Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Development ; 145(4)2018 02 21.
Article in English | MEDLINE | ID: mdl-29437780

ABSTRACT

Tooth agenesis is one of the predominant developmental anomalies in humans, usually affecting the permanent dentition generated by sequential tooth formation and, in most cases, caused by mutations perturbing epithelial Wnt/ß-catenin signaling. In addition, loss-of-function mutations in the Wnt feedback inhibitor AXIN2 lead to human tooth agenesis. We have investigated the functions of Wnt/ß-catenin signaling during sequential formation of molar teeth using mouse models. Continuous initiation of new teeth, which is observed after genetic activation of Wnt/ß-catenin signaling in the oral epithelium, was accompanied by enhanced expression of Wnt antagonists and a downregulation of Wnt/ß-catenin signaling in the dental mesenchyme. Genetic and pharmacological activation of mesenchymal Wnt/ß-catenin signaling negatively regulated sequential tooth formation, an effect partly mediated by Bmp4. Runx2, a gene whose loss-of-function mutations result in sequential formation of supernumerary teeth in the human cleidocranial dysplasia syndrome, suppressed the expression of Wnt inhibitors Axin2 and Drapc1 in dental mesenchyme. Our data indicate that increased mesenchymal Wnt signaling inhibits the sequential formation of teeth, and suggest that Axin2/Runx2 antagonistic interactions modulate the level of mesenchymal Wnt/ß-catenin signaling, underlying the contrasting dental phenotypes caused by human AXIN2 and RUNX2 mutations.


Subject(s)
Odontogenesis/genetics , Tooth/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Axin Protein/metabolism , Fluorescent Antibody Technique , In Situ Hybridization , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Wnt Signaling Pathway
2.
Calcif Tissue Int ; 109(2): 121-131, 2021 08.
Article in English | MEDLINE | ID: mdl-33743023

ABSTRACT

Treatment with intravenous bisphosphonate (BP) in children and adolescents with osteogenesis imperfecta (OI) started in Sweden in 1991. No human studies on the role of BP therapy in development of disturbances in tooth mineralization or tooth morphology have been published. The study cohort comprised 219 individuals who were divided into four groups: group 1, BP treatment onset before 2 years of age (n = 22); group 2, BP treatment onset between 2 and 6 years of age (n = 20); group 3, BP treatment onset between 6 and 10 years of age (n = 13); and a control group of patients with OI who had not received BP therapy (n = 164). The chi-square test was used in between-group comparisons of the prevalence of tooth agenesis. The prevalence of tooth agenesis was significantly higher in children who began BP treatment before the age of 2 years (group 1; 59%,) compared to the controls (10%; p < 0.001) and to children who had begun BP therapy between ages 2 and 6 years (group 2; 10%; p = 0.009) or between ages 6 and 10 years (group 3; 8%; p = 0.003). Different types of disturbances in the enamel formation were seen in 52 premolars, where 51 were seen in those who began BP treatment before the age of 2 years. To conclude, starting BP treatment before the age of 2 years increases the risk of abnormalities in tooth formation manifesting as morphological aberrations, tooth agenesis, and enamel defects.


Subject(s)
Osteogenesis Imperfecta , Tooth , Adolescent , Adult , Child , Child, Preschool , Diphosphonates/therapeutic use , Humans , Odontogenesis , Osteogenesis Imperfecta/complications , Osteogenesis Imperfecta/drug therapy , Sweden/epidemiology , Young Adult
3.
Stem Cells ; 37(9): 1238-1248, 2019 09.
Article in English | MEDLINE | ID: mdl-31145830

ABSTRACT

Continuous growth of the mouse incisor teeth is due to the life-long maintenance of epithelial stem cells (SCs) in their niche called cervical loop (CL). Several signaling factors regulate SC maintenance and/or their differentiation to achieve organ homeostasis. Previous studies indicated that Hedgehog signaling is crucial for both the maintenance of the SCs in the niche, as well as for their differentiation. How Hedgehog signaling regulates these two opposing cellular behaviors within the confinement of the CL remains elusive. In this study, we used in vitro organ and cell cultures to pharmacologically attenuate Hedgehog signaling. We analyzed expression of various genes expressed in the SC niche to determine the effect of altered Hedgehog signaling on the cellular hierarchy within the niche. These genes include markers of SCs (Sox2 and Lgr5) and transit-amplifying cells (P-cadherin, Sonic Hedgehog, and Yap). Our results show that Hedgehog signaling is a critical survival factor for SCs in the niche, and that the architecture and the diversity of the SC niche are regulated by multiple Hedgehog ligands. We demonstrated the presence of an additional Hedgehog ligand, nerve-derived Desert Hedgehog, secreted in the proximity of the CL. In addition, we provide evidence that Hedgehog receptors Ptch1 and Ptch2 elicit independent responses, which enable multimodal Hedgehog signaling to simultaneously regulate SC maintenance and differentiation. Our study indicates that the cellular hierarchy in the continuously growing incisor is a result of complex interplay of two Hedgehog ligands with functionally distinct Ptch receptors. Stem Cells 2019;37:1238-1248.


Subject(s)
Epithelial Cells/metabolism , Hedgehog Proteins/metabolism , Patched-1 Receptor/metabolism , Patched-2 Receptor/metabolism , Stem Cell Niche , Stem Cells/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Epithelial Cells/cytology , Hedgehog Proteins/genetics , Incisor/cytology , Mice, Knockout , Mice, Transgenic , Models, Biological , Patched-1 Receptor/genetics , Patched-2 Receptor/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction/genetics , Stem Cells/cytology
4.
Nature ; 513(7519): 551-4, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25079316

ABSTRACT

Mesenchymal stem cells occupy niches in stromal tissues where they provide sources of cells for specialized mesenchymal derivatives during growth and repair. The origins of mesenchymal stem cells have been the subject of considerable discussion, and current consensus holds that perivascular cells form mesenchymal stem cells in most tissues. The continuously growing mouse incisor tooth offers an excellent model to address the origin of mesenchymal stem cells. These stem cells dwell in a niche at the tooth apex where they produce a variety of differentiated derivatives. Cells constituting the tooth are mostly derived from two embryonic sources: neural crest ectomesenchyme and ectodermal epithelium. It has been thought for decades that the dental mesenchymal stem cells giving rise to pulp cells and odontoblasts derive from neural crest cells after their migration in the early head and formation of ectomesenchymal tissue. Here we show that a significant population of mesenchymal stem cells during development, self-renewal and repair of a tooth are derived from peripheral nerve-associated glia. Glial cells generate multipotent mesenchymal stem cells that produce pulp cells and odontoblasts. By combining a clonal colour-coding technique with tracing of peripheral glia, we provide new insights into the dynamics of tooth organogenesis and growth.


Subject(s)
Cell Differentiation , Cell Lineage , Incisor/cytology , Mesenchymal Stem Cells/cytology , Neuroglia/cytology , Animals , Cell Tracking , Clone Cells/cytology , Dental Pulp/cytology , Female , Incisor/embryology , Male , Mice , Models, Biological , Neural Crest/cytology , Odontoblasts/cytology , Regeneration , Schwann Cells/cytology
5.
Development ; 142(22): 3954-63, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26450968

ABSTRACT

Epithelial morphogenesis generates the shape of the tooth crown. This is driven by patterned differentiation of cells into enamel knots, root-forming cervical loops and enamel-forming ameloblasts. Enamel knots are signaling centers that define the positions of cusp tips in a tooth by instructing the adjacent epithelium to fold and proliferate. Here, we show that the forkhead-box transcription factor Foxi3 inhibits formation of enamel knots and cervical loops and thus the differentiation of dental epithelium in mice. Conditional deletion of Foxi3 (Foxi3 cKO) led to fusion of molars with abnormally patterned shallow cusps. Foxi3 was expressed in the epithelium, and its expression was reduced in the enamel knots and cervical loops and in ameloblasts. Bmp4, a known inducer of enamel knots and dental epithelial differentiation, downregulated Foxi3 in wild-type teeth. Using genome-wide gene expression profiling, we showed that in Foxi3 cKO there was an early upregulation of differentiation markers, such as p21, Fgf15 and Sfrp5. Different signaling pathway components that are normally restricted to the enamel knots were expanded in the epithelium, and Sostdc1, a marker of the intercuspal epithelium, was missing. These findings indicated that the activator-inhibitor balance regulating cusp patterning was disrupted in Foxi3 cKO. In addition, early molar bud morphogenesis and, in particular, formation of the suprabasal epithelial cell layer were impaired. We identified keratin 10 as a marker of suprabasal epithelial cells in teeth. Our results suggest that Foxi3 maintains dental epithelial cells in an undifferentiated state and thereby regulates multiple stages of tooth morphogenesis.


Subject(s)
Cell Differentiation/physiology , Epithelium/physiology , Forkhead Transcription Factors/metabolism , Molar/embryology , Morphogenesis/physiology , Signal Transduction/physiology , Tooth Crown/embryology , Animals , Bone Morphogenetic Protein 4/metabolism , Epithelium/metabolism , Fluorescent Antibody Technique , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Protein Array Analysis , Reverse Transcriptase Polymerase Chain Reaction
6.
Eur J Oral Sci ; 126 Suppl 1: 67-71, 2018 10.
Article in English | MEDLINE | ID: mdl-30178557

ABSTRACT

Remarkable breakthroughs in the fields of developmental biology and stem cell biology during the last 15 yr have led to a new level of understanding regarding how teeth develop and how stem cells can be programmed. As a result, the possibilities of growing new teeth and of tooth bioengineering have been explored. Currently, a great deal is known about how signaling molecules and genes regulate tooth development, and modern research using transgenic mouse models has demonstrated that it is possible to induce the formation of new teeth by tinkering with the signaling networks that govern early tooth development. A breakthrough in stem cell biology in 2006 opened up the possibility that a patient's own cells can be programmed to develop into pluripotent stem cells and used for building new tissues and organs. At present, active research in numerous laboratories around the world addresses the question of how to program the stem and progenitor cells to develop into tooth-specific cell types. Taken together, the remarkable progress in developmental and stem cell biology is now feeding hopes of growing new teeth in the dental clinic in the not-too-distant future.


Subject(s)
Bioengineering , Tooth, Artificial , Tooth/growth & development , Bioengineering/methods , Humans , Regeneration , Stem Cells/physiology , Tooth/physiology
7.
Development ; 140(7): 1424-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23462476

ABSTRACT

Tooth renewal is initiated from epithelium associated with existing teeth. The development of new teeth requires dental epithelial cells that have competence for tooth formation, but specific marker genes for these cells have not been identified. Here, we analyzed expression patterns of the transcription factor Sox2 in two different modes of successional tooth formation: tooth replacement and serial addition of primary teeth. We observed specific Sox2 expression in the dental lamina that gives rise to successional teeth in mammals with one round of tooth replacement as well as in reptiles with continuous tooth replacement. Sox2 was also expressed in the dental lamina during serial addition of mammalian molars, and genetic lineage tracing indicated that Sox2(+) cells of the first molar give rise to the epithelial cell lineages of the second and third molars. Moreover, conditional deletion of Sox2 resulted in hyperplastic epithelium in the forming posterior molars. Our results indicate that the Sox2(+) dental epithelium has competence for successional tooth formation and that Sox2 regulates the progenitor state of dental epithelial cells. The findings imply that the function of Sox2 has been conserved during evolution and that tooth replacement and serial addition of primary teeth represent variations of the same developmental process. The expression patterns of Sox2 support the hypothesis that dormant capacity for continuous tooth renewal exists in mammals.


Subject(s)
Biomarkers , Epithelial Cells/metabolism , Mammals , Reptiles , SOXB1 Transcription Factors/physiology , Tooth/growth & development , Animals , Biomarkers/metabolism , Cells, Cultured , Embryo, Mammalian , Female , Ferrets , Humans , Mammals/embryology , Mammals/genetics , Mammals/growth & development , Mice , Mice, Transgenic , Models, Biological , Pregnancy , Regeneration/genetics , Regeneration/physiology , Reptiles/genetics , Reptiles/growth & development , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tooth/embryology , Tooth/metabolism , Tooth/physiology
8.
Stem Cells ; 33(5): 1670-81, 2015 May.
Article in English | MEDLINE | ID: mdl-25693510

ABSTRACT

Continuous growth of rodent incisors relies on epithelial stem cells (SCs) located in the SC niche called labial cervical loop (LaCL). Here, we found a population of apoptotic cells residing in a specific location of the LaCL in mouse incisor. Activated Caspase 3 and Caspase 9, expressed in this location colocalized in part with Lgr5 in putative SCs. The addition of Caspase inhibitors to incisors ex vivo resulted in concentration dependent thickening of LaCL. To examine the role of Wnt signaling in regulation of apoptosis, we exposed the LaCL of postnatal day 2 (P2) mouse incisor ex vivo to BIO, a known activator of Wnt/ß-catenin signaling. This resulted in marked thinning of LaCL as well as enhanced apoptosis. We found that Wnt/ß-catenin signaling was intensely induced by BIO in the mesenchyme surrounding the LaCL, but, unexpectedly, no ß-catenin activity was detected in the LaCL epithelium either before or after BIO treatment. We discovered that the expression of Fgf10, an essential growth factor for incisor epithelial SCs, was dramatically downregulated in the mesenchyme around BIO-treated LaCL, and that exogenous Fgf10 could rescue the thinning of the LaCL caused by BIO. We conclude that the homeostasis of the epithelial SC population in the mouse incisor depends on a proper rate of apoptosis and that this apoptosis is controlled by signals from the mesenchyme surrounding the LaCL. Fgf10 is a key mesenchymal signal limiting apoptosis of incisor epithelial SCs and its expression is negatively regulated by Wnt/ß-catenin. Stem Cells 2015;33:1670-1681.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/cytology , Fibroblast Growth Factor 10/pharmacology , Homeostasis/drug effects , Mesoderm/metabolism , Stem Cells/metabolism , Tooth/cytology , Wnt Signaling Pathway/drug effects , Animals , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Incisor/cytology , Mesoderm/drug effects , Mice , Models, Biological , Receptors, G-Protein-Coupled/metabolism , Stem Cell Niche/drug effects , Stem Cells/cytology , Stem Cells/drug effects
9.
Development ; 139(19): 3487-97, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22949612

ABSTRACT

Teeth are found in almost all vertebrates, and they therefore provide a general paradigm for the study of epithelial organ development and evolution. Here, we review the developmental mechanisms underlying changes in tooth complexity and tooth renewal during evolution, focusing on recent studies of fish, reptiles and mammals. Mammals differ from other living vertebrates in that they have the most complex teeth with restricted capacity for tooth renewal. As we discuss, however, limited tooth replacement in mammals has been compensated for in some taxa by the evolution of continuously growing teeth, the development of which appears to reuse the regulatory pathways of tooth replacement.


Subject(s)
Biological Evolution , Odontogenesis/physiology , Regeneration/physiology , Tooth/embryology , Tooth/physiology , Animals , Humans , Models, Biological , Odontogenesis/genetics , Organ Size/genetics , Organogenesis/genetics , Organogenesis/physiology , Regeneration/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Tooth/growth & development , Tooth/metabolism
10.
Development ; 139(17): 3189-99, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22833125

ABSTRACT

Uncovering the origin and nature of phenotypic variation within species is the first step in understanding variation between species. Mouse models with altered activities of crucial signal pathways have highlighted many important genes and signal networks regulating the morphogenesis of complex structures, such as teeth. The detailed analyses of these models have indicated that the balanced actions of a few pathways regulating cell behavior modulate the shape and number of teeth. Currently, however, most mouse models studied have had gross alteration of morphology, whereas analyses of more subtle modification of morphology are required to link developmental studies to evolutionary change. Here, we have analyzed a signaling network involving ectodysplasin (Eda) and fibroblast growth factor 20 (Fgf20) that subtly affects tooth morphogenesis. We found that Fgf20 is a major downstream effector of Eda and affects Eda-regulated characteristics of tooth morphogenesis, including the number, size and shape of teeth. Fgf20 function is compensated for by other Fgfs, in particular Fgf9 and Fgf4, and is part of an Fgf signaling loop between epithelium and mesenchyme. We showed that removal of Fgf20 in an Eda gain-of-function mouse model results in an Eda loss-of-function phenotype in terms of reduced tooth complexity and third molar appearance. However, the extra anterior molar, a structure lost during rodent evolution 50 million years ago, was stabilized in these mice.


Subject(s)
Ectodysplasins/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Morphogenesis/physiology , Signal Transduction/physiology , Tooth/embryology , Animals , Biological Evolution , Galactosides , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization , Indoles , Luciferases , Mice , Microscopy, Confocal , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL