36 results on '"Sachiko Iseki"'
Search Results
2. How to make a tongue: Cellular and molecular regulation of muscle and connective tissue formation during mammalian tongue development
- Author
-
Hadeel Adel Al-Lami, Karen J. Liu, Sachiko Iseki, Martyn T. Cobourne, Guilherme M. Xavier, Christel Thauvin-Robinet, Anahid A. Birjandi, Department of Craniofacial Development & Orthodontics, London Dental Institute (LDI), King‘s College London-King‘s College London, Section of Molecular Craniofacial Embryology (Graduate School of Tokyo Medical and Dental University), Department of Orthodontics (University of Baghdad College of Dentistry), University of Baghdad, Centre de génétique - Centre de référence des maladies rares, anomalies du développement et syndromes malformatifs (CHU de Dijon), Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand (CHU Dijon), FHU TRANSLAD (CHU de Dijon), Lipides - Nutrition - Cancer [Dijon - U1231] (LNC), Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM), Equipe GAD (LNC - U1231), and Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Organogenesis ,Hedgehog signaling ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Bifid tongue ,Mesoderm ,Wnt ,03 medical and health sciences ,0302 clinical medicine ,Cranial neural crest ,Tongue ,Macroglossia ,medicine ,Animals ,Humans ,TGF-beta ,Hedgehog ,Mammals ,Aglossia ,Muscles ,Myogenesis ,Gene Expression Regulation, Developmental ,Cell Biology ,Anatomy ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Connective Tissue ,Neural Crest ,Embryology ,Gross anatomy ,medicine.symptom ,030217 neurology & neurosurgery ,Signal Transduction ,Developmental Biology - Abstract
IF 6.614 (2016); International audience; The vertebrate tongue is a complex muscular organ situated in the oral cavity and involved in multiple functions including mastication, taste sensation, articulation and the maintenance of oral health. Although the gross embryological contributions to tongue formation have been known for many years, it is only relatively recently that the molecular pathways regulating these processes have begun to be discovered. In particular, there is now evidence that the Hedgehog, TGF-Beta, Wnt and Notch signaling pathways all play an important role in mediating appropriate signaling interactions between the epithelial, cranial neural crest and mesodermal cell populations that are required to form the tongue. In humans, a number of congenital abnormalities that affect gross morphology of the tongue have also been described, occurring in isolation or as part of a developmental syndrome, which can greatly impact on the health and well-being of affected individuals. These anomalies can range from an absence of tongue formation (aglossia) through to diminutive (microglossia), enlarged (macroglossia) or bifid tongue. Here, we present an overview of the gross anatomy and embryology of mammalian tongue development, focusing on the molecular processes underlying formation of the musculature and connective tissues within this organ. We also survey the clinical presentation of tongue anomalies seen in human populations, whilst considering their developmental and genetic etiology.
- Published
- 2019
3. Cell lineage- and expression-based inference of the roles of forkhead box transcription factor Foxc2 in craniofacial development
- Author
-
Tri Vu Hoang, Kazushi Aoto, Masaki Takechi, Chisaki Akagawa, Toshiko Furutera, Worachat Namangkalakul, Tsutomu Iwamoto, Manami Takenoshita, Michiyo Miyashin, Tsutomu Kume, and Sachiko Iseki
- Subjects
0301 basic medicine ,Craniofacial abnormality ,Mesenchyme ,Organogenesis ,Mice, Transgenic ,In situ hybridization ,Winged Helix ,Biology ,Craniofacial Abnormalities ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Cell Lineage ,Craniofacial ,Transcription factor ,Loss function ,Gene Expression Regulation, Developmental ,Forkhead Transcription Factors ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Neural Crest ,biology.protein ,FOXC2 ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Background Foxc2 is a member of the winged helix/forkhead (Fox) box family of transcription factors. Loss of function of Foxc2 causes craniofacial abnormalities such as cleft palate and deformed cranial base, but its role during craniofacial development remains to be elucidated RESULTS: The contributions of Foxc2-positive and its descendant cells to the craniofacial structure at E18.5 were examined using a tamoxifen-inducible Cre driver mouse (Foxc2-CreERT2) crossed with the R26R-LacZ reporter mouse. Foxc2 expression at E8.5 is restricted to the cranial mesenchyme, contributing to specific components including the cranial base, sensory capsule, tongue, upper incisor, and middle ear. Expression at E10.5 was still positively regulated in most of those regions. In situ hybridization analysis of Foxc2 and its closely related gene, Foxc1, revealed that expression domains of these genes largely overlap in the cephalic mesenchyme. Meanwhile, the tongue expressed Foxc2 but not Foxc1, and its development was affected by the neural crest-specific deletion of Foxc2 in mice (Wnt1-Cre; Foxc2fl/fl ). Conclusions Foxc2 is expressed in cranial mesenchyme that contributes to specific craniofacial tissue components from an early stage, and it seems to be involved in their development in cooperation with Foxc1. Foxc2 also has its own role in tongue development. This article is protected by copyright. All rights reserved.
- Published
- 2021
4. Transcription factorFoxc1is involved in anterior part of cranial base formation
- Author
-
Masaki Takechi, Tsutomu Kume, Nandar Mya, Sachiko Iseki, Toshiko Furutera, and Shigeru Okuhara
- Subjects
0301 basic medicine ,Embryology ,Ossification ,Cartilage ,General Medicine ,Anatomy ,Biology ,Chondrogenesis ,Chondrocyte ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Pediatrics, Perinatology and Child Health ,Intramembranous ossification ,medicine ,Perichondrium ,Trabecular cartilage ,medicine.symptom ,Endochondral ossification ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The cranial base is a structure mainly formed through endochondral ossification and integrated into the craniofacial complex, which acts as an underlying platform for the developing brain. Foxc1 is an indispensable regulator during intramembranous and endochondral ossification. In this study, we found that the spontaneous loss of Foxc1 function in a mouse (congenital hydrocephalous), Foxc1ch/ch , demonstrated the anterior cranial base defects, including unossified presphenoid and lack of middle part of the basisphenoid bone. Hypoplastic presphenoid primordial cartilage (basal portion of the trabecular cartilage [bTB]) and a lack of the middle part of basisphenoid primordial cartilage (the hypophyseal cartilage) were consistently observed at earlier developmental stage. Foxc1 was expressed robustly and ubiquitously in undifferentiated mesenchyme of the cranial base-forming area in E11.0 wild-type fetuses. Once chondrogenesis commenced, the expression was downregulated and later limited to the perichondrium. Detection of transcripts of Collagen type2 A1 (Col2a1) revealed that both bTB and the anterior part of the hypophyseal cartilage developing anterior to the persistent epithelial stalk of the anterior lobe of the pituitary gland were suppressed in the Foxc1ch/ch . Proliferation activity of chondrocyte precursor cells was higher in the Foxc1ch/ch . Loss of Foxc1 function only in the neural crest cell lineage (Wnt1-cre;Foxc1ch/flox ) showed ossification of the posterior part of the hypophyseal cartilage derived from the mesoderm. These findings suggest that Foxc1 is an important regulator to further chondrogenesis and initiate the ossification of the presphenoid and basisphenoid bones.
- Published
- 2018
5. Foxc2CreERT2knock-in mice mark stage-specificFoxc2-expressing cells during mouse organogenesis
- Author
-
Kazushi Aoto, Nobuaki Yoshida, Naoyuki Miura, Paul A. Trainor, Mohammad Khaja Mafij Uddin, Hirotomo Saitsu, Tsutomu Kume, Sachiko Iseki, Mohammod Johirul Islam, and Mohammed Badrul Amin
- Subjects
0301 basic medicine ,Embryology ,biology ,Cardiac neural crest cells ,Mesenchyme ,Embryogenesis ,Cre recombinase ,Organogenesis ,General Medicine ,Embryonic stem cell ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Cranial neural crest ,medicine.anatomical_structure ,Pediatrics, Perinatology and Child Health ,Immunology ,biology.protein ,medicine ,FOXC2 ,Developmental Biology - Abstract
Foxc2, a member of the winged helix transcription factor family, is essential for eye, calvarial bone, cardiovascular and kidney development in mice. Nevertheless, how Foxc2-expressing cells and their descendent cells contribute to the development of these tissues and organs has not been elucidated. Here, we generated a Foxc2 knock-in (Foxc2CreERT2 ) mouse, in which administration of estrogen receptor antagonist tamoxifen induces nuclear translocation of Cre recombinase in Foxc2-expressing cells. By crossing with ROSA-LacZ reporter mice (Foxc2CreERT2 ; R26R), the fate of Foxc2 positive (Foxc2+ ) cells was analyzed through LacZ staining at various embryonic stages. We found Foxc2+ cell descendants in the supraoccipital and exoccipital bone in E18.5 embryos, when tamoxifen was administered at embryonic day (E) 8.5. Furthermore, Foxc2+ descendant cranial neural crest cells at E8-10 were restricted to the corneal mesenchyme, while Foxc2+ cell derived cardiac neural crest cells at E6-12 were found in the aorta, pulmonary trunk and valves, and endocardial cushions. Foxc2+ cell descendant contributions to the glomerular podocytes in the kidney were also observed following E6.5 tamoxifen treatment. Our results are consistent with previous reports of Foxc2 expression during early embryogenesis and the Foxc2CreERT2 mouse provides a tool to investigate spatiotemporal roles of Foxc2 and contributions of Foxc2+ expressing cells during mouse embryogenesis.
- Published
- 2017
6. Heterozygous mutation of the splicing factor Sf3b4 affects development of the axial skeleton and forebrain in mouse
- Author
-
Yuichi Hiraoka, Takako Usami, Takahiko Yamada, Harumi Ishikubo, Masaki Takechi, Norisuke Yokoyama, Sachiko Iseki, Tetsuya Yoda, Kiyoko Ogawa-Goto, Yuki Taga, Yoshikazu Hirate, Masami Kanai-Azuma, and Toshiko Furutera
- Subjects
0301 basic medicine ,Male ,Axial skeleton ,Mutant ,Calvaria ,Biology ,03 medical and health sciences ,Splicing factor ,Mice ,0302 clinical medicine ,Prosencephalon ,medicine ,Animals ,Skeleton ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Forebrain ,RNA splicing ,Knockout mouse ,Mutation ,Female ,RNA Splicing Factors ,Homeotic gene ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Background Splicing factor 3B subunit 4 (SF3B4) is a causative gene of an acrofacial dysostosis, Nager syndrome. Although in vitro analyses of SF3B4 have proposed multiple noncanonical functions unrelated to splicing, less information is available based on in vivo studies using model animals. Results We performed expression and functional analyses of Sf3b4 in mice. The mouse Sf3b4 transcripts were found from two-cell stage, and were ubiquitously present during embryogenesis with high expression levels in several tissues such as forming craniofacial bones and brain. In contrast, expression of a pseudogene-like sequence of mouse Sf3b4 (Sf3b4_ps) found by in silico survey was not detected up to embryonic day 10. We generated a Sf3b4 knockout mouse using CRISPR-Cas9 system. The homozygous mutant mouse of Sf3b4 was embryonic lethal. The heterozygous mutant of Sf3b4 mouse (Sf3b4+/- ) exhibited smaller body size compared to the wild-type from postnatal to adult period, as well as homeotic posteriorization of the vertebral morphology and flattened calvaria. The flattened calvaria appears to be attributable to mild microcephaly due to a lower cell proliferation rate in the forebrain. Conclusions Our study suggests that Sf3b4 controls anterior-posterior patterning of the axial skeleton and guarantees cell proliferation for forebrain development in mice.
- Published
- 2019
7. Temporospatial sonic hedgehog signalling is essential for neural crest-dependent patterning of the intrinsic tongue musculature
- Author
-
Karen J. Liu, Tomoko Sagai, Guilherme M. Xavier, Toshihiko Shiroishi, Sachiko Iseki, Martyn T. Cobourne, Hadeel Adel Al-Lami, Takanori Amano, Anahid A. Birjandi, and Shigeru Okuhara
- Subjects
Heterozygote ,animal structures ,Time Factors ,Hypoglossia ,Lingual septum ,Wnt1 Protein ,Ligands ,Mesoderm ,Tendons ,Neural crest ,Mice ,Cranial neural crest ,Tongue ,Transforming Growth Factor beta ,medicine ,Morphogenesis ,Animals ,Hedgehog Proteins ,Sonic hedgehog ,Molecular Biology ,Tendon ,Alleles ,Body Patterning ,Cell Proliferation ,biology ,Cilium ,Gene Expression Regulation, Developmental ,Proteins ,Tendon formation ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,Enhancer Elements, Genetic ,Phenotype ,Neural Crest ,embryonic structures ,biology.protein ,Female ,Gene Deletion ,Developmental Biology ,Signal Transduction - Abstract
The tongue is a highly specialised muscular organ with a complex anatomy required for normal function. We have utilised multiple genetic approaches to investigate local temporospatial requirements for sonic hedgehog (SHH) signalling during tongue development. Mice lacking a Shh cis-enhancer, MFCS4 (ShhMFCS4/−), with reduced SHH in dorsal tongue epithelium have perturbed lingual septum tendon formation and disrupted intrinsic muscle patterning, with these defects reproduced following global Shh deletion from E10.5 in pCag-CreERTM; Shhflox/flox embryos. SHH responsiveness was diminished in local cranial neural crest cell (CNCC) populations in both mutants, with SHH targeting these cells through the primary cilium. CNCC-specific deletion of orofaciodigital syndrome 1 (Ofd1), which encodes a ciliary protein, in Wnt1-Cre; Ofdfl/Y mice led to a complete loss of normal myotube arrangement and hypoglossia. In contrast, mesoderm-specific deletion of Ofd1 in Mesp1-Cre; Ofdfl/Y embryos resulted in normal intrinsic muscle arrangement. Collectively, these findings suggest key temporospatial requirements for local SHH signalling in tongue development (specifically, lingual tendon differentiation and intrinsic muscle patterning through signalling to CNCCs) and provide further mechanistic insight into the tongue anomalies seen in patients with disrupted hedgehog signalling.
- Published
- 2019
8. Developmental mechanisms of the tympanic membrane in mammals and non-mammalian amniotes
- Author
-
Shigeru Kuratani, Hiroki Kurihara, Tatsuya Hirasawa, Sachiko Iseki, Taro Kitazawa, Masaki Takechi, and Tamami Hirai
- Subjects
0301 basic medicine ,Embryology ,Mesenchyme ,Mesenchymal stem cell ,Thin layer ,Ectoderm ,General Medicine ,Anatomy ,Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Membrane ,Pediatrics, Perinatology and Child Health ,medicine ,Middle ear ,Endoderm ,030217 neurology & neurosurgery ,Pharyngeal arch ,Developmental Biology - Abstract
The tympanic membrane is a thin layer that originates from the ectoderm, endoderm, and mesenchyme. Molecular-genetic investigations have revealed that interaction between epithelial and mesenchymal cells in the pharyngeal arches is essential for development of the tympanic membrane. We have recently reported that developmental mechanisms underlying the tympanic membrane seem to be different between mouse and chicken, suggesting that the tympanic membrane evolved independently in mammals and non-mammalian amniotes. In this review, we summarize previous studies of tympanic membrane formation in the mouse. We also discuss its formation in amniotes from an evolutionary point of view.
- Published
- 2016
9. Foxc1is required for early stage telencephalic vascular development
- Author
-
Shizuko Ichinose, Masato S. Ota, Sachiko Iseki, Shigeru Okuhara, Thanit Prasitsak, and Mya Nandar
- Subjects
Plexus ,Angiogenesis ,Cerebrum ,Mesenchyme ,Neural tube ,Subventricular zone ,Anatomy ,Biology ,Cell biology ,Neuroepithelial cell ,medicine.anatomical_structure ,nervous system ,medicine ,Developmental Biology ,Blood vessel - Abstract
Background: The brain vascular system arises from the perineural vascular plexus (PNVP) which sprouts radially into the neuroepithelium and subsequently branches off laterally to form a secondary plexus in the subventricular zone (SVZ), the subventricular vascular plexus (SVP). The process of SVP formation remains to be fully elucidated. We investigated the role of Foxc1 in early stage vascular formation in the ventral telencephalon. Results: The Foxc1 loss of function mutant mouse, Foxc1ch/ch, showed enlarged telencephalon and hemorrhaging in the ventral telencephalon by embryonic day 11.0. The mutant demonstrated blood vessel dilation and aggregation of endothelial cells in the SVZ after the invasion of endothelial cells through the radial path, which lead to failure of SVP formation. During this early stage of vascular development, Foxc1 was expressed in endothelial cells and pericytes, as well as in cranial mesenchyme surrounding the neural tube. Correspondingly, abnormal deposition pattern of basement membrane proteins around the vessels and increased strong Vegfr2 staining dots were found in the aggregation sites. Conclusions: These observations reveal an essential role for Foxc1 in the early stage of vascular formation in the telencephalon. Developmental Dynamics 244:703–711, 2015. © 2015 Wiley Periodicals, Inc.
- Published
- 2015
10. Differing contributions of the first and second pharyngeal arches to tympanic membrane formation in the mouse and chick
- Author
-
Toshiko Furutera, Masaki Takechi, Filippo M. Rijli, Taro Kitazawa, Junko Takei, Sachiko Iseki, Takahiko Yamada, Shigeru Kuratani, Hiroki Kurihara, and Tri Vu Hoang
- Subjects
0301 basic medicine ,Second pharyngeal arch ,Tympanic Membrane ,Pharyngeal pouch ,Green Fluorescent Proteins ,Ear, Middle ,Chick Embryo ,Models, Biological ,Quail ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Ear canal ,Molecular Biology ,Homeodomain Proteins ,Mice, Knockout ,First pharyngeal arch ,biology ,Anatomy ,Embryo, Mammalian ,biology.organism_classification ,Surface ectoderm ,Branchial Region ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,Middle ear ,Amniote ,Chickens ,Ear Canal ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
We have proposed that independent origins of the tympanic membrane (TM), consisting of the external auditory meatus (EAM) and first pharyngeal pouch, are linked with distinctive middle ear structures in terms of dorsal-ventral patterning of the pharyngeal arches during amniote evolution. However, previous studies have suggested that the first pharyngeal arch (PA1) is crucial for TM formation in both the mouse and chick. In this study, we compare TM formation along the anterior-posterior axis in these animals using Hoxa2 expression as a marker of the second pharyngeal arch (PA2). In the chick, the EAM begins to invaginate at the surface ectoderm of PA2, not at the first pharyngeal cleft, and the entire TM forms in PA2. Chick-quail chimera that have lost PA2 and duplicated PA1 suggest that TM formation is achieved by developmental interaction between a portion of the EAM and the columella auris in PA2, and that PA1 also contributes to formation of the remaining part of the EAM. In contrast, in the mouse, TM formation highly associates with interdependent relationship between the EAM and tympanic ring in PA1.
- Published
- 2017
11. Difference in apical and basal growth of the frontal bone primordium inFoxc1ch/chmice
- Author
-
Akihiko Machida, Kiyoshi Harada, Sachiko Iseki, and Shigeru Okuhara
- Subjects
Bone sialoprotein ,Embryology ,biology ,Cell growth ,Mutant ,Meninges ,General Medicine ,Anatomy ,RUNX2 ,Basal (phylogenetics) ,medicine.anatomical_structure ,stomatognathic system ,embryonic structures ,Pediatrics, Perinatology and Child Health ,Cranial vault ,biology.protein ,medicine ,Primordium ,Developmental Biology - Abstract
The frontal and parietal bones form the major part of the calvarium and their primordia appear at the basolateral region of the head and grow apically. A spontaneous loss of Foxc1 function mutant mouse, congenital hydrocephalus (Foxc1(ch/ch)), results in congenital hydrocephalus accompanied by defects in the apical part of the skull vault. We found that during the initiation stage of apical growth of the frontal bone primordium in the Foxc1(ch/ch) mouse, the Runx2 expression domain extended only to the basal side and bone sialoprotein (Bsp) and N-cadherin expression domains appeared only in the basal region. Fluorescent dye (DiI) labeling of the frontal primordium by ex-utero surgery confirmed that apical extension of the frontal bone primordium of the mouse was severely retarded, while extension to the basal side underneath the brain was largely unaffected. Consistent with this observation, decreased cell proliferation activity was seen at the apical tip but not the basal tip of the frontal bone primordium as determined by double detection of Runx2 transcripts and BrdU incorporation. Furthermore, expression of the osteogenic-related genes Bmp4 and-7 was observed only in the basal part of the meninges during the initiation period of primordium growth. These results suggest that a loss of Foxc1 function affects skull bone formation of the apical region and that Bmp expression in the meninges might influence the growth of the calvarial bone primordium.
- Published
- 2014
12. FGF18 accelerates osteoblast differentiation by upregulatingBmp2expression
- Author
-
Sachiko Iseki, Noriko Tachikawa, Tomoko Nagayama, Shohei Kasugai, Masato S. Ota, and Shigeru Okuhara
- Subjects
Embryology ,Chemistry ,Mesenchyme ,Cellular differentiation ,Osteoblast ,General Medicine ,Anatomy ,FGF18 ,Fibroblast growth factor ,Bone morphogenetic protein ,Bone morphogenetic protein 2 ,Cell biology ,medicine.anatomical_structure ,Downregulation and upregulation ,Pediatrics, Perinatology and Child Health ,medicine ,Developmental Biology - Abstract
Fibroblast growth factor (FGF) signaling is involved in skeletal development. Among total 22 FGFs, it is suggested that FGF18 functions in promotion of osteoblast differentiation. In order to elucidate the mechanism of FGF18-dependent acceleration of osteogenesis, we implanted rhFGF18 soaked beads over mouse fetal coronal sutures using ex-utero surgery. The coronal suture area comprises the peripheries of the developing frontal and parietal bones, separated by the sutural mesenchyme. rhFGF18 accelerated osteogenesis by promoting connection of the frontal and parietal bone domains, resulting in elimination of the sutural mesenchyme. Expression of Fgf receptors, Fgfr1, -2 and -3 involved in skeletal development, was maintained or upregulated in the developing bone domains, consistent with enhanced osteogenesis. Bone morphogenetic protein (Bmp) 2 was specifically upregulated in the skeletogenic layer and the application of Bmp antagonist, rmNoggin, inhibited rhFGF18-dependent upregulation of osteoblast markers. These results suggest that FGF18 accelerates osteogenesis by upregulation of Bmp2 as well as maintenance or upregulation of Fgfr1, -2 and -3 expression in osteoblasts.
- Published
- 2013
13. Hes1 is required for the development of the superior cervical ganglion of sympathetic trunk and the carotid body
- Author
-
Sachiko Iseki, Tokio Katoh, Noriko Nemoto, Takayoshi Saitoh, and Yoko Kameda
- Subjects
Superior cervical ganglion ,Tyrosine 3-Monooxygenase ,Superior Cervical Ganglion ,Biology ,Mice ,Glomus cell ,medicine.artery ,Basic Helix-Loop-Helix Transcription Factors ,In Situ Nick-End Labeling ,medicine ,Animals ,Common carotid artery ,Homeodomain Proteins ,Mice, Knockout ,Carotid Body ,Neural crest ,Sympathetic trunk ,Anatomy ,medicine.disease ,Immunohistochemistry ,Hypoplasia ,medicine.anatomical_structure ,Neural Crest ,embryonic structures ,Transcription Factor HES-1 ,Carotid body ,Developmental Biology ,Artery - Abstract
Hes1 gene represses the expression of proneural basic helix–loop–helix (bHLH) factor Mash1, which is essential for the differentiation of the sympathetic ganglia and carotid body glomus cells. The sympathetic ganglia, carotid body, and common carotid artery in Wnt1-Cre/R26R double transgenic mice were intensely labeled by X-gal staining, i.e., the neural crest origin. The deficiency of Hes1 caused severe hypoplasia of the superior cervical ganglion (SCG). At embryonic day (E) 17.5–E18.5, the volume of the SCG in Hes1 null mutants was reduced to 26.4% of the value in wild-type mice. In 4 of 30 cases (13.3%), the common carotid artery derived from the third arch artery was absent in the null mutants, and the carotid body was not formed. When the common carotid artery was retained, the organ grew in the wall of the third arch artery and glomus cell precursors were provided from the SCG in the null mutants as well as in wild-types. However, the volume of carotid body in the null mutants was only 52.5% of the value in wild-types at E17.5–E18.5. These results suggest that Hes1 plays a critical role in regulating the development of neural crest derivatives in the mouse cervical region. Developmental Dynamics 241:1289–1300, 2012. V C 2012 Wiley Periodicals, Inc. Key findings: � Hes1 gene plays a role in maintaining the undifferentiated cells during development. � Hes1 null mutant embryos displayed a severe hypoplasia of the superior cervical ganglion (SCG) of sympathetic trunk. � Hes1 null mutants failed to form the common carotid artery as a partially penetrated phenotype (13.3%), resulting in the absence of the carotid body. � When the carotid artery was retained, the carotid body of the null mutants was smaller in size than that of wild types. � Neural crest derivatives, including the SCG, carotid body, and common carotid artery, were affected by the lack of Hes1.
- Published
- 2012
14. Expression of angiogenesis-related factors and inflammatory cytokines in placenta and umbilical vessels in pregnancies with preeclampsia and chorioamnionitis/funisitis
- Author
-
Atsuko Taki, Kikuko Oku, Motohiro Komaki, Mayumi Abe, Sachiko Iseki, Ikuo Morita, and Shuki Mizutani
- Subjects
Embryology ,Fetus ,Pregnancy ,medicine.medical_specialty ,business.industry ,Obstetrics ,General Medicine ,medicine.disease ,Chorioamnionitis ,Preeclampsia ,Proinflammatory cytokine ,Andrology ,Angiopoietin ,medicine.anatomical_structure ,Funisitis ,Placenta ,embryonic structures ,Pediatrics, Perinatology and Child Health ,cardiovascular system ,medicine ,business ,reproductive and urinary physiology ,Developmental Biology - Abstract
We hypothesized that gene expression in placenta and umbilical vessels are affected by intrauterine environment and some of the expression in umbilical vessels originating from the fetus could reflect fetal condition of these complicated pregnancies. Expression of angiogenesis-related factors and inflammatory cytokines were examined in placenta and umbilical vessels to clarify the effects of intrauterine environment of pregnancies complicated by preeclampsia and chorioamnionitis/funisitis. Forty-six preterm cesarean section deliveries were classified into three groups based on maternal condition during prenatal monitoring: preeclampsia (PE) (n = 11), chorioamnionitis/funisitis (CAM) (n = 8), and preterm control (PC) (n = 27). Angiogenesis-related factors and inflammatory cytokines in placentas, umbilical arteries and umbilical veins were analyzed by RT-PCR and immunohistochemistry. We demonstrated that Ang-2, Tie-2, and Dll4 increase in the placentas of PE compared to PC for the first time, and we confirmed the findings of previous reports showing the high expression of HIF-1α, sFlt-1, endoglin, leptin, and AT1R. Expression of angiogenesis-related factors, including HIF-1α, VEGF, angiopoietin, and TGF-β systems, and inflammatory cytokines, such as TNF-α and IL-6, increased in umbilical vessels of PE. Umbilical veins of CAM showed a higher Dll4 level than did PC. In preeclampsia, abnormal expressions of angiogenesis-related factors related to lifestyle diseases in adulthood were seen in the placenta and umbilical vessels as compared to PC. Chorioamnionitis/funisitis showed only upregulation of DII4 in umbilical veins.
- Published
- 2012
15. Effects of embryonic hypoxia on lip formation
- Author
-
Shigeru Okuhara, Sachiko Iseki, Yutaka Sato, Ryosuke Nagaoka, and Teruo Amagasa
- Subjects
Embryology ,Pathology ,medicine.medical_specialty ,Cleft Lip ,Mesenchyme ,Morphogenesis ,Apoptosis ,Biology ,Embryo Culture Techniques ,Mesoderm ,Mice ,chemistry.chemical_compound ,Downregulation and upregulation ,Pregnancy ,Mesenchymal cell proliferation ,medicine ,Animals ,Hypoxia ,Cell Proliferation ,Cell growth ,Gene Expression Regulation, Developmental ,General Medicine ,Hypoxia (medical) ,Embryonic stem cell ,Cell Hypoxia ,Lip ,Cell biology ,Mice, Inbred C57BL ,Vascular endothelial growth factor ,medicine.anatomical_structure ,chemistry ,Face ,Pediatrics, Perinatology and Child Health ,Female ,medicine.symptom ,Developmental Biology - Abstract
The upper lip is formed by the fusion of facial processes, a process in which many genetic and environmental factors are involved. Embryonic hypoxia is induced by uterine anemia and the administration of vasoconstrictors during pregnancy. To define the relationship between hypoxia and upper lip formation, hypoxic conditions were created in a whole embryo culture system. Hypoxic embryos showed a high frequency of impaired fusion, reflecting failure in the growth of the lateral nasal process (LNP). In hypoxic embryos, cell proliferation activity in the LNP mesenchyme was decreased following downregulation of genes that are involved in lip formation. We also observed upregulation of vascular endothelial growth factor expression along with the induction of apoptosis in the LNP. These results suggest that embryonic hypoxia during lip formation induces apoptosis in physiologically hypoxic regions, hypoxia-induced gene expression and downregulation of the genes involved in maxillofacial morphogenesis as immediate responses, followed by reduction of mesenchymal cell proliferation activity, resulting in insufficient growth of the facial processes. Birth Defects Research (Part A), 2012. © 2012 Wiley Periodicals, Inc.
- Published
- 2012
16. Disintegration of the medial epithelial seam: Is cell death important in palatogenesis?
- Author
-
Sachiko Iseki
- Subjects
Programmed cell death ,animal structures ,Mesenchymal stem cell ,Palatal shelves ,Cell Biology ,Anatomy ,Cell fate determination ,Biology ,Epithelium ,Cell biology ,medicine.anatomical_structure ,Apoptosis ,Nasal septum ,medicine ,Process (anatomy) ,Developmental Biology - Abstract
During palatogenesis, the palatal medial edge epithelium (MEE) forms the medial epithelial seam (MES) on adhesion of the opposing palatal shelves. The MES eventually disappears, leading to mesenchymal confluence of the palate and completion of palatogenesis. Failure of these processes results in cleft palate, one of the most common congenital anomalies in human affecting around one case in 500-2500 live births. The cell fate of MEE has been controversial for more than 20 years. Recent studies suggest that the disappearance of MES is a complex process involving cell death, epithelial-mesenchymal transition (EMT) and epithelial migration. Interestingly, transforming growth factor-β3 (Tgf β3) expression in MEE and the tip epithelium of the nasal septum begins just before palatal shelf reorientation and lasts until MES disruption, and several works including targeted disruption of the gene have indicated that the process appears to be regulated mainly by the TGFβ3-TGFβR signaling. However, how MEE cells choose their fate and how the cell fate is altered in response to cellular environment remains to be elucidated.
- Published
- 2011
17. Runx1 and Runx2 cooperate during sternal morphogenesis
- Author
-
Kenichi Shinomiya, Yasuhiro Takeuchi, Seiji Fukumoto, Shingo Sato, Keisuke Ae, Makiko Iwasaki, Koji Fujita, Hiroshi Itoh, Toshiyuki Ikeda, Fumiko Yano, Sachiko Iseki, Ayako Kimura, Hiroyuki Inose, Shu Takeda, Hiroshi Kawaguchi, Ung-il Chung, Takeshi Imamura, James F. Martin, and Tetsuya Jinno
- Subjects
Sternum ,Transgene ,Morphogenesis ,Core Binding Factor Alpha 1 Subunit ,SOX9 ,In situ hybridization ,Biology ,Bone and Bones ,Chondrocyte ,Mesoderm ,Mice ,chemistry.chemical_compound ,Chondrocytes ,medicine ,Animals ,Humans ,Cell Lineage ,Transgenes ,Molecular Biology ,Research Articles ,Homeodomain Proteins ,Mice, Knockout ,Stem Cells ,Mesenchymal stem cell ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell biology ,RUNX2 ,Cartilage ,medicine.anatomical_structure ,RUNX1 ,chemistry ,Core Binding Factor Alpha 2 Subunit ,embryonic structures ,Immunology ,SOXD Transcription Factors ,HeLa Cells ,Developmental Biology - Abstract
Chondrocyte differentiation is strictly regulated by various transcription factors, including Runx2 and Runx3; however, the physiological role of Runx1 in chondrocyte differentiation remains unknown. To examine the role of Runx1, we generated mesenchymal-cell-specific and chondrocyte-specific Runx1-deficient mice [Prx1 Runx1(f/f) mice and alpha1(II) Runx1(f/f) mice, respectively] to circumvent the embryonic lethality of Runx1-deficient mice. We then mated these mice with Runx2 mutant mice to obtain mesenchymal-cell-specific or chondrocyte-specific Runx1; Runx2 double-mutant mice [Prx1 DKO mice and alpha1(II) DKO mice, respectively]. Prx1 Runx1(f/f) mice displayed a delay in sternal development and Prx1 DKO mice completely lacked a sternum. By contrast, alpha1(II) Runx1(f/f) mice and alpha1(II) DKO mice did not show any abnormal sternal morphogenesis or chondrocyte differentiation. Notably, Runx1, Runx2 and the Prx1-Cre transgene were co-expressed specifically in the sternum, which explains the observation that the abnormalities were limited to the sternum. Histologically, mesenchymal cells condensed normally in the prospective sternum of Prx1 DKO mice; however, commitment to the chondrocyte lineage, which follows mesenchymal condensation, was significantly impaired. In situ hybridization analyses demonstrated that the expression of alpha1(II) collagen (Col2a1 - Mouse Genome Informatics), Sox5 and Sox6 in the prospective sternum of Prx1 DKO mice was severely attenuated, whereas Sox9 expression was unchanged. Molecular analyses revealed that Runx1 and Runx2 induce the expression of Sox5 and Sox6, which leads to the induction of alpha1(II) collagen expression via the direct regulation of promoter activity. Collectively, these results show that Runx1 and Runx2 cooperatively regulate sternal morphogenesis and the commitment of mesenchymal cells to become chondrocytes through the induction of Sox5 and Sox6.
- Published
- 2010
18. Development and tissue origins of the mammalian cranial base
- Author
-
Simon D. Bamforth, Gillian M. Morriss-Kay, Sachiko Iseki, Brandeis McBratney-Owen, and Bjorn R. Olsen
- Subjects
New Head Hypothesis ,Mesoderm ,Indoles ,Time Factors ,Wnt1 ,Morphogenesis ,Mice, Transgenic ,Mesp1 ,Wnt1 Protein ,Biology ,Article ,Chondrocranium ,Mice ,Neural crest ,03 medical and health sciences ,Tissue origins ,0302 clinical medicine ,Cranial vault ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Cell Lineage ,Cranial base ,Craniofacial ,Promoter Regions, Genetic ,Molecular Biology ,In Situ Hybridization ,Cranial development ,030304 developmental biology ,Skull Base ,0303 health sciences ,Staining and Labeling ,Cartilage ,Galactosides ,Cell Biology ,Anatomy ,Embryo, Mammalian ,Chondrogenesis ,Antigens, Differentiation ,Mice, Inbred C57BL ,medicine.anatomical_structure ,embryonic structures ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The vertebrate cranial base is a complex structure composed of bone, cartilage and other connective tissues underlying the brain; it is intimately connected with development of the face and cranial vault. Despite its central importance in craniofacial development, morphogenesis and tissue origins of the cranial base have not been studied in detail in the mouse, an important model organism. We describe here the location and time of appearance of the cartilages of the chondrocranium. We also examine the tissue origins of the mouse cranial base using a neural crest cell lineage cell marker, Wnt1-Cre/R26R, and a mesoderm lineage cell marker, Mesp1-Cre/R26R. The chondrocranium develops between E11 and E16 in the mouse, beginning with development of the caudal (occipital) chondrocranium, followed by chondrogenesis rostrally to form the nasal capsule, and finally fusion of these two parts via the midline central stem and the lateral struts of the vault cartilages. X-Gal staining of transgenic mice from E8.0 to 10 days post-natal showed that neural crest cells contribute to all of the cartilages that form the ethmoid, presphenoid, and basisphenoid bones with the exception of the hypochiasmatic cartilages. The basioccipital bone and non-squamous parts of the temporal bones are mesoderm derived. Therefore the prechordal head is mostly composed of neural crest-derived tissues, as predicted by the New Head Hypothesis. However, the anterior location of the mesoderm-derived hypochiasmatic cartilages, which are closely linked with the extra-ocular muscles, suggests that some tissues associated with the visual apparatus may have evolved independently of the rest of the “New Head”.
- Published
- 2008
19. Dual odontogenic origins develop at the early stage of rat maxillary incisor development
- Author
-
Sachiko Iseki, Kazuhiro Eto, Rungarun Kriangkrai, and Suconta Chareonvit
- Subjects
Embryology ,Fibroblast Growth Factor 8 ,Ectomesenchyme ,Embryonic Development ,Bone Morphogenetic Protein 4 ,Rats, Sprague-Dawley ,stomatognathic system ,Gene expression ,Maxilla ,Animals ,Hedgehog Proteins ,Sonic hedgehog ,Stage (cooking) ,Process (anatomy) ,Homeodomain Proteins ,MSX1 Transcription Factor ,Regulation of gene expression ,biology ,Gene Expression Regulation, Developmental ,Cell Biology ,Anatomy ,Rats ,Incisor ,stomatognathic diseases ,Bone morphogenetic protein 4 ,Bone Morphogenetic Proteins ,Trans-Activators ,biology.protein ,Odontogenesis ,Homeobox ,PAX9 Transcription Factor ,Transcription Factors ,Developmental Biology - Abstract
Developmental process of rat maxillary incisor has been studied through histological analysis and investigation of tooth-related gene expression patterns at initial tooth development. The tooth-related genes studied here are fibroblast growth factor-8 (Fgf-8), pituitary homeobox gene-2 (Pitx-2), sonic hedgehog (Shh), muscle segment homeobox-1 (Msx-1), paired box-9 (Pax-9) and bone morphogenetic protein-4 (Bmp-4). The genes are expressed in oral epithelium and/or ectomesenchyme at the stage of epithelial thickening to the early bud stage of tooth development. Both the histological observation and tooth-related gene expression patterns during early stage of maxillary incisor development demonstrate that dual odontogenic origins aligned medio-laterally in the medial nasal process develop, subsequently only single functional maxillary incisor dental placode forms. The cascade of tooth-related gene expression patterns in rat maxillary incisor studied here is quite similar to those of the previous studies in mouse mandibular molar, even though the origins of oral epithelium and ectomesenchyme involved in development of maxillary incisor and mandibular molar are different. Thus, we conclude that maxillary incisor and mandibular molar share a similar signaling control of Fgf-8, Pitx-2, Shh, Msx-1, Pax-9 and Bmp-4 genes at the stage of oral epithelial thickening to the early bud stage of tooth development.
- Published
- 2006
20. Study ofPax6 mutant rat revealed the association between upper incisor formation and midface formation
- Author
-
Koichi Yahagi, Rungarun Kriangkrai, Michio Fujiwara, Suconta Chareonvit, Kazuhiro Eto, and Sachiko Iseki
- Subjects
Time Factors ,PAX6 Transcription Factor ,Biology ,Epithelium ,Rats, Sprague-Dawley ,stomatognathic system ,Incisor ,Maxilla ,otorhinolaryngologic diseases ,medicine ,Animals ,Paired Box Transcription Factors ,Supernumerary ,Eye Proteins ,Process (anatomy) ,Homeodomain Proteins ,Facial cleft ,Wild type ,Gene Expression Regulation, Developmental ,Embryo ,Anatomy ,medicine.disease ,Rats ,Repressor Proteins ,stomatognathic diseases ,medicine.anatomical_structure ,Face ,Mutation ,PAX6 ,Developmental Biology - Abstract
In this study, we investigated the process of supernumerary upper incisor formation in the Pax6 mutant rat, rSey 2 /rSey 2 , which exhibits a facial cleft between the medial nasal and maxillary processes. Histological investigation and epithelial labeling studies of wild type rat embryos indicated that the upper incisor develops by fusion of two primary dental placodes (PDPs) in the medial nasal process with a contribution from the epithelium of the maxillary process. In the rSey 2 /rSey 2 embryo, both PDPs are formed but they stay apart, then subsequently these PDPs independently develop into upper incisor tooth buds. In order to examine if the failure of the two placodes to fuse is due to the cleft between the maxillary and medial nasal processes, maxillary and medial nasal process fusion was inhibited with a barrier in wild type embryos. This resulted in the maintenance of the two distinct PDPs. These results demonstrate that fusion of the facial processes reduces the number of odontogenic placodes and is required to assemble all components at one site for rat upper incisor formation. The results also provide further insight into the mechanism of supernumerary incisor formation in human cleft lip conditions. Developmental Dynamics 235:2134 –2143, 2006. © 2006 Wiley-Liss, Inc.
- Published
- 2006
21. Expression patterns of Twist and Fgfr1, -2 and -3 in the developing mouse coronal suture suggest a key role for Twist in suture initiation and biogenesis
- Author
-
Gillian M. Morriss-Kay, Andrew O.M. Wilkie, David Johnson, and Sachiko Iseki
- Subjects
Embryology ,animal structures ,Mesenchyme ,Gene Expression ,Biology ,Craniosynostosis ,Embryonic and Fetal Development ,Mice ,medicine ,Animals ,Receptor, Fibroblast Growth Factor, Type 3 ,Receptor, Fibroblast Growth Factor, Type 1 ,Receptor, Fibroblast Growth Factor, Type 2 ,Genetics ,Fibroblast growth factor receptor 1 ,Twist-Related Protein 1 ,Nuclear Proteins ,Receptor Protein-Tyrosine Kinases ,Cranial Sutures ,Protein-Tyrosine Kinases ,medicine.disease ,Receptors, Fibroblast Growth Factor ,Embryonic stem cell ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Fibroblast growth factor receptor ,Coronal suture ,Stem cell ,Haploinsufficiency ,Transcription Factors ,Developmental Biology - Abstract
Sutural growth depends on maintenance of a balance between proliferation of osteogenic stem cells and their differentiation to form new bone, so that the stem cell population is maintained until growth of the skull is complete. The identification of heterozygous mutations in FGFR1, -2 and -3 and TWIST as well as microdeletions of TWIST in human craniosynostosis syndromes has highlighted these genes as playing important roles in maintaining the suture as a growth centre. In contrast to Drosophila, a molecular relationship between human (or other vertebrate) TWIST and FGFR genes has not yet been established. TWIST mutations exert their effect via haploinsufficiency whereas FGFR mutations have a gain-of-function mechanism of action. To investigate the biological basis of FGFR signalling pathways in the developing calvarium we compared the expression patterns of Twist with those of Fgfr1, -2 and -3 in the fetal mouse coronal suture over the course of embryonic days 14-18, as the suture is initiated and matures. Our results show that: (1) Twist expression precedes that of Fgfr genes at the time of initiation of the coronal suture; (2) in contrast to Fgfr transcripts, which are localised within and around the developing bone domains, Twist is expressed by the midsutural mesenchyme cells. Twist expression domains show some overlap with those of Fgfr2, which is expressed in the most immature (proliferating) osteogenic tissue.
- Published
- 2000
22. Fgfr1 and Fgfr2 have distinct differentiation- and proliferation-related roles in the developing mouse skull vault
- Author
-
Sachiko Iseki, Andrew O.M. Wilkie, and Gillian M. Morriss-Kay
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Mesenchyme ,Gene Expression ,Biology ,Fibroblast growth factor ,Craniosynostosis ,Mice ,medicine ,Animals ,Receptor, Fibroblast Growth Factor, Type 1 ,Receptor, Fibroblast Growth Factor, Type 2 ,Molecular Biology ,Cartilage ,Skull ,Receptor Protein-Tyrosine Kinases ,Cell Differentiation ,Anatomy ,medicine.disease ,Receptors, Fibroblast Growth Factor ,Cell biology ,Mice, Inbred C57BL ,stomatognathic diseases ,medicine.anatomical_structure ,Fibroblast growth factor receptor ,embryonic structures ,biology.protein ,Coronal suture ,Osteonectin ,Stem cell ,Cell Division ,Developmental Biology - Abstract
Fibroblast growth factor receptors (FGFRs) play major roles in skeletogenesis, and activating mutations of the human FGFR1, FGFR2 and FGFR3 genes cause premature fusion of the skull bones (craniosynostosis). We have investigated the patterns of expression of Fgfr1, Fgfr2 and Fgfr3 in the fetal mouse head, with specific reference to their relationship to cell proliferation and differentiation in the frontal and parietal bones and in the coronal suture. Fgfr2 is expressed only in proliferating osteoprogenitor cells; the onset of differentiation is preceded by down-regulation of Fgfr2 and up-regulation of Fgfr1. Following up-regulation of the differentiation marker osteopontin, Fgfr1, osteonectin and alkaline phosphatase are down-regulated, suggesting that they are involved in the osteogenic differentiation process but not in maintaining the differentiated state. Fgfr3 is expressed in the cranial cartilage, including a plate of cartilage underlying the coronal suture, as well as in osteogenic cells, suggesting a dual role in skull development. Subcutaneous insertion of FGF2-soaked beads onto the coronal suture on E15 resulted in up-regulation of osteopontin and Fgfr1 in the sutural mesenchyme, down-regulation of Fgfr2, and inhibition of cell proliferation. This pattern was observed at 6 and 24 hours after bead insertion, corresponding to the timing and duration of FGF2 diffusion from the beads. We suggest (a) that a gradient of FGF ligand, from high levels in the differentiated region to low levels in the environment of the osteogenic stem cells, modulates differential expression of Fgfr1 and Fgfr2, and (b) that signalling through FGFR2 regulates stem cell proliferation whereas signalling through FGFR1 regulates osteogenic differentiation.
- Published
- 1999
23. Polarizing activity, Sonic hedgehog , and tooth development in embryonic and postnatal mouse
- Author
-
Judith L. Leatherman, Tomoichiro Yamaai, Eiki Koyama, Sachiko Iseki, Eleanor B. Golden, Hideyo Ohuchi, Hidefumi Yoshioka, Sumihare Noji, Tsutomu Nohno, Maurizio Pacifici, and Yoshio Hayashi
- Subjects
Molar ,biology ,Mesenchyme ,Enamel organ ,Anatomy ,Enamel knot ,Cell biology ,stomatognathic diseases ,medicine.anatomical_structure ,stomatognathic system ,Zone of polarizing activity ,Incisor ,medicine ,biology.protein ,Sonic hedgehog ,Ameloblast ,Developmental Biology - Abstract
Tooth development involves reciprocal epithelial-mesenchymal interactions, polarized growth, mesenchyme condensation, and complex morphogenetic events. Because these processes bear similarities to those occurring in the developing limb, we asked whether morphogenetic signals found in the limb also occur in the developing tooth. We grafted mouse embryo tooth germs to the anterior margin of host chick embryo wing buds and determined whether the dental tissues had polarizing activity. Indeed, the grafts induced supernumerary digits. Activity of both molar and incisor tooth germs increased from bud to cap stages and was maximal at late bell stage in newborn. With further development the polarizing activity began to decrease, became undetectable in adult molar mesenchyme but persisted in incisor mesenchyme, correlating with the fact that incisors grow throughout postnatal life while molars do not. When different portions of neonatal incisors were assayed, a clear proximo-distal gradient of activity was apparent, with maximal activity restricted to the most proximal portion where undifferentiated mesenchyme and enamel organ reside. In situ hybridizations demonstrated that prior to induction of supernumerary digits, the tooth germ grafts induced expression in host tissue of Hoxd-12 and Hoxd-13. In addition, whole-mount in situ hybridizations and immunohistochemistry showed that developing tooth germs express Sonic hedgehog (Shh). Shh expression was first detected in bud stage tooth germs; at later stages Shh transcripts were prominent in enamel knot and differentiating ameloblasts at the cuspal region. We concluded that tooth germs possess polarizing activity and produce polarizing factors such as Shh. As in the limb, these factor(s) and activity probably play key roles in establishing polarity and regulating morphogenesis during early tooth development. Given its subsequent association with differentiating ameloblasts, Shh probably participates also in cytogenetic events during odontogenesis. © 1996 Wiley-Liss, Inc.
- Published
- 1996
24. Retinoic Acid Treatment Induces the Ectopic Exporession of Retinoic Acid Receptor beta Gene and Excessive Cell Death in the Embryonic Mouse Face
- Author
-
Kazuhiro Eto, Noriko Osumi-Yamashita, Sumihare Noji, Sachiko Iseki, Hirofumi Doi, Tsutomu Nohno, Shigehiko Taniguchi, and Eiki Koyama
- Subjects
medicine.medical_specialty ,Programmed cell death ,Retinoic acid ,Morphogenesis ,Cell Biology ,In situ hybridization ,Biology ,Retinoic acid receptor ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Internal medicine ,embryonic structures ,Gene expression ,medicine ,Ectopic expression ,Receptor ,Developmental Biology - Abstract
Maternal treatment with 100 mg/kg of retinoic acid (RA) on day 9 of gestation in mice caused craniofacial abnormalities of the mandibulofacial dysostosis type. The abnormal morphology was attributed to the excessive cell death in the dorsal aspects of the maxillary and mandibular prominences of the first pharyngeal arch and in the proximal region of the mandibular prominence. To investigate the expression of the RA receptor (RAR) genes in abnormal face morphogenesis, in situ hybridization was performed. The distribution patterns of RAR α and γ transcripts were not altered in the treated embryos. By contrast, the teratogenic dose of RA increased the level of RAR β transcripts, as early as 3 hr after RA-treatment, in the regions where the RAR β expression is at a low level in normal development. The increase of RAR β transcripts was detected by 12 hr, and declined to the low level within 24 hr after the treatment. The regions where ectopic expression of RAR β gene was observed included the areas where the excessive cell death occurred 9–12 hr after RA-treatment. These results suggest that ectopic induction of RAR β by RA may lead to the excessive cell death, threfore may cause abnormal morphogenesis.
- Published
- 1992
25. 09-P026 Combined in silico and in vivo analyses reveal role of Hes1 in taste cell differentiation
- Author
-
Takashi Kondo, Kaori Kondo, Hiroshi Tanaka, Yoshiyuki Kaneko, Soichi Ogishima, Sachiko Iseki, and Masato S. Ota
- Subjects
Embryology ,Taste cell ,In vivo ,In silico ,HES1 ,Biology ,Cell biology ,Developmental Biology - Published
- 2009
- Full Text
- View/download PDF
26. 03-P117 Involvement of cis-regulatory element of sonic hedgehog in palatal development
- Author
-
Sachiko Iseki, Toshihiko Shiroishi, Shigeru Okuhara, and Tomoko Sagai
- Subjects
Embryology ,biology.protein ,Biology ,Sonic hedgehog ,Cis-regulatory element ,Cell biology ,Developmental Biology - Published
- 2009
- Full Text
- View/download PDF
27. Association of tenascin-W expression with mineralization in mouse calvarial development
- Author
-
Masahiro Saito, Koichi Ueda, Shigeru Okuhara, Sachiko Iseki, Ayako Mikura, and Masato S. Ota
- Subjects
endocrine system ,Embryology ,animal structures ,Biology ,Fibroblast growth factor ,Mineralization (biology) ,Andrology ,Frontal suture ,Extracellular matrix ,Mice ,Calcification, Physiologic ,Downregulation and upregulation ,Osteogenesis ,medicine ,Animals ,In Situ Hybridization ,Fetus ,Bone Development ,Osteoblasts ,Skull ,Osteoblast ,Cell Differentiation ,Tenascin ,General Medicine ,Anatomy ,musculoskeletal system ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Animals, Newborn ,Tenascin Family ,embryonic structures ,Pediatrics, Perinatology and Child Health ,Fibroblast Growth Factor 2 ,Developmental Biology - Abstract
Tenascin-W is a tenascin family member that forms part of a complex extracellular matrix, and previous studies have suggested its association with osteogenesis. In the present study we investigated the roles of tenascin-W in osteogenesis. We found that tenascin-W is expressed in osteoblasts at the edge of the developing bone domain prior to mineralization in mouse fetuses. Expression of tenascin-W was induced during the course of mineralization of the Kusa-A1 osteoblast cell line. In the interfrontal suture of postnatal mice, the anterior portion remains patent and the posterior portion closes by 4 weeks of age. Tenascin-W expression was downregulated at 1 week of age in the posterior frontal suture, whereas in the anterior suture, expression was maintained until the mice reached 4 weeks of age. Fibroblast growth factor 2 (FGF2)-bead application to the mouse fetal skull by ex-utero surgery accelerated osteoblast differentiation, but inhibited mineralization with a downregulation of tenascin-W expression. These results suggest that tenascin-W is involved in osteoblast maturation (i.e. mineralization).
- Published
- 2009
28. Impaired meningeal development in association with apical expansion of calvarial bone osteogenesis in the Foxc1 mutant
- Author
-
Kirsi Sainio, Shizuko Ichinose, Kazuhiro Eto, Philaiporn Vivatbutsiri, Sachiko Iseki, and Marjo K. Hytönen
- Subjects
Pathology ,medicine.medical_specialty ,Histology ,Dura mater ,Biology ,Cell morphology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Meninges ,Microscopy, Electron, Transmission ,medicine ,Animals ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,In Situ Hybridization ,030304 developmental biology ,0303 health sciences ,Bone Development ,Pia mater ,Skull ,Wild type ,Gene Expression Regulation, Developmental ,Forkhead Transcription Factors ,Cell Biology ,Anatomy ,Original Articles ,Immunohistochemistry ,Mice, Mutant Strains ,Apex (geometry) ,medicine.anatomical_structure ,Frontal bone ,Dura Mater ,Arachnoid ,030217 neurology & neurosurgery ,Gene Deletion ,Developmental Biology ,Hydrocephalus - Abstract
Loss of function of the mouse forkhead/winged helix transcription factor Foxc1 induces congenital hydrocephalus and impaired skull bone development due to failure of apical expansion of the bone. In this study we investigated meningeal development in the congenital hydrocephalus (ch) mouse with spontaneous loss of function mutant of Foxc1, around the period of initiation of skull bone apical expansion. In situ hybridization of Runx2 revealed active apical expansion of the frontal bone begins between embryonic day 13.5 and embryonic day 14.5 in the wild type, whereas expansion was inhibited in the mutant. Ultrastructural analysis revealed that three layers of the meninges begin to develop at E13.5 in the basolateral site of the head and subsequently progress to the apex in wild type. In ch homozygotes, although three layers were recognized at first at the basolateral site, cell morphology and structure of the layers became abnormal except for the pia mater, and arachnoidal and dural cells never differentiated in the apex. We identified meningeal markers for each layer and found that their expression was down-regulated in the mutant arachnoid and dura maters. These results suggest that there is a close association between meningeal development and the apical growth of the skull bones.
- Published
- 2008
29. Cell lineage in mammalian craniofacial mesenchyme
- Author
-
Toshiyuki Yoshida, Gillian M. Morriss-Kay, Sachiko Iseki, Yumiko Saga, and Philaiporn Vivatbutsiri
- Subjects
Embryology ,Mesoderm ,animal structures ,Ectomesenchyme ,Mesenchyme ,Biology ,Muscle Development ,Parietal Bone ,Mice ,Cell Movement ,medicine ,Animals ,Cell Lineage ,Mammals ,Neural fold ,Skull ,Neural crest ,Endothelial Cells ,Anatomy ,beta-Galactosidase ,medicine.anatomical_structure ,Connective Tissue ,Neural Crest ,embryonic structures ,Frontal Bone ,Eye development ,Neural crest cell migration ,Neural plate ,Developmental Biology - Abstract
We have analysed the contributions of neural crest and mesoderm to mammalian craniofacial mesenchyme and its derivatives by cell lineage tracing experiments in mouse embryos, using the permanent genetic markers Wnt1-cre for neural crest and Mesp1-cre for mesoderm, combined with the Rosa26 reporter. At the end of neural crest cell migration (E9.5) the two patterns are reciprocal, with a mutual boundary just posterior to the eye. Mesodermal cells expressing endothelial markers (angioblasts) are found not to respect this boundary; they are associated with the migrating neural crest from the 5-somite stage, and by E9.5 they form a pre-endothelial meshwork throughout the cranial mesenchyme. Mesodermal cells of the myogenic lineage also migrate with neural crest cells, as the branchial arches form. By E17.5 the neural crest-mesoderm boundary in the subectodermal mesenchyme becomes out of register with that of the underlying skeletogenic layer, which is between the frontal and parietal bones. At E13.5 the primordia of these bones lie basolateral to the brain, extending towards the vertex of the skull during the following 4–5 days. We used DiI labelling of the bone primordia in ex-utero E13.5 embryos to distinguish between two possibilities for the origin of the frontal and parietal bones: (1) recruitment from adjacent connective tissue or (2) proliferation of the original primordia. The results clearly demonstrated that the bone primordia extend vertically by intrinsic growth, without detectable recruitment of adjacent mesenchymal cells.
- Published
- 2008
30. Twist induces reversal of myotube formation
- Author
-
Mustafa Anayasa, Eleni Hjiantoniou, Paschalis Nicolaou, Ioannis Bantounas, James B. Uney, Leonidas A. Phylactou, Sachiko Iseki, and Masahiro Saito
- Subjects
Cancer Research ,Myogenesis ,Cellular differentiation ,Muscle Fibers, Skeletal ,Twist-Related Protein 1 ,Nuclear Proteins ,Cell Differentiation ,Cell Biology ,Biology ,Cell cycle ,MyoD ,Muscle Development ,Molecular biology ,Cell biology ,Twist transcription factor ,MyoD Protein ,Myocyte ,Animals ,Molecular Biology ,Transcription factor ,Developmental Biology - Abstract
Mammals possess reduced ability to regenerate lost tissue, compared with other vertebrates, which can regenerate through differentiation of precursor cells or de-differentiation. Mammalian multinucleated myotube formation is a differentiation process, which arises from the fusion of mononucleated myoblasts and is thought to be an irreversible process toward muscle formation. By overexpressing the Twist gene in terminally differentiated myotubes, we managed to induce reversal of cell differentiation. More specifically, following expression of the Twist gene, myotubes underwent morphological changes that caused them to cleave. This was accompanied by a reduction in the expression of certain myogenic markers. Interestingly, Twist overexpression also caused a reduction in the muscle transcription factor MyoD. Further experiments showed an increase in the cell cycle entry molecule, cyclin D1 and initiation of DNA synthesis, due to Twist overexpression. The exploitation of Twist-mediated reversal of differentiation and the study of its specific mechanism would be important in order to study mammalian cellular de-differentiation and determine its potential in muscle regeneration.
- Published
- 2007
31. Twist is required for establishment of the mouse coronal suture
- Author
-
Sachiko Iseki, Kazuhiro Eto, Leonidas A. Phylactou, Toshiyuki Yoshida, Isao Ishikawa, and James B. Uney
- Subjects
Histology ,animal structures ,Morpholino ,Blotting, Western ,Biology ,Craniosynostosis ,Twist transcription factor ,Mice ,Organ Culture Techniques ,medicine ,Animals ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,In Situ Hybridization ,Cell Proliferation ,Fibrous joint ,Osteoblasts ,Skull ,Twist-Related Protein 1 ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Osteoblast ,Cell Differentiation ,Cell Biology ,Anatomy ,Cranial Sutures ,Original Articles ,Acrocephalosyndactylia ,Oligonucleotides, Antisense ,medicine.disease ,Immunohistochemistry ,RUNX2 ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Coronal suture ,Developmental Biology ,Transcription Factors - Abstract
Cranial sutures are the growth centres of the skull, enabling expansion of the skull to accommodate rapid growth of the brain. Haploinsufficiency of the human TWIST gene function causes the craniosynostosis syndrome, Saethre-Chotzen syndrome (SCS), in which premature fusion of the coronal suture is a characteristic feature. Previous studies have indicated that Twist is expressed in the coronal suture during development, and therefore that it may play an important role in development and maintenance of the suture. The Twist-null mouse is lethal before the onset of osteogenesis, and the heterozygote exhibits coronal suture synostosis postnatally. In this study we investigated the function of Twist in the development of the mouse coronal suture, by inhibiting Twist synthesis using morpholino antisense oligonucleotides in calvarial organ culture. Decreased Twist production resulted in a narrow sutural space and fusion of bone domains within 48 h after the addition of the morpholino oligonucleotides. Proliferation activity in the sutural cells was decreased, and the expression of osteogenic marker genes such as Runx2 and Fgfr2 was up-regulated in the developing bone domain within 4 h. These results suggest that during establishment of the suture area, Twist is required for the regulation of sutural cell proliferation and osteoblast differentiation.
- Published
- 2005
32. Tissue origins and interactions in the mammalian skull vault
- Author
-
Gillian M. Morriss-Kay, Henry M. Sucov, Robert E. Maxson, Xiaobing Jiang, and Sachiko Iseki
- Subjects
Genetic Markers ,Physiology and anatomy ,animal structures ,Wnt1 ,Mice, Transgenic ,Tretinoin ,Wnt1 Protein ,Biology ,Mesoderm ,Parietal Bone ,Mice ,Meninges ,Osteogenesis ,Proto-Oncogene Proteins ,Cranial vault ,retinoic acid ,medicine ,Animals ,Molecular Biology ,Body Patterning ,Neural fold ,Ossification ,Skull ,Neural crest ,Brain ,Anatomy ,Cell Biology ,Cranial Sutures ,Zebrafish Proteins ,Biological Evolution ,Wnt Proteins ,medicine.anatomical_structure ,Neural Crest ,Intramembranous ossification ,embryonic structures ,Coronal suture ,medicine.symptom ,mouse development ,skull sutures ,Development (zoology) ,Neural plate ,Developmental Biology - Abstract
During mammalian evolution, expansion of the cerebral hemispheres was accompanied by expansion of the frontal and parietal bones of the skull vault and deployment of the coronal (fronto-parietal) and sagittal (parietal–parietal) sutures as major growth centres. Using a transgenic mouse with a permanent neural crest cell lineage marker, Wnt1-Cre/R26R, we show that both sutures are formed at a neural crest–mesoderm interface: the frontal bones are neural crest-derived and the parietal bones mesodermal, with a tongue of neural crest between the two parietal bones. By detailed analysis of neural crest migration pathways using X-gal staining, and mesodermal tracing by DiI labelling, we show that the neural crest–mesodermal tissue juxtaposition that later forms the coronal suture is established at E9.5 as the caudal boundary of the frontonasal mesenchyme. As the cerebral hemispheres expand, they extend caudally, passing beneath the neural crest–mesodermal interface within the dermis, carrying with them a layer of neural crest cells that forms their meningeal covering. Exposure of embryos to retinoic acid at E10.0 reduces this meningeal neural crest and inhibits parietal ossification, suggesting that intramembranous ossification of this mesodermal bone requires interaction with neural crest-derived meninges, whereas ossification of the neural crest-derived frontal bone is autonomous. These observations provide new perspectives on skull evolution and on human genetic abnormalities of skull growth and ossification.
- Published
- 2002
33. Fgfr2 and osteopontin domains in the developing skull vault are mutually exclusive and can be altered by locally applied FGF2
- Author
-
John K. Heath, Gillian M. Morriss-Kay, Kazuhiro Eto, T. Ishimaru, Sachiko Iseki, and Andrew O.M. Wilkie
- Subjects
musculoskeletal diseases ,medicine.medical_specialty ,Sialoglycoproteins ,Fibroblast growth factor ,Models, Biological ,Craniosynostosis ,Paracrine signalling ,Craniosynostoses ,Mice ,Osteogenesis ,Pregnancy ,Internal medicine ,Cranial vault ,medicine ,Animals ,Humans ,Osteopontin ,Receptor, Fibroblast Growth Factor, Type 2 ,Molecular Biology ,In Situ Hybridization ,Regulation of gene expression ,integumentary system ,biology ,Skull ,Gene Expression Regulation, Developmental ,Receptor Protein-Tyrosine Kinases ,Cell Differentiation ,Cranial Sutures ,medicine.disease ,Immunohistochemistry ,Receptors, Fibroblast Growth Factor ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,Fibroblast growth factor receptor ,embryonic structures ,Mutation ,biology.protein ,Female ,Fibroblast Growth Factor 2 ,Coronal suture ,Cell Division ,Developmental Biology ,Signal Transduction - Abstract
Mutations in the human fibroblast growth factor receptor type 2 (FGFR2) gene cause craniosynostosis, particularly affecting the coronal suture. We show here that, in the fetal mouse skull vault, Fgfr2 transcripts are most abundant at the periphery of the membrane bones; they are mutually exclusive with those of osteopontin (an early marker of osteogenic differentiation) but coincide with sites of rapid cell proliferation. Fibroblast growth factor type 2 (FGF2) protein, which has a high affinity for the FGFR2 splice variant associated with craniosynostosis, is locally abundant; immunohistochemical detection showed it to be present at low levels in Fgfr2 expression domains and at high levels in differentiated areas. Implantation of FGF2-soaked beads onto the fetal coronal suture by ex utero surgery resulted in ectopic osteopontin expression, encircled by Fgfr2 expression, after 48 hours. We suggest that increased FGF/FGFR signalling in the developing skull, whether due to FGFR2 mutation or to ectopic FGF2, shifts the cell proliferation/differentiation balance towards differentiation by enhancing the normal paracrine down-regulation of Fgfr2.
- Published
- 1997
34. P101. Cleft palate in compound heterozygote of sonic hedgehog and MFCS4
- Author
-
Toshihiko Shiroishi, Takanori Amano, Ryosuke Nagaoka, Shigeru Okuhara, Sachiko Iseki, and Tomoko Sagai
- Subjects
Cancer Research ,education.field_of_study ,animal structures ,biology ,Population ,Pharynx ,Cell Biology ,In situ hybridization ,Anatomy ,Epithelium ,Cell biology ,medicine.anatomical_structure ,Tongue ,embryonic structures ,medicine ,biology.protein ,Myocyte ,Sonic hedgehog ,Secondary palate ,education ,Molecular Biology ,Developmental Biology - Abstract
Although cleft palate is the most common birth defect of human, detailed molecular mechanism in it is not fully elucidated. Palate development is achieved through descending, reorientation and fusion of palatal shelves in association with neighboring organs including mandible and tongue. Sonic hedgehog (Shh) plays essential roles in midline formation and development of many organs such as central nerve system, limb and secondary palate. During palate development, expression of Shh is in the epithelia of tongue, pharynx and rugae. MFCS4 is one of the cis-regulatory sequences for Shh and it is exclusively activated in the epithelium of pharynx from E10.5 to E15.5. Although heterozygous deletion mutants of Shh (Shh+/−) or MFCS4 (MFCS4+/−) do not show cleft palate, compound heterozygotes that yield both Shh+/− and MFCS4+/− (Shh+/− MFCS4−/+) exhibit cleft palate with full penetrance. By analyzing this compound heterozygote, we aimed to clarify the function of Shh in palate development. Arrested development of palatal shelves of Shh+/− MFCS4−/+ in reorientation phase was found by histological observation. The failure of palatal shelves reorientation was rescued by rotary organ culture of Shh+/− MFCS4−/+ maxillae, suggesting the development of the tongue is disrupted. In situ hybridization for myogenic differentiation 1 (MyoD1) showed that the development of intrinsic and extrinsic lingual muscles was hypoplastic and orientation of genioglossus muscle was abnormal. These alterations of the tongue muscle development corresponded to expanded expression of stromal cell-derived factor 1 (Sdf-1) that recruits CXC chemokine receptor 4 (Cxcr4)-positive migrating myoblasts. These findings suggest that because myoblasts fail in proliferation, condensation and subsequent differentiation due to expanded and disrupted signaling between Sdf-1 and corresponding receptor Cxcr4, movement of lingual muscles is insufficient, which blocks palatal shelf reorientation. In human MFCS4, we have identified single nucleotide polymorphisms that are specific for the cleft palate population, further suggesting the involvement of Shh regulated by MFCS4 in palate development.
- Published
- 2010
35. P113. Effects of embryonic hypoxia on maxillofacial development
- Author
-
Shigeru Okuhara, Sachiko Iseki, Ryosuke Nagaoka, and Teruo Amagasa
- Subjects
Cancer Research ,Angiogenesis ,Morphogenesis ,Wnt signaling pathway ,Embryo ,Cell Biology ,Anatomy ,Biology ,Hypoxia (medical) ,Embryonic stem cell ,Cell biology ,Vascular endothelial growth factor ,stomatognathic diseases ,chemistry.chemical_compound ,chemistry ,medicine ,medicine.symptom ,Molecular Biology ,Transcription factor ,Developmental Biology - Abstract
It has been well known that many genetic and environmental factors are involved in maxillofacial development. Disturbance in these factors could result in congenital anomalies such as cleft lip and palate in human. Some environmental factors, administration of vasoconstrictors and uterine anemia during pregnancy, induce embryonic anemia, and it has been suggested that embryonic hypoxia inhibits development of circulatory organs, limb and maxillofacial organs. In this study we studied the effect of hypoxic condition on mouse lip formation. Lip forms by the fusion between facial processes, medial nasal, lateral nasal, and maxillary processes. We cultured mouse embryos at the beginning of facial process fusion stage by whole embryo culture system under standard oxygen supply and hypoxic conditions, and found that hypoxia induced the failure in the fusion, cleft lip, at higher frequency than standard oxygen supply. Then, we further investigated the effects of embryonic hypoxia on expression change of the genes that are associated with angiogenesis as a consequence of hypoxia and maxillofacial morphogenesis in the facial processes. Expression levels of vascular endothelial growth factor (VEGF) that is upregulated in response to hypoxic condition in vivo were significantly increased in the embryos cultured under hypoxic condition (hypoxic embryos). Transcription factors, Msx1 and Msx2, are expressed in the facial mesenchyme and required for proliferation and differentiation. These transcription factors were downregulated in the facial mesenchyme of hypoxic embryos. Active Wnt signaling has been shown to be involved in lip formation, and the amount of phosphor-Gsk3 alpha/beta, active Wnt signaling marker, were decreased in hypoxic embryos. These results suggest that embryonic hypoxia during the period of facial process fusion induces similar molecular response to adult hypoxic condition and affects expression levels of genes that are strongly associated with lip formation.
- Published
- 2010
36. Cranial anomaly of homozygous rSey rat is associated with a defect in the migration pathway of midbrain crest cells
- Author
-
Kazuhiro Eto, Hirofumi Doi, Youichirou Ninomiya, Shigeru Kuratani, Michio Fujiwara, Noriko Osumi-Yamashita, Kaori Aoki, Suconta Chareonvit, Sachiko Iseki, and Tadashi Watanabe
- Subjects
animal structures ,PAX6 Transcription Factor ,Ectomesenchyme ,Ectoderm ,Biology ,Rats, Mutant Strains ,Craniofacial Abnormalities ,Rats, Sprague-Dawley ,Cell Movement ,Mesencephalon ,Pregnancy ,otorhinolaryngologic diseases ,Nasal septum ,medicine ,Animals ,Paired Box Transcription Factors ,Craniofacial ,Eye Proteins ,In Situ Hybridization ,Homeodomain Proteins ,Homozygote ,Skull ,Gene Expression Regulation, Developmental ,Neural crest ,Medial nasal prominence ,Cell Biology ,Anatomy ,Rats ,DNA-Binding Proteins ,Repressor Proteins ,medicine.anatomical_structure ,Neural Crest ,embryonic structures ,Female ,Crest ,Developmental Biology ,Lateral nasal prominence - Abstract
Craniofacial development of vertebrates depends largely on neural crest contribution and each subdomain of the crest-derived ectomesenchyme follows its specific genetic control. The rat small eye (rSey) involves a mutation in the Pax-6 gene and the external feature of rSey homozygous embryos exhibits craniofacial defects in ocular and frontonasal regions. In order to identify the mechanism of craniofacial development, we examined the cranial morphology and migration of cephalic crest cells in rSey embryos. The chondrocranial defects of homozygous rSey embryos primarily consisted of spheno-orbital and ethmoidal anomalies. The former defects appeared to be brought about by the lack of the eye. In the ethmoid region, the nasal septum and the derivative of the medial nasal prominence were present, while the rest of the nasal capsule, as well as the nasal and lachrymal bones, were totally absent except for a pair of cartilaginous rods in place of the nasal capsule. This suggests that the primary cranial defect is restricted to the lateral nasal prominence derivatives. Dil labeling revealed the abnormal migration of crest cells specifically from the anterior midbrain to the lateral nasal prominence in homozygous rSey embryos. Pax-6 was not expressed in the crest cells but was strongly expressed in the frontonasal ectoderm. To determine whether or not this migratory defect actually resides in environmental cues, normal midbrain crest cells from wild-type embryos were labeled with Dil and were orthotopically injected into host rSey embryos. Migration of the donor crest cells into the lateral nasal prominence was abnormal in homozygous host embryos, while they migrated normally in wild-type or heterozygous embryos. Therefore, the cranial defects in rSey homozygous embryos are due to inappropriate substrate for crest cell migration towards the lateral nasal prominence, which consistently explains the cranial morphology of homozygous rSey embryos.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.