Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Cell Rep ; 42(6): 112643, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37318953

RESUMO

Morphogenesis of ectodermal organs, such as hair, tooth, and mammary gland, starts with the formation of local epithelial thickenings, or placodes, but it remains to be determined how distinct cell types and differentiation programs are established during ontogeny. Here, we use bulk and single-cell transcriptomics and pseudotime modeling to address these questions in developing hair follicles and epidermis and produce a comprehensive transcriptomic profile of cellular populations in the hair placode and interplacodal epithelium. We report previously unknown cell populations and marker genes, including early suprabasal and genuine interfollicular basal markers, and propose the identity of suprabasal progenitors. By uncovering four different hair placode cell populations organized in three spatially distinct areas, with fine gene expression gradients between them, we posit early biases in cell fate establishment. This work is accompanied by a readily accessible online tool to stimulate further research on skin appendages and their progenitors.


Assuntos
Folículo Piloso , Transcriptoma , Camundongos , Animais , Folículo Piloso/metabolismo , Transcriptoma/genética , Camundongos Transgênicos , Epiderme/metabolismo , Cabelo
2.
Calcif Tissue Int ; 109(2): 121-131, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33743023

RESUMO

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.


Assuntos
Osteogênese Imperfeita , Dente , Adolescente , Adulto , Criança , Pré-Escolar , Difosfonatos/uso terapêutico , Humanos , Odontogênese , Osteogênese Imperfeita/complicações , Osteogênese Imperfeita/tratamento farmacológico , Suécia/epidemiologia , Adulto Jovem
3.
iScience ; 23(7): 101329, 2020 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-32674056

RESUMO

The transcription factor Sox21 is expressed in the epithelium of developing teeth. The present study aimed to determine the role of Sox21 in tooth development. We found that disruption of Sox21 caused severe enamel hypoplasia, regional osteoporosis, and ectopic hair formation in the gingiva in Sox21 knockout incisors. Differentiation markers were lost in ameloblasts, which formed hair follicles expressing hair keratins. Molecular analysis and chromatin immunoprecipitation sequencing indicated that Sox21 regulated Anapc10, which recognizes substrates for ubiquitination-mediated degradation, and determined dental-epithelial versus hair follicle cell fate. Disruption of either Sox21 or Anapc10 induced Smad3 expression, accelerated TGF-ß1-induced promotion of epithelial-to-mesenchymal transition (EMT), and resulted in E-cadherin degradation via Skp2. We conclude that Sox21 disruption in the dental epithelium leads to the formation of a unique microenvironment promoting hair formation and that Sox21 controls dental epithelial differentiation and enamel formation by inhibiting EMT via Anapc10.

4.
Bone Rep ; 12: 100285, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32509933

RESUMO

Runt-related transcription factor 2 (Runx2) is an essential transcription factor for osteoblast differentiation, and is activated by mechanical stress to promote osteoblast function. Cleidocranial dysplasia (CCD) is caused by mutations of RUNX2, and CCD patients exhibit malocclusion and often need orthodontic treatment. However, treatment is difficult because of impaired tooth movement, the reason of which has not been clarified. We examined the amount of experimental tooth movement in Runx2+/- mice, the animal model of CCD, and investigated bone formation on the tension side of experimental tooth movement in vivo. Continuous stretch was conducted to bone marrow stromal cells (BMSCs) as an in vitro model of the tension side of tooth movement. Compared to wild-type littermates the Runx2+/- mice exhibited delayed experimental tooth movement, and osteoid formation and osteocalcin (OSC) mRNA expression were impaired in osteoblasts on the tension side of tooth movement. Runx2 heterozygous deficiency delayed stretch-induced increase of DNA content in BMSCs, and also delayed and reduced stretch-induced alkaline phosphatase (ALP) activity, OSC mRNA expression, and calcium content of BMSCs in osteogenic medium. Furthermore Runx2+/- mice exhibited delayed and suppressed expression of mammalian target of rapamycin (mTOR) and rapamycin-insensitive companion of mTOR (Rictor), essential factors of mTORC2, which is regulated by Runx2 to phosphorylate Akt to regulate cell proliferation and differentiation, in osteoblasts on the tension side of tooth movement in vivo and in vitro. Loss of half Runx2 gene dosage inhibited stretch-induced PI3K dependent mTORC2/Akt activity to promote BMSCs proliferation. Furthermore, Runx2+/- BMSCs in osteogenic medium exhibited delayed and suppressed stretch-induced expression of mTOR and Rictor. mTORC2 regulated stretch-elevated Runx2 and ALP mRNA expression in BMSCs in osteogenic medium. We conclude that Runx2+/- mice present a useful model of CCD patients for elucidation of the molecular mechanisms in bone remodeling during tooth movement, and that Runx2 plays a role in stretch-induced proliferation and osteogenesis in BMSCs via mTORC2 activation.

5.
Sci Rep ; 10(1): 4963, 2020 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-32188889

RESUMO

Enamel is secreted by ameloblasts derived from tooth epithelial stem cells (SCs). Humans cannot repair or regenerate enamel, due to early loss of tooth epithelial SCs. Contrarily in the mouse incisors, epithelial SCs are maintained throughout life and endlessly generate ameloblasts, and thus enamel. Here we isolated Sox2-GFP+ tooth epithelial SCs which generated highly cellular spheres following a novel in vitro strategy. This system enabled analysis of SC regulation by various signaling molecules, and supported the stimulatory and inhibitory roles of Shh and Bmp, respectively; providing better insight into the heterogeneity of the SCs. Further, we generated a novel mouse reporter, Enamelin-tdTomato for identification of ameloblasts in live tissues and cells, and used it to demonstrate presence of ameloblasts in the new 3D co-culture system of dental SCs. Collectively, our results provide means of generating 3D tooth epithelium from adult SCs which can be utilized toward future generation of enamel.


Assuntos
Ameloblastos/citologia , Diferenciação Celular , Células Epiteliais/citologia , Células-Tronco/citologia , Dente/citologia , Ameloblastos/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Células Epiteliais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Células-Tronco/metabolismo , Dente/metabolismo
6.
Front Immunol ; 11: 608223, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33552067

RESUMO

The continuously growing mouse incisor provides a fascinating model for studying stem cell regulation and organ renewal. In the incisor, epithelial and mesenchymal stem cells assure lifelong tooth growth. The epithelial stem cells reside in a niche known as the cervical loop. Mesenchymal stem cells are located in the nearby apical neurovascular bundle and in the neural plexus. So far, little is known about extracellular cues that are controlling incisor stem cell renewal and guidance. The extracellular matrix protein tenascin-W, also known as tenascin-N (TNN), is expressed in the mesenchyme of the pulp and of the periodontal ligament of the incisor, and is closely associated with collagen 3 fibers. Here, we report for the first time the phenotype of tenascin-W/TNN deficient mice, which in a C57BL/6N background exhibit a reduced body weight and lifespan. We found major defects in the alveolar bone and periodontal ligament of the growing rodent incisors, whereas molars were not affected. The alveolar bone around the incisor was replaced by a dense scar-like connective tissue, enriched with newly formed nerve fibers likely leading to periodontal pain, less food intake and reduced body weight. Using soft food to reduce mechanical load on the incisor partially rescued the phenotype. In situ hybridization and Gli1 reporter mouse experiments revealed decreased hedgehog signaling in the incisor mesenchymal stem cell compartment, which coordinates the development of mesenchymal stem cell niche. These results indicate that TNN deficiency in mice affects periodontal remodeling and increases nerve fiber branching. Through periodontal pain the food intake is reduced and the incisor renewal and the neurovascular sonic hedgehog secretion rate are reduced. In conclusion, tenascin-W/TNN seems to have a primary function in rapid periodontal tissue remodeling and a secondary function in mechanosensation.


Assuntos
Incisivo/metabolismo , Células-Tronco Mesenquimais/metabolismo , Doenças Periodontais/metabolismo , Ligamento Periodontal/metabolismo , Tenascina/metabolismo , Odontalgia/metabolismo , Animais , Colágeno Tipo III/metabolismo , Ingestão de Alimentos , Comportamento Alimentar , Predisposição Genética para Doença , Incisivo/crescimento & desenvolvimento , Incisivo/inervação , Mecanotransdução Celular , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças Periodontais/genética , Doenças Periodontais/fisiopatologia , Ligamento Periodontal/crescimento & desenvolvimento , Ligamento Periodontal/inervação , Fenótipo , Nicho de Células-Tronco , Tenascina/genética , Odontalgia/genética , Odontalgia/fisiopatologia , Proteína GLI1 em Dedos de Zinco/genética , Proteína GLI1 em Dedos de Zinco/metabolismo
7.
Stem Cells ; 37(9): 1238-1248, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31145830

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Proteínas Hedgehog/metabolismo , Receptor Patched-1/metabolismo , Receptor Patched-2/metabolismo , Nicho de Células-Tronco , Células-Tronco/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Células Cultivadas , Células Epiteliais/citologia , Proteínas Hedgehog/genética , Incisivo/citologia , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Receptor Patched-1/genética , Receptor Patched-2/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais/genética , Células-Tronco/citologia
8.
Eur J Oral Sci ; 126 Suppl 1: 67-71, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30178557

RESUMO

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.


Assuntos
Bioengenharia , Dente Artificial , Dente/crescimento & desenvolvimento , Bioengenharia/métodos , Humanos , Regeneração , Células-Tronco/fisiologia , Dente/fisiologia
9.
Development ; 145(4)2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29437780

RESUMO

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.


Assuntos
Odontogênese/genética , Dente/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Proteína Axina/metabolismo , Imunofluorescência , Hibridização In Situ , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Via de Sinalização Wnt
10.
J Cell Biol ; 214(6): 753-67, 2016 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-27621364

RESUMO

During organogenesis, cell fate specification and patterning are regulated by signaling centers, specialized clusters of morphogen-expressing cells. In many organs, initiation of development is marked by bud formation, but the cellular mechanisms involved are ill defined. Here, we use the mouse incisor tooth as a model to study budding morphogenesis. We show that a group of nonproliferative epithelial cells emerges in the early tooth primordium and identify these cells as a signaling center. Confocal live imaging of tissue explants revealed that although these cells reorganize dynamically, they do not reenter the cell cycle or contribute to the growing tooth bud. Instead, budding is driven by proliferation of the neighboring cells. We demonstrate that the activity of the ectodysplasin/Edar/nuclear factor κB pathway is restricted to the signaling center, and its inactivation leads to fewer quiescent cells and a smaller bud. These data functionally link the signaling center size to organ size and imply that the early signaling center is a prerequisite for budding morphogenesis.


Assuntos
Movimento Celular , Proliferação de Células , Células Epiteliais/fisiologia , Incisivo/embriologia , Animais , Ectodisplasinas/genética , Ectodisplasinas/metabolismo , Receptor Edar/genética , Receptor Edar/metabolismo , Células Epiteliais/metabolismo , Fase G1 , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Incisivo/metabolismo , Camundongos Transgênicos , Microscopia Confocal , Morfogênese , NF-kappa B/genética , NF-kappa B/metabolismo , Tamanho do Órgão , Fenótipo , Transdução de Sinais
11.
Curr Top Dev Biol ; 115: 157-86, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26589925

RESUMO

Reciprocal interactions between epithelial and mesenchymal tissues play a fundamental role in the morphogenesis of teeth and regulate all aspects of tooth development. Extensive studies on mouse tooth development over the past 25 years have uncovered the molecular details of the signaling networks mediating these interactions (reviewed by Jussila & Thesleff, 2012; Lan, Jia, & Jiang, 2014). Five conserved signaling pathways, namely, the Wnt, BMP, FGF, Shh, and Eda, are involved in the mediation of the successive reciprocal epithelial-mesenchymal cross talk which follows the general principle of morphogenetic interactions (Davidson, 1993). The pathways regulate the expression of transcription factors which confer the identity of dental epithelium and mesenchyme. The signals and transcription factors are integrated in complex signaling networks whose fine-tuning allows the generation of the variation in tooth morphologies. In this review, we describe the principles and molecular mechanisms of the epithelial-mesenchymal interactions regulating successive stages of tooth formation: (i) the initiation of tooth development, with special reference to the shift of tooth-forming potential from epithelium to mesenchyme; (ii) the morphogenesis of the tooth crown, focusing on the roles of epithelial signaling centers; (iii) the differentiation of odontoblasts and ameloblasts, which produce dentin and enamel, respectively; and (iv) the maintenance of dental stem cells, which support the continuous growth of teeth.


Assuntos
Epitélio/embriologia , Mesoderma/embriologia , Crista Neural/embriologia , Odontogênese , Dente/embriologia , Animais , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/citologia , Mesoderma/metabolismo , Crista Neural/citologia , Crista Neural/metabolismo , Transdução de Sinais/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Dente/citologia , Dente/metabolismo
12.
Development ; 142(22): 3954-63, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26450968

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Epitélio/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Dente Molar/embriologia , Morfogênese/fisiologia , Transdução de Sinais/fisiologia , Coroa do Dente/embriologia , Animais , Proteína Morfogenética Óssea 4/metabolismo , Epitélio/metabolismo , Imunofluorescência , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Análise Serial de Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Oral Facial Pain Headache ; 29(3): 297-307, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26244438

RESUMO

AIMS: To clarify the effect of estrogen and food hardness on condylar cartilage and the cartilage-bone interface. METHODS: A total of 56 rats were divided into four groups: (1) ovariectomized rats fed a normal (pressed pellet) food, (2) ovariectomized rats fed a soft (powder) food, (3) control rats fed a normal (pressed pellet) food, and (4) control rats fed a soft (powder) food. Some rats (n = 29) were sacrificed at the age of 67 days and others (n = 27) at the age of 87 days, and then 5-µm-thick sagittal paraffin sections were prepared from each temporomandibular joint (TMJ). Toluidine blue staining, in situ hybridization with type X collagen, terminal deoxynucleotidyl transferase and deoxyuridine triphosphate nick end labeling (TUNEL-assay), and tartrate-resistant acid phosphatase (TRAP) histochemistry were performed. Immunohistochemical analyses included cathepsin K, adiponectin, proliferating cell nuclear antigen (PCNA), and type X collagen staining. Analysis of variance and appropriate post-hoc tests were used in all analyses. RESULTS: Ovariectomy and normal food consistency increased the thickness of condylar cartilage (P < .001), PCNA expression (P < .001) and type X collagen expression (P < .001). Ovariectomy decreased the number (P < .05) and size of osteoclasts (P < .01). Soft food increased the number of cartilage cells stained positively against adiponectin (P < .01). CONCLUSION: Decreased estrogen level and normal food hardness increase the thickness of condylar cartilage by various mechanisms.


Assuntos
Cartilagem/metabolismo , Estrogênios/fisiologia , Alimentos , Animais , Feminino , Dureza , Côndilo Mandibular/metabolismo , Ratos
14.
Stem Cells ; 33(5): 1670-81, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25693510

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Células Epiteliais/citologia , Fator 10 de Crescimento de Fibroblastos/farmacologia , Homeostase/efeitos dos fármacos , Mesoderma/metabolismo , Células-Tronco/metabolismo , Dente/citologia , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Incisivo/citologia , Mesoderma/efeitos dos fármacos , Camundongos , Modelos Biológicos , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células-Tronco/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
15.
Nature ; 513(7519): 551-4, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25079316

RESUMO

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.


Assuntos
Diferenciação Celular , Linhagem da Célula , Incisivo/citologia , Células-Tronco Mesenquimais/citologia , Neuroglia/citologia , Animais , Rastreamento de Células , Células Clonais/citologia , Polpa Dentária/citologia , Feminino , Incisivo/embriologia , Masculino , Camundongos , Modelos Biológicos , Crista Neural/citologia , Odontoblastos/citologia , Regeneração , Células de Schwann/citologia
17.
Dev Cell ; 28(5): 588-602, 2014 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-24636260

RESUMO

Epithelial reorganization involves coordinated changes in cell shapes and movements. This restructuring occurs during formation of placodes, ectodermal thickenings that initiate the morphogenesis of epithelial organs including hair, mammary gland, and tooth. Signaling pathways in ectodermal placode formation are well known, but the cellular mechanisms have remained ill defined. We established imaging methodology for live visualization of embryonic skin explants during the first wave of hair placode formation. We found that the vast majority of placodal cells were nonproliferative throughout morphogenesis. We show that cell compaction and centripetal migration are the main cellular mechanisms associated with hair placode morphogenesis and that inhibition of actin remodeling suppresses placode formation. Stimulation of both ectodysplasin/NF-κB and Wnt/ß-catenin signaling increased cell motility and the number of cells committed to placodal fate. Thus, cell fate choices and morphogenetic events are controlled by the same molecular pathways, providing the framework for coordination of these two processes.


Assuntos
Movimento Celular , Proliferação de Células , Embrião de Mamíferos/citologia , Folículo Piloso/citologia , Queratinas/fisiologia , Morfogênese/fisiologia , Animais , Western Blotting , Ciclo Celular , Ectodisplasinas/fisiologia , Embrião de Mamíferos/metabolismo , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Folículo Piloso/embriologia , Folículo Piloso/metabolismo , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Microscopia de Fluorescência , NF-kappa B/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
19.
Differentiation ; 87(1-2): 32-43, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24393477

RESUMO

Mammalian tooth development is characterized by formation of primary teeth that belong to different tooth classes and are later replaced by a single set of permanent teeth. The first primary teeth are initiated from the primary dental lamina, and the replacement teeth from the successional dental lamina at the lingual side of the primary teeth. An interdental lamina connects the primary tooth germs together. Most mammalian tooth development research is done on mouse, which does not have teeth in all tooth classes, does not replace its teeth, and does not develop an interdental lamina. We have used the ferret (Mustela putorius furo) as a model animal to elucidate the morphological changes and gene expression during the development of the interdental lamina and the initiation of primary teeth. In addition we have analyzed cell-cell signaling taking place in the interdental lamina as well as in the successional lamina during tooth replacement. By 3D reconstructions of serial histological sections we observed that the morphogenesis of the interdental lamina and the primary teeth are intimately linked. Expression of Pitx2 and Foxi3 in the interdental lamina indicates that it has odontogenic identity, and there is active signaling taking place in the interdental lamina. Bmp4 is coexpressed with the stem cell factor Sox2 at its lingual aspect suggesting that the interdental lamina may retain competence for tooth initiation. We show that when tooth replacement is initiated there is Wnt pathway activity in the budding successional lamina and adjacent mesenchyme but no active Fgf or Eda signaling. Genes associated with human tooth replacement phenotypes, including Runx2 and Il11rα, are mostly expressed in the mesenchyme around the successional lamina in the ferret. Our results highlight the importance of the dental lamina in the mammalian tooth development during the initiation of both primary and replacement teeth.


Assuntos
Furões/crescimento & desenvolvimento , Mesoderma/crescimento & desenvolvimento , Odontogênese/genética , Dente/crescimento & desenvolvimento , Animais , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição Forkhead/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Humanos , Camundongos , Fatores de Transcrição SOXB1/genética , Transdução de Sinais/genética , Fatores de Transcrição/biossíntese , Via de Sinalização Wnt/genética , Proteína Homeobox PITX2
20.
Eur J Oral Sci ; 121(6): 509-16, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24148099

RESUMO

Ameloblastomas are locally invasive odontogenic tumors that exhibit a high rate of recurrence and often associate with the third molars. They are suggested to originate from dental epithelium because the tumor cells resemble epithelial cells of developing teeth. Expression of the transcription factor SOX2 has been previously localized in epithelial stem and progenitor cells in developing teeth as well as in various tumors. Here, we show that SOX2 is expressed in the epithelial cells of follicular and plexiform ameloblastomas. SOX2 was localized in the dental lamina of developing human primary molars. It was also expressed in the fragmented dental lamina associated with the third molars and in the epithelium budding from its posterior aspect in mice. However, no SOX2 expression was detected in either Hertwig's epithelial root sheath directing the formation of roots or in the epithelial cell rests of Malassez covering the completed roots. SOX2 was associated with supernumerary tooth formation in odontoma-like tumors induced by Wnt signal activation in mice. We propose that SOX2 functions in maintaining the progenitor state of epithelium in ameloblastomas and that ameloblastomas may originate from SOX2-expressing dental lamina epithelium.


Assuntos
Ameloblastoma/metabolismo , Epitélio/metabolismo , Neoplasias Maxilomandibulares/metabolismo , Odontogênese/fisiologia , Tumores Odontogênicos/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Células-Tronco/metabolismo , Adolescente , Adulto , Idoso , Ameloblastoma/patologia , Animais , Biomarcadores/metabolismo , Epitélio/patologia , Feminino , Humanos , Hibridização In Situ , Neoplasias Maxilomandibulares/patologia , Masculino , Mandíbula/patologia , Camundongos , Camundongos Mutantes , Pessoa de Meia-Idade , Dente Serotino/crescimento & desenvolvimento , Dente Serotino/metabolismo , Tumores Odontogênicos/patologia , RNA Mensageiro/metabolismo , Raiz Dentária/crescimento & desenvolvimento , Raiz Dentária/metabolismo , Dente Supranumerário/embriologia , Via de Sinalização Wnt/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA