Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Tipo de estudio
Intervalo de año de publicación
1.
J Periodontol ; 93(1): 146-155, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34021604

RESUMEN

BACKGROUND: The immune checkpoint programmed cell death 1 (PD-1): PD-1 ligand 1 (PD-L1) pathway plays a crucial role in maintaining immune tolerance and preventing tissue damages by excessive immune responses. PD-L1 is physiologically expressed and upregulated in keratinocytes (KCs) in the oral cavity. We here investigated the contribution of PD-L1 that was overexpressed in gingival basal KCs in a ligature-induced periodontitis model. METHODS: Wild-type (WT) BALB/c and K14/PD-L1 transgenic (tg) mice, in which PD-L1 was overexpressed in basal KCs under control of the keratin 14 promoter, were used. To induce periodontitis, a 9-0 silk ligature was placed around the upper right second molar, and lipopolysaccharide from Porphyromonas gingivalis was applied on the suture. Gingival tissues were collected on day 7, after which histological analyses were performed, including by hematoxylin and eosin and tartrate-resistant acid phosphate staining (TRAP) and quantitative PCR for proinflammatory cytokines and bone metabolism-related genes. Alveolar bone loss at 7 weeks after ligature placement was assessed by micro-computed tomography analysis. RESULTS: PD-L1 was overexpressed in the basal KCs of all gingival epithelia in K14/PD-L1tg mice. Early ligature-induced periodontal inflammation, as assessed based on histological changes, elevation of proinflammatory cytokine (IL-1ß, IL-6, TNF-α) expression, periodontal ligament degeneration, and osteoclastogenesis as assessed by Rankl and Opg expression and TRAP+ cells, was markedly impaired in K14/PD-L1tg mice. Alveolar bone resorption at a late time point was also clearly minimized in K14/PD-L1tg mice. CONCLUSION: Overexpression of PD-L1 in gingival basal keratinocytes in K14/PD-L1tg mice reduces periodontal inflammation and alveolar bone resorption in a ligature-induced periodontitis model.


Asunto(s)
Pérdida de Hueso Alveolar , Periodontitis , Pérdida de Hueso Alveolar/genética , Animales , Antígeno B7-H1 , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Periodontitis/metabolismo , Receptor de Muerte Celular Programada 1 , Microtomografía por Rayos X
2.
Front Physiol ; 11: 532, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32581832

RESUMEN

During palatogenesis, the palatal shelves first grow vertically on either side of the tongue before changing their direction of growth to horizontal. The extracellular matrix (ECM) plays an important role in these dynamic changes in palatal shelf morphology. Tenascin-C (TNC) is an ECM glycoprotein that shows unique expression in the posterior part of the palatal shelf, but little is known about the regulation of TNC expression. Since transforming growth factor-beta-3 (TGF-ß3) and sonic hedgehog (SHH) signaling are known to play important roles in palatogenesis, we investigated whether TGF-ß3 and SHH are involved in the regulation of TNC expression in the developing palate. TGF-ß3 increased the expression of TNC mRNA and protein in primary mouse embryonic palatal mesenchymal cells (MEPM) obtained from palatal mesenchyme dissected at embryonic day 13.5-14.0. Interestingly, immunohistochemistry experiments revealed that TNC expression was diminished in K14-cre;Tgfbr2 fl/fl mice that lack the TGF-ß type II receptor in palatal epithelial cells and exhibit cleft soft palate, whereas TNC expression was maintained in Wnt1-cre;Tgfbr2 fl/fl mice that lack the TGF-ß type II receptor in palatal mesenchymal cells and exhibit a complete cleft palate. SHH also increased the expression of TNC mRNA and protein in MEPM cells. However, although TGF-ß3 up-regulated TNC mRNA and protein expression in O9-1 cells (a cranial neural crest cell line), SHH did not. Furthermore, TGF-ß inhibited the expression of osteoblastic differentiation markers (osterix and alkaline phosphatase) and induced the expression of fibroblastic markers (fibronectin and periostin) in O9-1 cells, whereas SHH did not affect the expression of osteoblastic and fibroblastic markers in O9-1 cells. However, immunohistochemistry experiments showed that TNC expression was diminished in the posterior palatal shelves of Shh-/+ ;MFCS4 +/- mice, which have deficient SHH signaling in the posterior palatal epithelium. Taken together, our findings support the proposal that TGF-ß and SHH signaling in palatal epithelium co-ordinate the expression of TNC in the posterior palatal mesenchyme through a paracrine mechanism. This signal cascade may work in the later stage of palatogenesis when cranial neural crest cells have differentiated into fibroblast-like cells. The spatiotemporal regulation of ECM-related proteins by TGF-ß and SHH signaling may contribute not only to tissue construction but also to cell differentiation or determination along the anterior-posterior axis of the palatal shelves.

3.
Development ; 146(21)2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31719045

RESUMEN

The tongue is a highly specialised muscular organ with a complex anatomy required for normal function. We have utilised multiple genetic approaches to investigate local temporospatial requirements for sonic hedgehog (SHH) signalling during tongue development. Mice lacking a Shh cis-enhancer, MFCS4 (ShhMFCS4/-), with reduced SHH in dorsal tongue epithelium have perturbed lingual septum tendon formation and disrupted intrinsic muscle patterning, with these defects reproduced following global Shh deletion from E10.5 in pCag-CreERTM; Shhflox/flox embryos. SHH responsiveness was diminished in local cranial neural crest cell (CNCC) populations in both mutants, with SHH targeting these cells through the primary cilium. CNCC-specific deletion of orofaciodigital syndrome 1 (Ofd1), which encodes a ciliary protein, in Wnt1-Cre; Ofdfl/Y mice led to a complete loss of normal myotube arrangement and hypoglossia. In contrast, mesoderm-specific deletion of Ofd1 in Mesp1-Cre; Ofdfl/Y embryos resulted in normal intrinsic muscle arrangement. Collectively, these findings suggest key temporospatial requirements for local SHH signalling in tongue development (specifically, lingual tendon differentiation and intrinsic muscle patterning through signalling to CNCCs) and provide further mechanistic insight into the tongue anomalies seen in patients with disrupted hedgehog signalling.


Asunto(s)
Tipificación del Cuerpo , Proteínas Hedgehog/metabolismo , Cresta Neural/citología , Transducción de Señal , Lengua/embriología , Alelos , Animales , Proliferación Celular , Elementos de Facilitación Genéticos , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Heterocigoto , Ligandos , Mesodermo/metabolismo , Ratones , Morfogénesis/genética , Fenotipo , Proteínas/metabolismo , Tendones/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo , Proteína Wnt1/metabolismo
4.
Congenit Anom (Kyoto) ; 58(5): 158-166, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29322554

RESUMEN

The cranial base is a structure mainly formed through endochondral ossification and integrated into the craniofacial complex, which acts as an underlying platform for the developing brain. Foxc1 is an indispensable regulator during intramembranous and endochondral ossification. In this study, we found that the spontaneous loss of Foxc1 function in a mouse (congenital hydrocephalous), Foxc1ch/ch , demonstrated the anterior cranial base defects, including unossified presphenoid and lack of middle part of the basisphenoid bone. Hypoplastic presphenoid primordial cartilage (basal portion of the trabecular cartilage [bTB]) and a lack of the middle part of basisphenoid primordial cartilage (the hypophyseal cartilage) were consistently observed at earlier developmental stage. Foxc1 was expressed robustly and ubiquitously in undifferentiated mesenchyme of the cranial base-forming area in E11.0 wild-type fetuses. Once chondrogenesis commenced, the expression was downregulated and later limited to the perichondrium. Detection of transcripts of Collagen type2 A1 (Col2a1) revealed that both bTB and the anterior part of the hypophyseal cartilage developing anterior to the persistent epithelial stalk of the anterior lobe of the pituitary gland were suppressed in the Foxc1ch/ch . Proliferation activity of chondrocyte precursor cells was higher in the Foxc1ch/ch . Loss of Foxc1 function only in the neural crest cell lineage (Wnt1-cre;Foxc1ch/flox ) showed ossification of the posterior part of the hypophyseal cartilage derived from the mesoderm. These findings suggest that Foxc1 is an important regulator to further chondrogenesis and initiate the ossification of the presphenoid and basisphenoid bones.


Asunto(s)
Colágeno Tipo II/genética , Factores de Transcripción Forkhead/genética , Cresta Neural/crecimiento & desarrollo , Base del Cráneo/crecimiento & desarrollo , Animales , Condrocitos/metabolismo , Condrogénesis/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mesodermo/crecimiento & desarrollo , Ratones , Cresta Neural/metabolismo , Base del Cráneo/metabolismo
5.
Dev Dyn ; 244(5): 703-11, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25733312

RESUMEN

BACKGROUND: The brain vascular system arises from the perineural vascular plexus (PNVP) which sprouts radially into the neuroepithelium and subsequently branches off laterally to form a secondary plexus in the subventricular zone (SVZ), the subventricular vascular plexus (SVP). The process of SVP formation remains to be fully elucidated. We investigated the role of Foxc1 in early stage vascular formation in the ventral telencephalon. RESULTS: The Foxc1 loss of function mutant mouse, Foxc1(ch/ch) , showed enlarged telencephalon and hemorrhaging in the ventral telencephalon by embryonic day 11.0. The mutant demonstrated blood vessel dilation and aggregation of endothelial cells in the SVZ after the invasion of endothelial cells through the radial path, which lead to failure of SVP formation. During this early stage of vascular development, Foxc1 was expressed in endothelial cells and pericytes, as well as in cranial mesenchyme surrounding the neural tube. Correspondingly, abnormal deposition pattern of basement membrane proteins around the vessels and increased strong Vegfr2 staining dots were found in the aggregation sites. CONCLUSIONS: These observations reveal an essential role for Foxc1 in the early stage of vascular formation in the telencephalon.


Asunto(s)
Circulación Cerebrovascular/fisiología , Embrión de Mamíferos , Factores de Transcripción Forkhead/metabolismo , Telencéfalo , Animales , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/embriología , Factores de Transcripción Forkhead/genética , Ratones , Ratones Mutantes , Telencéfalo/irrigación sanguínea , Telencéfalo/embriología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
6.
Congenit Anom (Kyoto) ; 54(3): 172-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24417671

RESUMEN

The frontal and parietal bones form the major part of the calvarium and their primordia appear at the basolateral region of the head and grow apically. A spontaneous loss of Foxc1 function mutant mouse, congenital hydrocephalus (Foxc1(ch/ch)), results in congenital hydrocephalus accompanied by defects in the apical part of the skull vault. We found that during the initiation stage of apical growth of the frontal bone primordium in the Foxc1(ch/ch) mouse, the Runx2 expression domain extended only to the basal side and bone sialoprotein (Bsp) and N-cadherin expression domains appeared only in the basal region. Fluorescent dye (DiI) labeling of the frontal primordium by ex-utero surgery confirmed that apical extension of the frontal bone primordium of the mouse was severely retarded, while extension to the basal side underneath the brain was largely unaffected. Consistent with this observation, decreased cell proliferation activity was seen at the apical tip but not the basal tip of the frontal bone primordium as determined by double detection of Runx2 transcripts and BrdU incorporation. Furthermore, expression of the osteogenic-related genes Bmp4 and-7 was observed only in the basal part of the meninges during the initiation period of primordium growth. These results suggest that a loss of Foxc1 function affects skull bone formation of the apical region and that Bmp expression in the meninges might influence the growth of the calvarial bone primordium.


Asunto(s)
Factores de Transcripción Forkhead/genética , Hueso Frontal/embriología , Hidrocefalia/embriología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Proliferación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Hueso Frontal/anomalías , Hueso Frontal/patología , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hidrocefalia/genética , Hidrocefalia/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoblastos/fisiología , Osteogénesis
7.
Congenit Anom (Kyoto) ; 53(2): 83-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23751042

RESUMEN

Fibroblast growth factor (FGF) signaling is involved in skeletal development. Among total 22 FGFs, it is suggested that FGF18 functions in promotion of osteoblast differentiation. In order to elucidate the mechanism of FGF18-dependent acceleration of osteogenesis, we implanted rhFGF18 soaked beads over mouse fetal coronal sutures using ex-utero surgery. The coronal suture area comprises the peripheries of the developing frontal and parietal bones, separated by the sutural mesenchyme. rhFGF18 accelerated osteogenesis by promoting connection of the frontal and parietal bone domains, resulting in elimination of the sutural mesenchyme. Expression of Fgf receptors, Fgfr1, -2 and -3 involved in skeletal development, was maintained or upregulated in the developing bone domains, consistent with enhanced osteogenesis. Bone morphogenetic protein (Bmp) 2 was specifically upregulated in the skeletogenic layer and the application of Bmp antagonist, rmNoggin, inhibited rhFGF18-dependent upregulation of osteoblast markers. These results suggest that FGF18 accelerates osteogenesis by upregulation of Bmp2 as well as maintenance or upregulation of Fgfr1, -2 and -3 expression in osteoblasts.


Asunto(s)
Proteína Morfogenética Ósea 2/biosíntesis , Diferenciación Celular , Factores de Crecimiento de Fibroblastos/biosíntesis , Osteoblastos/citología , Animales , Desarrollo Óseo/genética , Proteína Morfogenética Ósea 2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Osteoblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Regulación hacia Arriba
8.
Birth Defects Res A Clin Mol Teratol ; 94(4): 215-22, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22434669

RESUMEN

The upper lip is formed by the fusion of facial processes, a process in which many genetic and environmental factors are involved. Embryonic hypoxia is induced by uterine anemia and the administration of vasoconstrictors during pregnancy. To define the relationship between hypoxia and upper lip formation, hypoxic conditions were created in a whole embryo culture system. Hypoxic embryos showed a high frequency of impaired fusion, reflecting failure in the growth of the lateral nasal process (LNP). In hypoxic embryos, cell proliferation activity in the LNP mesenchyme was decreased following downregulation of genes that are involved in lip formation. We also observed upregulation of vascular endothelial growth factor expression along with the induction of apoptosis in the LNP. These results suggest that embryonic hypoxia during lip formation induces apoptosis in physiologically hypoxic regions, hypoxia-induced gene expression and downregulation of the genes involved in maxillofacial morphogenesis as immediate responses, followed by reduction of mesenchymal cell proliferation activity, resulting in insufficient growth of the facial processes.


Asunto(s)
Labio Leporino/etiología , Hipoxia , Labio/embriología , Labio/fisiología , Animales , Apoptosis , Hipoxia de la Célula/fisiología , Proliferación Celular , Técnicas de Cultivo de Embriones , Cara/embriología , Cara/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Mesodermo , Ratones , Ratones Endogámicos C57BL , Embarazo
9.
Congenit Anom (Kyoto) ; 49(2): 77-84, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19489959

RESUMEN

Tenascin-W is a tenascin family member that forms part of a complex extracellular matrix, and previous studies have suggested its association with osteogenesis. In the present study we investigated the roles of tenascin-W in osteogenesis. We found that tenascin-W is expressed in osteoblasts at the edge of the developing bone domain prior to mineralization in mouse fetuses. Expression of tenascin-W was induced during the course of mineralization of the Kusa-A1 osteoblast cell line. In the interfrontal suture of postnatal mice, the anterior portion remains patent and the posterior portion closes by 4 weeks of age. Tenascin-W expression was downregulated at 1 week of age in the posterior frontal suture, whereas in the anterior suture, expression was maintained until the mice reached 4 weeks of age. Fibroblast growth factor 2 (FGF2)-bead application to the mouse fetal skull by ex-utero surgery accelerated osteoblast differentiation, but inhibited mineralization with a downregulation of tenascin-W expression. These results suggest that tenascin-W is involved in osteoblast maturation (i.e. mineralization).


Asunto(s)
Calcificación Fisiológica , Cráneo/embriología , Cráneo/metabolismo , Tenascina/metabolismo , Animales , Animales Recién Nacidos , Desarrollo Óseo , Diferenciación Celular , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis
10.
Artículo en Inglés | MEDLINE | ID: mdl-17336108

RESUMEN

OBJECTIVE: The object of this study was to elucidate the kinetics of receptor activator of NFkB ligand (RANKL), RANK, osteoprotegerin (OPG), and cytokine expressions in experimentally induced rat periapical lesions. STUDY DESIGN: The mRNA expressions of RANKL, RANK, OPG, and cytokines in experimentally induced rat periapical lesions were evaluated by real-time PCR. The lesions were induced in male Wistar rats (n = 48, 5 weeks of age) by unsealed pulp exposure of the lower first molars. RESULTS: Expression of RANKL was up-regulated at the beginning of lesion expansion, and expression ratio of RANKL against OPG, a competitor of RANKL, peaked at 2 and 3 weeks. Expression of inflammatory cytokines, such as TNF-alpha, IL-1alpha, and IL-1beta also increased at this stage, suggesting contribution of synergic effects of RANKL and proinflammatory cytokine signaling to lesion expansion. Most of RANKL+ cells were fibroblastic, but few of them were T cells. CONCLUSION: Expression of RANKL and proinflammatory cytokines was correlated with periapical lesion expansion.


Asunto(s)
Citocinas/biosíntesis , Osteoprotegerina/biosíntesis , Periodontitis Periapical/metabolismo , Ligando RANK/biosíntesis , Receptor Activador del Factor Nuclear kappa-B/biosíntesis , Fosfatasa Ácida/metabolismo , Animales , Técnicas para Inmunoenzimas , Interleucina-10/biosíntesis , Interleucina-1alfa/biosíntesis , Interleucina-1beta/biosíntesis , Isoenzimas/metabolismo , Cinética , Masculino , Periodontitis Periapical/patología , Ratas , Ratas Wistar , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fosfatasa Ácida Tartratorresistente , Factor de Necrosis Tumoral alfa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...