Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
Arch Oral Biol ; 155: 105797, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37633030

RESUMO

OBJECTIVE: This study aimed to evaluate the role of ruxolitinib in the interferon beta (IFN-ß) mediated osteoblast differentiation using human dental pulp stem cells (hDPSCs). DESIGN: hDPSCs from five deciduous teeth of healthy patients were stimulated by adding human recombinant IFN-ß protein (1 or 2 ng/ml) to the osteogenic differentiation induction medium. Substrate formation was determined using Alizarin Red staining, calcium concentration, and osteoblast marker expression levels. Ruxolitinib was used to inhibit the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway. Apoptosis was detected using terminal deoxynucleotidyl nick-end labeling (TUNEL) staining, and necroptosis was detected using propidium iodide staining and phosphorylated mixed lineage kinase domain-like protein (pMLKL) expression. RESULTS: In the IFN-ß-treated group, substrate formation was inhibited by a reduction in alkaline phosphatase (ALP) expression in a concentration-dependent manner. Although the proliferation potency was unchanged between the IFN-ß-treated and control groups, the cell number was significantly reduced in the experimental group. TUNEL-positive cell number was not significantly different; however, the protein level of necroptosis markers, interleukin-6 (IL-6) and pMLKL were significantly increased in the substrate formation. Cell number and ALP expression level were improved in the group administered ruxolitinib, a JAK-STAT inhibitor. Additionally, ruxolitinib significantly suppressed IL-6 and pMLKL levels. CONCLUSION: Ruxolitinib interfered with the IFN-ß-mediated necroptosis and osteogenic differentiation via the JAK-STAT pathway.

2.
Genesis ; 61(1-2): e23509, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36622051

RESUMO

Craniofacial anomalies (CFAs) are a diverse group of disorders affecting the shapes of the face and the head. Malformation of the cranial base in humans leads CFAs, such as midfacial hypoplasia and craniosynostosis. These patients have significant burdens associated with breathing, speaking, and chewing. Invasive surgical intervention is the current primary option to correct these structural deficiencies. Understanding molecular cellular mechanism for craniofacial development would provide novel therapeutic options for CFAs. In this study, we found that enhanced bone morphogenetic protein (BMP) signaling in cranial neural crest cells (NCCs) (P0-Cre;caBmpr1a mice) causes premature fusion of intersphenoid synchondrosis (ISS) resulting in leading to short snouts and hypertelorism. Histological analyses revealed reduction of proliferation and higher cell death in ISS at postnatal day 3. We demonstrated to prevent the premature fusion of ISS in P0-Cre;caBmpr1a mice by injecting a p53 inhibitor Pifithrin-α to the pregnant mother from E15.5 to E18.5, resulting in rescue from short snouts and hypertelorism. We further demonstrated to prevent premature fusion of cranial sutures in P0-Cre;caBmpr1a mice by injecting Pifithrin-α through E8.5 to E18.5. These results suggested that enhanced BMP-p53-induced cell death in cranial NCCs causes premature fusion of ISS and sutures in time-dependent manner.


Assuntos
Anormalidades Craniofaciais , Base do Crânio , Proteínas Morfogenéticas Ósseas/metabolismo , Crista Neural/metabolismo , Crista Neural/patologia , Proliferação de Células , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Masculino , Feminino , Animais , Camundongos , Animais Recém-Nascidos , Transdução de Sinais , Apoptose , Condrócitos/metabolismo , Proteínas Smad/metabolismo , Ligação Proteica , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Gravidez , Base do Crânio/anormalidades , Base do Crânio/metabolismo , Base do Crânio/patologia , Hipertelorismo/metabolismo , Hipertelorismo/patologia
3.
Pathol Int ; 71(12): 803-813, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34587661

RESUMO

Diamond-Blackfan anemia (DBA) is a genetic disorder caused by mutations in genes encoding ribosomal proteins and characterized by erythroid aplasia and various physical abnormalities. Although accumulating evidence suggests that defective ribosome biogenesis leads to p53-mediated apoptosis in erythroid progenitor cells, little is known regarding the underlying causes of the physical abnormalities. In this study, we established induced pluripotent stem cells from a DBA patient with RPL5 haploinsufficiency. These cells retained the ability to differentiate into osteoblasts and chondrocytes. However, RPL5 haploinsufficiency impaired the production of mucins and increased apoptosis in differentiated chondrocytes. Increased expression of the pro-apoptotic genes BAX and CASP9 further indicated that RPL5 haploinsufficiency triggered p53-mediated apoptosis in chondrocytes. Murine double minute 2 (MDM2), the primary negative regulator of p53, plays a crucial role in erythroid aplasia in DBA patient. We found the phosphorylation level of MDM2 was significantly decreased in RPL5 haploinsufficient chondrocytes. In stark contrast, we found no evidence that RPL5 haploinsufficiency impaired osteogenesis. Collectively, our data support a model in which RPL5 haploinsufficiency specifically induces p53-mediated apoptosis in chondrocytes through MDM2 inhibition, which leads to physical abnormalities in DBA patients.


Assuntos
Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/patologia , Haploinsuficiência , Proteínas Ribossômicas/genética , Animais , Apoptose/genética , Criança , Condrócitos/patologia , Marcadores Genéticos , Humanos , Células-Tronco Pluripotentes Induzidas , Masculino , Camundongos , Osteogênese/genética
4.
Biofactors ; 47(6): 992-1015, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34418170

RESUMO

This study aimed to reveal the possible mechanisms by which O-linked-N-acetylglucosaminylation (O-GlcNAcylation) regulates osteoblast differentiation using a series of bioinformatics-oriented experiments. To examine the influence of O-GlcNAcylation levels on osteoblast differentiation, osteoblastic MC3T3-E1 cells were treated with O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) inhibitors. Correlations between the levels of O-GlcNAcylation and the expression of osteogenic markers as well as OGT were evaluated by qPCR and western blotting. The O-GlcNAcylated proteins assumed to correlate with Runx2 expression were retrieved from several public databases and used for further bioinformatics analysis. Following the findings of the bioinformatics analysis, intracellular calcium ([Ca2+ ]i ) was monitored in the cells treated with OGT and OGA inhibitors using a confocal laser-scanning microscope (CLS). The interaction effect between O-GlcNAcylation and [Ca2+ ]i on osteogenic marker expression was determined using stable OGT knockdown MC3T3-E1 cells. O-GlcNAcylation was positively associated with osteoblast differentiation. The time-course profile of global O-GlcNAcylated proteins showed a distinctive pattern with different molecular weights during osteoblast differentiation. The expression pattern of several O-GlcNAcylated proteins was significantly similar to that of Runx2 expression. Bioinformatic analysis of the retrieved Runx2-related-O-GlcNAcylated-proteins revealed the importance of [Ca2+ ]i . CLS showed that alteration of O-GlcNAcylation rapidly changed [Ca2+ ]i in MC3T3-E1 cells. O-GlcNAcylation and [Ca2+ ]i showed an interaction effect on the expression of osteogenic markers. OGT knockdown disrupted the [Ca2+ ]i -induced expression changes of osteogenic markers. O-GlcNAcylation interacts with [Ca2+ ]i and elicits osteoblast differentiation by regulating the expression of osteogenic markers.


Assuntos
Sinalização do Cálcio/fisiologia , Diferenciação Celular/fisiologia , Biologia Computacional/métodos , N-Acetilglucosaminiltransferases/metabolismo , Osteoblastos/metabolismo , Osteogênese/fisiologia , Animais , Camundongos , Modelos Animais
5.
Genesis ; 56(6-7): e23220, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-30134066

RESUMO

Craniosynostosis is defined as congenital premature fusion of one or more cranial sutures. While the genetic basis for about 30% of cases is known, the causative genes for the diverse presentations of the remainder of cases are unknown. The recently discovered cranial suture stem cell population affords an opportunity to identify early signaling pathways that contribute to craniosynostosis. We previously demonstrated that enhanced BMP signaling in neural crest cells (caA3 mutants) leads to premature cranial suture fusion resulting in midline craniosynostosis. Since enhanced mTOR signaling in neural crest cells leads to craniofacial bone lesions, we investigated the extent to which mTOR signaling is involved in the pathogenesis of BMP-mediated craniosynostosis by affecting the suture stem cell population. Our results demonstrate a loss of suture stem cells in the caA3 mutant mice by the newborn stage. We have found increased activation of mTOR signaling in caA3 mutant mice during embryonic stages, but not at the newborn stage. Our study demonstrated that inhibition of mTOR signaling via rapamycin in a time specific manner partially rescued the loss of the suture stem cell population. This study provides insight into how enhanced BMP signaling regulates suture stem cells via mTOR activation.


Assuntos
Craniossinostoses/genética , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/efeitos dos fármacos , Animais , Proteínas Morfogenéticas Ósseas/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/fisiologia , Suturas Cranianas/embriologia , Craniossinostoses/tratamento farmacológico , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Crista Neural/metabolismo , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/metabolismo , Crânio/embriologia
6.
Genesis ; 56(3): e23093, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29411501

RESUMO

BMP signaling plays a critical role in craniofacial development. Augmentation of BMPR1A signaling through neural crest-specific expression of constitutively active Bmpr1a (caBmpr1a) results in craniofacial deformities in mice. To investigate whether deletion of Tak1 may rescue the craniofacial deformities caused by enhanced Smad-dependent signaling through caBMPR1A, we generated embryos to activate transcription of caBmpr1a transgene and ablate Tak1 in neural crest derivatives at the same time. We found that deformities of the double mutant mice showed more severe than those with each single mutation, including median facial cleft and cleft palate. We found higher levels of cell death in the medial nasal and the lateral nasal processes at E10.5 in association with higher levels of p53 in the double mutant embryos. We also found higher levels of pSmad1/5/9 in the lateral nasal processes at E10.5 in the double mutant embryos. Western analyses revealed that double mutant embryos showed similar degrees of upregulation of pSmad1/5/9 with caBmpr1a or Tak1-cKO embryos while the double mutant embryos showed higher levels of phospho-p38 than caBmpr1a or Tak1-cKO embryos at E17.5, but not at E10.5. It suggested that deletion of Tak1 aggravates the craniofacial deformities of the caBmpr1a mutants by increasing p53 and phospho-p38 at different stage of embryogenesis.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Morte Celular/genética , Anormalidades Craniofaciais/genética , Estudos de Associação Genética , Genótipo , MAP Quinase Quinase Quinases/genética , Mutação , Animais , Apoptose/genética , Biomarcadores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Anormalidades Craniofaciais/diagnóstico , Idade Gestacional , Imuno-Histoquímica , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Transdução de Sinais , Proteínas Smad/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Sci Rep ; 7: 44522, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28300208

RESUMO

Whole-organ regeneration has great potential for the replacement of dysfunctional organs through the reconstruction of a fully functional bioengineered organ using three-dimensional cell manipulation in vitro. Recently, many basic studies of whole-tooth replacement using three-dimensional cell manipulation have been conducted in a mouse model. Further evidence of the practical application to human medicine is required to demonstrate tooth restoration by reconstructing bioengineered tooth germ using a postnatal large-animal model. Herein, we demonstrate functional tooth restoration through the autologous transplantation of bioengineered tooth germ in a postnatal canine model. The bioengineered tooth, which was reconstructed using permanent tooth germ cells, erupted into the jawbone after autologous transplantation and achieved physiological function equivalent to that of a natural tooth. This study represents a substantial advancement in whole-organ replacement therapy through the transplantation of bioengineered organ germ as a practical model for future clinical regenerative medicine.


Assuntos
Medicina Regenerativa , Engenharia Tecidual , Germe de Dente/transplante , Dente/transplante , Animais , Engenharia Biomédica/tendências , Cães , Humanos , Odontogênese/fisiologia , Regeneração/fisiologia , Células-Tronco , Dente/crescimento & desenvolvimento , Erupção Dentária , Germe de Dente/fisiologia , Reimplante Dentário , Transplante Autólogo/métodos
8.
Bone ; 91: 20-9, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27373501

RESUMO

INTRODUCTION: The intercellular network of cell-cell communication among osteocytes is mediated by gap junctions. Gap junctional intercellular communication (GJIC) is thought to play an important role in the integration and synchronization of bone remodeling. To further understand the mechanism of bone development it is important to quantify the difference in the GJIC capacity of young and developmentally mature osteocytes. MATERIALS AND METHODS: We first established an embryonic chick calvaria growth model to show the growth of the calvaria in embryos at 13 to 21days of age. We then applied a fluorescence recovery after photobleaching (FRAP) technique to compare the difference in the GJIC capacity of young osteocytes with that of developmentally mature osteocytes. Finally, we quantified the dye (Calcein) diffusion from the FRAP data using a mathematic model of simple diffusion which was also used to identify simple diffusion GJIC pattern cells (fitted model) and accelerated diffusion GJIC pattern cells (non-fitted model). RESULTS: The relationship between the longest medial-lateral length of the calvaria (frontal bone) and the embryonic age fit a logarithmic growth model: length=5.144×ln(day)-11.340. The morphometric data during osteocyte differentiation showed that the cellular body becomes more spindle-shaped and that the cell body volume decreased by approximately 22% with an increase in the length of the processes between the cells. However, there were no significant differences in the cellular body surface area or in the distance between the mass centres of the cells. The dye-displacement rate in young osteocytes was significantly higher than that in developmentally mature osteocytes: dye displacement only occurred in 26.88% of the developmentally mature osteocytes, while it occurred in 64.38% of the young osteocytes. Additionally, in all recovered osteocytes, 36% of the developmentally mature osteocytes comprised non-fitted model cells while 53.19% of the young osteocytes were the non-fitted model, which indicates the active transduction of dye molecules. However, there were no statistically significant differences between the young and developmentally mature osteocytes with regard to the diffusion coefficient, permeability coefficient, or permeance of the osteocyte processes, which were 3.93±3.77 (×10(-8)cm(2)/s), 5.12±4.56 (×10(-5)cm(2)/s) and 2.99±2.47 (×10(-13)cm(2)/s) (mean±SD), respectively. CONCLUSIONS: These experiments comprehensively quantified the GJIC capacity in the embryonic chick calvaria and indicated that the cell-cell communication capacity of the osteocytes in the embryonic chick calvaria was related to their development.


Assuntos
Comunicação Celular , Diferenciação Celular , Junções Comunicantes/metabolismo , Osteócitos/citologia , Osteócitos/metabolismo , Crânio/citologia , Crânio/embriologia , Animais , Permeabilidade da Membrana Celular , Embrião de Galinha , Recuperação de Fluorescência Após Fotodegradação , Modelos Biológicos
9.
Cytokine Growth Factor Rev ; 27: 129-39, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26747371

RESUMO

BMP signaling is one of the key pathways regulating craniofacial development. It is involved in the early patterning of the head, the development of cranial neural crest cells, and facial patterning. It regulates development of its mineralized structures, such as cranial bones, maxilla, mandible, palate, and teeth. Targeted mutations in the mouse have been instrumental to delineate the functional involvement of this signaling network in different aspects of craniofacial development. Gene polymorphisms and mutations in BMP pathway genes have been associated with various non-syndromic and syndromic human craniofacial malformations. The identification of intricate cellular interactions and underlying molecular pathways illustrate the importance of local fine-regulation of Bmp signaling to control proliferation, apoptosis, epithelial-mesenchymal interactions, and stem/progenitor differentiation during craniofacial development. Thus, BMP signaling contributes both to shape and functionality of our facial features. BMP signaling also regulates postnatal craniofacial growth and is associated with dental structures life-long. A more detailed understanding of BMP function in growth, homeostasis, and repair of postnatal craniofacial tissues will contribute to our ability to rationally manipulate this signaling network in the context of tissue engineering.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Anormalidades Craniofaciais/embriologia , Transição Epitelial-Mesenquimal , Crista Neural/embriologia , Transdução de Sinais , Animais , Anormalidades Craniofaciais/patologia , Face/anormalidades , Face/embriologia , Face/patologia , Humanos , Camundongos , Crista Neural/patologia
10.
PLoS One ; 10(3): e0121938, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25812134

RESUMO

We herein describe a novel procedure for dentin regeneration that mimics the biological processes of tooth development in nature. The canonical Wnt signaling pathway is an important regulator of the Dentin sialophosphoprotein (Dspp) expression. Our approach mimics the biological processes underlying tooth development in nature and focuses on the activation of canonical Wnt signaling to trigger the natural process of dentinogenesis. The coronal portion of the dentin and the underlying pulp was removed from the first molars. We applied lithium chloride (LiCl), an activator of canonical Wnt signaling, on the amputated pulp surface to achieve transdifferentiation toward odontoblasts from the surrounding pulpal cells. MicroCT and microscopic analyses demonstrated that the topical application of LiCl induced dentin repair, including the formation of a complete dentin bridge. LiCl-induced dentin is a tubular dentin in which the pulp cells are not embedded within the matrix, as in primary dentin. In contrast, a dentin bridge was not induced in the control group treated with pulp capping with material carriers alone, although osteodentin without tubular formation was induced at a comparatively deeper position from the pulp exposure site. We also evaluated the influence of LiCl on differentiation toward odontoblasts in vitro. In the mDP odontoblast cell line, LiCl activated the mRNA expression of Dspp, Axin2 and Kallikrein 4 (Klk4) and downregulated the Osteopontin (Osp) expression. These results provide a scientific basis for the biomimetic regeneration of dentin using LiCl as a new capping material to activate dentine regeneration.


Assuntos
Polpa Dentária/efeitos dos fármacos , Polpa Dentária/fisiologia , Dentina/fisiologia , Dentinogênese/efeitos dos fármacos , Cloreto de Lítio/administração & dosagem , Administração Tópica , Animais , Polpa Dentária/diagnóstico por imagem , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Expressão Gênica , Masculino , Camundongos , Modelos Animais , Dente Molar/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ratos , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Microtomografia por Raio-X
11.
Development ; 142(7): 1357-67, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25742798

RESUMO

Bone morphogenetic protein (BMP) signaling plays many roles in skull morphogenesis. We have previously reported that enhanced BMP signaling through the BMP type IA receptor (BMPR1A) in cranial neural crest cells causes craniosynostosis during postnatal development. Additionally, we observed that 55% of Bmpr1a mutant mice show neonatal lethality characterized by a distended gastrointestinal tract. Here, we show that severely affected mutants exhibit defective nasal cartilage, failure of fusion between the nasal septum and the secondary palate, and higher levels of phosphorylated SMAD1 and SMAD5 in the nasal tissue. TUNEL demonstrated an increase in apoptosis in both condensing mesenchymal tissues and cartilage of the nasal region in mutants. The levels of p53 (TRP53) tumor suppressor protein were also increased in the same tissue. Injection of pifithrin-α, a chemical inhibitor of p53, into pregnant mice prevented neonatal lethality while concomitantly reducing apoptosis in nasal cartilage primordia, suggesting that enhanced BMP signaling induces p53-mediated apoptosis in the nasal cartilage. The expression of Bax and caspase 3, downstream targets of p53, was increased in the mutants; however, the p53 expression level was unchanged. It has been reported that MDM2 interacts with p53 to promote degradation. We found that the amount of MDM2-p53 complex was decreased in all mutants, and the most severely affected mutants had the largest decrease. Our previous finding that the BMP signaling component SMAD1 prevents MDM2-mediated p53 degradation coupled with our new data indicate that augmented BMP signaling induces p53-mediated apoptosis by prevention of p53 degradation in developing nasal cartilage. Thus, an appropriate level of BMP signaling is required for proper craniofacial morphogenesis.


Assuntos
Apoptose , Proteínas Morfogenéticas Ósseas/metabolismo , Morfogênese , Cartilagens Nasais/embriologia , Crista Neural/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Benzotiazóis/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Fatores de Crescimento de Fibroblastos/metabolismo , Integrases/metabolismo , Mesoderma/efeitos dos fármacos , Mesoderma/embriologia , Mesoderma/patologia , Camundongos , Morfogênese/efeitos dos fármacos , Mutação/genética , Cartilagens Nasais/anormalidades , Cartilagens Nasais/metabolismo , Cartilagens Nasais/patologia , Mucosa Nasal/metabolismo , Crista Neural/efeitos dos fármacos , Crista Neural/embriologia , Nariz/embriologia , Ligação Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Tolueno/análogos & derivados , Tolueno/farmacologia
12.
Dev Biol ; 400(2): 202-9, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25722188

RESUMO

BMP signaling mediated by ACVR1 plays a critical role for development of multiple structures including the cardiovascular and skeletal systems. While deficient ACVR1 signaling impairs normal embryonic development, hyperactive ACVR1 function (R206H in humans and Q207D mutation in mice, ca-ACVR1) results in formation of heterotopic ossification (HO). We developed a mouse line, which conditionally expresses ca-ACVR1 with Nfatc1-Cre(+) transgene. Mutant mice developed ectopic cartilage and bone at the distal joints of the extremities including the interphalangeal joints and hind limb ankles as early as P4 in the absence of trauma or exogenous bone morphogenetic protein (BMP) administration. Micro-CT showed that even at later time points (up to P40), cartilage and bone development persisted at the affected joints most prominently in the ankle. Interestingly, this phenotype was not present in areas of bone outside of the joints - tibia are normal in mutants and littermate controls away from the ankle. These findings demonstrate that this model may allow for further studies of heterotopic ossification, which does not require the use of stem cells, direct trauma or activation with exogenous Cre gene administration.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Modelos Animais de Doenças , Ossificação Heterotópica/genética , Transdução de Sinais , Receptores de Ativinas Tipo I/genética , Animais , Camundongos , Mutação , Fatores de Transcrição NFATC , Osteoblastos/metabolismo , Osteogênese
13.
Dev Dyn ; 244(3): 488-96, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25410786

RESUMO

BACKGROUND: The rodent salivary gland is not fully developed at birth and the cellular definitive differentiation takes place postnatally. However, little is known about its molecular mechanism. RESULTS: Here we provide the loss-of-function genetic evidence that Runx signaling affects postnatal development of the submandibular gland (SMG). Core binding factor ß (Cbfb) is a cotranscription factor which forms a heterodimer with Runx proteins. Cbfb was specifically expressed in the duct epithelium, specifically in the SMG. Epithelial Cbfb deficiency resulted in decrease in the size of the SMG and in the saliva secretion on postnatal day 35. The Cbfb mutant SMG specifically exhibited involution of the granular convoluted tubules (GCT), with a down-regulated expression of its marker genes, such as Klk1, Ngf, and Egf. The induction of GCT is under the control of androgens, and the Cbfb mutant SMG demonstrated down-regulated expression of Crisp3, an androgen-dependent transcript. Because the circulating testosterone or tissue dihydrotestosterone levels were not affected in the Cbfb mutants, it appears that Runx/Cbfb signaling regulate androgen receptor pathway, but does not affect the circulating testosterone levels or the enzymatic conversion to DHT. CONCLUSIONS: Runx signaling is important in the postnatal development of androgen-dependent GCT in the SMG.


Assuntos
Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/metabolismo , Transdução de Sinais/fisiologia , Glândula Submandibular/embriologia , Animais , Subunidades alfa de Fatores de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/genética , Fator de Crescimento Epidérmico/biossíntese , Fator de Crescimento Epidérmico/genética , Camundongos , Camundongos Transgênicos , Mutação , Fator de Crescimento Neural/biossíntese , Fator de Crescimento Neural/genética , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Proteínas e Peptídeos Salivares/biossíntese , Proteínas e Peptídeos Salivares/genética , Proteínas de Plasma Seminal/biossíntese , Proteínas de Plasma Seminal/genética , Glândula Submandibular/citologia
14.
Bone ; 53(1): 204-15, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23246671

RESUMO

Bone cells respond to mechanical stimuli by producing a variety of biological signals, and one of the earliest events is intracellular calcium ([Ca(2+)](i)) mobilization. Our recently developed ex vivo live [Ca(2+)](i) imaging system revealed that bone cells in intact bone explants showed autonomous [Ca(2+)](i) oscillations, and osteocytes specifically modulated these oscillations through gap junctions. However, the behavior and connectivity of the [Ca(2+)](i) signaling networks in mechanotransduction have not been investigated in intact bone. We herein introduce a novel fluid-flow platform for probing cellular signaling networks in live intact bone, which allows the application of capillary-driven flow just on the bone explant surface while performing real-time fluorogenic monitoring of the [Ca(2+)](i) changes. In response to the flow, the percentage of responsive cells was increased in both osteoblasts and osteocytes, together with upregulation of c-fos expression in the explants. However, enhancement of the peak relative fluorescence intensity was not evident. Treatment with 18 α-GA, a reversible inhibitor of gap junction, significantly blocked the [Ca(2+)](i) responsiveness in osteocytes without exerting any major effect in osteoblasts. On the contrary, such treatment significantly decreased the flow-activated oscillatory response frequency in both osteoblasts and osteocytes. The stretch-activated membrane channel, when blocked by Gd(3+), is less affected in the flow-induced [Ca(2+)](i) response. These findings indicated that flow-induced mechanical stimuli accompanied the activation of the autonomous [Ca(2+)](i) oscillations in both osteoblasts and osteocytes via gap junction-mediated cell-cell communication and hemichannel. Although how the bone sense the mechanical stimuli in vivo still needs to be elucidated, the present study suggests that cell-cell signaling via augmented gap junction and hemichannel-mediated [Ca(2+)](i) mobilization could be involved as an early signaling event in mechanotransduction.


Assuntos
Osso e Ossos/metabolismo , Sinalização do Cálcio , Estresse Psicológico , Animais , Sequência de Bases , Embrião de Galinha , Primers do DNA , Genes fos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Biol Chem ; 287(15): 12217-29, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22351753

RESUMO

Cell surface heparan sulfate (HS) is an essential regulator of cell signaling and development. HS traps signaling molecules, like Wnt in the glycosaminoglycan side chains of HS proteoglycans (HSPGs), and regulates their functions. Endosulfatases Sulf1 and Sulf2 are secreted at the cell surface to selectively remove 6-O-sulfate groups from HSPGs, thereby modifying the affinity of cell surface HSPGs for its ligands. This study provides molecular evidence for the functional roles of HSPG sulfation and desulfation in dentinogenesis. We show that odontogenic cells are highly sulfated on the cell surface and become desulfated during their differentiation to odontoblasts, which produce tooth dentin. Sulf1/Sulf2 double null mutant mice exhibit a thin dentin matrix and short roots combined with reduced expression of dentin sialophosphoprotein (Dspp) mRNA, encoding a dentin-specific extracellular matrix precursor protein, whereas single Sulf mutants do not show such defective phenotypes. In odontoblast cell lines, Dspp mRNA expression is potentiated by the activation of the Wnt canonical signaling pathway. In addition, pharmacological interference with HS sulfation promotes Dspp mRNA expression through activation of Wnt signaling. On the contrary, the silencing of Sulf suppresses the Wnt signaling pathway and subsequently Dspp mRNA expression. We also show that Wnt10a protein binds to cell surface HSPGs in odontoblasts, and interference with HS sulfation decreases the binding affinity of Wnt10a for HSPGs, which facilitates the binding of Wnt10a to its receptor and potentiates the Wnt signaling pathway, thereby up-regulating Dspp mRNA expression. These results demonstrate that Sulf-mediated desulfation of cellular HSPGs is an important modification that is critical for the activation of the Wnt signaling in odontoblasts and for production of the dentin matrix.


Assuntos
Dentinogênese , Heparitina Sulfato/fisiologia , Animais , Células Cultivadas , Dentina/crescimento & desenvolvimento , Dentina/metabolismo , Dentina/patologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteoglicanas de Heparan Sulfato/metabolismo , Heparitina Sulfato/metabolismo , Camundongos , Camundongos Knockout , Dente Molar/crescimento & desenvolvimento , Dente Molar/metabolismo , Dente Molar/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Odontoblastos/metabolismo , Fenótipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Sulfatases/genética , Sulfatases/metabolismo , Sulfotransferases/genética , Sulfotransferases/metabolismo , Anormalidades Dentárias/enzimologia , Anormalidades Dentárias/genética , Transcrição Gênica , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas Wnt/fisiologia , Via de Sinalização Wnt
16.
Stem Cells ; 29(11): 1792-803, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21898689

RESUMO

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


Assuntos
Diferenciação Celular/fisiologia , Subunidade beta de Fator de Ligação ao Core/metabolismo , Incisivo/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Proliferação de Células , Subunidades alfa de Fatores de Ligação ao Core/genética , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/genética , Fator 10 de Crescimento de Fibroblastos/genética , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator 3 de Crescimento de Fibroblastos/genética , Fator 3 de Crescimento de Fibroblastos/metabolismo , Fator 9 de Crescimento de Fibroblastos/genética , Fator 9 de Crescimento de Fibroblastos/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Microtomografia por Raio-X
17.
Differentiation ; 79(2): 74-83, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19945209

RESUMO

The periodontal ligament (PDL) comprises adult stem cells, which are responsible for periodontal tissue regeneration. In the present study, we investigated the specific profile of the stem cells in the human PDL. Microscopic analysis demonstrated that PDL cells showed a fibroblastic appearance, forming flat and loose aggregates. PDL cells expressed embryonic stem cell-associated antigens (SSEA-1, SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, OCT4, NANOG, SOX2, and REX1, and alkaline phosphatase activity), as well as conventional mesenchymal stem cell markers. When PDL cells were cultured in the presence of all-trans-retinoic acid, the numbers of SSEA-3+ and SSEA-4+ PDL cells were significantly decreased, while that of SSEA-1+ was increased. SSEA-4+ PDL cells showed a greater telomere length and growth rate. SSEA-4+ PDL cells exhibited the potential to generate specialized cells derived from three embryonic germ layers: mesodermal (adipocytes, osteoblasts, and chondrocytes), ectodermal (neurons), and endodermal (hepatocytes) lineages. Our findings demonstrated that SSEA-4, a major antigen to distinguish human embryonic stem cells, could also be used to identify multipotent stem cells in the PDL. Hence, SSEA-4+ human PDL cells appear to be a promising source of stem cells for regenerative medicine.


Assuntos
Células-Tronco Multipotentes/citologia , Ligamento Periodontal/citologia , Antígenos Embrionários Estágio-Específicos/metabolismo , Adulto , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Humanos , Antígenos CD15/metabolismo , Células-Tronco Multipotentes/metabolismo , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA