Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Surg Radiol Anat ; 39(7): 735-739, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28078367

RESUMEN

PURPOSE: Lingual foramina can be observed between the lingual aspects of the mandible in humans. A sublingual artery is thought to exist in sublingual space and a submental artery in submaxillary space, which pierce the mandible through lingual foramina. During surgery for oral implant placement between apices of the mental foramen, it is important to determine the existence and positioning of lingual foramina. The purpose of this study was to investigate the positions of lingual foramina in relation to the mylohyoid muscle and vertical position of the mylohyoid line using cone-beam computed tomography (CBCT) images. METHODS: We examined 20 formalin-perfused cadavers. The mylohyoid muscle was dissected and marked with a silicone tube, then CBCT images were obtained to evaluate the relationship of that muscle with lingual foramina. RESULTS: We observed 37 lingual foramina in the 20 cadavers. As for vertical positioning, 16 lingual foramina were found in sublingual space, while in horizontal positioning, 6 were found in the anterior region of sublingual space. The ratio of vertical distance from the inferior margin to the mylohyoid line and mental spine was lower in the anterior region as compared to the posterior region. CONCLUSION: In this study, lingual foramina were found to commonly exist in sublingual space above the mylohyoid muscle and pierce the mesial side. For evaluation of the vertical position of the mylohyoid line, it is better to use the stable mental spine rather than the alveolar process.


Asunto(s)
Mandíbula/anatomía & histología , Suelo de la Boca/anatomía & histología , Anciano de 80 o más Años , Tomografía Computarizada de Haz Cónico , Femenino , Humanos , Masculino , Mandíbula/diagnóstico por imagen , Suelo de la Boca/diagnóstico por imagen , Músculo Esquelético/anatomía & histología
2.
Case Rep Obstet Gynecol ; 2015: 170343, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26180648

RESUMEN

Background. Although radiation exposure is of great concern to expecting patients, little information is available on the fetal radiation dose associated with prophylactic internal iliac artery balloon occlusion (IIABO). Here we estimated the fetal radiation dose associated with prophylactic IIABO in Caesarean section (CS). Cases. We report our experience with the IIABO procedure in three consecutive patients with suspected placenta previa/accreta. Fetal radiation dose measurements were conducted prior to each CS by using an anthropomorphic phantom. Based on the simulated value, we calculated the fetal radiation dose as the absorbed dose. We found that the fetal radiation doses ranged from 12.88 to 31.6 mGy. The fetal radiation dose during the prophylactic IIABOs did not exceed 50 mGy. Conclusion. The IIABO procedure could result in a very small increase in the risk of harmful effects to the fetus.

3.
Surg Radiol Anat ; 37(3): 255-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25026999

RESUMEN

The present study sought to clarify the course of the phrenic nerve and its correlation with anatomical landmarks in the neck region. We examined 17 cadavers (30 sides). In each, the phrenic nerves was dissected from the lateral side of the neck, and its position within the triangle formed by the mastoid process and sternal and acromial ends of the clavicle was determined. The point where the phrenic nerve arises in the posterior triangle was found to be similar to the point where the cutaneous blanches of the cervical plexus emerge at the middle of the posterior border of the sternocleidomastoid muscle. In the supraclavian triangle, the phrenic nerve crosses the anterior border of the anterior scalene muscle near Erb's point where the superficial point is 2-3 cm superior from the clavicle and posterior border of the sternocleidomastoid muscle. The phrenic nerve arises in the posterior triangle near the nerve point, then descends to the anterior surface of the anterior scalene muscle in the supraclavian triangle. It is necessary to be aware of the supraclavian triangle below Erb's point during neck dissection procedures.


Asunto(s)
Disección del Cuello/métodos , Músculos del Cuello/cirugía , Nervio Frénico/anatomía & histología , Cadáver , Disección/métodos , Femenino , Humanos , Masculino , Músculos del Cuello/inervación , Nervio Frénico/cirugía
4.
Biochem Biophys Res Commun ; 434(2): 287-92, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23537649

RESUMEN

Estrogen is a key factor to induce the sexually dimorphic nucleus (SDN) in the preoptic area (POA) of the rat brain. Identification of estrogen-dependent signaling pathways at SDN in POA during the critical period is a prerequisite for elucidating the mechanism. In the present study, we treated female rats with/without 17ß-estradiol (E2) at birth, designated as postnatal day 1 (P1), and prepared total RNA from brain slices containing SDN for DNA microarray analysis. Among the estrogen-responsive genes identified, protein kinase C-delta (PKC-δ) was significantly up-regulated by E2 at P5. We examined the downstream effectors of PKC-δ protein by Western blotting and found an E2-induced PKC-δ/Rac1/PAK1/LIMK1/cofilin pathway. In the pathway, E2 suppressed the phosphorylation (inactive form) of cofilin. This result was supported by immunohistochemistry, where the phosphorylation/dephosphorylation of cofilin occurred at SDN, which suggests that cell migration is a cue to create sexual dimorphism in POA.


Asunto(s)
Actinas/metabolismo , Movimiento Celular , Cofilina 1/metabolismo , Estradiol/farmacología , Área Preóptica/efectos de los fármacos , Caracteres Sexuales , Animales , Animales Recién Nacidos , Western Blotting , Cofilina 1/genética , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Femenino , Inmunohistoquímica , Quinasas Lim/genética , Quinasas Lim/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Embarazo , Área Preóptica/embriología , Área Preóptica/metabolismo , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo , Ratas , Ratas Wistar , Transducción de Señal , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
5.
Neural Regen Res ; 8(2): 149-55, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25206485

RESUMEN

Estrogen receptor α is widely distributed in the rat brain, but the tissue- or target-specificity of the estrogen receptor α gene promoters remains unknown. In the present study, we used transgenic rats expressing enhanced green fluorescent protein under the control of the estrogen receptor α 0/B promoter to examine expression driven by this promoter in two significant nuclei that regulate cardiovascular activity, the area postrema and the nucleus tractus solitarius. Immunohistochemistry showed that enhanced green fluorescent protein-labeled cells were distributed in the area postrema and the nucleus tractus solitarius of both female and male transgenic rats, and a neural network of enhanced green fluorescent protein-positive fibers was seen between the area postrema and the nucleus tractus solitarius. The number of enhanced green fluorescent protein-labeled cells in the area postrema of female rats was significantly higher than in the males, but no significant difference was found in the number of enhanced green fluorescent protein-labeled cells in the nucleus tractus solitarius. The sex differences in the number of enhanced green fluorescent protein-labeled cells in the area postrema was not affected after ovariectomy or 17ß-estradiol benzoate treatment in adult rats. Our results suggest that the effects of estrogen in the area postrema are related to the expression of estrogen receptor α under the control of the 0/B promoter, and changes in the sex hormone environment in the adult period do not affect estrogen receptor α expression in the area postrema or the nucleus tractus solitarius.

6.
J Steroid Biochem Mol Biol ; 124(1-2): 38-46, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21251978

RESUMEN

We re-examined mouse ERα mRNA variants using rapid amplification of cDNA ends (RACE) and RT-PCR. Our analysis showed the presence of several mRNA variants containing unique 5'- or 3'-nucleotide sequences. We mapped the cDNA sequences on the mouse genome, and identified four novel 3'-terminal and 5'-leader exons in the intronic region between exons 4 and 5. RT-PCR analysis revealed that the expression patterns of the C-terminally truncated ERα products (CTERPs) were similar to that of Wild-type ERα and that the N-terminally truncated ERα products (NTERPs) appeared to have different expression profiles. Moreover, we constructed expression vectors and analyzed the subcellular localization and the transcriptional activation abilities of the variant proteins in transfected HEK293 cells using immunocytochemistry and luciferase reporter assay. The CTERP variants localized in the nuclei and constitutively activated estrogen response element (ERE)-driven promoters, while the NTERP variant was located in the extra-nuclear regions and had no ability to activate the ERE promoters in the presence or absence of 10 nM estradiol. Our results indicate that the mouse ERα gene is more complex than previously thought in terms of genomic organization and that alternative splicing and alternative usage of intronic promoters contribute to the remarkable diversity of ERα mRNAs and proteins.


Asunto(s)
Receptor alfa de Estrógeno/genética , Empalme Alternativo , Animales , Mapeo Cromosómico/métodos , Receptor alfa de Estrógeno/biosíntesis , Exones , Femenino , Células HEK293 , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Isoformas de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Técnica del ADN Polimorfo Amplificado Aleatorio , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
7.
Endocrinology ; 151(4): 1923-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20185767

RESUMEN

The volume of the sexually dimorphic nucleus of the preoptic area (SDN-POA) is two to four times larger in male rats than in females; however, the mechanism for the establishment of sexual dimorphism and the function of this nucleus is almost unknown. Perinatal estrogen can cause sexual dimorphism via the estrogen receptor alpha (ERalpha). Recently, transgenic rats were generated that express enhanced green fluorescent protein (EGFP) under the control of the ERalpha gene promoter 0/B to tag ERalpha-positive neurons in the brain. In the present study, we examined whether this EGFP expression could be a marker for the SDN-POA in adults. EGFP-labeled cells were distributed in the core of the SDN-POA (0/B-SDN) of male and female transgenic rats, in accordance with the Nissl staining and immunoreactivity for the SDN marker, calbindin. They were also immunoreactive for ERalpha. The core was bigger in volume and contained more 0/B-SDN neurons in males than in females. The EGFP-tagged cells were packed more densely in the female core than that in males. Subcutaneous injection of 100 mug 17beta-estradiol to females on the day of birth, or orchidectomy of male neonates, reversed the sexually dimorphic phenotype of the volume of the 0/B-SDN, despite not affecting the cell number. We suggest that this EGFP expression in the SDN-POA could be a useful marker to clarify the sexual differentiation and function of the SDN-POA. Moreover, the ERalpha gene promoter 0/B plays a key role in the organization of the sexual differentiation of the SDN-POA.


Asunto(s)
Receptor alfa de Estrógeno/genética , Neuronas/metabolismo , Área Preóptica/metabolismo , Regiones Promotoras Genéticas/genética , Caracteres Sexuales , Animales , Calbindinas , Recuento de Células , Estradiol/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Masculino , Neuronas/efectos de los fármacos , Área Preóptica/efectos de los fármacos , Ratas , Ratas Transgénicas , Ratas Wistar , Proteína G de Unión al Calcio S100/metabolismo
8.
Eur J Neurosci ; 28(5): 849-61, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18691332

RESUMEN

Inhibitory synaptic transmission mediated by gamma-aminobutyric acid (GABA)(A) receptors is involved in regulation of experience-dependent cortical plasticity. However, little information is available on their presynaptic and postsynaptic developmental profiles. The present study aims to investigate the developmental changes of miniature and unitary inhibitory postsynaptic currents (mIPSCs and uIPSCs) in mouse barrel cortex. mIPSCs recorded from supragranular pyramidal neurons showed a gradual increase in frequency during postnatal days 6-15 (PD6-15) followed by a marked increase at PD16-20, and mIPSCs frequency reached a plateau at about PD21-30. The amplitude of mIPSCs showed a transient decrease at PD10-12 followed by an increase during PD13-30, and reached a plateau at about PD30. Their decay time constant progressively decreased during the first 30 days postnatally, and reached a steady level at about PD30. Paired recordings from interneurons and synaptically coupled target pyramidal cells revealed that uIPSC amplitude increased with age up to PD30. In contrast, failure rate and coefficient of variation decreased during PD7-15 and showed little change at a later stage. Short-term depression induced by presynaptic stimulation at 33 Hz progressively decreased during PD6-20, and was stabilized at about PD21-30. Quantal analysis revealed that the number of release sites increased with age up to PD30, while the release probability increased during PD6-12 and then reached a plateau level. These results suggest that the number of release sites and release probability of GABA and GABA(A)-mediated IPSC kinetics show distinct developmental profiles, which could play roles in regulating the onset and offset of critical periods for experience-dependent cortical plasticity.


Asunto(s)
Inhibición Neural/fisiología , Células Piramidales/metabolismo , Receptores de GABA-A/metabolismo , Corteza Somatosensorial/crecimiento & desarrollo , Corteza Somatosensorial/metabolismo , Transmisión Sináptica/fisiología , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Potenciales Postsinápticos Inhibidores/fisiología , Interneuronas/citología , Interneuronas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Células Piramidales/citología , Corteza Somatosensorial/citología , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Factores de Tiempo
9.
Neurosci Lett ; 436(1): 35-9, 2008 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-18358606

RESUMEN

Estrogen plays critical roles in the neuroendocrine system of adult female rats through separate actions, respectively, in the preoptic area (POA) and the ventromedial nucleus of the hypothalamus (VMH). Seven-week-old rats were treated with/without estrogen after they were ovariectomized, and four estrogen-responsive, neuronal system-related genes, encoding alpha4 neuronal nicotinic acetylcholine receptor (Chrna4), GABA(A) receptor delta (Gabrd), serotonin receptor 6 (Htr6), and GABA transporter 2 (Slc6a13), were investigated by real-time RT-PCR and Western blot analyses to examine their differential regulation by estrogen between the anterior part containing POA and the posterior part containing VMH. We further examined Bax, Bcl2, and Prkce, the former two genes to be involved in the gene expression network of Chrna4 and the latter gene, that of Gabrd. The regulation of Bax and Bcl2 by estrogen differed between the anterior and posterior parts. The results demonstrated differential regulation of these neuronal system-related genes by estrogen between the anterior and posterior parts of the hypothalamus and suggested the roles of gene expression networks for the respective genes in the neuroendocrine system of adult female rats.


Asunto(s)
Estrógenos/metabolismo , Regulación de la Expresión Génica/fisiología , Hipotálamo/metabolismo , Animales , Western Blotting , Femenino , Proteínas Transportadoras de GABA en la Membrana Plasmática/biosíntesis , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , ARN Mensajero/análisis , Ratas , Ratas Wistar , Receptores de GABA-A/biosíntesis , Receptores de GABA-A/genética , Receptores Nicotínicos/biosíntesis , Receptores Nicotínicos/genética , Receptores de Serotonina/biosíntesis , Receptores de Serotonina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Brain Res Mol Brain Res ; 139(1): 42-51, 2005 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-15953656

RESUMEN

Transgenic rats expressing enhanced green fluorescent protein (EGFP) under the control of an estrogen receptor (ER) alpha promoter were generated to tag ERalpha-positive neurons in the brain. Two transgenes, one containing sequences for promoter A and DsRed and the other containing sequences for promoter 0/B and EGFP, were injected simultaneously into Wistar rat zygotes. Twenty-two founders with both transgenes were identified. Ten lines of these founders expressed the EGFP tag in the brains of their first filial generation, whereas none similarly expressed the DsRed tag. In two lines selected for the brightness of the EGFP fluorescence in their brains, tagged cells showed essentially the same patterns. Tagged cells were in the preoptic area (POA), bed nucleus of the stria terminalis (BNST), hypothalamic arcuate nucleus and medial amygdala. ERalpha-immunoreactive neurons were identified in all of these structures by immunohistochemistry. In ovariectomized females, approximately 75% of the EGFP-fluorescent cells in the POA-BNST were immunoreactive for ERalpha. In the POA-BNST, ovariectomy increased the number of EGFP-immunopositive cells and estrogen supplementation reversed this effect, indicating that the promoter 0/B is involved in estrogen-induced downregulation of ERalpha. EGFP was also present in cells in the cerebral cortex and hippocampus, which have not previously been associated with endocrine regulation. Conversely, only a few cells were tagged in the hypothalamic ventromedial nucleus, which contained many ERalpha-immunoreactive neurons. This discrepancy could have arisen as a result of differential promoter usage.


Asunto(s)
Regulación hacia Abajo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Regiones Promotoras Genéticas , Prosencéfalo , Animales , Animales Modificados Genéticamente , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Masculino , Ovariectomía , Prosencéfalo/anatomía & histología , Prosencéfalo/metabolismo , Ratas , Ratas Wistar
12.
Endocrinology ; 144(8): 3297-300, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12865305

RESUMEN

The regulatory mechanisms controlling gene expression of GnRH subtypes, particularly of the evolutionarily conserved GnRH2, remain speculative. To address this issue, we have successfully coupled the anatomic specificity of immunofluorescently defined "cell picking" with the sensitivity of real-time quantitative RT-PCR (RT-Q-RT-PCR), which enabled us to examine the presence and quantity of GnRH mRNAs in individual neurons. Here, using RT-Q-RT-PCR, we report change in the levels of transcripts of GnRH subtypes in individual neurons harvested from the brain of mature and immature males of tilapia, Oreochromis niloticus. The levels of GnRH1 mRNA per cell and the percentage of neurons expressing GnRH1 transcripts exceeding 0.05 x 10(2) fg/cell were significantly higher in mature males (44.2%) compared with immature males (4.7%). In contrast, there was no difference in mRNA levels and the percentage of cells expressing GnRH2 and GnRH3 between the two reproductive states. Thus, using a novel approach that enables immunofluorescently labeled single-cell RT-Q-RT-PCR analysis of GnRH neurons, we present evidence that shows preoptic GnRH1 is important for gonadal maturation, whereas GnRH2 and GnRH3 might have supplementary roles in reproductive behaviors or nonreproductive functions. Furthermore, we speculate that the use of this method will allow the identification and quantification of known and unknown genes in single GnRH neurons, which would greatly facilitate our understanding of the complex interactions that govern the physiology of individual cells of GnRH variants in vertebrate species.


Asunto(s)
Encéfalo/citología , Cíclidos/metabolismo , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/análisis , Hormona Liberadora de Gonadotropina/genética , Neuronas/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Envejecimiento , Animales , Recuento de Células , Cíclidos/anatomía & histología , Técnica del Anticuerpo Fluorescente , Hormona Liberadora de Gonadotropina/fisiología , Masculino , Bulbo Olfatorio/citología , Área Preóptica/citología , ARN Mensajero/análisis , Tegmento Mesencefálico/citología , Testículo/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...