11,160 results on '"ectoderm"'
Search Results
152. New Transcription Factor Study Results from First Affiliated Hospital of Shandong First Medical University Described (Transcriptional differential analysis of ocular surface ectoderm and surface ectoderm).
- Subjects
TRANSCRIPTION factors ,ECTODERM ,SURFACE analysis ,CELL cycle regulation ,PROTEIN genetics - Abstract
Researchers from the First Affiliated Hospital of Shandong First Medical University in China conducted a study to identify transcriptional differences between the ocular surface ectoderm (OSE) and surface ectoderm (SE) using RNA-seq. They analyzed differentially expressed genes (DEGs) between OSE and SE cells and performed gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. The study found that OSE cells had specific gene expression patterns related to ion transport, axon development, and synaptic transmission regulation. The findings provide insights into the development and induction of OSE and corneal epithelial cells. [Extracted from the article]
- Published
- 2024
153. CFL1-dependent dynamicity of surface ectoderm filopodia-like protrusions increases neurulation zippering speed in mice (Updated March 23, 2024).
- Subjects
ECTODERM ,NEURAL tube ,DEVELOPMENTAL biology ,NEURAL tube defects - Abstract
According to a preprint abstract, researchers have found that the protein CFL1 plays a crucial role in the closure of the neural tube in mice embryos. The study shows that CFL1 is necessary for the proper localization of F-actin in the cranial neuroepithelium, and its absence leads to dysmorphic headfolds and incomplete closure of the neural tube. Additionally, CFL1 expression in the surface ectoderm enhances the speed of zippering during spinal neurulation, and embryos lacking surface ectoderm CFL1 have a decreased zippering speed and an increased incidence of spina bifida. These findings suggest that CFL1 regulates the cellular-level dynamics of filopodial extensions, which in turn affects the tissue-level zippering speed required for complete neural tube closure. [Extracted from the article]
- Published
- 2024
154. Phenotypic and Genotypic Features of Thai Patients With Nonsyndromic Tooth Agenesis and WNT10A Variants.
- Author
-
Kanchanasevee, Charinya, Sriwattanapong, Kanokwan, Theerapanon, Thanakorn, Thaweesapphithak, Sermporn, Chetruengchai, Wanna, Porntaveetus, Thantrira, and Shotelersuk, Vorasuk
- Subjects
HYPODONTIA ,ASIANS ,IMPACTION of teeth ,INCISORS - Abstract
Tooth agenesis is one of the most common orodental anomalies that demonstrate phenotypic and genotypic heterogeneity with a prevalence of 2.5%–7%. Mutations in WNT10A have been proposed to be the most common cause of nonsyndromic tooth agenesis (NSTA). The aim of this study was to characterize the dental features and genetic variants of NSTA in a Thai population. We recruited 13 unrelated patients with NSTA who attended the Faculty of Dentistry, Chulalongkorn University, Thailand, from 2017 to 2019. All 13 underwent whole exome sequencing that identified likely pathogenic genetic variants, all in WNT10A , in five patients. All five patients had second premolar agenesis, while three also had absent or peg-shaped upper lateral incisors. Patient 1 possessed a novel heterozygous duplication, c.916_918dupAAC (p.Asn306dup) in WNT10A. Patients 2 and 3 harbored a heterozygous and homozygous c.637G > A (p.Gly213Ser) in WNT10A , respectively. Patients 4 possessed a heterozygous c.511C > T (p.Arg171Cys) in WNT10A. Patient 5 harbored a homozygous c.511C > T (p.Arg171Cys) in WNT10A and a novel heterozygous c.413A > T (p.Asn138Ile) in EDARADD , suggesting digenic inheritance. We recruited another 18 family members of these five patients. Out of 23 participants, homozygous WNT10A variants were identified in 2 patients and heterozygous variants in 17 individuals. Both homozygous patients had NSTA. Eight out of 17 heterozygous individuals (8/17) had NSTA or a peg-shaped lateral incisor, indicating a 47% penetrance of the heterozygous variants or 53% (10/19) penetrance of either homozygous or heterozygous variants in WNT10A. The frequencies of the c.511C > T in our in-house 1,876 Thai exome database, Asian populations, and non-Asian populations were 0.016, 0.005–0.033, and 0.001, respectively; while those of the c.637G > A were 0.016, 0.004–0.029, and 0.000, respectively. In conclusion, our study reports two novel variants with one each in WNT10A and EDARADD , expanding the genotypic spectra of NSTA. Second premolar agenesis is a common phenotype in affected individuals with variants in WNT10A ; however, its penetrance is incomplete. Lastly, the different frequencies of WNT10A variants, c.511C > T and c.637G > A, in diverse populations might contribute to the prevalence range of NSTA between continents. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
155. Mcrs1 interacts with Six1 to influence early craniofacial and otic development.
- Author
-
Neilson, Karen M., Keer, Stephanie, Bousquet, Nicole, Macrorie, Olivia, Majumdar, Himani D., Kenyon, Kristy L., Alfandari, Dominique, and Moody, Sally A.
- Subjects
- *
NEURAL crest , *GENE expression , *TRANSCRIPTION factors , *ECTODERM , *BRANCHIAL arch - Abstract
The Six1 transcription factor plays a major role in craniofacial development. Mutations in SIX1 and its co-factor, EYA1 , are causative for about 50% of Branchio-otic/Branchio-oto-renal syndrome (BOR) patients, who are characterized by variable craniofacial, otic and renal malformations. We previously screened for other proteins that might interact with Six1 to identify additional genes that may play a role in BOR, and herein characterize the developmental role of one of them, Microspherule protein 1 (Mcrs1). We found that in cultured cells, Mcrs1 bound to Six1 and in both cultured cells and embryonic ectoderm reduced Six1-Eya1 transcriptional activation. Knock-down of Mcrs1 in embryos caused an expansion of the domains of neural plate genes and two genes expressed in both the neural plate and neural crest (zic1 , zic2). In contrast, two other genes expressed in pre-migratory neural crest (foxd3 , sox9) were primarily reduced. Cranial placode genes showed a mixture of expanded and diminished expression domains. At larval stages, loss of Mcrs1 resulted in a significant reduction of otic vesicle gene expression concomitant with a smaller otic vesicle volume. Experimentally increasing Mcrs1 above endogenous levels favored the expansion of neural border and neural crest gene domains over cranial placode genes; it also reduced otic vesicle gene expression but not otic vesicle volume. Co-expression of Mcrs1 and Six1 as well as double knock-down and rescue experiments establish a functional interaction between Mcrs1 and Six1 in the embryo, and demonstrate that this interaction has an important role in the development of craniofacial tissues including the otic vesicle. • Mcrs1 binds to Six1 and alters transcriptional activation. • Knockdown of Mcrs1 alters cranial neural gene expression domains. • Knockdown of Mcrs1 reduces the size of the otic vesicle. • Mcrs1 and Six1 interact to effect craniofacial development. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
156. Accelerated differentiation of human pluripotent stem cells into neural lineages via an early intermediate ectoderm population.
- Author
-
Walsh, Patrick, Truong, Vincent, Nayak, Sushmita, Saldías Montivero, Marietta, Low, Walter C., Parr, Ann M., and Dutton, James R.
- Subjects
CELL differentiation ,PLURIPOTENT stem cells ,ECTODERM ,CELLULAR therapy ,ANTIOXIDANTS - Abstract
Differentiation of human pluripotent stem cells (hPSCs) into ectoderm provides neurons and glia useful for research, disease modeling, drug discovery, and potential cell therapies. In current protocols, hPSCs are traditionally differentiated into an obligate rostro‐dorsal ectodermal fate expressing PAX6 after 6 to 12 days in vitro when protected from mesendoderm inducers. This rate‐limiting step has performed a long‐standing role in hindering the development of rapid differentiation protocols for ectoderm‐derived cell types, as any protocol requires 6 to 10 days in vitro to simply initiate. Here, we report efficient differentiation of hPSCs into a naive early ectodermal intermediate within 24 hours using combined inhibition of bone morphogenic protein and fibroblast growth factor signaling. The induced population responds immediately to morphogen gradients to upregulate rostro‐caudal neurodevelopmental landmark gene expression in a generally accelerated fashion. This method can serve as a new platform for the development of novel, rapid, and efficient protocols for the manufacture of hPSC‐derived neural lineages. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
157. Sublingual Dermoid Cysts of Oral Cavity.
- Author
-
THOTA, RAMVIHARI, PERIASAMY, SENTHILNATHAN, and N., MAHATHI
- Subjects
- *
DERMOID cysts , *DENTAL caries , *EPIDERMAL cyst , *ECTODERM , *CHILD patients - Abstract
Dermoid cysts in the oral cavity are very rare and unusual lesions. Their etiology is not yet clear but can be associated with trapped cells as a result of the inclusion error resulting in the development of the ectoderm, mesoderm, and endoderm tissues. The aim of this case report is to evidence the presence of a dermoid cyst in the floor of the mouth of a pediatric patient which was surgically removed. In the present case, the lesion showed soft consistency, regular borders, smooth surface, and the same color as the adjacent mucosa, asymptomatic, and measuring 3 × 3 cm in its greatest diameter. The initial diagnosis was ranula in consequence of the similarity with clinical characteristics and localization. After surgical removal of the lesion, a fibrotic capsule was identified with a friable material with intensive yellow color. The microscopic exam revealed a cystic lesion with a cavity lined by squamous stratified epithelium hyper orthokeratinized. Cutaneous attachments, such as sebaceous glands and hair follicles, were present in connective adjacent tissue. Surgical intervention is elective in these situations. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
158. Optic vesicle morphogenesis requires primary cilia.
- Author
-
Fiore, Luciano, Takata, Nozomu, Acosta, Sandra, Ma, Wanshu, Pandit, Tanushree, Oxendine, Michael, and Oliver, Guillermo
- Subjects
- *
CILIA & ciliary motion , *JOUBERT syndrome , *ECTODERM , *EMBRYOS , *EPITHELIUM , *MORPHOGENESIS - Abstract
Arl13b is a gene known to regulate ciliogenesis. Functional alterations in this gene's activity have been associated with Joubert syndrome. We found that in Arl13 null mouse embryos the orientation of the optic cup is inverted, such that the lens is abnormally surrounded by an inverted optic cup whose retina pigmented epithelium is oddly facing the surface ectoderm. Loss of Arl13b leads to the disruption of optic vesicle's patterning and expansion of ventral fates. We show that this phenotype is consequence of miss-regulation of Sonic hedgehog (Shh) signaling and demonstrate that the Arl13b −/− eye phenotype can be rescued by deletion of Gli2, a downstream effector of the Shh pathway. This work identified an unexpected role of primary cilia during the morphogenetic movements required for the formation of the eye. • Arl13b is a gene known to regulate ciliogenesis. • Arl13 null mouse embryos the orientation of the optic cup is inverted. • The phenotype is consequence of miss-regulation of Sonic hedgehog signaling. • The Arl13b −/− eye phenotype can be rescued by deletion of Gli2. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
159. Tbx2 mediates dorsal patterning and germ layer suppression through inhibition of BMP/GDF and Activin/Nodal signaling.
- Author
-
Reich, Shoshana, Kayastha, Peter, Teegala, Sushma, and Weinstein, Daniel C.
- Subjects
- *
EPIBLAST , *ACTIVIN , *MESODERM , *DNA-binding proteins , *ECTODERM , *GROWTH regulators - Abstract
Background: Members of the T-box family of DNA-binding proteins play a prominent role in the differentiation of the three primary germ layers. VegT, Brachyury, and Eomesodermin function as transcriptional activators and, in addition to directly activating the transcription of endoderm- and mesoderm-specific genes, serve as regulators of growth factor signaling during induction of these germ layers. In contrast, the T-box gene, tbx2, is expressed in the embryonic ectoderm, where Tbx2 functions as a transcriptional repressor and inhibits mesendodermal differentiation by the TGFβ ligand Activin. Tbx2 misexpression also promotes dorsal ectodermal fate via inhibition of the BMP branch of the TGFβ signaling network. Results: Here, we report a physical association between Tbx2 and both Smad1 and Smad2, mediators of BMP and Activin/Nodal signaling, respectively. We perform structure/function analysis of Tbx2 to elucidate the roles of both Tbx2-Smad interaction and Tbx2 DNA-binding in germ layer suppression. Conclusion: Our studies demonstrate that Tbx2 associates with intracellular mediators of the Activin/Nodal and BMP/GDF pathways. We identify a novel repressor domain within Tbx2, and have determined that Tbx2 DNA-binding activity is required for repression of TGFβ signaling. Finally, our data also point to overlapping yet distinct mechanisms for Tbx2-mediated repression of Activin/Nodal and BMP/GDF signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
160. Rspo2 antagonizes FGF signaling during vertebrate mesoderm formation and patterning.
- Author
-
Reis, Alice H. and Sokol, Sergei Y.
- Subjects
- *
MESODERM , *EMBRYOLOGY , *WNT signal transduction , *ECTODERM , *XENOPUS , *SIGNAL processing - Abstract
R-spondins are a family of secreted proteins that play important roles in embryonic development and cancer. R-spondins have been shown to modulate the Wnt pathway; however, their involvement in other developmental signaling processes have remained largely unstudied. Here, we describe a novel function of Rspo2 in FGF pathway regulation in vivo. Overexpressed Rspo2 inhibited elongation of Xenopus ectoderm explants and Erk1 activation in response to FGF. By contrast, the constitutively active form of Mek1 stimulated Erk1 even in the presence of Rspo2, suggesting that Rspo2 functions upstream of Mek1. The observed inhibition of FGF signaling was accompanied by the downregulation of the FGF target genes tbxt/brachyury and cdx4, which mediate anterioposterior axis specification. Importantly, these target genes were upregulated in Rspo2-depleted explants. The FGF inhibitory activity was mapped to the thrombospondin type 1 region, contrasting the known function of the Furin-like domains in Wnt signaling. Further domain analysis revealed an unexpected intramolecular interaction that might control Rspo2 signaling output. We conclude that, in addition to its role in Wnt signaling, Rspo2 acts as an FGF antagonist during mesoderm formation and patterning. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
161. EMBRYONIC DEVELOPMENT OF THE INNER EAR IN THE QUAIL BIRD COTURNIX COTARNIX (LINNAEUS, 1758).
- Author
-
Abed, Asmaa Basheer
- Subjects
QUAILS ,EMBRYOLOGY ,EPIDERMIS ,ECTODERM ,MESOTHELIUM - Abstract
The study concerned the embryonic development of inner ear of Quail Coturnix coturnix (Linnaeus, 1758). The study is done in the period between (20 February and 20 October) in Department of Biology, College of Education for Pure Science (Ibn Al-Haitham), University of Baghdsd. Ten adult quail Coturnix coturnix (Linnaeus, 1758). The first sign of the formation of sensory part of the ear is evident in the embryo of the quail bird at 38 hours incubation. At this stage apairs of thickening on the sides of hind brain is known as the otic placodes of the proximal epidermis in the head region, these two placodes fall below the general level of the apparent ectoderm by invagination process at (40) hours incubation to be walls of pair spaces known as auditory pits where the walls are in the first composition is continuos with surface ectoderm and open wide open, after that the presure is sparated from the surface ectoderm. The ectoderm then clears the surface to become auditory vesicles located between ectoderm and hind brain (Myeloncephalon) where the hearing pressure deepens and grows with in the mesothelium, where the ectoderm separates when the fetus reaches (48) hours incibation, and becomes close to the lateral wall of the hind brain (Myeloncephalon). [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
162. Congenital pituitary hypoplasia model demonstrates hypothalamic OTX2 regulation of pituitary progenitor cells.
- Author
-
Ryusaku Matsumoto, Hidetaka Suga, Takashi Aoi, Hironori Bando, Hidenori Fukuoka, Genzo Iguchi, Satoshi Narumi, Tomonobu Hasegawa, Keiko Muguruma 10 11, Wataru Ogawa, and Yutaka Takahashi
- Subjects
- *
PROGENITOR cells , *INDUCED pluripotent stem cells , *HORMONE deficiencies , *HYPOTHALAMUS , *PITUITARY diseases , *ECTODERM - Abstract
Pituitary develops from oral ectoderm in contact with adjacent ventral hypothalamus. Impairment in this process results in congenital pituitary hypoplasia (CPH); however, there have been no human disease models for CPH thus far, prohibiting the elucidation of the underlying mechanisms. In this study, we established a disease model of CPH using patient-derived induced pluripotent stem cells (iPSCs) and 3D organoid technique, in which oral ectoderm and hypothalamus develop simultaneously. Interestingly, patient iPSCs with a heterozygous mutation in the orthodenticle homeobox 2 (OTX2) gene showed increased apoptosis in the pituitary progenitor cells, and the differentiation into pituitary hormone-producing cells was severely impaired. As an underlying mechanism, OTX2 in hypothalamus, not in oral ectoderm, was essential for progenitor cell maintenance by regulating LHX3 expression in oral ectoderm via FGF10 expression in the hypothalamus. Convincingly, the phenotype was reversed by the correction of the mutation, and the haploinsufficiency of OTX2 in control iPSCs revealed a similar phenotype, demonstrating that this mutation was responsible. Thus, we established an iPSC-based congenital pituitary disease model, which recapitulated interaction between hypothalamus and oral ectoderm and demonstrated the essential role of hypothalamic OTX2. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
163. Dorsoventral decoupling of Hox gene expression underpins the diversification of molluscs.
- Author
-
Pin Huan, Qian Wang, Sujian Tan, and Baozhong Liu
- Subjects
- *
HOMEOBOX genes , *GENE expression , *MOLLUSKS , *ECTODERM , *GASTROPODA - Abstract
In contrast to the Hox genes in arthropods and vertebrates, those in molluscs show diverse expression patterns with differences reported among lineages. Here, we investigate 2 phylogenetically distant molluscs, a gastropod and a polyplacophoran, and show that the Hox expression in both species can be divided into 2 categories. The Hox expression in the ventral ectoderm generally shows a canonical staggered pattern comparable to the patterns of other bilaterians and likely contributes to ventral patterning, such as neurogenesis. The other category of Hox expression on the dorsal side is strongly correlated with shell formation and exhibits lineage-specific characteristics in each class of mollusc. This generalized model of decoupled dorsoventral Hox expression is compatible with known Hox expression data from other molluscan lineages and may represent a key characteristic of molluscan Hox expression. These results support the concept of widespread staggered Hox expression in Mollusca and reveal aspects that may be related to the evolutionary diversification of molluscs. We propose that dorsoventral decoupling of Hox expression allowed lineage-specific dorsal and ventral patterning, which may have facilitated the evolution of diverse body plans in different molluscan lineages. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
164. Swelling in the floor of the mouth: A diagnostic dilemma.
- Author
-
Datta, Ginni and Yadav, Ankita
- Subjects
EPIDERMAL cyst ,EPIBLAST ,DILEMMA ,MOUTH ,ECTODERM - Abstract
Epidermoid cysts are abnormal structures originating from abnormal ectoderm and are benign and slow growing. These are found anywhere in the body. They arise during embryonic fusion due to entrapment of epithelial remnants and sometimes due to faulty implantation of the epithelium. The cysts can be epidermoid (squamous epithelium), true dermoid (skin apppendages), and teratoid (all germ layers). They are usually diagnosed by fine-needle cytological aspiration and require imaging to plan line of treatment. Diagnostic dilemma is always an issue as cystic lesions are commonly found in the head-and-neck region. Here, we present a case of large epidermoid cyst involving sublingual and submandibular region. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
165. PRDM1 controls the sequential activation of neural, neural crest and sensory progenitor determinants.
- Author
-
Prajapati, Ravindra S., Hintze, Mark, and Streit, Andrea
- Subjects
- *
NEURAL crest , *EMBRYOLOGY , *EPIBLAST , *ECTODERM - Abstract
During early embryogenesis, the ectoderm is rapidly subdivided into neural, neural crest and sensory progenitors. How the onset of lineage determinants and the loss of pluripotency markers are temporally and spatially coordinated in vivo is still debated. Here, we identify a crucial role for the transcription factor PRDM1 in the orderly transition from epiblast to defined neural lineages in chick. PRDM1 is initially expressed broadly in the entire epiblast, but becomes gradually restricted as cell fates are specified. We find that PRDM1 is required for the loss of some pluripotency markers and the onset of neural, neural crest and sensory progenitor specifier genes. PRDM1 directly activates their expression by binding to their promoter regions and recruiting the histone demethylase Kdm4a to remove repressive histone marks. However, once neural lineage determinants become expressed, they in turn repress PRDM1, whereas prolonged PRDM1 expression inhibits neural, neural crest and sensory progenitor genes, suggesting that its downregulation is necessary for cells to maintain their identity. Therefore, PRDM1 plays multiple roles during ectodermal cell fate allocation. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
166. Localization of Tfap2β , Casq2 , Penk , Zic1 , and Zic3 Expression in the Developing Retina, Muscle, and Sclera of the Embryonic Mouse Eye.
- Author
-
Wan, Yong, White, Casey, Robert, Nadine, Rogers, Matthew B., and Szabo-Rogers, Heather L.
- Subjects
PROTEIN expression ,ECTODERM ,MESENCHYME ,TRANSCRIPTION factors ,NEURAL development - Abstract
Optic development involves sequential interactions between several different tissue types, including the overlying ectoderm, adjacent mesoderm, and neural crest mesenchyme and the neuroectoderm. In an ongoing expression screen, we identified that Tfap2β, Casq2, Penk, Zic1, and Zic3 are expressed in unique cell types in and around the developing eye. Tfap2β, Zic1, and Zic3 are transcription factors, Casq2 is a calcium binding protein and Penk is a neurotransmitter. Tfap2β, Zic1, and Zic3 have reported roles in brain and craniofacial development, while Casq2 and Penk have unknown roles. These five genes are expressed in the major tissue types in the eye, including the muscles, nerves, cornea, and sclera. Penk expression is found in the sclera and perichondrium. At E12.5 and E15.5, the extra-ocular muscles express Casq2, the entire neural retina expresses Zic1, and Zic3 is expressed in the optic disk and lip of the optic cup. The expression of Tfap2β expanded from corneal epithelium to the neural retina between E12.5 to E15.5. These genes are expressed in similar domains as Hedgehog (Gli1, and Ptch1) and the Wnt (Lef1) pathways. The expression patterns of these five genes warrant further study to determine their role in eye morphogenesis: [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
167. Xenopus Sox3 activates sox2 and geminin and indirectly represses Xvent2 expression to induce neural progenitor formation at the expense of non-neural ectodermal derivatives
- Author
-
Rogers, Crystal D, Harafuji, Naoe, Archer, Tenley, Cunningham, Doreen D, and Casey, Elena S
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Genetics ,Neurosciences ,Stem Cell Research ,Pediatric ,Stem Cell Research - Embryonic - Non-Human ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Animals ,Biomarkers ,Bone Morphogenetic Proteins ,Carrier Proteins ,Cell Differentiation ,Cell Proliferation ,Ectoderm ,Embryo ,Nonmammalian ,Gene Expression Regulation ,Developmental ,Homeodomain Proteins ,Neurons ,SOXB1 Transcription Factors ,Signal Transduction ,Stem Cells ,Transcription Factors ,Xenopus Proteins ,Xenopus laevis ,Sox3 ,Sox2 ,Neural progenitors ,Neural induction ,Geminin ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
The SRY-related, HMG box SoxB1 transcription factors are highly homologous, evolutionarily conserved proteins that are expressed in neuroepithelial cells throughout neural development. SoxB1 genes are down-regulated as cells exit the cell-cycle to differentiate and are considered functionally redundant in maintaining neural precursor populations. However, little is known about Sox3 function and its mode of action during primary neurogenesis. Using gain and loss-of-function studies, we analyzed Sox3 function in detail in Xenopus early neural development and compared it to that of Sox2. Through these studies we identified the first targets of a SoxB1 protein during primary neurogenesis. Sox3 functions as an activator to induce expression of the early neural genes, sox2 and geminin in the absence of protein synthesis and to indirectly inhibit the Bmp target Xvent2. As a result, Sox3 increases cell proliferation, delays neurogenesis and inhibits epidermal and neural crest formation to expand the neural plate. Our studies indicate that Sox3 and 2 have many similar functions in this process including the ability to activate expression of geminin in naïve ectodermal explants. However, there are some differences; Sox3 activates the expression of sox2, while Sox2 does not activate expression of sox3 and sox3 is uniquely expressed throughout the ectoderm prior to neural induction suggesting a role in neural competence. With morpholino-mediated knockdown of Sox3, we demonstrate that it is required for induction of neural tissue by BMP inhibition. Together these data indicate that Sox3 has multiple roles in early neural development including as a factor required for nogginmediated neural induction.
- Published
- 2009
168. Neural Crest
- Author
-
Thattaliyath, Bijoy, Hutson, Mary, Rickert-Sperling, Silke, editor, Kelly, Robert G., editor, and Driscoll, David J., editor
- Published
- 2016
- Full Text
- View/download PDF
169. First-Trimester Embryology: An Overview
- Author
-
Jones, Cresta W., Penzkover, Deborah, Pollard, Rachel, Kuhlmann, Randall S., and Abramowicz, Jacques S., editor
- Published
- 2016
- Full Text
- View/download PDF
170. Craniofacial Anomalies
- Author
-
Chu, Eugenia, Wong, Brian J.-F, editor, Arnold, Michelle G., editor, and Boeckmann, Jacob O., editor
- Published
- 2016
- Full Text
- View/download PDF
171. Patterning of the Brain, Neural Proliferation, and Migration
- Author
-
Lagercrantz, Hugo and Lagercrantz, Hugo
- Published
- 2016
- Full Text
- View/download PDF
172. Neural plate progenitors give rise to both anterior and posterior pituitary cells.
- Author
-
Chen, Qiyu, Leshkowitz, Dena, Li, Hanjie, van Impel, Andreas, Schulte-Merker, Stefan, Amit, Ido, Rizzoti, Karine, and Levkowitz, Gil
- Subjects
- *
PITUITARY gland , *ANTERIOR pituitary gland , *CELL differentiation , *PROGENITOR cells , *ECTODERM , *TRANSCRIPTOMES - Abstract
The pituitary is the master neuroendocrine gland, which regulates body homeostasis. It consists of the anterior pituitary/adenohypophysis harboring hormones producing cells and the posterior pituitary/neurohypophysis, which relays the passage of hormones from the brain to the periphery. It is accepted that the adenohypophysis originates from the oral ectoderm (Rathke's pouch), whereas the neural ectoderm contributes to the neurohypophysis. Single-cell transcriptomics of the zebrafish pituitary showed that cyp26b1 -positive astroglial pituicytes of the neurohypophysis and prop1 -positive adenohypophyseal progenitors expressed common markers implying lineage relatedness. Genetic tracing identifies that, in contrast to the prevailing dogma, neural plate precursors of zebrafish (her4.3 +) and mouse (Sox1+) contribute to both neurohypophyseal and a subset of adenohypophyseal cells. Pituicyte-derived retinoic-acid-degrading enzyme Cyp26b1 fine-tunes differentiation of prop1+ progenitors into hormone-producing cells. These results challenge the notion that adenohypophyseal cells are exclusively derived from non-neural ectoderm and demonstrate that crosstalk between neuro- and adeno-hypophyseal cells affects differentiation of pituitary cells. [Display omitted] • Pituicytes, the resident neurohypophyseal astroglia, are conserved among vertebrates • Pituicytes and adenohypophyseal progenitors are closely related cell types • Neuroectodermal cells contribute to both neurohypophyseal and subsets of adenohypophyseal cells • Pituicyte-derived Cyp26b1 limits transition of Prop1+ progenitors into endocrine cells The adenohypophysis and neurohypophysis that constitute the pituitary gland are derived from the pre-placodal ectoderm and the neuro-ectoderm, respectively. However, Chen et al. demonstrate that subsets of adenohypophyseal cells are derived from the neuroectoderm of zebrafish and mouse, and that neuroectoderm-derived astroglial pituicytes refine differentiation of adenohypophyseal progenitors. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
173. Editorial highlights.
- Author
-
Trainor, Paul A.
- Subjects
EMBRYOLOGY ,DEVELOPMENTAL biology ,ECTODERM ,XENOPUS ,PERIODICAL articles - Abstract
This document is an editorial highlights article from the journal Developmental Dynamics. It discusses recent advances in the field of developmental biology. The article highlights three specific studies: one on Xenopus ectoderm patterning, one on pharyngeal development in mice, and one on the expression of adamts9 in zebrafish. These studies provide new insights into the genetic and molecular mechanisms that regulate embryonic development and could have implications for understanding human syndromes and diseases. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
174. A Molecular Analysis of Neural Olfactory Placode Differentiation in Human Pluripotent Stem Cells
- Author
-
Rebecca L. Bricker, Uchit Bhaskar, Rossella Titone, Melanie A. Carless, and Tiziano Barberi
- Subjects
Neurons ,Pluripotent Stem Cells ,Neural Crest ,Ectoderm ,Humans ,Cell Biology ,Hematology ,Hormones ,Developmental Biology - Abstract
During embryonic development, the olfactory sensory neurons (OSNs) and the gonadotropic-releasing hormone neurons (GNRHNs) migrate from the early nasal cavity, known as the olfactory placode, to the brain. Defects in the development of OSNs and GNRHNs result in neurodevelopmental disorders such as anosmia and congenital hypogonadotropic hypogonadism, respectively. Treatments do not restore the defective neurons in these disorders, and as a result, patients have a diminished sense of smell or a gonadotropin hormone deficiency. Human pluripotent stem cells (hPSCs) can produce any cell type in the body; therefore, they are an invaluable tool for cell replacement therapies. Transplantation of olfactory placode progenitors, derived from hPSCs, is a promising therapeutic to replace OSNs and GNRHNs and restore tissue function. Protocols to generate olfactory placode progenitors are limited, and thus, we describe, in this study, a novel in vitro model for olfactory placode differentiation in hPSCs, which is capable of producing both OSNs and GNRHNs. Our study investigates the major developmental signaling factors that recapitulate the embryonic development of the olfactory tissue. We demonstrate that induction of olfactory placode in hPSCs requires bone morphogenetic protein inhibition, wingless/integrated protein inhibition, retinoic acid inhibition, transforming growth factor alpha activation, and fibroblast growth factor 8 activation. We further show that the protocol transitions hPSCs through the anterior pan-placode ectoderm and neural ectoderm regions in early development while preventing neural crest and non-neural ectoderm regions. Finally, we demonstrate production of OSNs and GNRHNs by day 30 of differentiation. Our study is the first to report on OSN differentiation in hPSCs.
- Published
- 2022
175. Quality criteria for in vitro human pluripotent stem cell-derived models of tissue-based cells
- Author
-
Francesca Pistollato, Anna Bal-Price, Sandra Coecke, Surat Parvatam, David Pamies, Katherine Czysz, Jie Hao, Kehkooi Kee, Adrian Kee Keong Teo, Shuaishuai Niu, Anja Wilmes, Lena Smirnova, Christian Freund, Christine Mummery, Glyn Stacey, Molecular and Computational Toxicology, and AIMMS
- Subjects
Pluripotent Stem Cells ,GCCP ,Invitrotoxicology ,Induced Pluripotent Stem Cells ,Endoderm ,Cell Culture Techniques ,Reproducibility of Results ,Humanpluripotentstemcells ,iPSCs ,Cell Differentiation ,Quality control criteria ,Readiness level ,Toxicology ,Mesoderm ,SDG 3 - Good Health and Well-being ,Differentiation ,Ectoderm ,Humans ,Human pluripotent stem cells ,Qualitycontrolcriteria ,Readinesslevel ,3D ,In vitro toxicology - Abstract
The advent of the technology to isolate or generate human pluripotent stem cells provided the potential to develop a wide range of human models that could enhance understanding of mechanisms underlying human development and disease. These systems are now beginning to mature and provide the basis for the development of in vitro assays suitable to understand the biological processes involved in the multi-organ systems of the human body, and will improve strategies for diagnosis, prevention, therapies and precision medicine. Induced pluripotent stem cell lines are prone to phenotypic and genotypic changes and donor/clone dependent variability, which means that it is important to identify the most appropriate characterization markers and quality control measures when sourcing new cell lines and assessing differentiated cell and tissue culture preparations for experimental work. This paper considers those core quality control measures for human pluripotent stem cell lines and evaluates the state of play in the development of key functional markers for their differentiated cell derivatives to promote assurance of reproducibility of scientific data derived from pluripotent stem cell-based systems.
- Published
- 2022
176. Threshold-dependent BMP-mediated repression: a model for a conserved mechanism that patterns the neuroectoderm.
- Author
-
Mizutani, Claudia Mieko, Meyer, Néva, Roelink, Henk, and Bier, Ethan
- Subjects
Neurons ,Chick Embryo ,Ectoderm ,Animals ,Drosophila melanogaster ,Drosophila Proteins ,Bone Morphogenetic Proteins ,Transcription Factors ,Crosses ,Genetic ,Evolution ,Molecular ,Signal Transduction ,Gene Expression Regulation ,Developmental ,Body Patterning ,Models ,Biological ,Developmental Biology ,Biological Sciences ,Medical and Health Sciences ,Agricultural and Veterinary Sciences - Abstract
Subdivision of the neuroectoderm into three rows of cells along the dorsal-ventral axis by neural identity genes is a highly conserved developmental process. While neural identity genes are expressed in remarkably similar patterns in vertebrates and invertebrates, previous work suggests that these patterns may be regulated by distinct upstream genetic pathways. Here we ask whether a potential conserved source of positional information provided by the BMP signaling contributes to patterning the neuroectoderm. We have addressed this question in two ways: First, we asked whether BMPs can act as bona fide morphogens to pattern the Drosophila neuroectoderm in a dose-dependent fashion, and second, we examined whether BMPs might act in a similar fashion in patterning the vertebrate neuroectoderm. In this study, we show that graded BMP signaling participates in organizing the neural axis in Drosophila by repressing expression of neural identity genes in a threshold-dependent fashion. We also provide evidence for a similar organizing activity of BMP signaling in chick neural plate explants, which may operate by the same double negative mechanism that acts earlier during neural induction. We propose that BMPs played an ancestral role in patterning the metazoan neuroectoderm by threshold-dependent repression of neural identity genes.
- Published
- 2006
177. The Polycomb group protein Eed protects the inactive X-chromosome from differentiation-induced reactivation
- Author
-
Kalantry, Sundeep, Mills, Kyle C, Yee, Della, Otte, Arie P, Panning, Barbara, and Magnuson, Terry
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research ,Genetics ,Stem Cell Research - Embryonic - Non-Human ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,CDX2 Transcription Factor ,Cell Differentiation ,Cell Line ,Cells ,Cultured ,Ectoderm ,Embryo ,Mammalian ,Endoderm ,Enhancer of Zeste Homolog 2 Protein ,Epigenesis ,Genetic ,Gene Expression ,Green Fluorescent Proteins ,Heterochromatin ,Histone-Lysine N-Methyltransferase ,Histones ,Homeodomain Proteins ,In Situ Hybridization ,Fluorescence ,Methylation ,Mice ,Mice ,Knockout ,Mice ,Transgenic ,Polycomb Repressive Complex 2 ,Polycomb-Group Proteins ,Proteins ,RNA ,Long Noncoding ,RNA ,Untranslated ,Receptor ,Fibroblast Growth Factor ,Type 2 ,Repressor Proteins ,T-Box Domain Proteins ,Transcription Factors ,Trophoblasts ,X Chromosome ,X Chromosome Inactivation ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
The Polycomb group (PcG) encodes an evolutionarily conserved set of chromatin-modifying proteins that are thought to maintain cellular transcriptional memory by stably silencing gene expression. In mouse embryos that are mutated for the PcG protein Eed, X-chromosome inactivation (XCI) is not stably maintained in extra-embryonic tissues. Eed is a component of a histone-methyltransferase complex that is thought to contribute to stable silencing in undifferentiated cells due to its enrichment on the inactive X-chromosome in cells of the early mouse embryo and in stem cells of the extra-embryonic trophectoderm lineage. Here, we demonstrate that the inactive X-chromosome in Eed(-/-) trophoblast stem cells and in cells of the trophectoderm-derived extra-embryonic ectoderm in Eed(-/-) embryos remain transcriptionally silent, despite lacking the PcG-mediated histone modifications that normally characterize the facultative heterochromatin of the inactive X-chromosome. Whereas undifferentiated Eed(-/-) trophoblast stem cells maintained XCI, reactivation of the inactive X-chromosome occurred when these cells were differentiated. These results indicate that PcG complexes are not necessary to maintain transcriptional silencing of the inactive X-chromosome in undifferentiated stem cells. Instead, PcG proteins seem to propagate cellular memory by preventing transcriptional activation of facultative heterochromatin during differentiation.
- Published
- 2006
178. cSox3 expression and neurogenesis in the epibranchial placodes
- Author
-
Abu-Elmagd, Muhammad
- Subjects
572.8 ,Chick ,Development ,Ectoderm - Published
- 2001
179. Neural induction in Xenopus: requirement for ectodermal and endomesodermal signals via Chordin, Noggin, beta-Catenin, and Cerberus.
- Author
-
Kuroda, Hiroki, Wessely, Oliver, and De Robertis, EM
- Subjects
Central Nervous System ,Brain ,Neurons ,Cell Line ,Ectoderm ,Mesoderm ,Organizers ,Embryonic ,Animals ,Xenopus ,Glycoproteins ,Intercellular Signaling Peptides and Proteins ,Proteins ,Xenopus Proteins ,Carrier Proteins ,Cytoskeletal Proteins ,Trans-Activators ,Oligonucleotides ,Antisense ,RNA ,RNA ,Messenger ,Oligonucleotides ,Cell Transplantation ,Signal Transduction ,Cell Differentiation ,Gene Expression Regulation ,Developmental ,Cell Lineage ,Time Factors ,Female ,Male ,beta Catenin ,Organizers ,Embryonic ,Antisense ,Messenger ,Gene Expression Regulation ,Developmental ,Developmental Biology ,Biological Sciences ,Medical and Health Sciences ,Agricultural and Veterinary Sciences - Abstract
The origin of the signals that induce the differentiation of the central nervous system (CNS) is a long-standing question in vertebrate embryology. Here we show that Xenopus neural induction starts earlier than previously thought, at the blastula stage, and requires the combined activity of two distinct signaling centers. One is the well-known Nieuwkoop center, located in dorsal-vegetal cells, which expresses Nodal-related endomesodermal inducers. The other is a blastula Chordin- and Noggin-expressing (BCNE) center located in dorsal animal cells that contains both prospective neuroectoderm and Spemann organizer precursor cells. Both centers are downstream of the early beta-Catenin signal. Molecular analyses demonstrated that the BCNE center was distinct from the Nieuwkoop center, and that the Nieuwkoop center expressed the secreted protein Cerberus (Cer). We found that explanted blastula dorsal animal cap cells that have not yet contacted a mesodermal substratum can, when cultured in saline solution, express definitive neural markers and differentiate histologically into CNS tissue. Transplantation experiments showed that the BCNE region was required for brain formation, even though it lacked CNS-inducing activity when transplanted ventrally. Cell-lineage studies demonstrated that BCNE cells give rise to a large part of the brain and retina and, in more posterior regions of the embryo, to floor plate and notochord. Loss-of-function experiments with antisense morpholino oligos (MO) showed that the CNS that forms in mesoderm-less Xenopus embryos (generated by injection with Cerberus-Short [CerS] mRNA) required Chordin (Chd), Noggin (Nog), and their upstream regulator beta-Catenin. When mesoderm involution was prevented in dorsal marginal-zone explants, the anterior neural tissue formed in ectoderm was derived from BCNE cells and had a complete requirement for Chd. By injecting Chd morpholino oligos (Chd-MO) into prospective neuroectoderm and Cerberus morpholino oligos (Cer-MO) into prospective endomesoderm at the 8-cell stage, we showed that both layers cooperate in CNS formation. The results suggest a model for neural induction in Xenopus in which an early blastula beta-Catenin signal predisposes the prospective neuroectoderm to neural induction by endomesodermal signals emanating from Spemann's organizer.
- Published
- 2004
180. Neural Induction in Xenopus: Requirement for Ectodermal and Endomesodermal Signals via Chordin, Noggin, β-Catenin, and Cerberus
- Author
-
Kuroda, Hiroki, Wessely, Oliver, and Robertis, EM De
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Underpinning research ,1.1 Normal biological development and functioning ,Neurological ,Animals ,Brain ,Carrier Proteins ,Cell Differentiation ,Cell Line ,Cell Lineage ,Cell Transplantation ,Central Nervous System ,Cytoskeletal Proteins ,Ectoderm ,Female ,Gene Expression Regulation ,Developmental ,Glycoproteins ,Intercellular Signaling Peptides and Proteins ,Male ,Mesoderm ,Neurons ,Oligonucleotides ,Oligonucleotides ,Antisense ,Organizers ,Embryonic ,Proteins ,RNA ,RNA ,Messenger ,Signal Transduction ,Time Factors ,Trans-Activators ,Xenopus ,Xenopus Proteins ,beta Catenin ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Developmental Biology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
The origin of the signals that induce the differentiation of the central nervous system (CNS) is a long-standing question in vertebrate embryology. Here we show that Xenopus neural induction starts earlier than previously thought, at the blastula stage, and requires the combined activity of two distinct signaling centers. One is the well-known Nieuwkoop center, located in dorsal-vegetal cells, which expresses Nodal-related endomesodermal inducers. The other is a blastula Chordin- and Noggin-expressing (BCNE) center located in dorsal animal cells that contains both prospective neuroectoderm and Spemann organizer precursor cells. Both centers are downstream of the early beta-Catenin signal. Molecular analyses demonstrated that the BCNE center was distinct from the Nieuwkoop center, and that the Nieuwkoop center expressed the secreted protein Cerberus (Cer). We found that explanted blastula dorsal animal cap cells that have not yet contacted a mesodermal substratum can, when cultured in saline solution, express definitive neural markers and differentiate histologically into CNS tissue. Transplantation experiments showed that the BCNE region was required for brain formation, even though it lacked CNS-inducing activity when transplanted ventrally. Cell-lineage studies demonstrated that BCNE cells give rise to a large part of the brain and retina and, in more posterior regions of the embryo, to floor plate and notochord. Loss-of-function experiments with antisense morpholino oligos (MO) showed that the CNS that forms in mesoderm-less Xenopus embryos (generated by injection with Cerberus-Short [CerS] mRNA) required Chordin (Chd), Noggin (Nog), and their upstream regulator beta-Catenin. When mesoderm involution was prevented in dorsal marginal-zone explants, the anterior neural tissue formed in ectoderm was derived from BCNE cells and had a complete requirement for Chd. By injecting Chd morpholino oligos (Chd-MO) into prospective neuroectoderm and Cerberus morpholino oligos (Cer-MO) into prospective endomesoderm at the 8-cell stage, we showed that both layers cooperate in CNS formation. The results suggest a model for neural induction in Xenopus in which an early blastula beta-Catenin signal predisposes the prospective neuroectoderm to neural induction by endomesodermal signals emanating from Spemann's organizer.
- Published
- 2004
181. Top-Down Inhibition of BMP Signaling Enables Robust Induction of hPSCs Into Neural Crest in Fully Defined, Xeno-free Conditions
- Author
-
James O.S. Hackland, Tom J.R. Frith, Oliver Thompson, Ana Marin Navarro, Martin I. Garcia-Castro, Christian Unger, and Peter W. Andrews
- Subjects
neural crest ,BMP ,ectoderm ,human pluripotent stem cells ,embryonic stem cells ,top-down inhibition ,TDi ,endogenous signal control ,fully defined differentiation ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Defects in neural crest development have been implicated in many human disorders, but information about human neural crest formation mostly depends on extrapolation from model organisms. Human pluripotent stem cells (hPSCs) can be differentiated into in vitro counterparts of the neural crest, and some of the signals known to induce neural crest formation in vivo are required during this process. However, the protocols in current use tend to produce variable results, and there is no consensus as to the precise signals required for optimal neural crest differentiation. Using a fully defined culture system, we have now found that the efficient differentiation of hPSCs to neural crest depends on precise levels of BMP signaling, which are vulnerable to fluctuations in endogenous BMP production. We present a method that controls for this phenomenon and could be applied to other systems where endogenous signaling can also affect the outcome of differentiation protocols.
- Published
- 2017
- Full Text
- View/download PDF
182. Differentiation of Human Induced Pluripotent Stem Cells to Mammary-like Organoids
- Author
-
Ying Qu, Bingchen Han, Bowen Gao, Shikha Bose, Yiping Gong, Kolja Wawrowsky, Armando E. Giuliano, Dhruv Sareen, and Xiaojiang Cui
- Subjects
3D culture ,alveolar structure ,ectoderm ,iPSC ,mammary-like cells ,organoid ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Human induced pluripotent stem cells (iPSCs) can give rise to multiple cell types and hold great promise in regenerative medicine and disease-modeling applications. We have developed a reliable two-step protocol to generate human mammary-like organoids from iPSCs. Non-neural ectoderm-cell-containing spheres, referred to as mEBs, were first differentiated and enriched from iPSCs using MammoCult medium. Gene expression profile analysis suggested that mammary gland function-associated signaling pathways were hallmarks of 10-day differentiated mEBs. We then generated mammary-like organoids from 10-day mEBs using 3D floating mixed gel culture and a three-stage differentiation procedure. These organoids expressed common breast tissue, luminal, and basal markers, including estrogen receptor, and could be induced to produce milk protein. These results demonstrate that human iPSCs can be directed in vitro toward mammary lineage differentiation. Our findings provide an iPSC-based model for studying regulation of normal mammary cell fate and function as well as breast disease development.
- Published
- 2017
- Full Text
- View/download PDF
183. Xenopus Dusp6 modulates FGF signaling to precisely pattern pre-placodal ectoderm
- Author
-
Kohei Tsukano, Takayoshi Yamamoto, Tomoko Watanabe, and Tatsuo Michiue
- Subjects
Homeodomain Proteins ,Neural Plate ,Xenopus laevis ,Dual Specificity Phosphatase 6 ,Neural Crest ,Ectoderm ,Animals ,Gene Expression Regulation, Developmental ,Humans ,Cell Biology ,Xenopus Proteins ,Molecular Biology ,Developmental Biology - Abstract
Pre-placodal ectoderm (PPE), a horseshoe-shaped narrow region formed during early vertebrate development, gives rise to multiple types of sensory organs and ganglia. For PPE induction, a certain level of FGF signal activation is required. However, it is difficult to reproducibly induce the narrow region with variations in gene expression, including FGF, among individuals. An intracellular regulatory factor of FGF signaling, Dusp6, is expressed by FGF signal activation and inactivates a downstream regulator, ERK1/2, in adult tissues; however, its role in early development is not well known. Here, we reveal that Dusp6 is expressed in an FGF-dependent manner in Xenopus PPE. Gain- and loss-of-function experiments showed that Dusp6 is required for expression of a PPE gene, Six1, and patterning of adjacent regions, neural plate, and neural crest. To reveal the importance of Dusp6 in variable FGF production, we performed Dusp6 knockdown with FGF-bead implantation, which resulted in varying Six1 expression patterns. Taken together, these results suggest that Dusp6 is required for PPE formation and that it contributes to the robust patterning of PPE by mediating FGF signaling.
- Published
- 2022
184. Gene expression analysis of the <scp> Xenopus laevis </scp> early limb bud proximodistal axis
- Author
-
Daniel T. Hudson, Jessica S. Bromell, Robert C. Day, Tyler McInnes, Joanna M. Ward, and Caroline W. Beck
- Subjects
Limb Buds ,Gene Expression Regulation, Developmental ,Gene Expression ,Tretinoin ,Extremities ,Nerve Tissue Proteins ,Xenopus Proteins ,Mesoderm ,Fibroblast Growth Factors ,DNA-Binding Proteins ,Xenopus laevis ,Ectoderm ,Animals ,Developmental Biology - Abstract
Limb buds develop as bilateral outgrowths of the lateral plate mesoderm and are patterned along three axes. Current models of proximal to distal patterning of early amniote limb buds suggest that two signals, a distal organizing signal from the apical epithelial ridge (AER, Fgfs) and an opposing proximal (retinoic acid [RA]) act early on pattern this axis.Transcriptional analysis of stage 51 Xenopus laevis hindlimb buds sectioned along the proximal-distal axis showed that the distal region is distinct from the rest of the limb. Expression of capn8.3, a novel calpain, was located in cells immediately flanking the AER. The Wnt antagonist Dkk1 was AER-specific in Xenopus limbs. Two transcription factors, sall1 and zic5, were expressed in distal mesenchyme. Zic5 has no described association with limb development. We also describe expression of two proximal genes, gata5 and tnn, not previously associated with limb development. Differentially expressed genes were associated with Fgf, Wnt, and RA signaling as well as differential cell adhesion and proliferation.We identify new candidate genes for early proximodistal limb patterning. Our analysis of RA-regulated genes supports a role for transient RA gradients in early limb bud in proximal-to-distal patterning in this anamniote model organism.
- Published
- 2022
185. Data on microRNAs and microRNA-targeted mRNAs in Xenopus ectoderm
- Author
-
Vrutant V. Shah, Benjamin Soibam, Ruth A. Ritter, Ashley Benham, Jamina Oomen, and Amy K. Sater
- Subjects
microRNA ,Neural ,Ectoderm ,Xenopus ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Science (General) ,Q1-390 - Abstract
Small RNAs from early neural (i.e., Noggin-expressing, or NOG) and epidermal (expressing a constitutively active BMP4 receptor, CABR) ectoderm in Xenopus laevis were sequenced to identify microRNAs (miRs) expressed in each tissue. Argonaute-associated mRNAs were isolated and sequenced to identify genes that are regulated by microRNAs in these tissues. Interactions between these ectodermal miRs and selected miR-regulated mRNAs were predicted using the PITA algorithm; PITA predictions for over 600 mRNAs are presented. All sequencing data are available at NCBI (NCBI Bioproject Accession number: PRJNA325834). This article accompanies the manuscript “MicroRNAs and ectodermal specification I. Identification of miRs and miR-targeted mRNAs in early anterior neural and epidermal ectoderm” (V.V. Shah, B. Soibam, R.A. Ritter, A. Benham, J. Oomen, A.K. Sater, 2016) [1].
- Published
- 2016
- Full Text
- View/download PDF
186. Cell fate and signalling in chick limb bud development
- Author
-
Vargesson, Neil Andrew
- Subjects
572.8 ,Ectoderm ,Mesenchymal-epithelial interactions - Abstract
The chick limb develops from a bud of apparently homogeneous mesenchyme cells encased in ectoderm. Mesenchymal-epithelial interactions and mesenchymal signalling pathways are important in controlling the patterning and positioning of the skeletal elements and other tissues of the limb. I have produced detailed and comprehensive fatemaps for mesenchyme and apical ridge of a stage 20 chick wing bud. The mesenchyme fate maps show that the majority of the limb including the digits arises from posterior subapical mesenchyme. Fatemaps of the apical ridge show that apical ridge and mesenchyme do not remain in concert as development proceeds; the apical ridge expands more anteriorly than mesenchyme. The detailed behaviour of cells in the fate maps is not consistent with the model of vertebrate limb evolution which suggests an anterior bending of the posterior metapterygial axis led to the evolution of the hand plate. I have used these fate maps to investigate the relationship between cell lineage and gene expression. I show that the Hoxa-13 and Hoxd-13 expression domains expand by different mechanisms. I investigated activation of 5' HoxA genes in response to FGF-4, and show that Hoxa-13 gene expression may require FGF-4 for elaboration but not initiation. I have also used the fate maps to investigate cell fate in regulating and 'regenerating' limbs. I show that regulation and regeneration can only operate in the progress zone. Furthermore I show polarising signals act over a very short distance in reprogramming cells in the anterior tip to produce posterior digits. Finally I investigated expression of genes in the Notch signalling pathway in chick limb outgrowth and patterning. I show that genes in the Notch signalling pathway are expressed at different stages of chick limb development, and are implicated in bud outgrowth, formation of the musculature and vasculature and in digit spacing and positioning.
- Published
- 1998
187. In vitro Characteristics of Heterogeneous Equine Hoof Progenitor Cell Isolates
- Author
-
Qingqiu Yang, Vanessa Marigo Rocha Pinto, Wei Duan, Erica E. Paxton, Jenna H. Dessauer, William Ryan, and Mandi J. Lopez
- Subjects
mesoderm ,ectoderm ,ultrastructure ,scaffold ,horse ,stem ,Biotechnology ,TP248.13-248.65 - Abstract
Damage to an ectodermal-mesodermal interface like that in the equine hoof and human finger nail bed can permanently alter tissue structure and associated function. The purpose of this study was to establish and validate in vitro culture of primary progenitor cell isolates from the ectodermal-mesodermal tissue junction in equine hooves, the stratum internum, with and without chronic inflammation known to contribute to lifelong tissue defects. The following were evaluated in hoof stratum internum cell isolates up to 5 cell passages (P): expansion capacity by cell doublings and doubling time; plasticity with multi-lineage differentiation and colony-forming unit (CFU) frequency percentage; immunophenotype with immunocytochemistry and flow cytometry; gene expression with RT-PCR; and ultrastructure with transmission electron microscopy. The presence of keratin (K)14, 15 and K19 as well as cluster of differentiation (CD)44 and CD29 was determined in situ with immunohistochemistry. To confirm in vivo extracellular matrix (ECM) formation, cell-scaffold (polyethylene glycol/poly-L-lactic acid and tricalcium phosphate/hydroxyapatite) constructs were evaluated with scanning electron microscopy 9 weeks after implantation in athymic mice. Cultured cells had characteristic progenitor cell morphology, expansion, CFU frequency percentage and adipocytic, osteoblastic, and neurocytic differentiation capacity. CD44, CD29, K14, K15 and K19 proteins were present in native hoof stratum internum. Cultured cells also expressed K15, K19 and desmogleins 1 and 3. Gene expression of CD105, CD44, K14, K15, sex determining region Y-box 2 (SOX2) and octamer-binding transcription factor 4 (OCT4) was confirmed in vitro. Cultured cells had large, eccentric nuclei, elongated mitochondria, and intracellular vacuoles. Scaffold implants with cells contained fibrous ECM 9 weeks after implantation compared to little or none on acellular scaffolds. In vitro expansion and plasticity and in vivo ECM deposition of heterogeneous, immature cell isolates from the ectodermal-mesodermal tissue interface of normal and chronically inflamed hooves are typical of primary cell isolates from other adult tissues, and they appear to have both mesodermal and ectodermal qualities in vitro. These results establish a unique cell culture model to target preventative and restorative therapies for ectodermal-mesodermal tissue junctions.
- Published
- 2019
- Full Text
- View/download PDF
188. "Beyond transcription: How post-transcriptional mechanisms drive neural crest EMT".
- Author
-
Guzman-Espinoza M, Kim M, Ow C, and Hutchins EJ
- Subjects
- Ectoderm, Neural Tube, Cell Movement physiology, Gene Expression Regulation, Developmental, Neural Crest metabolism, Epithelial-Mesenchymal Transition genetics
- Abstract
The neural crest is a stem cell population that originates from the ectoderm during the initial steps of nervous system development. Neural crest cells delaminate from the neuroepithelium by undergoing a spatiotemporally regulated epithelial-mesenchymal transition (EMT) that proceeds in a coordinated wave head-to-tail to exit from the neural tube. While much is known about the transcriptional programs and membrane changes that promote EMT, there are additional levels of gene expression control that neural crest cells exert at the level of RNA to control EMT and migration. Yet, the role of post-transcriptional regulation, and how it drives and contributes to neural crest EMT, is not well understood. In this mini-review, we explore recent advances in our understanding of the role of post-transcriptional regulation during neural crest EMT., (© 2023 The Authors. genesis published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
189. In vitro modeling of cranial placode differentiation: Recent advances, challenges, and perspectives.
- Author
-
Griffin C and Saint-Jeannet JP
- Subjects
- Animals, Humans, Vertebrates, Cell Differentiation, Signal Transduction, Gene Expression Regulation, Developmental, Skull, Ectoderm
- Abstract
Cranial placodes are transient ectodermal thickenings that contribute to a diverse array of organs in the vertebrate head. They develop from a common territory, the pre-placodal region that over time segregates along the antero-posterior axis into individual placodal domains: the adenohypophyseal, olfactory, lens, trigeminal, otic, and epibranchial placodes. These placodes terminally differentiate into the anterior pituitary, the lens, and contribute to sensory organs including the olfactory epithelium, and inner ear, as well as several cranial ganglia. To study cranial placodes and their derivatives and generate cells for therapeutic purposes, several groups have turned to in vitro derivation of placodal cells from human embryonic stem cells (hESCs) or induced pluripotent stem cells (hiPSCs). In this review, we summarize the signaling cues and mechanisms involved in cranial placode induction, specification, and differentiation in vivo, and discuss how this knowledge has informed protocols to derive cranial placodes in vitro. We also discuss the benefits and limitations of these protocols, and the potential of in vitro cranial placode modeling in regenerative medicine to treat cranial placode-related pathologies., Competing Interests: Declaration of competing interest The authors report no competing interests. The authors alone are responsible for the content and writing of this article., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
190. Tissues and signals with true organizer properties in craniofacial development.
- Author
-
Tophkhane SS and Richman JM
- Subjects
- Chick Embryo, Animals, Mice, Jaw, Neural Crest, Endothelins, Body Patterning, Chickens, Ectoderm
- Abstract
Transplantation experiments have shown that a true organizer provides instructive signals that induce and pattern ectopic structures in the responding tissue. Here, we review craniofacial experiments to identify tissues with organizer properties and signals with organizer properties. In particular, we evaluate whether transformation of identity took place in the mesenchyme. Using these stringent criteria, we find the strongest evidence for the avian foregut ectoderm. Transplanting a piece of quail foregut endoderm to a host chicken embryo caused ectopic beaks to form derived from chicken mesenchyme. The beak identity, whether upper or lower as well as orientation, was controlled by the original anterior-posterior position of the donor endoderm. There is also good evidence that the nasal pit is necessary and sufficient for lateral nasal patterning. Finally, we review signals that have organizer properties on their own without the need for tissue transplants. Mouse germline knockouts of the endothelin pathway result in transformation of identity of the mandible into a maxilla. Application of noggin-soaked beads to post-migratory neural crest cells transforms maxillary identity. This suggests that endothelin or noggin rich ectoderm could be organizers (not tested). In conclusion, craniofacial, neural crest-derived mesenchyme is competent to respond to tissues with organizer properties, also originating in the head. In future, we can exploit such well defined systems to dissect the molecular changes that ultimately lead to patterning of the upper and lower jaw., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
191. Mechanical control of neural plate folding by apical domain alteration.
- Author
-
Matsuda M, Rozman J, Ostvar S, Kasza KE, and Sokol SY
- Subjects
- Nervous System, Ectoderm, Morphogenesis, Neural Plate, Neural Tube
- Abstract
Vertebrate neural tube closure is associated with complex changes in cell shape and behavior, however, the relative contribution of these processes to tissue folding is not well understood. At the onset of Xenopus neural tube folding, we observed alternation of apically constricted and apically expanded cells. This apical domain heterogeneity was accompanied by biased cell orientation along the anteroposterior axis, especially at neural plate hinges, and required planar cell polarity signaling. Vertex models suggested that dispersed isotropically constricting cells can cause the elongation of adjacent cells. Consistently, in ectoderm, cell-autonomous apical constriction was accompanied by neighbor expansion. Thus, a subset of isotropically constricting cells may initiate neural plate bending, whereas a 'tug-of-war' contest between the force-generating and responding cells reduces its shrinking along the body axis. This mechanism is an alternative to anisotropic shrinking of cell junctions that are perpendicular to the body axis. We propose that apical domain changes reflect planar polarity-dependent mechanical forces operating during neural folding., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
192. Missing pieces of the pituitary puzzle: participation of extra-adenohypophyseal placode-lineage cells in the adult pituitary gland.
- Author
-
Kato Y, Yoshida S, and Kato T
- Subjects
- Pituitary Gland, Ectoderm, Neural Crest, Pituitary Gland, Anterior, Pituitary Gland, Posterior
- Abstract
The pituitary gland is a major endocrine tissue composing of two distinct entities, the adenohypophysis (anterior pituitary, cranial placode origin) and the neurohypophysis (posterior pituitary, neural ectoderm origin), and plays important roles in maintaining vital homeostasis. This tissue is maintained by a slow, consistent cell-renewal system of adult stem/progenitor cells. Recent accumulating evidence shows that neural crest-, head mesenchyme-, and endoderm lineage cells invade during pituitary development and contribute to the maintenance of the adult pituitary gland. Based on these novel observations, this article discusses whether these lineage cells are involved in pituitary organogenesis, maintenance, regeneration, dysplasia, or tumors., (© 2023. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
193. Prechordal structures act cooperatively in early trabeculae development of gnathostome skull.
- Author
-
da Cunha JI, Barauna AMD, and Garcez RC
- Subjects
- Neural Crest, Ectoderm, Head, Hedgehog Proteins genetics, Hedgehog Proteins metabolism, Skull
- Abstract
The vertebrate skull is formed by mesoderm and neural crest (NC) cells. The mesoderm contributes to the skull chordal domain, with the notochord playing an essential role in this process. The NC contributes to the skull prechordal domain, prompting investigation into the embryonic structures involved in prechordal neurocranium cartilage formation. The trabeculae cartilage, a structure of the prechordal neurocranium, arises at the convergence of prechordal plate (PCP), ventral midline (VM) cells of the diencephalon, and dorsal oral ectoderm. This study examines the molecular participation of these embryonic structures in gnathostome trabeculae development. PCP-secreted SHH induces its expression in VM cells of the diencephalon, initiating a positive feedback loop involving SIX3 and GLI1. SHH secreted by the VM cells of the diencephalon acts on the dorsal oral ectoderm, stimulating condensation of NC cells to form trabeculae. SHH from the prechordal region affects the expression of SOX9 in NC cells. BMP7 and SHH secreted by PCP induce NKX2.1 expression in VM cells of the diencephalon, but this does not impact trabeculae formation. Molecular cooperation between PCP, VM cells of the diencephalon, and dorsal oral ectoderm is crucial for craniofacial development by NC cells in the prechordal domain., Competing Interests: Declaration of competing interest The authors declare no conflict of interest., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
194. Making a head: Neural crest and ectodermal placodes in cranial sensory development
- Author
-
Alison Koontz, Hugo A. Urrutia, and Marianne E. Bronner
- Subjects
Neural Crest ,Organogenesis ,Ectoderm ,Vertebrates ,Animals ,Gene Expression Regulation, Developmental ,Cell Biology ,Article ,Developmental Biology - Abstract
During development of the vertebrate sensory system, many important components like the sense organs and cranial sensory ganglia arise within the head and neck. Two progenitor populations, the neural crest, and cranial ectodermal placodes, contribute to these developing vertebrate peripheral sensory structures. The interactions and contributions of these cell populations to the development of the lens, olfactory, otic, pituitary gland, and cranial ganglia are vital for appropriate peripheral nervous system development. Here, we review the origins of both neural crest and placode cells at the neural plate border of the early vertebrate embryo and investigate the molecular and environmental signals that influence specification of different sensory regions. Finally, we discuss the underlying molecular pathways contributing to the complex vertebrate sensory system from an evolutionary perspective, from basal vertebrates to amniotes.
- Published
- 2023
195. Ectodermal Wnt signaling, cell fate determination, and polarity of the skate gill arch skeleton
- Author
-
Rees, Jenaid M, Sleight, Victoria A, Clark, Stephen J, Nakamura, Tetsuya, Gillis, J Andrew, Rees, Jenaid M [0000-0001-9434-5445], Gillis, J Andrew [0000-0003-2062-3777], and Apollo - University of Cambridge Repository
- Subjects
Gills ,Evolutionary Biology ,General Immunology and Microbiology ,General Neuroscience ,skate leucoraja erinacea ,General Medicine ,General Biochemistry, Genetics and Molecular Biology ,pharyngeal arch ,Shh ,Wnt ,Branchial Region ,FOS: Biological sciences ,Ectoderm ,Animals ,skeleton ,Hedgehog Proteins ,Skates, Fish ,Other ,Wnt Signaling Pathway ,Research Article ,Developmental Biology ,gill arch - Abstract
Peer reviewed: True, Acknowledgements: With thanks to Dr Kate Criswell and Dr Christine Hirschberger for advice, and to the University of Cambridge Wellcome PhD. Programme in Developmental Mechanisms. The authors were funded by a Wellcome PhD studentship (214953/Z/18/Z) to JMR, and by a Royal Society University Research Fellowship (UF130182 and URF\R\191007) and Royal Society Research Grant (RG140377) to JAG., The gill skeleton of cartilaginous fishes (sharks, skates, rays, and holocephalans) exhibits a striking anterior–posterior polarity, with a series of fine appendages called branchial rays projecting from the posterior margin of the gill arch cartilages. We previously demonstrated in the skate (Leucoraja erinacea) that branchial rays derive from a posterior domain of pharyngeal arch mesenchyme that is responsive to Sonic hedgehog (Shh) signaling from a distal gill arch epithelial ridge (GAER) signaling centre. However, how branchial ray progenitors are specified exclusively within posterior gill arch mesenchyme is not known. Here, we show that genes encoding several Wnt ligands are expressed in the ectoderm immediately adjacent to the skate GAER, and that these Wnt signals are transduced largely in the anterior arch environment. Using pharmacological manipulation, we show that inhibition of Wnt signalling results in an anterior expansion of Shh signal transduction in developing skate gill arches, and in the formation of ectopic anterior branchial ray cartilages. Our findings demonstrate that ectodermal Wnt signalling contributes to gill arch skeletal polarity in skate by restricting Shh signal transduction and chondrogenesis to the posterior arch environment and highlights the importance of signalling interactions at embryonic tissue boundaries for cell fate determination in vertebrate pharyngeal arches.
- Published
- 2023
196. Otic Induction
- Author
-
Ohnishi, Hiroe, Sakamoto, Tatsunori, and Ito, Juichi, editor
- Published
- 2014
- Full Text
- View/download PDF
197. Esrrb function is required for proper primordial germ cell development in presomite stage mouse embryos.
- Author
-
Okamura, Eiichi, Tam, Oliver H., Posfai, Eszter, Li, Lingyu, Cockburn, Katie, Lee, Cheryl Q.E., Garner, Jodi, and Rossant, Janet
- Subjects
- *
EMBRYOS , *NUCLEAR receptors (Biochemistry) , *GERM cells , *KNOCKOUT mice , *STEM cells , *MICE , *ECTODERM - Abstract
Estrogen related receptor beta (Esrrb) is an orphan nuclear receptor that is required for self-renewal and pluripotency in mouse embryonic stem (ES) cells. However, in the early post-implantation mouse embryo, Esrrb is specifically expressed in the extraembryonic ectoderm (ExE) and plays a crucial role in trophoblast development. Previous studies showed that Esrrb is also required to maintain trophoblast stem (TS) cells, the in vitro stem cell model of the early trophoblast lineage. In order to identify regulatory targets of Esrrb in vivo , we performed microarray analysis of Esrrb -null versus wild-type post-implantation ExE, and identified 30 genes down-regulated in Esrrb -mutants. Among them is Bmp4 , which is produced by the ExE and known to be critical for primordial germ cell (PGC) specification in vivo. We further identified an enhancer region bound by Esrrb at the Bmp4 locus by performing Esrrb ChIP-seq and luciferase reporter assay using TS cells. Finally, we established a knockout mouse line in which the enhancer region was deleted using CRISPR/Cas9 technology. Both Esrrb -null embryos and enhancer knockout embryos expressed lower levels of Bmp4 in the ExE, and had reduced numbers of PGCs. These results suggested that Esrrb functions as an upstream factor of Bmp4 in the ExE, regulating proper PGC development in mice. • Esrrb binds to a critical enhancer region at Bmp4 locus in trophoblast stem cells. • Esrrb knockout mouse embryos have reduced numbers of primordial germ cells. • Esrrb-binding Bmp4 enhancer knockout mice show sub-fertile phenotype. • Esrrb acts as an upstream regulator of Bmp4 expression in extraembryonic ectoderm. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
198. A novel self-organizing embryonic stem cell system reveals signaling logic underlying the patterning of human ectoderm.
- Author
-
Britton, George, Heemskerk, Idse, Hodge, Rachel, Qutub, Amina A., and Warmflash, Aryeh
- Subjects
- *
EMBRYONIC stem cells , *ECTODERM , *WNT signal transduction , *LOGIC - Abstract
During development, the ectoderm is patterned by a combination of BMP and WNT signaling. Research in model organisms has provided substantial insight into this process, however, there are currently no systems to study ectodermal patterning in humans. Further, the complexity of neural plate border specification has made it difficult to transition from discovering the genes involved to deeper mechanistic understanding. Here, we develop an in vitro model of human ectodermal patterning, in which hESCs self-organize to form robust and quantitatively reproducible patterns corresponding to the complete medial-lateral axis of the embryonic ectoderm. Using this platform, we show that the duration of endogenous WNT signaling is a crucial control parameter, and that cells sense relative levels of BMP and WNT signaling in making fate decisions. These insights allowed us to develop an improved protocol for placodal differentiation. Thus, our platform is a powerful tool for studying human ectoderm patterning and for improving directed differentiation protocols. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
199. Tbx1 and Foxi3 genetically interact in the pharyngeal pouch endoderm in a mouse model for 22q11.2 deletion syndrome.
- Author
-
Hasten, Erica and Morrow, Bernice E.
- Subjects
- *
ENDODERM , *SUBCLAVIAN artery , *EXTRACELLULAR matrix proteins , *NOTCH genes , *THYMUS , *PARATHYROID glands - Abstract
We investigated whether Tbx1, the gene for 22q11.2 deletion syndrome (22q11.2DS) and Foxi3, both required for segmentation of the pharyngeal apparatus (PA) to individual arches, genetically interact. We found that all Tbx1+/-;Foxi3+/- double heterozygous mouse embryos had thymus and parathyroid gland defects, similar to those in 22q11.2DS patients. We then examined Tbx1 and Foxi3 heterozygous, null as well as conditional Tbx1Cre and Sox172A-iCre/+ null mutant embryos. While Tbx1Cre/+;Foxi3f/f embryos had absent thymus and parathyroid glands, Foxi3-/- and Sox172A-iCre/+;Foxi3f/f endoderm conditional mutant embryos had in addition, interrupted aortic arch type B and retroesophageal origin of the right subclavian artery, which are all features of 22q11.2DS. Tbx1Cre/+;Foxi3f/f embryos had failed invagination of the third pharyngeal pouch with greatly reduced Gcm2 and Foxn1 expression, thereby explaining the absence of thymus and parathyroid glands. Immunofluorescence on tissue sections with E-cadherin and ZO-1 antibodies in wildtype mouse embryos at E8.5-E10.5, revealed that multilayers of epithelial cells form where cells are invaginating as a normal process. We noted that excessive multilayers formed in Foxi3-/-, Sox172A-iCre/+;Foxi3f/f as well as Tbx1 null mutant embryos where invagination should have occurred. Several genes expressed in the PA epithelia were downregulated in both Tbx1 and Foxi3 null mutant embryos including Notch pathway genes Jag1, Hes1, and Hey1, suggesting that they may, along with other genes, act downstream to explain the observed genetic interaction. We found Alcam and Fibronectin extracellular matrix proteins were reduced in expression in Foxi3 null but not Tbx1 null embryos, suggesting that some, but not all of the downstream mechanisms are shared. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
200. Neuroblastoma Characteristics and Embryonic Origin of The Primary Lesion Site: A SEER Study.
- Author
-
Jiaxi He, Shaochang Wu, Na Zhu, Chunjiao Rong, Tielun Yan, and Xiumei Yan
- Subjects
- *
METASTASIS , *NEUROBLASTOMA , *PUBLIC health surveillance , *RETROSPECTIVE studies , *DESCRIPTIVE statistics , *GERMINOMA - Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.