastrula, a bilateral vortex-like counter-rotating cell flow, called 'polonaise movements', appears along the midline. Here, through experimental manipulations, we addressed relationships between the polonaise movements and morphogenesis of the primitive streak, the earliest midline structure in amniotes. Suppression of the Wnt/planar cell polarity (PCP) signaling pathway maintains the polonaise movements along a deformed primitive streak. Mitotic arrest leads to diminished extension and development of the primitive streak and maintains the early phase of the polonaise movements. Ectopically induced Vg1, an axis-inducing morphogen, generates the polonaise movements, aligned to the induced midline, but disturbs the stereotypical cell flow pattern at the authentic midline. Despite the altered cell flow, induction and extension of the primitive streak are preserved along both authentic and induced midlines. Finally, we show that ectopic axis-inducing morphogen, Vg1, is capable of initiating the polonaise movements without concomitant PS extension under mitotic arrest conditions. These results are consistent with a model wherein primitive streak morphogenesis is required for the maintenance of the polonaise movements, but the polonaise movements are not necessarily responsible for primitive streak morphogenesis. Our data describe a previously undefined relationship between the large-scale cell flow and midline morphogenesis in gastrulation.

4.
J Clin Invest ; 133(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36637912

ABSTRACT

Mutations of G protein-coupled receptors (GPCRs) cause various human diseases, but the mechanistic details are limited. Here, we establish p.E303K in the gene encoding the endothelin receptor type A (ETAR/EDNRA) as a recurrent mutation causing mandibulofacial dysostosis with alopecia (MFDA), with craniofacial changes similar to those caused by p.Y129F. Mouse models carrying either of these missense mutations exhibited a partial maxillary-to-mandibular transformation, which was rescued by deleting the ligand endothelin 3 (ET3/EDN3). Pharmacological experiments confirmed the causative ETAR mutations as gain of function, dependent on ET3. To elucidate how an amino acid substitution far from the ligand binding site can increase ligand affinity, we used molecular dynamics (MD) simulations. E303 is located at the intracellular end of transmembrane domain 6, and its replacement by a lysine increased flexibility of this portion of the helix, thus favoring G protein binding and leading to G protein-mediated enhancement of agonist affinity. The Y129F mutation located under the ligand binding pocket reduced the sodium-water network, thereby affecting the extracellular portion of helices in favor of ET3 binding. These findings provide insight into the pathogenesis of MFDA and into allosteric mechanisms regulating GPCR function, which may provide the basis for drug design targeting GPCRs.


Subject(s)
Mandibulofacial Dysostosis , Animals , Mice , Humans , Mandibulofacial Dysostosis/genetics , Gain of Function Mutation , Ligands , Binding Sites , Mutation , Receptors, G-Protein-Coupled/genetics , Protein Binding , Alopecia/genetics , Allosteric Site
5.
Dev Dyn ; 2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36038963

ABSTRACT

BACKGROUND: The somatopleure serves as the primordium of the amnion, an extraembryonic membrane surrounding the embryo. Recently, we have reported that amniogenic somatopleural cells (ASCs) not only form the amnion but also migrate into the embryo and differentiate into cardiomyocytes and vascular endothelial cells. However, detailed differentiation processes and final distributions of these intra-embryonic ASCs (hereafter referred to as iASCs) remain largely unknown. RESULTS: By quail-chick chimera analysis, we here show that iASCs differentiate into various cell types including cardiomyocytes, smooth muscle cells, cardiac interstitial cells, and vascular endothelial cells. In the pharyngeal region, they distribute selectively into the thyroid gland and differentiate into vascular endothelial cells to form intra-thyroid vasculature. Explant culture experiments indicated sequential requirement of fibroblast growth factor (FGF) and vascular endothelial growth factor (VEGF) signaling for endothelial differentiation of iASCs. Single-cell transcriptome analysis further revealed heterogeneity and the presence of hemangioblast-like cell population within ASCs, with a switch from FGF to VEGF receptor gene expression. CONCLUSION: The present study demonstrates novel roles of ASCss especially in heart and thyroid development. It will provide a novel clue for understanding the cardiovascular development of amniotes from embryological and evolutionary perspectives.

6.
Methods Mol Biol ; 2206: 103-127, 2021.
Article in English | MEDLINE | ID: mdl-32754814

ABSTRACT

For more than 2000 years, the avian embryo has helped scientists understand questions of developmental and cell biology. As early as 350 BC Aristotle described embryonic development inside a chicken egg (Aristotle, Generation of animals. Loeb Classical Library (translated), vol. 8, 1943). In the seventeenth century, Marcello Malpighi, referred to as the father of embryology, first diagramed the microscopic morphogenesis of the chick embryo, including extensive characterization of the cardiovascular system (Pearce Eur Neurol 58(4):253-255, 2007; West, Am J Physiol Lung Cell Mol Physiol 304(6):L383-L390, 2016). The ease of accessibility to the embryo and similarity to mammalian development have made avians a powerful system among model organisms. Currently, a unique combination of classical and modern techniques is employed for investigation of the vascular system in the avian embryo. Here, we will introduce the essential techniques of embryonic manipulation for experimental study in vascular biology.


Subject(s)
Chickens/physiology , Neovascularization, Physiologic/physiology , Quail/physiology , Animals , Chick Embryo , Embryo, Mammalian/physiology , Embryonic Development/physiology , Models, Animal
7.
Sci Rep ; 7(1): 8955, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827655

ABSTRACT

The somatopleure is the amniotic primordium in amniote development, but its boundary to the embryonic body at early embryonic stages and the fate of cells constituting this structure are not well characterized. It also remains unclear how cells behave during the demarcation between intra- and extra-embryonic tissues. Here we identify cellular alignments, which indicate two streams towards the sites of dorsal amniotic closure and ventral thoracic wall formation. A subpopulation of mesodermal cells moving ventrally from the somatopleural region adjacent to the base of the head fold enter the body of the embryo and distribute to the thoracic wall, pharyngeal arches and heart. These cells are induced to differentiate into vascular endothelial cells and cardiomyocytes possibly by FGF and BMP signaling, respectively. These results indicate that the somatopleure acting as the amniotic primordium also serves as a source of embryonic cells, which may contribute to cardiovascular development.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cardiovascular System/cytology , Fibroblast Growth Factors/metabolism , Germ Layers/cytology , Animals , Birds , Cardiovascular System/embryology , Cell Differentiation , Cell Lineage , Cells, Cultured , Chick Embryo , Ectoderm/cytology , Endothelial Cells/cytology , Mesoderm/cytology , Myocytes, Cardiac/cytology , Signal Transduction
8.
Dev Biol ; 409(1): 72-83, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26506449

ABSTRACT

Thyroid development and formation vary among species, but in most species the thyroid morphogenesis consists of five stages: specification, budding, descent, bilobation and folliculogenesis. The detailed mechanisms of these stages have not been fully clarified. During early development, the cranial neural crest (CNC) contributes to the thyroid gland. The removal of the postotic CNC (corresponding to rhombomeres 6, 7 and 8, also known as the cardiac neural crest) results in abnormalities of the cardiovascular system, thymus, parathyroid glands, and thyroid gland. To investigate the influence of the CNC on thyroid bilobation process, we divided the CNC into two regions, the postotic CNC and the preotic CNC (from the mesencephalon to rhombomere 5) regions and examined. We found that preotic CNC-ablated embryos had a unilateral thyroid lobe, and confirmed the presence of a single lobe or the absence of lobes in postotic CNC-ablated chick embryos. The thyroid anlage in each region-ablated embryos was of a normal size at the descent stage, but at a later stage, the thyroid in preotic CNC-ablated embryos was of a normal size, conflicting with a previous report in which the thyroid was reduced in size in the postotic CNC-ablated embryos. The postotic CNC cells differentiated into connective tissues of the thyroid in quail-to-chick chimeras. In contrast, the preotic CNC cells did not differentiate into connective tissues of the thyroid. We found that preotic CNC cells encompassed the thyroid anlage from the specification stage to the descent stage. Finally, we found that endothelin-1 and endothelin type A receptor-knockout mice and bosentan (endothelin receptor antagonist)-treated chick embryos showed bilobation anomalies that included single-lobe formation. Therefore, not only the postotic CNC, but also the preotic CNC plays an important role in thyroid morphogenesis.


Subject(s)
Neural Crest/cytology , Skull/cytology , Thyroid Gland/embryology , Animals , Bosentan , Branchial Region/blood supply , Cell Movement , Chick Embryo , Chickens , Endothelin-1/metabolism , Mice , Morphogenesis , Neovascularization, Physiologic , Quail , Signal Transduction , Sulfonamides
9.
Mech Dev ; 130(11-12): 553-66, 2013.
Article in English | MEDLINE | ID: mdl-23933587

ABSTRACT

Endothelin-1 (Edn1), originally identified as a vasoconstrictor peptide, is involved in the development of cranial/cardiac neural crest-derived tissues and organs. In craniofacial development, Edn1 binds to Endothelin type-A receptor (Ednra) to induce homeobox genes Dlx5/Dlx6 and determines the mandibular identity in the first pharyngeal arch. However, it remains unsolved whether this pathway is also critical for pharyngeal arch artery development to form thoracic arteries. Here, we show that the Edn1/Ednra signaling is involved in pharyngeal artery development by controlling the fate of neural crest cells through a Dlx5/Dlx6-independent mechanism. Edn1 and Ednra knock-out mice demonstrate abnormalities in pharyngeal arch artery patterning, which include persistent first and second pharyngeal arteries, resulting in additional branches from common carotid arteries. Neural crest cell labeling with Wnt1-Cre transgene and immunostaining for smooth muscle cell markers revealed that neural crest cells abnormally differentiate into smooth muscle cells at the first and second pharyngeal arteries of Ednra knock-out embryos. By contrast, Dlx5/Dlx6 knockout little affect the development of pharyngeal arch arteries and coronary arteries, the latter of which is also contributed by neural crest cells through an Edn-dependent mechanism. These findings indicate that the Edn1/Ednra signaling regulates neural crest differentiation to ensure the proper patterning of pharyngeal arch arteries, which is independent of the regional identification of the pharyngeal arches along the dorsoventral axis mediated by Dlx5/Dlx6.


Subject(s)
Arteries/metabolism , Body Patterning/genetics , Branchial Region/metabolism , Endothelin-1/genetics , Neural Crest/metabolism , Receptors, Endothelin/genetics , Animals , Arteries/abnormalities , Branchial Region/abnormalities , Cell Differentiation , Embryo, Mammalian , Endothelin-1/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homologous Recombination , Integrases/genetics , Integrases/metabolism , Mice , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neural Crest/abnormalities , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Receptors, Endothelin/metabolism , Signal Transduction
10.
Nat Commun ; 3: 1267, 2012.
Article in English | MEDLINE | ID: mdl-23232397

ABSTRACT

Neural crest cells constitute a multipotent cell population that gives rise to diverse cell lineages. The neural crest arising from the postotic hindbrain is known as the 'cardiac' neural crest, and contributes to the great vessels and outflow tract endocardial cushions, but the neural crest contribution to structures within the heart remains largely controversial. Here we demonstrate that neural crest cells from the preotic region migrate into the heart and differentiate into coronary artery smooth muscle cells. Preotic neural crest cells preferentially distribute to the conotruncal region and interventricular septum. Ablation of the preotic neural crest causes abnormalities in coronary septal branch and orifice formation. Mice and chicks lacking endothelin signalling show similar abnormalities in the coronary artery, indicating its involvement in neural crest-dependent coronary artery formation. This is the first report that reveals the preotic neural crest contribution to heart development and smooth muscle heterogeneity within a coronary artery.


Subject(s)
Coronary Vessels/embryology , Endothelins/physiology , Muscle, Smooth, Vascular/embryology , Neural Crest/embryology , Signal Transduction/physiology , Animals , Chick Embryo , Coronary Vessels/growth & development , Coronary Vessels/physiology , Coturnix/embryology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/physiology , Neural Crest/cytology , Neural Crest/physiology
11.
Gene Expr Patterns ; 11(7): 371-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21565284

ABSTRACT

The endothelin (Edn) system plays pleiotropic roles in renal function and various disease processes through two distinct G protein-coupled receptors, Edn receptors type-A (Ednra) and type-B (Ednrb). However, difficulties in the accurate identification of receptor-expressing cells in situ have made it difficult to dissect their diverse action in renal (patho)physiology. We have recently established mouse lines in which lacZ and EGFP are 'knocked-in' to the Ednra locus to faithfully mark Ednra-expressing cells. Here we analyzed these mice for their expression in the kidney to characterize Ednra-expressing cells. Ednra expression was first observed in undifferentiated mesenchymal cells around the ureteric bud at E12.5. Thereafter, Ednra expression was widely observed in vascular smooth muscle cells, JG cells and mesenchymal cells in the interstitium. After growth, the expression became confined to vascular smooth muscle cells, pericytes and renin-producing JG cells. By contrast, most cells in the nephron and vascular endothelial cells did not express Ednra. These results indicate that Ednra expression may be linked with non-epithelial fate determination and differentiation of metanephric mesenchyme. Ednra-lacZ/EGFP knock-in mice may serve as a useful tool in studies on renal function and pathophysiology of various renal diseases.


Subject(s)
Kidney/cytology , Kidney/metabolism , Receptor, Endothelin A/genetics , Animals , Cell Differentiation , Gene Expression Regulation , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Nephrons/cytology , Nephrons/metabolism , Pericytes/cytology , Pericytes/metabolism
12.
Development ; 137(22): 3823-33, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20929948

ABSTRACT

The avian and mammalian heart originates from two distinct embryonic regions: an early differentiating first heart field and a dorsomedially located second heart field. It remains largely unknown when and how these subdivisions of the heart field divide into regions with different fates. Here, we identify in the mouse a subpopulation of the first (crescent-forming) field marked by endothelin receptor type A (Ednra) gene expression, which contributes to chamber myocardium through a unique type of cell behavior. Ednra-lacZ/EGFP-expressing cells arise in the ventrocaudal inflow region of the early linear heart tube, converge to the midline, move anteriorly along the outer curvature and give rise to chamber myocardium mainly of the left ventricle and both atria. This movement was confirmed by fluorescent dye-labeling and transplantation experiments. The Ednra-lacZ/EGFP-expressing subpopulation is characterized by the presence of Tbx5-expressing cells. Ednra-null embryonic hearts often demonstrate hypoplasia of the ventricular wall, low mitotic activity and decreased Tbx5 expression with reciprocal expansion of Tbx2 expression. Conversely, endothelin 1 stimulates ERK phosphorylation and Tbx5 expression in the early embryonic heart. These results indicate that early Ednra expression defines a subdomain of the first heart field contributing to chamber formation, in which endothelin 1/Ednra signaling is involved. The present finding provides an insight into how subpopulations within the crescent-forming (first) heart field contribute to the coordination of heart morphogenesis through spatiotemporally defined cell movements.


Subject(s)
Gene Expression Regulation, Developmental , Heart/embryology , Myocardium/metabolism , Organogenesis , Receptor, Endothelin A/metabolism , Animals , Embryo, Mammalian/metabolism , Endothelins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knock-In Techniques , Heart Ventricles/embryology , Mice , Mitosis , Phosphorylation , Receptor, Endothelin A/genetics , T-Box Domain Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 105(48): 18806-11, 2008 Dec 02.
Article in English | MEDLINE | ID: mdl-19017795

ABSTRACT

Articulated jaws are highly conserved structures characteristic of gnathostome evolution. Epithelial-mesenchymal interactions within the first pharyngeal arch (PA1) instruct cephalic neural crest cells (CNCCs) to form the different skeletal elements of the jaws. The endothelin-1 (Edn1)/endothelin receptor type-A (Ednra)-->Dlx5/6-->Hand2 signaling pathway is necessary for lower jaw formation. Here, we show that the Edn1 signaling is sufficient for the conversion of the maxillary arch to mandibular identity. Constitutive activation of Ednra induced the transformation of upper jaw, maxillary, structures into lower jaw, mandibular, structures with duplicated Meckel's cartilage and dermatocranial jaws constituted by 4 dentary bones. Misexpression of Hand2 in the Ednra domain caused a similar transformation. Skeletal transformations are accompanied by neuromuscular remodeling. Ednra is expressed by most CNCCs, but its constitutive activation affects predominantly PA1. We conclude that after migration CNCCs are not all equivalent, suggesting that their specification occurs in sequential steps. Also, we show that, within PA1, CNCCs are competent to form both mandibular and maxillary structures and that an Edn1 switch is responsible for the choice of either morphogenetic program.


Subject(s)
Endothelin-1/metabolism , Mandible/embryology , Maxilla/embryology , Receptor, Endothelin A/metabolism , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning , Endothelin-1/genetics , Gene Expression Regulation, Developmental , Mandible/anatomy & histology , Mandible/metabolism , Maxilla/anatomy & histology , Maxilla/metabolism , Mice , Mice, Transgenic , Receptor, Endothelin A/genetics
14.
Development ; 135(4): 755-65, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18199583

ABSTRACT

The endothelin (Edn) system comprises three ligands (Edn1, Edn2 and Edn3) and their G-protein-coupled type A (Ednra) and type B (Ednrb) receptors. During embryogenesis, the Edn1/Ednra signaling is thought to regulate the dorsoventral axis patterning of pharyngeal arches via Dlx5/Dlx6 upregulation. To further clarify the underlying mechanism, we have established mice in which gene cassettes can be efficiently knocked-in into the Ednra locus using recombinase-mediated cassette exchange (RMCE) based on the Cre-lox system. The first homologous recombination introducing mutant lox-flanked Neo resulted in homeotic transformation of the lower jaw to an upper jaw, as expected. Subsequent RMCE-mediated knock-in of lacZ targeted its expression to the cranial/cardiac neural crest derivatives as well as in mesoderm-derived head mesenchyme. Knock-in of Ednra cDNA resulted in a complete rescue of craniofacial defects of Ednra-null mutants. By contrast, Ednrb cDNA could not rescue them except for the most distal pharyngeal structures. At early stages, the expression of Dlx5, Dlx6 and their downstream genes was downregulated and apoptotic cells distributed distally in the mandible of Ednrb-knock-in embryos. These results, together with similarity in craniofacial defects between Ednrb-knock-in mice and neural-crest-specific Galpha(q)/Galpha(11)-deficient mice, indicate that the dorsoventral axis patterning of pharyngeal arches is regulated by the Ednra-selective, G(q)/G(11)-dependent signaling, while the formation of the distal pharyngeal region is under the control of a G(q)/G(11)-independent signaling, which can be substituted by Ednrb. This RMCE-mediated knock-in system can serve as a useful tool for studies on gene functions in craniofacial development.


Subject(s)
Branchial Region/embryology , Endothelin-1/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Mutagenesis, Insertional , Receptor, Endothelin A/metabolism , Recombinases/metabolism , Signal Transduction , Animals , Branchial Region/metabolism , Craniofacial Abnormalities , DNA, Complementary , Embryo, Mammalian/abnormalities , Embryo, Mammalian/enzymology , Embryonic Development , Gene Expression Regulation, Developmental , Mesoderm/embryology , Mesoderm/enzymology , Mice , Mice, Inbred C57BL , Models, Biological , Muscle, Skeletal/abnormalities , Neural Crest/embryology , Neural Crest/enzymology , Phenotype , Receptor, Endothelin A/deficiency , Receptor, Endothelin A/genetics , Receptor, Endothelin B/metabolism , beta-Galactosidase/metabolism
15.
Dev Growth Differ ; 47(8): 501-10, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16287482

ABSTRACT

Musashi-1 (Msi-1) is an RNA-binding protein that plays key roles in the maintenance of neural stem cell states and in their differentiation into neural cells. Msi-1 has also been proposed as a candidate marker gene of mammalian intestinal stem cells and their immediate lineages. In this study, we examined Msi-1 expression in the small intestine and the stomach of both chicken and mouse during embryonic, fetal and postnatal development. In addition, we analyzed the expression of c-hairy-1, a chicken homologue of mouse Hes1, and assessed the proliferative activity of the cells expressing both of these factors. Significantly, during the development of these digestive organs in both species Msi-1 expression showed dynamic changes, suggesting that it is important for digestive organ development, particularly for epithelial differentiation. Based on our observations of the expression patterns of Msi-1 and c-hairy-1 in the adult small intestine, we speculate that Msi-1 is also a stem cell marker of the chicken small intestinal epithelium.


Subject(s)
Avian Proteins/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Cycle Proteins/biosynthesis , Homeodomain Proteins/biosynthesis , Intestine, Small/cytology , Intestine, Small/embryology , Nerve Tissue Proteins/biosynthesis , RNA-Binding Proteins/biosynthesis , Stomach/cytology , Stomach/embryology , Animals , Avian Proteins/genetics , Avian Proteins/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , Chick Embryo , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Intestine, Small/metabolism , Mice , Nerve Tissue Proteins/genetics , Proventriculus/cytology , Proventriculus/embryology , Proventriculus/metabolism , Proventriculus/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology , Transcription Factor HES-1
SELECTION OF CITATIONS
SEARCH DETAIL
...