Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Lasers Surg Med ; 56(5): 496-507, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38650443

RESUMEN

OBJECTIVES: The aim of this work is to assess the performance of multimodal spectroscopic approach combined with single core optical fiber for detection of bladder cancer during surgery in vivo. METHODS: Multimodal approach combines diffuse reflectance spectroscopy (DRS), fluorescence spectroscopy in the visible (405 nm excitation) and near-infrared (NIR) (690 nm excitation) ranges, and high-wavenumber Raman spectroscopy. All four spectroscopic methods were combined in a single setup. For 21 patients with suspected bladder cancer or during control cystoscopy optical spectra of bladder cancer, healthy bladder wall tissue and/or scars were measured. Classification of cancerous and healthy bladder tissue was performed using machine learning methods. RESULTS: Statistically significant differences in relative total haemoglobin content, oxygenation, scattering, and visible fluorescence intensity were found between tumor and normal tissues. The combination of DRS and visible fluorescence spectroscopy allowed detecting cancerous tissue with sensitivity and specificity of 78% and 91%, respectively. The addition of features extracted from NIR fluorescence and Raman spectra did not improve the quality of classification. CONCLUSIONS: This study demonstrates that multimodal spectroscopic approach allows increasing sensitivity and specificity of bladder cancer detection in vivo. The developed approach does not require special probes and can be used with single-core optical fibers applied for laser surgery.


Asunto(s)
Espectrometría de Fluorescencia , Espectrometría Raman , Neoplasias de la Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/patología , Humanos , Espectrometría Raman/métodos , Femenino , Espectrometría de Fluorescencia/métodos , Masculino , Anciano , Persona de Mediana Edad , Sensibilidad y Especificidad , Cistoscopía , Anciano de 80 o más Años , Espectroscopía Infrarroja Corta/métodos
2.
BJU Int ; 126(1): 159-167, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32277557

RESUMEN

OBJECTIVE: To investigate the effects of laser temporal pulse shaping of the super-pulse thulium fibre laser (SPTFL) and to compare these in controlled in vitro conditions with various holmium: yttrium aluminium garnet (Ho:YAG) pulse delivery modes. MATERIALS AND METHODS: The SPTFL (Urolase SP, IRE-Polus, Fryazino, Russia), with an emission wavelength of 1.94 µm, and a Ho:YAG laser (P120H; Lumenis, Yokneam, Israel) with Moses technology were compared. Pulse shape, stone retropulsion and ablation efficiency were evaluated using BegoStones and compared for each laser mode: short (SP), long (LP), and Moses pulse (MP) for Ho:YAG, regular pulse (RP) and dual pulse (DP) for SPTFL. RESULTS: The Ho:YAG SP mode exhibited an asymmetrical pulse shape, with a steep leading slope and a much more gradual trailing slope, without any flat section. Pulses generated by the SPTFL were significantly longer and therefore had lower peak power than those generated by the Ho:YAG laser at equivalent energy settings. Retropulsion for the holmium:YAG LP and MP modes was similar and lower than that for the SP mode, but higher than for the SPTFL (all P ≤ 0.02), with an average stone displacement approximately four times and two times lower for SPTFL as compared to the Ho:YAG laser. Comparison of ablation volumes indicated that the SPTFL induced significantly higher (twofold) ablation than the Ho:YAG laser. CONCLUSIONS: The magnitude and initial velocity of stone retropulsion decreased with longer pulse duration and lower pulse peak power, without sacrificing ablation efficiency. These observations are manifest when comparing the Ho:YAG laser with the SPTFL. The novel SPTFL provides greater versatility and control of pulse variables than the Ho:YAG laser. Further clinical investigation of practical benefits achievable with pulse-shaping SPTFL modes is warranted.


Asunto(s)
Terapia por Láser/métodos , Láseres de Estado Sólido/uso terapéutico , Litotripsia por Láser/métodos , Cálculos Urinarios/cirugía , Holmio , Humanos , Tulio
3.
World J Urol ; 38(2): 497-503, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31055626

RESUMEN

PURPOSE: A superpulse (500 W peak power) thulium fiber laser operating at a 1940 nm wavelength, suitable for lithotripsy, has recently been developed. The goal of this study was to compare stone fragmentation and dusting performance of the prototype superpulse thulium fiber laser with leading commercially available, high-power holmium:YAG lithotripters (wavelength 2100 nm) in a controlled in vitro environment. METHODS: Two experimental setups were designed for investigating stone ablation rates and retropulsion effects, respectively. In addition, the ablation setup enabled water temperature measurements during stone fragmentation in the laser-stone interaction zone. Human uric acid (UA) and calcium oxalate monohydrate (COM) stones were used for ablation experiments, whereas standard BegoStone phantoms were utilized in retropulsion experiments. The laser settings were matched in terms of pulse energy, pulse repetition rate, and average power. RESULTS: At equivalent settings, thulium fiber laser ablation rates were higher than those for holmium:YAG laser in both dusting mode (threefold for COM stones and 2.5-fold for UA stones) and fragmentation mode (twofold for UA stones). For single-pulse retropulsion experiments, the threshold for onset of stone retropulsion was two to four times higher for thulium fiber laser. The holmium:YAG laser generated significantly stronger retropulsion effects at equal pulse energies. The water temperature elevation near the laser-illuminated volume did not differ between the two lasers. CONCLUSIONS: Distinctive features of the thulium fiber laser (optimal wavelength and long pulse duration) resulted in faster stone ablation and lower retropulsion in comparison to the holmium:YAG laser.


Asunto(s)
Láseres de Estado Sólido/uso terapéutico , Litotripsia por Láser/métodos , Fantasmas de Imagen , Cálculos Urinarios/terapia , Diseño de Equipo , Holmio , Humanos , Tulio
4.
Dev Biol ; 444 Suppl 1: S297-S307, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29571612

RESUMEN

Mutations that disrupt the inwardly rectifying potassium channel Kir2.1 lead to Andersen-Tawil syndrome that includes periodic paralysis, cardiac arrhythmia, cognitive deficits, craniofacial dysmorphologies and limb defects. The molecular mechanism that underlies the developmental consequences of inhibition of these channels has remained a mystery. We show that while loss of Kir2.1 function does not affect expression of several early facial patterning genes, the domain in which Pou3f3 is expressed in the maxillary arch is reduced. Pou3f3 is important for development of the jugal and squamosal bones. The reduced expression domain of Pou3f3 is consistent with the reduction in the size of the squamosal and jugal bones in Kcnj2KO/KO animals, however it does not account for the diverse craniofacial defects observed in Kcnj2KO/KO animals. We show that Kir2.1 function is required in the cranial neural crest for morphogenesis of several craniofacial structures including palate closure. We find that while the palatal shelves of Kir2.1-null embryos elevate properly, they are reduced in size due to decreased proliferation of the palatal mesenchyme. While we find no reduction in expression of BMP ligands, receptors, and associated Smads in this setting, loss of Kir2.1 reduces the efficacy of BMP signaling as shown by the reduction of phosphorylated Smad 1/5/8 and reduced expression of BMP targets Smad6 and Satb2.


Asunto(s)
Cara/embriología , Canales de Potasio de Rectificación Interna/fisiología , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/fisiología , Anomalías Craneofaciales/embriología , Regulación de la Expresión Génica/genética , Ratones , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/fisiología , Cresta Neural/metabolismo , Cresta Neural/fisiología , Factores del Dominio POU/fisiología , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Transducción de Señal , Cráneo/embriología , Factores de Transcripción/metabolismo
5.
PLoS Genet ; 10(1): e1004074, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24497835

RESUMEN

During vertebrate craniofacial development, neural crest cells (NCCs) contribute to most of the craniofacial pharyngeal skeleton. Defects in NCC specification, migration and differentiation resulting in malformations in the craniofacial complex are associated with human craniofacial disorders including Treacher-Collins Syndrome, caused by mutations in TCOF1. It has been hypothesized that perturbed ribosome biogenesis and resulting p53 mediated neuroepithelial apoptosis results in NCC hypoplasia in mouse Tcof1 mutants. However, the underlying mechanisms linking ribosome biogenesis and NCC development remain poorly understood. Here we report a new zebrafish mutant, fantome (fan), which harbors a point mutation and predicted premature stop codon in zebrafish wdr43, the ortholog to yeast UTP5. Although wdr43 mRNA is widely expressed during early zebrafish development, and its deficiency triggers early neural, eye, heart and pharyngeal arch defects, later defects appear fairly restricted to NCC derived craniofacial cartilages. Here we show that the C-terminus of Wdr43, which is absent in fan mutant protein, is both necessary and sufficient to mediate its nucleolar localization and protein interactions in metazoans. We demonstrate that Wdr43 functions in ribosome biogenesis, and that defects observed in fan mutants are mediated by a p53 dependent pathway. Finally, we show that proper localization of a variety of nucleolar proteins, including TCOF1, is dependent on that of WDR43. Together, our findings provide new insight into roles for Wdr43 in development, ribosome biogenesis, and also ribosomopathy-induced craniofacial phenotypes including Treacher-Collins Syndrome.


Asunto(s)
Disostosis Mandibulofacial/genética , Cresta Neural/crecimiento & desarrollo , Proteínas Nucleares/genética , Ribosomas/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Apoptosis/genética , Cartílago/crecimiento & desarrollo , Cartílago/metabolismo , Diferenciación Celular/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular , Disostosis Mandibulofacial/etiología , Disostosis Mandibulofacial/patología , Ratones , Cresta Neural/citología , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Mapas de Interacción de Proteínas/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/metabolismo
6.
J Physiol ; 594(12): 3245-70, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-26864374

RESUMEN

KEY POINTS: Xenopus laevis craniofacial development is a good system for the study of Andersen-Tawil Syndrome (ATS)-associated craniofacial anomalies (CFAs) because (1) Kcnj2 is expressed in the nascent face; (2) molecular-genetic and biophysical techniques are available for the study of ion-dependent signalling during craniofacial morphogenesis; (3) as in humans, expression of variant Kcnj2 forms in embryos causes a muscle phenotype; and (4) variant forms of Kcnj2 found in human patients, when injected into frog embryos, cause CFAs in the same cell lineages. Forced expression of WT or variant Kcnj2 changes the normal pattern of Vmem (resting potential) regionalization found in the ectoderm of neurulating embryos, and changes the normal pattern of expression of ten different genetic regulators of craniofacial development, including markers of cranial neural crest and of placodes. Expression of other potassium channels and two different light-activated channels, all of which have an effect on Vmem , causes CFAs like those induced by injection of Kcnj2 variants. In contrast, expression of Slc9A (NHE3), an electroneutral ion channel, and of GlyR, an inactive Cl(-) channel, do not cause CFAs, demonstrating that correct craniofacial development depends on a pattern of bioelectric states, not on ion- or channel-specific signalling. Using optogenetics to control both the location and the timing of ion flux in developing embryos, we show that affecting Vmem of the ectoderm and no other cell layers is sufficient to cause CFAs, but only during early neurula stages. Changes in Vmem induced late in neurulation do not affect craniofacial development. We interpret these data as strong evidence, consistent with our hypothesis, that ATS-associated CFAs are caused by the effect of variant Kcnj2 on the Vmem of ectodermal cells of the developing face. We predict that the critical time is early during neurulation, and the critical cells are the ectodermal cranial neural crest and placode lineages. This points to the potential utility of extant, ion flux-modifying drugs as treatments to prevent CFAs associated with channelopathies such as ATS. ABSTRACT: Variants in potassium channel KCNJ2 cause Andersen-Tawil Syndrome (ATS); the induced craniofacial anomalies (CFAs) are entirely unexplained. We show that KCNJ2 is expressed in Xenopus and mouse during the earliest stages of craniofacial development. Misexpression in Xenopus of KCNJ2 carrying ATS-associated mutations causes CFAs in the same structures affected in humans, changes the normal pattern of membrane voltage potential regionalization in the developing face and disrupts expression of important craniofacial patterning genes, revealing the endogenous control of craniofacial patterning by bioelectric cell states. By altering cells' resting potentials using other ion translocators, we show that a change in ectodermal voltage, not tied to a specific protein or ion, is sufficient to cause CFAs. By adapting optogenetics for use in non-neural cells in embryos, we show that developmentally patterned K(+) flux is required for correct regionalization of the resting potentials and for establishment of endogenous early gene expression domains in the anterior ectoderm, and that variants in KCNJ2 disrupt this regionalization, leading to the CFAs seen in ATS patients.


Asunto(s)
Síndrome de Andersen/genética , Anomalías Craneofaciales/genética , Canales de Potasio de Rectificación Interna/genética , Animales , Embrión de Mamíferos , Larva , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/anomalías , Optogenética , ARN Mensajero/genética , Xenopus laevis
7.
Genesis ; 49(4): 360-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21225658

RESUMEN

Zebrafish craniofacial, skeletal, and tooth development closely resembles that of higher vertebrates. Our goal is to identify viable adult zebrafish mutants that can be used as models for human mineralized craniofacial, dental, and skeletal system disorders. We used a large-scale forward-genetic chemical N-ethyl-nitroso-urea mutagenesis screen to identify 17 early lethal homozygous recessive mutants with defects in craniofacial cartilage elements, and 7 adult homozygous recessive mutants with mineralized tissue phenotypes including craniofacial shape defects, fused sutures, dysmorphic or missing skeletal elements, scoliosis, and neural arch defects. One mutant displayed both an early lethal homozygous phenotype and an adult heterozygous phenotype. These results extend the utility of the zebrafish model beyond the embryo to study human bone and cartilage disorders.


Asunto(s)
Cartílago/anomalías , Anomalías Craneofaciales/genética , Modelos Animales de Enfermedad , Fenotipo , Azul Alcián , Animales , Antraquinonas , Etilnitrosourea , Genes Recesivos/genética , Mutagénesis , Pez Cebra
8.
J Huntingtons Dis ; 10(3): 405-412, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34397420

RESUMEN

HDinHD (Huntington's Disease in High Definition; HDinHD.org) is an open online portal for the HD research community that presents a synthesized view of HD-related scientific data. Here, we present a broad overview of HDinHD and highlight the newly launched HDinHD Explorer tool that enables researchers to discover and explore a wide range of diverse yet interconnected HD-related data. We demonstrate the utility of HDinHD Explorer through data mining of a single collection of newly released in vivo therapeutic intervention study reports alongside previously published reports.


Asunto(s)
Enfermedad de Huntington , Humanos , Enfermedad de Huntington/genética
9.
BMC Dev Biol ; 10: 48, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20459789

RESUMEN

BACKGROUND: Growth factors and their receptors are mediators of organogenesis and must be tightly regulated in a temporal and spatial manner for proper tissue morphogenesis. Intracellular regulators of growth factor signaling pathways provide an additional level of control. Members of the Sprouty family negatively regulate receptor tyrosine kinase pathways in several developmental contexts. To gain insight into the role of Spry1 in neural crest development, we analyzed the developmental effects of conditional expression of Spry1 in neural crest-derived tissues. RESULTS: Here we report that conditional expression of Spry1 in neural crest cells causes defects in craniofacial and cardiac development in mice. Spry1;Wnt1-Cre embryos die perinatally and exhibit facial clefting, cleft palate, cardiac and cranial nerve defects. These defects appear to be the result of decreased proliferation and increased apoptosis of neural crest and neural crest-derived cell populations. In addition, the domains of expression of several key transcription factors important to normal craniofacial and cardiac development including AP2, Msx2, Dlx5, and Dlx6 were reduced in Spry1;Wnt1-Cre transgenic embryos. CONCLUSION: Collectively, these data suggest that Spry1 is an important regulator of craniofacial and cardiac morphogenesis and perturbations in Spry1 levels may contribute to congenital disorders involving tissues of neural crest origin.


Asunto(s)
Anomalías Craneofaciales/embriología , Cardiopatías Congénitas/embriología , Proteínas de la Membrana/metabolismo , Cresta Neural/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Apoptosis , Proliferación Celular , Técnicas de Silenciamiento del Gen , Corazón , Ratones
10.
Dev Biol ; 318(2): 323-34, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18471809

RESUMEN

Haploinsufficiency of the transcription factor TWIST1 is associated with Saethre-Chotzen Syndrome and is manifested by craniosynostosis, which is the premature closure of the calvaria sutures. Previously, we found that Twist1 forms functional homodimers and heterodimers that have opposing activities. Our data supported a model that within the calvaria sutures Twist1 homodimers (T/T) reside in the osteogenic fronts while Twist1/E protein heterodimers (T/E) are in the mid-sutures. Twist1 haploinsufficiency alters the balance between these dimers, favoring an increase in homodimer formation throughout the sutures. The data we present here further supports this model and extends it to integrate the Twist1 dimers with the pathways that are known to regulate cranial suture patency. This data provides the first evidence of a functional link between Twist1 and the FGF pathway, and indicates that differential regulation of FGF signaling by T/T and T/E dimers plays a central role in governing cranial suture patency. Furthermore, we show that inhibition of FGF signaling prevents craniosynostosis in Twist1(+/-) mice, demonstrating that inhibition of a signaling pathway that is not part of the initiating mutation can prevent suture fusion in a relevant genetic model of craniosynostosis.


Asunto(s)
Suturas Craneales/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Craneosinostosis/metabolismo , Dimerización , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Osteoblastos/citología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
11.
Eur J Cell Biol ; 83(7): 327-35, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15503856

RESUMEN

Since little is known about the function of polypeptide growth factors as regulators of multiple cell cycles, we compared the ability of FGF1, PDGF-AB and serum to induce a second round of DNA synthesis in Swiss 3T3 cells previously exposed to either FGF1, PDGF-AB or serum during the first cell cycle using [14C]- and [3H]thymidine in a double labeling system to distinguish between the first and second cell cycles. Surprisingly, we observed that cells exposed to either FGF1 or PDGF-AB in the first cell cycle were unable to synthesize DNA in response to FGF1 or PDGF-AB in the second cell cycle; yet these cells responded well to serum as a second cycle mitogen. Interestingly, while cells exposed to either FGF1 or PDGF-AB in the second cycle displayed normal receptor-mediated signaling and expressed cyclin D and E, they, like senescent fibroblasts and endothelial cells, failed to express cyclin A, and the continuous exposure of cells to either FGF1 or PDGF-AB resulted in a decrease in the kinase activity of the cyclin E/cdk2 complex. In addition, an increased association of this complex was observed with p21 CIP in an FGF1-dependent manner as well as with p27 KIP in a PDGF-AB-dependent manner. Lastly, the downregulation of p21 expression using an antisense strategy was able to partially rescue the replicative response of Swiss 3T3 cells to FGF1 in the second cycle. These data suggest that (i) FGF1 and PDGF-AB may limit their mitogenic effect to a single cell cycle, (ii) entry into the second round of replication is serum dependent and (iii) the self-limiting nature of FGF1 and PDGF-AB correlates with the accumulation of the cdk inhibitors, p21 and p27, respectively.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Ciclina E/biosíntesis , Ciclinas/biosíntesis , ADN/biosíntesis , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Quinasas CDC2-CDC28/biosíntesis , Bovinos , Proteínas de Ciclo Celular/biosíntesis , Medio de Cultivo Libre de Suero , Ciclina D , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ratones , Transducción de Señal/efectos de los fármacos , Células 3T3 Swiss , Proteínas Supresoras de Tumor/biosíntesis
12.
Gene Expr Patterns ; 12(3-4): 130-5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22300525

RESUMEN

Rb1 is a tumor suppressor gene that regulates cell cycle progression through interactions with E2F transcription factors. In recent years, new roles for Rb1 in regulating cellular differentiation have also emerged. For example, it has been shown that Rb1 regulates osteoblast differentiation in a cell cycle independent manner, by binding to the transcription factor Runx2, and facilitating the up-regulation of late bone differentiation markers. Based on the facts that Runx2 also functions in tooth development, and that little is known about potential roles for Rb1 in mammalian tooth development, here we evaluated the expression of Rb1 mRNA in developmentally staged mouse teeth. Our data show that Rb1 mRNA is expressed in both dental epithelial and dental mesenchymal progenitor cells. In addition, Rb1 mRNA appears upregulated in differentiating ameloblasts and odontoblasts, suggesting roles for Rb1 in tooth differentiation.


Asunto(s)
Proteína de Retinoblastoma/metabolismo , Diente/embriología , Ameloblastos/metabolismo , Animales , Diferenciación Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Odontoblastos/metabolismo , Odontogénesis/genética , Osteogénesis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína de Retinoblastoma/genética , Factores de Transcripción/genética , Regulación hacia Arriba
13.
J Mol Histol ; 43(3): 281-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22476877

RESUMEN

Retinoblastoma protein (pRb) phosphorylation plays a central role in mediating cell cycle G1/S stage transition, together with E2F transcription factors. The binding of pRb to E2F is thought to be controlled by the sequential and cumulative phosphorylation of pRb at various amino acids. In addition to well characterized roles as a tumor suppressor, pRb has more recently been implicated in osteoprogenitor and other types of stem cell maintenance, proliferation and differentiation, thereby influencing the morphogenesis of developing organs. In this study, we present data characterizing the expression of pRb and three phosphorylated pRb (ppRb) isoforms-ppRbS780, ppRbS795, ppRbS807/811-in developmentally staged mouse molar and incisor teeth. Our results reveal distinct developmental expression patterns for individual ppRb isoforms in dental epithelial and dental mesenchymal cell differentiation, suggesting discrete functions in tooth development.


Asunto(s)
Factores de Transcripción E2F/genética , Regulación del Desarrollo de la Expresión Génica , Incisivo/metabolismo , Diente Molar/metabolismo , Fosfoproteínas/genética , Proteína de Retinoblastoma/genética , Animales , Diferenciación Celular , Factores de Transcripción E2F/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Incisivo/citología , Incisivo/embriología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Diente Molar/citología , Diente Molar/embriología , Odontogénesis/genética , Fosfoproteínas/metabolismo , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína de Retinoblastoma/metabolismo , Factores de Tiempo
14.
Int J Dev Biol ; 55(6): 641-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21769775

RESUMEN

Evidence from various in vitro gain and loss of function studies indicate that the bHLH transcription factor Twist1 negatively regulates chondrocyte differentiation; however limited information regarding Twist1 function in postnatal cartilage development and maintenance is available. Twist1 expression within the postnatal growth plate is restricted to immature, proliferating chondrocytes, and is significantly decreased or absent in hypertrophic chondrocytes. In order to examine the effect of maintaining the expression of Twist1 at later stages of chondocyte differentiation, we used type II collagen Cre (Col2-Cre) mice to activate a Cre-inducible Twist1 transgene specifically in chondrocytes (Col2-Twist1). At two weeks, postnatal growth was inhibited in Col2-Twist1 mice, as evidenced by limb shortening. Histological examination revealed abnormal growth plate structure, characterized by poor columnar organization of proliferating cartilaginous cells, decreased cellularity, and expansion of the hypertrophic zone. Moreover, structural defects within the growth plates of Col2-Twist1 transgenic mice included abnormal vascular invasion and focal regions of bony formation. Quantitative analysis of endochondral bone formation via micro-computed topography revealed impaired trabecular bone formation in the hindlimbs of Col2-Twist1 transgenic mice at various timepoints of postnatal development. Taken together, these findings indicate that regulated Twist1 expression contributes to growth plate organization and endochondral ossification to modulate postnatal longitudinal bone growth.


Asunto(s)
Condrocitos/metabolismo , Enanismo/metabolismo , Placa de Crecimiento/anomalías , Proteínas Nucleares/biosíntesis , Proteína 1 Relacionada con Twist/biosíntesis , Animales , Desarrollo Óseo/genética , Diferenciación Celular , Condrogénesis , Colágeno Tipo II , Enanismo/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Genotipo , Placa de Crecimiento/crecimiento & desarrollo , Placa de Crecimiento/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Osteogénesis , Proteína 1 Relacionada con Twist/genética
15.
Methods Cell Biol ; 101: 225-48, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21550447

RESUMEN

The zebrafish has emerged as an important model for vertebrate development as it relates to human health and disease. Work in this system has provided significant insights into the variety of genetic signals that direct the cellular activities and tissue interactions necessary for proper assembly of the pharyngeal skeleton. Unfortunately our understanding of craniofacial development beyond embryonic stages is far less complete. Stated another way, we know a great deal about the early patterning of the skull, but we know comparatively little about how mature craniofacial shape is determined and maintained over time. Here we propose ways to expand the current molecular genetic paradigm beyond the embryo to gain an understanding of the processes and mechanisms that guide growth and remodeling of mineralized craniofacial, skeletal, and dental tissues. First, we discuss sources of adult mutant phenotypes that can be used to study of postembryonic development. Next, we review salient quantitative methods that are necessary to define complex adult phenotypes. We also discuss how other organismal systems can be used to inform and complement studies in zebrafish. We conclude by discussing the implications for such studies within the context of furthering an understanding of the etiology and pathophysiology of human craniofacial malformations, as well as informing an understanding of adaptive craniofacial variation among natural populations.


Asunto(s)
Maxilares/fisiología , Desarrollo Maxilofacial/fisiología , Pez Cebra/crecimiento & desarrollo , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Desarrollo Maxilofacial/genética , Morfogénesis/genética , Morfogénesis/fisiología , Pez Cebra/genética
16.
Dev Dyn ; 235(5): 1345-57, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16502419

RESUMEN

Saethre-Chotzen syndrome is associated with haploinsufficiency of the basic-helix-loop-helix (bHLH) transcription factor TWIST1 and is characterized by premature closure of the cranial sutures, termed craniosynostosis; however, the mechanisms underlying this defect are unclear. Twist1 has been shown to play both positive and negative roles in mesenchymal specification and differentiation, and here we show that the activity of Twist1 is dependent on its dimer partner. Twist1 forms both homodimers (T/T) and heterodimers with E2A E proteins (T/E) and the relative level of Twist1 to the HLH inhibitor Id proteins determines which dimer forms. On the basis of the expression patterns of Twist1 and Id1 within the cranial sutures, we hypothesized that Twist1 forms homodimers in the osteogenic fronts and T/E heterodimers in the mid-sutures. In support of this hypothesis, we have found that genes regulated by T/T homodimers, such as FGFR2 and periostin, are expressed in the osteogenic fronts, whereas genes regulated by T/E heterodimers, such as thrombospondin-1, are expressed in the mid-sutures. The ratio between these dimers is altered in the sutures of Twist1+/- mice, favoring an increase in homodimers and an expansion of the osteogenic fronts. Of interest, the T/T to T/E ratio is greater in the coronal versus the sagittal suture, and this finding may contribute to making the coronal suture more susceptible to fusion due to TWIST haploinsufficiency. Importantly, we were able to inhibit suture fusion in Twist1+/- mice by modulating the balance between these dimers toward T/E formation, by either increasing the expression of E2A E12 or by decreasing Id expression. Therefore, we have identified dimer partner selection as an important mediator of Twist1 function and provide a mechanistic understanding of craniosynostosis due to TWIST haploinsufficiency.


Asunto(s)
Suturas Craneales/embriología , Suturas Craneales/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/química , Proteína 1 Relacionada con Twist/genética , Animales , Animales Recién Nacidos , Línea Celular , Suturas Craneales/química , Craneosinostosis/genética , Craneosinostosis/metabolismo , Dimerización , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Ratones Transgénicos , Proteínas Nucleares/deficiencia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Proteína 1 Relacionada con Twist/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA