Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Development ; 151(2)2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38108472

RESUMO

Nerves play important roles in organ development and tissue homeostasis. Stem/progenitor cells differentiate into different cell lineages responsible for building the craniofacial organs. The mechanism by which nerves regulate stem/progenitor cell behavior in organ morphogenesis has not yet been comprehensively explored. Here, we use tooth root development in mouse as a model to investigate how sensory nerves regulate organogenesis. We show that sensory nerve fibers are enriched in the dental papilla at the initiation of tooth root development. Through single cell RNA-sequencing analysis of the trigeminal ganglion and developing molar, we reveal several signaling pathways that connect the sensory nerve with the developing molar, of which FGF signaling appears to be one of the important regulators. Fgfr2 is expressed in the progenitor cells during tooth root development. Loss of FGF signaling leads to shortened roots with compromised proliferation and differentiation of progenitor cells. Furthermore, Hh signaling is impaired in Gli1-CreER;Fgfr2fl/fl mice. Modulation of Hh signaling rescues the tooth root defects in these mice. Collectively, our findings elucidate the nerve-progenitor crosstalk and reveal the molecular mechanism of the FGF-SHH signaling cascade during tooth root morphogenesis.


Assuntos
Dente , Animais , Camundongos , Dente Molar , Morfogênese/genética , Odontogênese/genética , Raiz Dentária
2.
Development ; 150(5)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36825984

RESUMO

Craniofacial morphogenesis requires complex interactions involving different tissues, signaling pathways, secreted factors and organelles. The details of these interactions remain elusive. In this study, we have analyzed the molecular mechanisms and homeostatic cellular activities governing soft palate development to improve regenerative strategies for individuals with cleft palate. We have identified canonical Wnt signaling as a key signaling pathway primarily active in cranial neural crest (CNC)-derived mesenchymal cells surrounding soft palatal myogenic cells. Using Osr2-Cre;ß-cateninfl/fl mice, we show that Wnt signaling is indispensable for mesenchymal cell proliferation and subsequently for myogenesis through mediating ciliogenesis. Specifically, we have identified that Wnt signaling directly regulates expression of the ciliary gene Ttll3. Impaired ciliary disassembly leads to differentiation defects in mesenchymal cells and indirectly disrupts myogenesis through decreased expression of Dlk1, a mesenchymal cell-derived pro-myogenesis factor. Moreover, we show that siRNA-mediated reduction of Ttll3 expression partly rescues mesenchymal cell proliferation and myogenesis in the palatal explant cultures from Osr2-Cre;ß-cateninfl/fl embryos. This study highlights the role of Wnt signaling in palatogenesis through the control of ciliary homeostasis, which establishes a new mechanism for Wnt-regulated craniofacial morphogenesis.


Assuntos
Fissura Palatina , Via de Sinalização Wnt , Camundongos , Animais , Via de Sinalização Wnt/fisiologia , Palato , Fissura Palatina/genética , Diferenciação Celular , Palato Mole , Homeostase , Regulação da Expressão Gênica no Desenvolvimento
3.
Genesis ; 62(1): e23582, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38069547

RESUMO

Tfap2b, a pivotal transcription factor, plays critical roles within neural crest cells and their derived lineage. To unravel the intricate lineage dynamics and contribution of these Tfap2b+ cells during craniofacial development, we established a Tfap2b-CreERT2 knock-in transgenic mouse line using the CRISPR-Cas9-mediated homologous direct repair. By breeding with tdTomato reporter mice and initiating Cre activity through tamoxifen induction at distinct developmental time points, we show the Tfap2b lineage within the key neural crest-derived domains, such as the facial mesenchyme, midbrain, cerebellum, spinal cord, and limbs. Notably, the migratory neurons stemming from the dorsal root ganglia are visible subsequent to Cre activity initiated at E8.5. Intriguingly, Tfap2b+ cells, serving as the progenitors for limb development, show activity predominantly commencing at E10.5. Across the mouse craniofacial landscape, Tfap2b exhibits a widespread presence throughout the facial organs. Here we validate its role as a marker of progenitors in tooth development and have confirmed that this process initiates from E12.5. Our study not only validates the Tfap2b-CreERT2 transgenic line, but also provides a powerful tool for lineage tracing and genetic targeting of Tfap2b-expressing cells and their progenitor in a temporally and spatially regulated manner during the intricate process of development and organogenesis.


Assuntos
Sistemas CRISPR-Cas , Tamoxifeno , Camundongos , Animais , Tamoxifeno/farmacologia , Camundongos Transgênicos , Proteína Vermelha Fluorescente , Integrases/genética , Integrases/metabolismo
4.
Development ; 148(8)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33766930

RESUMO

Stem cells self-renew or give rise to transit-amplifying cells (TACs) that differentiate into specific functional cell types. The fate determination of stem cells to TACs and their transition to fully differentiated progeny is precisely regulated to maintain tissue homeostasis. Arid1a, a core component of the switch/sucrose nonfermentable complex, performs epigenetic regulation of stage- and tissue-specific genes that is indispensable for stem cell homeostasis and differentiation. However, the functional mechanism of Arid1a in the fate commitment of mesenchymal stem cells (MSCs) and their progeny is not clear. Using the continuously growing adult mouse incisor model, we show that Arid1a maintains tissue homeostasis through limiting proliferation, promoting cell cycle exit and differentiation of TACs by inhibiting the Aurka-Cdk1 axis. Loss of Arid1a overactivates the Aurka-Cdk1 axis, leading to expansion of the mitotic TAC population but compromising their differentiation ability. Furthermore, the defective homeostasis after loss of Arid1a ultimately leads to reduction of the MSC population. These findings reveal the functional significance of Arid1a in regulating the fate of TACs and their interaction with MSCs to maintain tissue homeostasis.


Assuntos
Aurora Quinase A/metabolismo , Proteína Quinase CDC2/metabolismo , Proteínas de Ligação a DNA/metabolismo , Incisivo/embriologia , Células-Tronco Mesenquimais/metabolismo , Mitose , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Aurora Quinase A/genética , Proteína Quinase CDC2/genética , Proteínas de Ligação a DNA/genética , Camundongos , Camundongos Transgênicos , Fatores de Transcrição/genética
5.
Development ; 148(2)2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33323370

RESUMO

The control of size and shape is an important part of regulatory process during organogenesis. Tooth formation is a highly complex process that fine-tunes the size and shape of the tooth, which are crucial for its physiological functions. Each tooth consists of a crown and one or more roots. Despite comprehensive knowledge of the mechanism that regulates early tooth crown development, we have limited understanding of the mechanism regulating root patterning and size during development. Here, we show that Ror2-mediated non-canonical Wnt signaling in the dental mesenchyme plays a crucial role in cell proliferation, and thereby regulates root development size in mouse molars. Furthermore, Cdc42 acts as a potential downstream mediator of Ror2 signaling in root formation. Importantly, activation of Cdc42 can restore cell proliferation and partially rescue the root development size defects in Ror2 mutant mice. Collectively, our findings provide novel insights into the function of Ror2-mediated non-canonical Wnt signaling in regulating tooth morphogenesis, and suggest potential avenues for dental tissue engineering.


Assuntos
Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Raiz Dentária/embriologia , Raiz Dentária/metabolismo , Via de Sinalização Wnt , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Feminino , Masculino , Mesoderma/embriologia , Camundongos , Camundongos Mutantes , Morfogênese , Odontoblastos/citologia , Odontoblastos/metabolismo , Raiz Dentária/citologia
6.
PLoS Genet ; 17(2): e1009320, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33596195

RESUMO

Mammalian tooth crown formation has long served as a model for investigating how patterning and morphogenesis are orchestrated during development. However, the mechanism underlying root patterning and morphogenesis remains poorly understood. In this study, we find that Lhx6 labels a subpopulation of root progenitor cells in the apical dental mesenchyme, which is closely associated with furcation development. Loss of Lhx6 leads to furcation and root number defects, indicating that Lhx6 is a key root patterning regulator. Among the multiple cellular events regulated by Lhx6 is the odontoblast fate commitment of progenitor cells, which it controls in a cell-autonomous manner. Specifically, Lhx6 loss leads to elevated expression of the Wnt antagonist Sfrp2 and down-regulation of Wnt signaling in the furcation region, while overactivation of Wnt signaling in Lhx6+ progenitor cells partially restore the furcation defects in Lhx6-/- mice. Collectively, our findings have important implications for understanding organ morphogenesis and future strategies for tooth root regeneration.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Homeodomínio LIM/genética , Células-Tronco Mesenquimais/metabolismo , Dente Molar/metabolismo , Morfogênese/genética , Proteínas do Tecido Nervoso/genética , Raiz Dentária/metabolismo , Fatores de Transcrição/genética , Via de Sinalização Wnt/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Feminino , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Genéticos , Dente Molar/citologia , Dente Molar/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/metabolismo , Raiz Dentária/citologia , Raiz Dentária/crescimento & desenvolvimento , Fatores de Transcrição/metabolismo
7.
Dev Biol ; 468(1-2): 110-132, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32692983

RESUMO

BCOR is a critical regulator of human development. Heterozygous mutations of BCOR in females cause the X-linked developmental disorder Oculofaciocardiodental syndrome (OFCD), and hemizygous mutations of BCOR in males cause gestational lethality. BCOR associates with Polycomb group proteins to form one subfamily of the diverse Polycomb repressive complex 1 (PRC1) complexes, designated PRC1.1. Currently there is limited understanding of differing developmental roles of the various PRC1 complexes. We therefore generated a conditional exon 9-10 knockout Bcor allele and a transgenic conditional Bcor expression allele and used these to define multiple roles of Bcor, and by implication PRC1.1, in mouse development. Females heterozygous for Bcor exhibiting mosaic expression due to the X-linkage of the gene showed reduced postnatal viability and had OFCD-like defects. By contrast, Bcor hemizygosity in the entire male embryo resulted in embryonic lethality by E9.5. We further dissected the roles of Bcor, focusing on some of the tissues affected in OFCD through use of cell type specific Cre alleles. Mutation of Bcor in neural crest cells caused cleft palate, shortening of the mandible and tympanic bone, ectopic salivary glands and abnormal tongue musculature. We found that defects in the mandibular region, rather than in the palate itself, led to palatal clefting. Mutation of Bcor in hindlimb progenitor cells of the lateral mesoderm resulted in 2/3 syndactyly. Mutation of Bcor in Isl1-expressing lineages that contribute to the heart caused defects including persistent truncus arteriosus, ventricular septal defect and fetal lethality. Mutation of Bcor in extraembryonic lineages resulted in placental defects and midgestation lethality. Ubiquitous over expression of transgenic Bcor isoform A during development resulted in embryonic defects and midgestation lethality. The defects we have found in Bcor mutants provide insights into the etiology of the OFCD syndrome and how BCOR-containing PRC1 complexes function in development.


Assuntos
Catarata/congênito , Embrião de Mamíferos , Defeitos dos Septos Cardíacos , Microftalmia , Complexo Repressor Polycomb 1 , Proteínas Repressoras , Animais , Catarata/embriologia , Catarata/genética , Catarata/patologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/patologia , Defeitos dos Septos Cardíacos/embriologia , Defeitos dos Septos Cardíacos/genética , Defeitos dos Septos Cardíacos/patologia , Camundongos , Microftalmia/embriologia , Microftalmia/genética , Microftalmia/patologia , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
8.
Development ; 144(14): 2560-2569, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28576771

RESUMO

Signaling pathways are used reiteratively in different developmental processes yet produce distinct cell fates through specific downstream transcription factors. In this study, we used tooth root development as a model with which to investigate how the BMP signaling pathway regulates transcriptional complexes to direct the fate determination of multipotent mesenchymal stem cells (MSCs). We first identified the MSC population supporting mouse molar root growth as Gli1+ cells. Using a Gli1-driven Cre-mediated recombination system, our results provide the first in vivo evidence that BMP signaling activity is required for the odontogenic differentiation of MSCs. Specifically, we identified the transcription factors Pax9, Klf4, Satb2 and Lhx8 as being downstream of BMP signaling and expressed in a spatially restricted pattern that is potentially involved in determining distinct cellular identities within the dental mesenchyme. Finally, we found that overactivation of one key transcription factor, Klf4, which is associated with the odontogenic region, promotes odontogenic differentiation of MSCs. Collectively, our results demonstrate the functional significance of BMP signaling in regulating MSC fate during root development and shed light on how BMP signaling can achieve functional specificity in regulating diverse organ development.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem da Célula/genética , Linhagem da Célula/fisiologia , Feminino , Redes Reguladoras de Genes , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Camundongos Transgênicos , Odontoblastos/citologia , Odontoblastos/metabolismo , Odontogênese/genética , Odontogênese/fisiologia , Regeneração/genética , Regeneração/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/genética , Nicho de Células-Tronco/fisiologia , Raiz Dentária/citologia , Raiz Dentária/crescimento & desenvolvimento , Raiz Dentária/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína GLI1 em Dedos de Zinco/genética , Proteína GLI1 em Dedos de Zinco/metabolismo
9.
Development ; 144(21): 4037-4045, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28982687

RESUMO

Craniofacial development depends on cell-cell interactions, coordinated cellular movement and differentiation under the control of regulatory gene networks, which include the distal-less (Dlx) gene family. However, the functional significance of Dlx5 in patterning the oropharyngeal region has remained unknown. Here, we show that loss of Dlx5 leads to a shortened soft palate and an absence of the levator veli palatini, palatopharyngeus and palatoglossus muscles that are derived from the 4th pharyngeal arch (PA); however, the tensor veli palatini, derived from the 1st PA, is unaffected. Dlx5-positive cranial neural crest (CNC) cells are in direct contact with myoblasts derived from the pharyngeal mesoderm, and Dlx5 disruption leads to altered proliferation and apoptosis of CNC and muscle progenitor cells. Moreover, the FGF10 pathway is downregulated in Dlx5-/- mice, and activation of FGF10 signaling rescues CNC cell proliferation and myogenic differentiation in these mutant mice. Collectively, our results indicate that Dlx5 plays crucial roles in the patterning of the oropharyngeal region and development of muscles derived from the 4th PA mesoderm in the soft palate, likely via interactions between CNC-derived and myogenic progenitor cells.


Assuntos
Padronização Corporal , Região Branquial/embriologia , Comunicação Celular , Fator 10 de Crescimento de Fibroblastos/metabolismo , Proteínas de Homeodomínio/metabolismo , Boca/embriologia , Mioblastos/citologia , Crista Neural/citologia , Crânio/embriologia , Animais , Região Branquial/metabolismo , Diferenciação Celular , Proliferação de Células , Regulação para Baixo/genética , Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Camundongos Knockout , Desenvolvimento Muscular , Mioblastos/metabolismo , Crista Neural/metabolismo , Palato/embriologia , Palato/metabolismo , Transdução de Sinais , Crânio/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
10.
Dev Biol ; 441(1): 191-203, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29981310

RESUMO

Cleft palate is one of the most common craniofacial congenital defects in humans. It is associated with multiple genetic and environmental risk factors, including mutations in the genes encoding signaling molecules in the sonic hedgehog (Shh) pathway, which are risk factors for cleft palate in both humans and mice. However, the function of Shh signaling in the palatal epithelium during palatal fusion remains largely unknown. Although components of the Shh pathway are localized in the palatal epithelium, specific inhibition of Shh signaling in palatal epithelium does not affect palatogenesis. We therefore utilized a hedgehog (Hh) signaling gain-of-function mouse model, K14-Cre;R26SmoM2, to uncover the role of Shh signaling in the palatal epithelium during palatal fusion. In this study, we discovered that constitutive activation of Hh signaling in the palatal epithelium results in submucous cleft palate and persistence of the medial edge epithelium (MEE). Further investigation revealed that precise downregulation of Shh signaling is required at a specific time point in the MEE during palatal fusion. Upregulation of Hh signaling in the palatal epithelium maintains the proliferation of MEE cells. This may be due to a dysfunctional p63/Irf6 regulatory loop. The resistance of MEE cells to apoptosis is likely conferred by enhancement of a cell adhesion network through the maintenance of p63 expression. Collectively, our data illustrate that persistent Hh signaling in the palatal epithelium contributes to the etiology and pathogenesis of submucous cleft palate through its interaction with a p63/Irf6-dependent biological regulatory loop and through a p63-induced cell adhesion network.


Assuntos
Embrião de Mamíferos/metabolismo , Células Epiteliais/metabolismo , Proteínas Hedgehog/metabolismo , Palato/embriologia , Transdução de Sinais/fisiologia , Animais , Adesão Celular/fisiologia , Embrião de Mamíferos/citologia , Células Epiteliais/citologia , Proteínas Hedgehog/genética , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Camundongos , Camundongos Transgênicos , Palato/citologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transativadores/genética , Transativadores/metabolismo
11.
Hum Mol Genet ; 26(5): 860-872, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28069795

RESUMO

Ciliopathies are pleiotropic human diseases resulting from defects of the primary cilium, and these patients often have cleft lip and palate. IFT88 is required for the assembly and function of the primary cilia, which mediate the activity of key developmental signaling pathways. Through whole exome sequencing of a family of three affected siblings with isolated cleft lip and palate, we discovered that they share a novel missense mutation in IFT88 (c.915G > C, p.E305D), suggesting this gene should be considered a candidate for isolated orofacial clefting. In order to evaluate the function of IFT88 in regulating craniofacial development, we generated Wnt1-Cre;Ift88fl/fl mice to eliminate Ift88 specifically in cranial neural crest (CNC) cells. Wnt1-Cre;Ift88fl/flpups died at birth due to severe craniofacial defects including bilateral cleft lip and palate and tongue agenesis, following the loss of the primary cilia in the CNC-derived palatal mesenchyme. Loss of Ift88 also resulted in a decrease in neural crest cell proliferation during early stages of palatogenesis as well as a downregulation of the Shh signaling pathway in the palatal mesenchyme. Importantly, Osr2KI-Cre;Ift88fl/flmice, in which Ift88 is lost specifically in the palatal mesenchyme, exhibit isolated cleft palate. Taken together, our results demonstrate that IFT88 has a highly conserved function within the primary cilia of the CNC-derived mesenchyme in the lip and palate region in mice and is a strong candidate as an orofacial clefting gene in humans.


Assuntos
Fenda Labial/genética , Desenvolvimento Embrionário/genética , Proteínas Supressoras de Tumor/genética , Proteína Wnt1/genética , Animais , Proliferação de Células/genética , Fenda Labial/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/crescimento & desenvolvimento , Mesoderma/patologia , Camundongos , Crista Neural/crescimento & desenvolvimento , Crista Neural/metabolismo , Crista Neural/patologia , Palato/crescimento & desenvolvimento , Palato/patologia , Transdução de Sinais , Proteínas Supressoras de Tumor/biossíntese , Proteína Wnt1/biossíntese
12.
Development ; 142(19): 3374-82, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26293299

RESUMO

Coordination between the Hertwig's epithelial root sheath (HERS) and apical papilla (AP) is crucial for proper tooth root development. The hedgehog (Hh) signaling pathway and Nfic are both involved in tooth root development; however, their relationship has yet to be elucidated. Here, we establish a timecourse of mouse molar root development by histological staining of sections, and we demonstrate that Hh signaling is active before and during root development in the AP and HERS using Gli1 reporter mice. The proper pattern of Hh signaling activity in the AP is crucial for the proliferation of dental mesenchymal cells, because either inhibition with Hh inhibitors or constitutive activation of Hh signaling activity in transgenic mice leads to decreased proliferation in the AP and shorter roots. Moreover, Hh activity is elevated in Nfic(-/-) mice, a root defect model, whereas RNA sequencing and in situ hybridization show that the Hh attenuator Hhip is downregulated. ChIP and RNAscope analyses suggest that Nfic binds to the promoter region of Hhip. Treatment of Nfic(-/-) mice with Hh inhibitor partially restores cell proliferation, AP growth and root development. Taken together, our results demonstrate that an Nfic-Hhip-Hh signaling pathway is crucial for apical papilla growth and proper root formation. This discovery provides insight into the molecular mechanisms regulating tooth root development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Hedgehog/metabolismo , Morfogênese/fisiologia , Fatores de Transcrição NFI/metabolismo , Transdução de Sinais/fisiologia , Raiz Dentária/crescimento & desenvolvimento , Animais , Sequência de Bases , Imunoprecipitação da Cromatina , Primers do DNA/genética , Galactosídeos , Hibridização In Situ , Indóis , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Dente Molar/crescimento & desenvolvimento , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de RNA , Tamoxifeno , Microtomografia por Raio-X , Proteína GLI1 em Dedos de Zinco
13.
Genesis ; 54(9): 490-6, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27381449

RESUMO

Pax9 encodes a paired-box homeodomain (Pax) transcription factor and is critical for the development of multiple organs. Using CrispR/Cas9-mediated homologous directed repair (HDR), we generated a new Pax9-CreER knock-in mouse line in which the CreER(T2) fusion protein is produced after synthesis of endogenous Pax9 protein. We found that tdTomato reporter expression in Pax9-CreER;tdTomato reporter mice is detectable in a similar pattern to the endogenous Pax9 expression, faithfully recapitulating the Pax9 expression domains throughout the embryo and in the adult mouse. At early embryonic stages, the tdTomato reporter is expressed first in the pharyngeal pouch region and later in the craniofacial mesenchyme, somites, limbs, and lingual papillae in the adult tongue. These results demonstrate that this new Pax9-CreER knock-in mouse line can be used for lineage tracing and genetic targeting of Pax9-expressing cells and their progeny in a temporally and spatially controlled manner during development and organogenesis.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Introdução de Genes/métodos , Animais , Integrases/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição PAX9/genética , Tamoxifeno/farmacologia , Ativação Transcricional/efeitos dos fármacos , Proteína Vermelha Fluorescente
14.
Proc Natl Acad Sci U S A ; 108(16): 6503-8, 2011 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-21464310

RESUMO

In many adult tissues, mesenchymal stem cells (MSCs) are closely associated with perivascular niches and coexpress many markers in common with pericytes. The ability of pericytes to act as MSCs, however, remains controversial. By using genetic lineage tracing, we show that some pericytes differentiate into specialized tooth mesenchyme-derived cells--odontoblasts--during tooth growth and in response to damage in vivo. As the pericyte-derived mesenchymal cell contribution to odontoblast differentiation does not account for all cell differentiation, we identify an additional source of cells with MSC-like properties that are stimulated to migrate toward areas of tissue damage and differentiate into odontoblasts. Thus, although pericytes are capable of acting as a source of MSCs and differentiating into cells of mesenchymal origin, they do so alongside other MSCs of a nonpericyte origin. This study identifies a dual origin of MSCs in a single tissue and suggests that the pericyte contribution to MSC-derived mesenchymal cells in any given tissue is variable and possibly dependent on the extent of the vascularity.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais , Odontoblastos , Pericitos , Regeneração/fisiologia , Dente , Animais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Transgênicos , Odontoblastos/citologia , Odontoblastos/fisiologia , Pericitos/citologia , Pericitos/fisiologia , Dente/citologia , Dente/crescimento & desenvolvimento
15.
Cell Stem Cell ; 31(6): 904-920.e6, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38703771

RESUMO

Mesenchymal stem cells (MSCs) reside in niches to maintain tissue homeostasis and contribute to repair and regeneration. Although the physiological functions of blood and lymphatic vasculature are well studied, their regulation of MSCs as niche components remains largely unknown. Using adult mouse incisors as a model, we uncover the role of Trp53 in regulating vascular composition through THBS2 to maintain mesenchymal tissue homeostasis. Loss of Trp53 in GLI1+ progeny increases arteries and decreases other vessel types. Platelet-derived growth factors from arteries deposit in the MSC region and interact with PDGFRA and PDGFRB. Significantly, PDGFRA+ and PDGFRB+ cells differentially contribute to defined cell lineages in the adult mouse incisor. Collectively, our results highlight Trp53's importance in regulating the vascular niche for MSCs. They also shed light on how different arterial cells provide unique cues to regulate MSC subpopulations and maintain their heterogeneity. Furthermore, they provide mechanistic insight into MSC-vasculature crosstalk.


Assuntos
Incisivo , Células-Tronco Mesenquimais , Transdução de Sinais , Proteína Supressora de Tumor p53 , Animais , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Proteína Supressora de Tumor p53/metabolismo , Incisivo/citologia , Incisivo/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo
16.
Plast Reconstr Surg ; 153(3): 637-646, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-37224290

RESUMO

BACKGROUND: The standard graft material for alveolar cleft repair (ACR) is autogenous iliac crest. A promising alternative potential graft adjunct-newborn human umbilical cord mesenchymal stem cells (h-UCMSCs)-has yet to be explored in vivo. Their capacity for self-renewal, multipotent differentiation, and proliferation allows h-UCMSCs to be harnessed for regenerative medicine. This study sought to evaluate the efficacy of using tissue-derived h-UCMSCs and their osteogenic capabilities to improve ACR in a murine model. METHODS: Foxn1 mice were separated into three groups with the following calvarial defects: no treatment (empty defect; n = 6), poly(D,L-lactide-co-glycolide) (PLGA) scaffold ( n = 6), or h-UCMSCs with PLGA ( n = 4). Bilateral 2-mm-diameter parietal bone critical-sized defects were created using a dental drill. Microcomputed tomography (microCT) imaging was performed 1, 2, 3, and 4 weeks postoperatively. The mice were euthanized 4 weeks postoperatively for RNAScope, immunohistochemical, and histological analysis. RESULTS: No mice experienced complications during the follow-up period. MicroCT imaging and histological analysis demonstrated that the no-treatment and PLGA-only defects remained patent without significant defect size differences across groups. In contrast, the h-UCMSCs with PLGA group had significantly greater bone fill on microCT and histological analysis. CONCLUSIONS: This study demonstrates a successful calvarial defect model for the investigation of h-UCMSC-mediated osteogenesis and bone repair. Evidence reveals that PLGA alone has neither short-term effects on bone formation nor any unwanted side effects, making it an attractive scaffold. Further investigation using h-UCMSCs with PLGA in larger animals is warranted to advance future translation to patients requiring ACR. CLINICAL RELEVANCE STATEMENT: The authors' results demonstrate a successful murine calvarial defect model for the investigation of h-UCMSC-mediated osteogenesis and bone repair, and they provide preliminary evidence for the safe and efficacious use of this graft adjunct in alveolar cleft repair.


Assuntos
Osteogênese , Alicerces Teciduais , Humanos , Camundongos , Animais , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Microtomografia por Raio-X , Regeneração Óssea , Células-Tronco , Diferenciação Celular , Cordão Umbilical , Crânio/cirurgia , Crânio/patologia
17.
Bone Res ; 12(1): 37, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38910207

RESUMO

Stem/progenitor cells differentiate into different cell lineages during organ development and morphogenesis. Signaling pathway networks and mechanotransduction are important factors to guide the lineage commitment of stem/progenitor cells during craniofacial tissue morphogenesis. Here, we used tooth root development as a model to explore the roles of FGF signaling and mechanotransduction as well as their interaction in regulating the progenitor cell fate decision. We show that Fgfr1 is expressed in the mesenchymal progenitor cells and their progeny during tooth root development. Loss of Fgfr1 in Gli1+ progenitors leads to hyperproliferation and differentiation, which causes narrowed periodontal ligament (PDL) space with abnormal cementum/bone formation leading to ankylosis. We further show that aberrant activation of WNT signaling and mechanosensitive channel Piezo2 occurs after loss of FGF signaling in Gli1-CreER;Fgfr1fl/fl mice. Overexpression of Piezo2 leads to increased osteoblastic differentiation and decreased Piezo2 leads to downregulation of WNT signaling. Mechanistically, an FGF/PIEZO2/WNT signaling cascade plays a crucial role in modulating the fate of progenitors during root morphogenesis. Downregulation of WNT signaling rescues tooth ankylosis in Fgfr1 mutant mice. Collectively, our findings uncover the mechanism by which FGF signaling regulates the fate decisions of stem/progenitor cells, and the interactions among signaling pathways and mechanotransduction during tooth root development, providing insights for future tooth root regeneration.


Assuntos
Fatores de Crescimento de Fibroblastos , Mecanotransdução Celular , Raiz Dentária , Via de Sinalização Wnt , Animais , Via de Sinalização Wnt/fisiologia , Raiz Dentária/crescimento & desenvolvimento , Raiz Dentária/metabolismo , Raiz Dentária/citologia , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Camundongos , Diferenciação Celular , Células-Tronco/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Canais Iônicos
18.
Nat Commun ; 15(1): 4614, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38816354

RESUMO

ARID1B haploinsufficiency in humans causes Coffin-Siris syndrome, associated with developmental delay, facial dysmorphism, and intellectual disability. The role of ARID1B has been widely studied in neuronal development, but whether it also regulates stem cells remains unknown. Here, we employ scRNA-seq and scATAC-seq to dissect the regulatory functions and mechanisms of ARID1B within mesenchymal stem cells (MSCs) using the mouse incisor model. We reveal that loss of Arid1b in the GLI1+ MSC lineage disturbs MSCs' quiescence and leads to their proliferation due to the ectopic activation of non-canonical Activin signaling via p-ERK. Furthermore, loss of Arid1b upregulates Bcl11b, which encodes a BAF complex subunit that modulates non-canonical Activin signaling by directly regulating the expression of activin A subunit, Inhba. Reduction of Bcl11b or non-canonical Activin signaling restores the MSC population in Arid1b mutant mice. Notably, we have identified that ARID1B suppresses Bcl11b expression via specific binding to its third intron, unveiling the direct inter-regulatory interactions among BAF subunits in MSCs. Our results demonstrate the vital role of ARID1B as an epigenetic modifier in maintaining MSC homeostasis and reveal its intricate mechanistic regulatory network in vivo, providing novel insights into the linkage between chromatin remodeling and stem cell fate determination.


Assuntos
Proteínas de Ligação a DNA , Células-Tronco Mesenquimais , Proteínas Repressoras , Transdução de Sinais , Animais , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proliferação de Células , Ativinas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Humanos , Proteína GLI1 em Dedos de Zinco
19.
Dev Biol ; 367(2): 140-53, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22562112

RESUMO

Rodent incisors are capable of growing continuously and the renewal of dental epithelium giving rise to enamel-forming ameloblasts and dental mesenchyme giving rise to dentin-forming odontoblasts and pulp cells is achieved by stem cells residing at their proximal ends. Although the dental epithelial stem cell niche (cervical loop) is well characterized, little is known about the dental mesenchymal stem cell niche. Ring1a/b are the core Polycomb repressive complex1 (PRC1) components that have recently also been found in a protein complex with BcoR (Bcl-6 interacting corepressor) and Fbxl10. During mouse incisor development, we found that genes encoding members of the PRC1 complex are strongly expressed in the incisor apical mesenchyme in an area that contains the cells with the highest proliferation rate in the tooth pulp, consistent with a location for transit amplifying cells. Analysis of Ring1a(-/-);Ring1b(cko/cko) mice showed that loss of Ring1a/b postnatally results in defective cervical loops and disturbances of enamel and dentin formation in continuously growing incisors. To further characterize the defect found in Ring1a(-/-);Ring1b(cko/cko) mice, we demonstrated that cell proliferation is dramatically reduced in the apical mesenchyme and cervical loop epithelium of Ring1a(-/-);Ring1b(cko/cko) incisors in comparison to Ring1a(-/-);Ring1b(fl/fl)cre- incisors. Fgf signaling and downstream targets that have been previously shown to be important in the maintenance of the dental epithelial stem cell compartment in the cervical loop are downregulated in Ring1a(-/-);Ring1b(cko/cko) incisors. In addition, expression of other genes of the PRC1 complex is also altered. We also identified an essential postnatal requirement for Ring1 proteins in molar root formation. These results show that the PRC1 complex regulates the transit amplifying cell compartment of the dental mesenchymal stem cell niche and cell differentiation in developing mouse incisors and is required for molar root formation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Incisivo/citologia , Incisivo/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas Repressoras/metabolismo , Nicho de Células-Tronco/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Esmalte Dentário/citologia , Esmalte Dentário/crescimento & desenvolvimento , Esmalte Dentário/metabolismo , Dentina/citologia , Dentina/crescimento & desenvolvimento , Dentina/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Incisivo/anormalidades , Incisivo/crescimento & desenvolvimento , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Transdução de Sinais , Nicho de Células-Tronco/genética , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética
20.
Nat Commun ; 14(1): 344, 2023 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-36670126

RESUMO

Mesenchymal stem cells (MSCs) reside in microenvironments, referred to as niches, which provide structural support and molecular signals. Sensory nerves are niche components in the homeostasis of tissues such as skin, bone marrow and hematopoietic system. However, how the sensory nerve affects the behavior of MSCs remains largely unknown. Here we show that the sensory nerve is vital for mesenchymal tissue homeostasis and maintenance of MSCs in the continuously growing adult mouse incisor. Loss of sensory innervation leads to mesenchymal disorder and a decrease in MSCs. Mechanistically, FGF1 from the sensory nerve directly acts on MSCs by binding to FGFR1 and activates the mTOR/autophagy axis to sustain MSCs. Modulation of mTOR/autophagy restores the MSCs and rescues the mesenchymal tissue disorder of Fgfr1 mutant mice. Collectively, our study provides insights into the role of sensory nerves in the regulation of MSC homeostasis and the mechanism governing it.


Assuntos
Células-Tronco Mesenquimais , Camundongos , Animais , Células-Tronco Mesenquimais/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Autofagia/fisiologia , Medula Óssea/metabolismo , Homeostase , Nicho de Células-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA