Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7(1): 12063, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28935865

ABSTRACT

Ectothermal reptiles have internal pigmentation, which is not seen in endothermal birds and mammals. Here we show that the development of the dorsal neural tube-derived melanoblasts in turtle Trachemys scripta is regulated by similar mechanisms as in other amniotes, but significantly later in development, during the second phase of turtle trunk neural crest emigration. The development of melanoblasts coincided with a morphological change in the dorsal neural tube between stages mature G15 and G16. The melanoblasts delaminated and gathered in the carapacial staging area above the neural tube at G16, and differentiated into pigment-forming melanocytes during in vitro culture. The Mitf-positive melanoblasts were not restricted to the dorsolateral pathway as in birds and mammals but were also present medially through the somites similarly to ectothermal anamniotes. This matched a lack of environmental barrier dorsal and lateral to neural tube and the somites that is normally formed by PNA-binding proteins that block entry to medial pathways. PNA-binding proteins may also participate in the patterning of the carapacial pigmentation as both the migratory neural crest cells and pigment localized only to PNA-free areas.


Subject(s)
Melanocytes/metabolism , Neural Crest/metabolism , Neural Tube/metabolism , Turtles/metabolism , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Cells, Cultured , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Melanocytes/cytology , Neural Crest/cytology , Neural Crest/embryology , Neural Tube/cytology , Neural Tube/embryology , SOXE Transcription Factors/genetics , Turtles/embryology , Turtles/genetics
2.
Int J Cardiol ; 187: 519-24, 2015.
Article in English | MEDLINE | ID: mdl-25846664

ABSTRACT

INTRODUCTION: India carries the greatest burden of global non-communicable diseases (NCDs). Poverty is strongly associated with NCDs but there are few prevalence studies which have measured poverty in India, particularly in rural settings. METHODS: In Kerala, India, a population of 113,462 individuals was identified. The "Epidemiology of Non-communicable Diseases in Rural Areas" (ENDIRA) study was conducted via ASHAs (Accredited Social Health Activists). Standardised questionnaires were used in household interviews of individuals ≥18years during 2012 to gather sociodemographic, lifestyle and medical data for this population. The Government of Kerala definition of "the poverty line" was used. The association between below poverty line (BPL) status, NCDs and risk factors was analysed in multivariable regression models. RESULTS: 84,456 adults were included in the analyses (25.4% below the poverty line). The prevalence of NCDs was relatively common: myocardial infarction (MI) 1.4%, stroke 0.3%, respiratory diseases 5.0%, and cancer 1.1%. BPL status was not associated with age (p=0.96) or gender (p=0.26). Compared with those above the poverty line (APL), the BPL group was less likely to have diabetes, hypertension or dyslipidaemia (p<0.0001), and more likely to smoke (p<0.0001). Compared with APL, BPL was associated with stroke (OR 1.33, 1.04-1.69; p=0.02) and respiratory disease (OR 1.23, 1.15-1.32; p<0.0001) in multivariable analyses, but not MI or cancer. CONCLUSIONS: In rural Kerala, BPL status was associated with stroke and respiratory diseases, but not with MI and cancer although it was associated with smoking status, compared with above poverty line status.


Subject(s)
Cardiovascular Diseases/epidemiology , Diabetes Mellitus/epidemiology , Hypertension/epidemiology , Poverty/statistics & numerical data , Smoking/epidemiology , Stroke/epidemiology , Adult , Aged , Aged, 80 and over , Female , Health Services Accessibility/economics , Health Services Accessibility/statistics & numerical data , Humans , India/epidemiology , Male , Middle Aged , Regression Analysis , Risk Factors , Rural Population/statistics & numerical data , Socioeconomic Factors , Surveys and Questionnaires
3.
J Biomed Mater Res B Appl Biomater ; 102(6): 1181-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24425148

ABSTRACT

Biodegradable materials have been used as wound closure materials. It is important for these materials to enhance wound healing when the wound is vulnerable, and maintain wound closure until the wound is heal. This article studies the degradation process of bioresorbable magnesium micro-clips for wound closure in voice/laryngeal microsurgery. A novel computational approach is proposed to model degradation of the biodegradable micro-clips. The degradation process that considers both material and geometry of the device as well as its deployment is modeled as an energy minimization problem that is iteratively solved using active contour and incremental finite element methods. Strain energy of the micro-clip during degradation is calculated with the stretching and bending functions in the active contour formulation. The degradation rate is computed from strain energy using a transformation formulation. By relating strain energy to material degradation, the degradation rates and geometries of the micro-clip during degradation can be represented using a simulated degradation map. Computer simulation of the degradation of the micro-clip presented in the study is validated by in vivo and in vitro experiments.


Subject(s)
Absorbable Implants , Magnesium , Materials Testing , Models, Biological , Sutures , Wound Healing , Animals , Swine
4.
Front Physiol ; 3: 121, 2012.
Article in English | MEDLINE | ID: mdl-22563320

ABSTRACT

Greig cephalopolysyndactyly syndrome (GCPS) is an autosomal dominant disorder with polydactyly and syndactyly of the limbs and a broad spectrum of craniofacial abnormalities. Craniosynostosis of the metopic suture (interfrontal suture in mice) is an important but rare feature associated with GCPS. GCPS is caused by mutations in the transcription factor GLI3, which regulates Hedgehog signaling. The Gli3 loss-of-function (Gli3(Xt-J/Xt-J)) mouse largely phenocopies the human syndrome with the mice exhibiting polydactyly and several craniofacial abnormalities. Here we show that Gli3(Xt-J/Xt-J) mice exhibit ectopic ossification in the interfrontal suture and in the most severe cases the suture fuses already prior to birth. We show that abnormalities in frontal bones occur early in calvarial development, before the establishment of the interfrontal suture. It provides a model for the metopic suture pathology that can occur in GCPS.

5.
J Biol Chem ; 287(25): 21429-38, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22547067

ABSTRACT

Mutations in the gene encoding the zinc finger transcription factor GLI3 (GLI-Kruppel family member 3) have been identified in patients with Grieg cephalopolysyndactyly syndrome in which premature fusion of calvarial suture (craniosynostosis) is an infrequent but important feature. Here, we show that Gli3 acts as a repressor in the developing murine calvaria and that Dlx5, Runx2 type II isoform (Runx2-II), and Bmp2 are expressed ectopically in the calvarial mesenchyme, which results in aberrant osteoblastic differentiation in Gli3-deficient mouse (Gli3(Xt-J/Xt-J)) and resulted in craniosynostosis. At the same time, enhanced activation of phospho-Smad1/5/8 (pSmad1/5/8), which is a downstream mediator of canonical Bmp signaling, was observed in Gli3(Xt-J/Xt-J) embryonic calvaria. Therefore, we generated Gli3;Runx2 compound mutant mice to study the effects of decreasing Runx2 dosage in a Gli3(Xt-J/Xt-J) background. Gli3(Xt-J/Xt-J) Runx2(+/-) mice have neither craniosynostosis nor additional ossification centers in interfrontal suture and displayed a normalization of Dlx5, Runx2-II, and pSmad1/5/8 expression as well as sutural mesenchymal cell proliferation. These findings suggest a novel role for Gli3 in regulating calvarial suture development by controlling canonical Bmp-Smad signaling, which integrates a Dlx5/Runx2-II cascade. We propose that targeting Runx2 might provide an attractive way of preventing craniosynostosis in patients.


Subject(s)
Acrocephalosyndactylia/embryology , Core Binding Factor Alpha 1 Subunit/metabolism , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Skull/embryology , Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/prevention & control , Animals , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Matrix Metalloproteinases, Secreted/genetics , Matrix Metalloproteinases, Secreted/metabolism , Mesoderm/embryology , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Osteoblasts/metabolism , Smad Proteins , Zinc Finger Protein Gli3
6.
Stem Cells ; 29(11): 1792-803, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21898689

ABSTRACT

Rodent incisors grow continuously throughout life, and epithelial progenitor cells are supplied from stem cells in the cervical loop. We report that epithelial Runx genes are involved in the maintenance of epithelial stem cells and their subsequent continuous differentiation and therefore growth of the incisors. Core binding factor ß (Cbfb) acts as a binding partner for all Runx proteins, and targeted inactivation of this molecule abrogates the activity of all Runx complexes. Mice deficient in epithelial Cbfb produce short incisors and display marked underdevelopment of the cervical loop and suppressed epithelial Fgf9 expression and mesenchymal Fgf3 and Fgf10 expression in the cervical loop. In culture, FGF9 protein rescues these phenotypes. These findings indicate that epithelial Runx functions to maintain epithelial stem cells and that Fgf9 may be a target gene of Runx signaling. Cbfb mutants also lack enamel formation and display downregulated Shh mRNA expression in cells differentiating into ameloblasts. Furthermore, Fgf9 deficiency results in a proximal shift of the Shh expressing cell population and ectopic FGF9 protein suppresses Shh expression. These findings indicate that Shh as well as Fgf9 expression is maintained by Runx/Cbfb but that Fgf9 antagonizes Shh expression. The present results provide the first genetic evidence that Runx/Cbfb genes function in the maintenance of stem cells in developing incisors by activating Fgf signaling loops between the epithelium and mesenchyme. In addition, Runx genes also orchestrate continuous proliferation and differentiation by maintaining the expression of Fgf9 and Shh mRNA.


Subject(s)
Cell Differentiation/physiology , Core Binding Factor beta Subunit/metabolism , Incisor/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Core Binding Factor beta Subunit/genetics , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 3/genetics , Fibroblast Growth Factor 3/metabolism , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
7.
Hum Mol Genet ; 19(17): 3457-67, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20570969

ABSTRACT

Gli3 is a zinc-finger transcription factor whose activity is dependent on the level of hedgehog (Hh) ligand. Hh signaling has key roles during endochondral ossification; however, its role in intramembranous ossification is still unclear. In this study, we show that Gli3 performs a dual role in regulating both osteoprogenitor proliferation and osteoblast differentiation during intramembranous ossification. We discovered that Gli3Xt-J/Xt-J mice, which represent a Gli3-null allele, exhibit craniosynostosis of the lambdoid sutures and that this is accompanied by increased osteoprogenitor proliferation and differentiation. These cellular changes are preceded by ectopic expression of the Hh receptor Patched1 and reduced expression of the transcription factor Twist1 in the sutural mesenchyme. Twist1 is known to delay osteogenesis by binding to and inhibiting the transcription factor Runx2. We found that Runx2 expression in the lambdoid suture was altered in a pattern complimentary to that of Twist1. We therefore propose that loss of Gli3 results in a Twist1-, Runx2-dependent expansion of the sutural osteoprogenitor population as well as enhanced osteoblastic differentiation which results in a bony bridge forming between the parietal and interparietal bones. We show that FGF2 will induce Twist1, normalize osteoprogenitor proliferation and differentiation and rescue the lambdoid suture synostosis in Gli3Xt-J/Xt-J mice. Taken together, we define a novel role for Gli3 in osteoblast development; we describe the first mouse model of lambdoid suture craniosynostosis and show how craniosynostosis can be rescued in this model.


Subject(s)
Cell Differentiation , Cell Proliferation , Craniosynostoses/physiopathology , Kruppel-Like Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Osteogenesis , Skull/abnormalities , Stem Cells/cytology , Animals , Craniosynostoses/embryology , Craniosynostoses/genetics , Craniosynostoses/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , Humans , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Skull/cytology , Skull/embryology , Skull/metabolism , Stem Cells/metabolism , Zinc Finger Protein Gli3
8.
J Exp Zool B Mol Dev Evol ; 312B(4): 351-60, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19205045

ABSTRACT

Fibroblast growth factor receptor 2 (Fgfr2) has two splice variants IIIb and IIIc, which are unique in function and localization. Signalling through Fgfr2IIIb controls epithelial-mesenchymal interactions, which regulate morphogenesis during the development of several organs including the palate and tooth. In this study, we confirm that molar tooth development in Fgfr2IIIb(-/-) mice is arrested early in development and that the molar teeth of Fgf10(-/-) mice develop through all the normal stages of morphogenesis. We show that the molar phenotype of Fgfr2IIIb(-/-) mice is, in part, owing to reduced cell proliferation in both epithelial and mesenchymal compartments. We also show that the developing molar teeth of Fgf10(-/-) mice exhibit reduced cell proliferation. However, this reduction is not sufficient to arrest molar development. Recent evidence has indicated that Fgfr2IIIb/Fgf10 signalling is active in the calvaria in some pathological situations as heterozygous deletion of Fgfr2 exon IIIc in mice leads to ectopic expression of Fgfr2IIIb in the calvarial bones and causes craniosynostosis. Here, we investigate the mRNA expression of Fgfr2IIIb and Fgfr2IIIc as well as their ligands Fgf3, -7 and -10 in the developing murine tooth, palate and calvaria. We show that Fgf7 is expressed in the calvarial mesenchyme adjacent to the developing frontal bone and Fgf10 is expressed by osteoprogenitors in the developing frontal bone condensation. Taken together, we highlight the overlapping roles of Fgfr2IIIb/Fgf10 signalling in controlling epithelial-mesenchymal interactions during normal palate and tooth morphogenesis and how elevated signalling through Fgfr2IIIb/Fgf10 solely within the mesenchyme can result in abnormal calvarial morphogenesis.


Subject(s)
Morphogenesis , Receptor, Fibroblast Growth Factor, Type 2/physiology , Signal Transduction , Skull/growth & development , Tooth/growth & development , Animals , Cell Proliferation , Face , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/physiology , Mice , Mice, Transgenic
9.
Dev Biol ; 320(1): 39-48, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18572158

ABSTRACT

The Tbx1 gene is a transcriptional regulator involved in the DiGeorge syndrome, which affects normal facial and tooth development. Several clinical reports point to a common enamel defect in the teeth of patients with DiGeorge syndrome. Here, we have analyzed the expression, regulation, and function of Tbx1 during mouse molar development. Tbx1 expression is restricted to epithelial cells that give rise to the enamel producing ameloblasts and correlates with proliferative events. Tbx1 expression in epithelium requires mesenchyme-derived signals: dental mesenchyme induces expression of Tbx1 in recombined dental and non-dental epithelia. Bead implantation experiments show that FGF molecules are able to maintain epithelial Tbx1 expression during odontogenesis. Expression of Tbx1 in dental epithelium of FGF receptor 2b(-/-) mutant mice is downregulated, showing a genetic link between FGF signaling and Tbx1 in teeth. Forced expression of Tbx1 in dental explants activates amelogenin expression. These results indicate that Tbx1 expression in developing teeth is under control of FGF signaling and correlates with determination of the ameloblast lineage.


Subject(s)
Ameloblasts/cytology , Cell Lineage , Fibroblast Growth Factors/metabolism , Morphogenesis , Signal Transduction , T-Box Domain Proteins/metabolism , Tooth/embryology , Ameloblasts/drug effects , Amelogenin/genetics , Amelogenin/metabolism , Animals , Cell Lineage/drug effects , Cell Proliferation/drug effects , Electroporation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelium/drug effects , Epithelium/embryology , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/embryology , Mice , Mice, Inbred C57BL , Models, Biological , Morphogenesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction/drug effects , T-Box Domain Proteins/genetics , Tooth/cytology , Tooth/drug effects
10.
Dev Biol ; 311(2): 335-46, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17931618

ABSTRACT

In this study we have addressed the fundamental question of what cellular mechanisms control the growth of the calvarial bones and conversely, what is the fate of the sutural mesenchymal cells when calvarial bones approximate to form a suture. There is evidence that the size of the osteoprogenitor cell population determines the rate of calvarial bone growth. In calvarial cultures we reduced osteoprogenitor cell proliferation; however, we did not observe a reduction in the growth of parietal bone to the same degree. This discrepancy prompted us to study whether suture mesenchymal cells participate in the growth of the parietal bones. We found that mesenchymal cells adjacent to the osteogenic fronts of the parietal bones could differentiate towards the osteoblastic lineage and could become incorporated into the growing bone. Conversely, mid-suture mesenchymal cells did not become incorporated into the bone and remained undifferentiated. Thus mesenchymal cells have different fate depending on their position within the suture. In this study we show that continued proliferation of osteoprogenitors in the osteogenic fronts is the main mechanism for calvarial bone growth, but importantly, we show that suture mesenchyme cells can contribute to calvarial bone growth. These findings help us understand the mechanisms of intramembranous ossification in general, which occurs not only during cranial and facial bone development but also in the surface periosteum of most bones during modeling and remodeling.


Subject(s)
Bone Development/physiology , Cell Differentiation/physiology , Cranial Sutures , Mesoderm/cytology , Skull , Animals , Biomarkers/metabolism , Cell Lineage , Cell Proliferation , Cranial Sutures/cytology , Cranial Sutures/embryology , Cranial Sutures/growth & development , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Gene Expression Regulation, Developmental , Mice , Osteoblasts/cytology , Osteoblasts/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Skull/cytology , Skull/embryology , Skull/growth & development , Stem Cells/cytology , Stem Cells/physiology
11.
Biochem Biophys Res Commun ; 357(4): 1084-9, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17466949

ABSTRACT

The role of Hedgehog signaling in human basal cell hyperplasia formation and its progressing towards tumorigenesis was investigated. Hedgehog signaling members including PTCH1, GLI1, GLI2, and GLI3 were found co-localized with p63 expression in most hyperplastic basal cells, but rarely in normal basal cells, suggesting Hedgehog involvement in basal cell hyperplasia formation. Both CK-14 and CK-8 markers were found co-localized in the majority of hyperplastic basal cells, but relatively few in the normal basal cells, indicating a Hedgehog-promoted transitory differentiation. Furthermore, CK-14 and PTCH1 were found co-localized with CD44 in the hyerplastic basal cells, in a way similar to the CD44 co-localization with PTCH1 and GLI1 in the cancer cells. Together, the present study indicated Hedgehog involvement in forming basal cell hyperplasia and its progressing towards cancer, presumably by transforming the normal basal stem cells into the cancer stem cells where persistent Hedgehog activation might be mandatory for tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Hedgehog Proteins/metabolism , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Stem Cells/metabolism , Stem Cells/pathology , Cell Transformation, Neoplastic/pathology , Humans , Male , Prostate/metabolism , Prostate/pathology , Tumor Cells, Cultured
12.
Gene Expr Patterns ; 6(2): 206-12, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16168717

ABSTRACT

Hedgehog signalling regulates morphogenesis of many developing organs. Sonic hedgehog (Shh) signalling has been shown to regulate the growth and morphogenesis of the palatal shelves prior to their elevation and fusion. Here, we show that Shh expression is limited to a thickened palatal oral epithelium prior to palatal shelf elevation. After palatal shelf elevation above the tongue, Shh is expressed only in small areas of thickened palatal oral epithelium that corresponded to developing rugae. The receptor Ptc1 and a regulator of Hh signalling Hhip1 are expressed in the mesenchyme adjacent to the palatal oral epithelium so that the highest level of transcripts localize to the palatal mesenchyme surrounding the Shh-expressing thickened epithelium. Smoothened and transcriptional effectors Gli1-3, and Hh regulator Gas1 are expressed widely in the palatal mesenchyme. No differences were found in the expression patterns of Hh pathway members along the anterior-posterior axis of the developing palate.


Subject(s)
Palate/embryology , Palate/metabolism , Trans-Activators/genetics , Animals , Body Patterning/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , GPI-Linked Proteins , Gene Expression Regulation, Developmental , Hedgehog Proteins , In Situ Hybridization , Kruppel-Like Transcription Factors/genetics , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Zinc Finger Protein Gli3 , rab GTP-Binding Proteins/genetics
13.
Curr Mol Med ; 5(7): 699-722, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16305494

ABSTRACT

Advances in developmental biology combined with progress in human genetics are helping us decipher how the craniofacial region develops and how the consequences of misdirected development result in malformation. This review describes the molecular etiology of a number of craniofacial developmental anomalies. The more common craniofacial anomalies cleft lip and palate and craniosynostosis, as well as cleidocranial dysplasia, hemifacial microsomia, holoprosencephaly, enlarged parietal foramina, Treacher Collins syndrome and cherubism are discussed.


Subject(s)
Craniofacial Abnormalities/pathology , Morphogenesis , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/pathology , Animals , Cleft Palate/pathology , Face/abnormalities , Face/pathology , Holoprosencephaly/pathology , Humans , Radiography
14.
Dev Dyn ; 234(1): 28-35, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16028273

ABSTRACT

The development of both the tooth and palate requires coordinated bone morphogenetic protein (BMP) and fibroblast growth factor (FGF) signalling between epithelial and mesenchymal tissues. Here, we demonstrate that transcription factors Twist and Snail are downstream targets of FGF signalling, that Id1 and Msx2 are downstream targets of BMP signalling, and that Msx1 is regulated by both signalling pathways during tooth and palate development. We show that Twist and Snail expression in the mesenchyme is regulated by the overlying epithelium and that exogenous FGF4 in tooth and FGF2 in palate can mimic this regulation in isolated mesenchymal explants. Ids act in a dominant-negative manner to inhibit the function of other transcription factors such as Twist and Snail. FGF and BMP signalling can regulate development antagonistically, and we suggest that FGF-regulated Twist and Snail and BMP-regulated Id1 may mediate these antagonistic effects during both tooth and palate development.


Subject(s)
Molar/embryology , Palate/embryology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Proteins/physiology , DNA-Binding Proteins/metabolism , Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , MSX1 Transcription Factor/metabolism , Mesoderm/metabolism , Mice , RNA, Messenger/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics
15.
Dev Dyn ; 233(3): 847-52, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15906377

ABSTRACT

Calvarial bone and suture development is under complex regulation where bone morphogenetic protein (Bmp) and fibroblast growth factor (Fgf) signalling interact with Msx2/Twist and Noggin and regulate frontal bone primordia proliferation and suture fusion, respectively. We have shown previously that the winged helix transcription factor Foxc1, which is necessary for calvarial bone development, is required for the Bmp regulation of Msx2. We now show that FGF2 regulates the expression of Foxc1, indicating that Foxc1 integrates Bmp and Fgf signalling pathways. We also show that Foxc1 is not needed for the acquisition of osteogenic potential or for the differentiation of osteoblasts. The expression of Fgf receptors and Twist were normal in Foxc1-deficient calvarial mesenchyme, and ectopic FGF2 was able to induce the expression Osteopontin. Furthermore, we demonstrate that Foxc1 does not participate in the regulation of Noggin expression. Our findings indicate that Foxc1 integrates the Bmp and Fgf signalling pathways independently of Twist or Noggin. This signalling network is essential for the correct patterning and growth of calvarial bones.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Carrier Proteins/metabolism , Fibroblast Growth Factor 2/pharmacology , Nuclear Proteins/metabolism , Signal Transduction , Skull/embryology , Transcription Factors/metabolism , Animals , Cell Differentiation , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Humans , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis , Signal Transduction/drug effects , Skull/cytology , Skull/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Twist-Related Protein 1
16.
Eur J Hum Genet ; 13(2): 139-49, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15483646

ABSTRACT

Structural chromosome abnormalities have aided in gene identification for over three decades. Delineation of the deletion sizes and rearrangements allows for phenotype/genotype correlations and ultimately assists in gene identification. In this report, we have delineated the precise rearrangements in four subjects with deletions, duplications, and/or triplications of 1p36 and compared the regions of imbalance to two cases recently published. Fluorescence in situ hybridization (FISH) analysis revealed the size, order, and orientation of the duplicated/triplicated segments in each subject. We propose a premeiotic model for the formation of these complex rearrangements in the four newly ascertained subjects, whereby a deleted chromosome 1 undergoes a combination of multiple breakage-fusion-bridge (BFB) cycles and inversions to produce a chromosome arm with a complex rearrangement of deleted, duplicated and triplicated segments. In addition, comparing the six subjects' rearrangements revealed a region of overlap that when triplicated is associated with craniosynostosis and when deleted is associated with large, late-closing anterior fontanels. Within this region are the MMP23A and -B genes. We show MMP23 gene expression at the cranial sutures and we propose that haploinsufficiency results in large, late-closing anterior fontanels and overexpression results in craniosynostosis. These data emphasize the important role of cytogenetics in investigating and uncovering the etiologies of human genetic disease, particularly cytogenetic imbalances that reveal potentially dosage-sensitive genes.


Subject(s)
Chromosome Disorders/genetics , Chromosomes, Human, Pair 1/genetics , Cranial Sutures , Gene Duplication , Gene Expression Regulation/genetics , Sequence Deletion/genetics , Animals , Chromosome Breakage/genetics , Chromosome Disorders/pathology , Chromosome Disorders/physiopathology , Chromosome Inversion/genetics , Cranial Sutures/pathology , Cranial Sutures/physiopathology , Female , Gene Dosage , Humans , Male , Mice
17.
J Clin Invest ; 113(12): 1692-700, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15199404

ABSTRACT

Classical research has suggested that early palate formation develops via epithelial-mesenchymal interactions, and in this study we reveal which signals control this process. Using Fgf10-/-, FGF receptor 2b-/- (Fgfr2b-/-), and Sonic hedgehog (Shh) mutant mice, which all exhibit cleft palate, we show that Shh is a downstream target of Fgf10/Fgfr2b signaling. Our results demonstrate that mesenchymal Fgf10 regulates the epithelial expression of Shh, which in turn signals back to the mesenchyme. This was confirmed by demonstrating that cell proliferation is decreased not only in the palatal epithelium but also in the mesenchyme of Fgfr2b-/- mice. These results reveal a new role for Fgf signaling in mammalian palate development. We show that coordinated epithelial-mesenchymal interactions are essential during the initial stages of palate development and require an Fgf-Shh signaling network.


Subject(s)
Cleft Palate/metabolism , Epithelium/physiology , Fibroblast Growth Factors/metabolism , Mesoderm/physiology , Palate/embryology , Receptors, Fibroblast Growth Factor/metabolism , Animals , Cell Division/physiology , Cleft Palate/etiology , Culture Techniques , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/genetics , Hedgehog Proteins , In Situ Hybridization , In Situ Nick-End Labeling , Mice , Mice, Knockout , Palate/cytology , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Trans-Activators/genetics , Trans-Activators/metabolism
18.
Development ; 130(24): 6131-42, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14597577

ABSTRACT

The flat bones of the vertebrate skull vault develop from two migratory mesenchymal cell populations, the cranial neural crest and paraxial mesoderm. At the onset of skull vault development, these mesenchymal cells emigrate from their sites of origin to positions between the ectoderm and the developing cerebral hemispheres. There they combine, proliferate and differentiate along an osteogenic pathway. Anomalies in skull vault development are relatively common in humans. One such anomaly is familial calvarial foramina, persistent unossified areas within the skull vault. Mutations in MSX2 and TWIST are known to cause calvarial foramina in humans. Little is known of the cellular and developmental processes underlying this defect. Neither is it known whether MSX2 and TWIST function in the same or distinct pathways. We trace the origin of the calvarial foramen defect in Msx2 mutant mice to a group of skeletogenic mesenchyme cells that compose the frontal bone rudiment. We show that this cell population is reduced not because of apoptosis or deficient migration of neural crest-derived precursor cells, but because of defects in its differentiation and proliferation. We demonstrate, in addition, that heterozygous loss of Twist function causes a foramen in the skull vault similar to that caused by loss of Msx2 function. Both the quantity and proliferation of the frontal bone skeletogenic mesenchyme are reduced in Msx2-Twist double mutants compared with individual mutants. Thus Msx2 and Twist cooperate in the control of the differentiation and proliferation of skeletogenic mesenchyme. Molecular epistasis analysis suggests that Msx2 and Twist do not act in tandem to control osteoblast differentiation, but function at the same epistatic level.


Subject(s)
DNA-Binding Proteins/metabolism , Frontal Bone/embryology , Homeodomain Proteins/metabolism , Mesoderm/physiology , Neural Crest/cytology , Nuclear Proteins/metabolism , Transcription Factors , Animals , Apoptosis , Biomarkers , DNA-Binding Proteins/genetics , Frontal Bone/abnormalities , Frontal Bone/anatomy & histology , Frontal Bone/physiology , Gene Expression Regulation, Developmental , Gestational Age , Homeodomain Proteins/genetics , Humans , Mesoderm/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neural Crest/physiology , Nuclear Proteins/genetics , Twist-Related Protein 1
19.
Dev Biol ; 262(1): 75-87, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14512019

ABSTRACT

Calvarial bones form by direct ossification of mesenchyme. This requires condensation of mesenchymal cells which then proliferate and differentiate into osteoblasts. Congenital hydrocephalus (ch) mutant mice lack the forkhead/winged helix transcription factor Foxc1. In ch mutant mice, calvarial bones remain rudimentary at the sites of initial osteogenic condensations. In this study, we have localized the ossification defect in ch mutants to the calvarial mesenchyme, which lacks the expression of transcription factors Msx2 and Alx4. This lack of expression is associated with a reduction in the proliferation of osteoprogenitor cells. We have previously shown that BMP induces Msx2 in calvarial mesenchyme (Development 125, 1241-1251, 1998). Here, we show that BMP also induces Alx4 in this tissue. We also show that BMP-induced expression of Msx2 and Alx4 requires Foxc1. We therefore suggest that Foxc1 regulates BMP-mediated osteoprogenitor proliferation and that this regulation is required for the progression of osteogenesis beyond the initial condensations in calvarial bone development.


Subject(s)
Bone Development , Proteins/physiology , Skull/embryology , Transcription Factors/physiology , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/pharmacology , Cell Division , DNA-Binding Proteins/analysis , Forkhead Transcription Factors , Hydrocephalus/etiology , Mice , Phenotype , Proteins/analysis , Transcription Factors/analysis
20.
Eur J Orthod ; 25(2): 139-48, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12737212

ABSTRACT

The development and growth of the skull is a co-ordinated process involving many different tissues that interact with each other to form a complex end result. When normal development is disrupted, debilitating pathological conditions, such as craniosynostosis (premature calvarial suture fusion) and cleidocranial dysplasia (delayed suture closure), can result. It is known that mutations in the fibroblast growth factor receptors 1, 2, and 3(FGFR1, 2, and 3), as well as the transcription factors MSX2 and TWIST cause craniosynostosis, and that mutations in the transcription factor RUNX2 (CBFA1) cause cleidocranial dysplasia. However, relatively little is known about the development of the calvaria: where and when these genes are active during normal calvarial development, how these genes may interact in the developing calvaria, and the disturbances that may occur to cause these disorders. In this work an attempt has been made to address some of these questions from a basic biological perspective. The expression patterns of the above-mentioned genes in the developing mouse skull are detailed. The microdissection and in vitro culture techniques have begun the task of identifying Fgfrs, Msx2, and Twist interacting in intricate signalling pathways that if disrupted could lead to craniosynostosis.


Subject(s)
Cranial Sutures/embryology , Craniosynostoses/genetics , Neoplasm Proteins , Protein-Tyrosine Kinases , Skull/embryology , Animals , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit , Craniosynostoses/embryology , Culture Techniques , DNA-Binding Proteins/genetics , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Integrin-Binding Sialoprotein , Mesoderm/metabolism , Mice , Molecular Biology , Mutation/genetics , Nuclear Proteins/genetics , Osteoblasts/metabolism , Osteogenesis/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 1 , Receptor, Fibroblast Growth Factor, Type 2 , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/genetics , Sialoglycoproteins/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Twist-Related Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...