Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 232
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gastrointest Endosc ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38851456

RESUMEN

BACKGROUND AND AIMS: Despite the benefits of artificial intelligence (AI) in small bowel (SB) capsule endoscopy (CE) image reading, information on its application in the stomach and SB CE is lacking. METHODS: In this multicenter, retrospective diagnostic study, gastric imaging data were added to the deep learning (DL)-based SmartScan (SS), which has been described previously. A total of 1,069 magnetically controlled gastrointestinal (GI) CE examinations (comprising 2,672,542 gastric images) were used in the training phase for recognizing gastric pathologies, producing a new AI algorithm named SS Plus. 342 fully automated, magnetically controlled CE (FAMCE) examinations were included in the validation phase. The performance of both senior and junior endoscopists with both the SS Plus-Assisted Reading (SSP-AR) and conventional reading (CR) modes was assessed. RESULTS: SS Plus was designed to recognize 5 types of gastric lesions and 17 types of SB lesions. SS Plus reduced the number of CE images required for review to 873.90 (1000) (median, IQR 814.50-1,000) versus 44,322.73 (42,393) (median, IQR 31,722.75-54,971.25) for CR. Furthermore, with SSP-AR, endoscopists took 9.54 min (8.51) (median, IQR 6.05-13.13) to complete the CE video reading. In the 342 CE videos, SS Plus identified 411 gastric and 422 SB lesions, whereas 400 gastric and 368 intestinal lesions were detected with CR. Moreover, junior endoscopists remarkably improved their CE image reading ability with SSP-AR. CONCLUSIONS: Our study shows that the newly upgraded DL-based algorithm SS Plus can detect GI lesions and help improve the diagnostic performance of junior endoscopists in interpreting CE videos.

2.
Future Oncol ; 19(15): 1063-1072, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37283023

RESUMEN

Aim: To evaluate independent risk factors specific for early-stage nasopharyngeal carcinoma (NPC). Methods: A total of 566 patients with early-stage NPC from 2004 to 2019 were identified using the Surveillance, Epidemiology and End Results database. Results: Older ages (70-79 and >80 years) were independent risk factors, with hazard ratios of 1.961 and 5.011, respectively. The hazard ratio for early-stage NPC in Asian and Pacific Islander residents (0.475) was lower than that for White residents. A tumor size <3 cm was a protective factor for overall and cancer-specific survival in the current study. Conclusion: In patients with early-stage NPC, age >70 years, race and tumor size were independent prognosticators for cancer-specific survival.


Asunto(s)
Neoplasias Nasofaríngeas , Humanos , Estados Unidos/epidemiología , Anciano , Carcinoma Nasofaríngeo/patología , Pronóstico , Neoplasias Nasofaríngeas/diagnóstico , Neoplasias Nasofaríngeas/epidemiología , Neoplasias Nasofaríngeas/terapia , Modelos de Riesgos Proporcionales , Factores de Riesgo , Estadificación de Neoplasias
3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 54(3): 482-490, 2023 May.
Artículo en Zh | MEDLINE | ID: mdl-37248572

RESUMEN

Gastrointestinal microecology (GM) system is composed of normal gut microbiota and its living environment. The impact of GM on human health and many diseases has been widely studied. The impact of GM system on tumors is mainly reflected in the remodeling of the tumor microenvironment (TME). TME, a special microenvironment that tumors live in, can regulate the characteristics of tumor cells and affect the occurrence and development of tumors through intercellular contact and the secretion of cytokines. At present, cancer stem cell (CSC) model is considered an important theory that explains the origin and malignant progression of tumors. The formation and proliferation of CSC usually represent increased tumor invasion, metastasis, and chemotherapy resistance, resulting in poor clinical prognosis in patients. Therefore, it is important to study the role and mechanism through which GM system affects the acquisition of CSC characteristics through remodeling TME, thereby affecting tumor invasion, metastasis, and chemotherapy resistance. Studies on this topic are of great significance for clinical understanding of tumor malignant progression and improving tumor treatment outcomes. However, due to the low content of single bacteria in the gastrointestinal model, high heterogeneity, and difficulty in tracing distant metastasis, there are still great limitations in the previous research. Herein, we reviewed the research progress in the effect of GM remodeling of TME on the acquisition of tumor stemness, tumor invasion and metastasis, and the resistance to chemotherapy.


Asunto(s)
Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/tratamiento farmacológico , Células Madre Neoplásicas
4.
BMC Neurosci ; 22(1): 18, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33752606

RESUMEN

BACKGROUND: The SCN11A gene, encoded Nav1.9 TTX resistant sodium channels, is a main effector in peripheral inflammation related pain in nociceptive neurons. The role of SCN11A gene in the auditory system has not been well characterized. We therefore examined the expression of SCN11A in the murine cochlea, the morphological and physiological features of Nav1.9 knockout (KO) ICR mice. RESULTS: Nav1.9 expression was found in the primary afferent endings beneath the inner hair cells (IHCs). The relative quantitative expression of Nav1.9 mRNA in modiolus of wild-type (WT) mice remains unchanged from P0 to P60. The number of presynaptic CtBP2 puncta in Nav1.9 KO mice was significantly lower than WT. In addition, the number of SGNs in Nav1.9 KO mice was also less than WT in the basal turn, but not in the apical and middle turns. There was no lesion in the somas and stereocilia of hair cells in Nav1.9 KO mice. Furthermore, Nav1.9 KO mice showed higher and progressive elevated ABR threshold at 16 kHz, and a significant increase in CAP thresholds. CONCLUSIONS: These data suggest a role of Nav1.9 in regulating the function of ribbon synapses and the auditory nerves. The impairment induced by Nav1.9 gene deletion mimics the characters of cochlear synaptopathy.


Asunto(s)
Nervio Coclear/patología , Pérdida Auditiva Sensorineural/genética , Canal de Sodio Activado por Voltaje NAV1.9/genética , Sinapsis/patología , Animales , Nervio Coclear/metabolismo , Eliminación de Gen , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patología , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/patología , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Sinapsis/metabolismo
5.
Semin Thromb Hemost ; 47(3): 240-253, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33794549

RESUMEN

Cell surface proteoglycans are important constituents of the glycocalyx and participate in cell-cell and cell-extracellular matrix (ECM) interactions, enzyme activation and inhibition, and multiple signaling routes, thereby regulating cell proliferation, survival, adhesion, migration, and differentiation. Heparanase, the sole mammalian heparan sulfate degrading endoglycosidase, acts as an "activator" of HS proteoglycans, thus regulating tissue hemostasis. Heparanase is a multifaceted enzyme that together with heparan sulfate, primarily syndecan-1, drives signal transduction, immune cell activation, exosome formation, autophagy, and gene transcription via enzymatic and nonenzymatic activities. An important feature is the ability of heparanase to stimulate syndecan-1 shedding, thereby impacting cell behavior both locally and distally from its cell of origin. Heparanase releases a myriad of HS-bound growth factors, cytokines, and chemokines that are sequestered by heparan sulfate in the glycocalyx and ECM. Collectively, the heparan sulfate-heparanase axis plays pivotal roles in creating a permissive environment for cell proliferation, differentiation, and function, often resulting in the pathogenesis of diseases such as cancer, inflammation, endotheliitis, kidney dysfunction, tissue fibrosis, and viral infection.


Asunto(s)
Enfermedad , Glucuronidasa/metabolismo , Heparitina Sulfato/metabolismo , Humanos
6.
FASEB J ; 34(1): 1169-1181, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914631

RESUMEN

BHLHE40, a member of the basic helix-loop-helix transcription factor family, has been reported to play an important role in inflammatory diseases. However, the regulation and function of BHLHE40 in Helicobacter pylori (H pylori)-associated gastritis is unknown. We observed that gastric BHLHE40 was significantly elevated in patients and mice with H pylori infection. Then, we demonstrate that H pylori-infected GECs express BHLHE40 via cagA-ERK pathway. BHLHE40 translocates to cell nucleus, and then binds to cagA protein-activated p-STAT3 (Tyr705). The complex increases chemotactic factor CXCL12 expression (production). Release of CXCL12 from GECs fosters CD4+ T cell infiltration in the gastric mucosa. Our results identify the cagA-BHLHE40-CXCL12 axis that contributes to inflammatory response in gastric mucosa during H pylori infection.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Quimiocina CXCL12/metabolismo , Células Epiteliales/metabolismo , Gastritis/microbiología , Infecciones por Helicobacter/metabolismo , Proteínas de Homeodominio/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Núcleo Celular/metabolismo , Femenino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Gastritis/metabolismo , Regulación de la Expresión Génica , Helicobacter pylori , Humanos , Inflamación , Ratones , Ratones Endogámicos C57BL , Estómago/microbiología , Regulación hacia Arriba
7.
FASEB J ; 33(4): 5018-5033, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30596522

RESUMEN

Cathepsin C (CtsC) functions as a central coordinator for activation of many serine proteases in immune cells. However, CtsC expression in gastric epithelial cells and its role in Helicobacter pylori infection remain unclear. Real-time PCR, Western blot, and immunohistochemistry analyses identified that CtsC was decreased in gastric mucosa of H. pylori-infected patients and mice. Isolated gastric epithelial cells and cell lines were stimulated with H. pylori and/or TGF-ß1 showed that down-regulation of CtsC in gastric epithelial cells largely depended on H. pylori cagA via Src/ERK and Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathways, and the effect could be synergistically augmented by TGF-ß1 in an autocrine manner. In human gastric mucosa, CtsC expression was negatively correlated with bacteria colonization; accordingly, provision of exogenous active CtsC overwhelmed H. pylori persistence in gastric mucosa of mice. In the presence of active CtsC, isolated human neutrophils activated via NF-κB pathway with augmented bactericidal capacity in vitro. We also found that neutrophils activated and cleared bacteria in active CtsC-injected mice and that there was no bactericidal capacity in mice that were simultaneously neutrophil-depleted by Ly6G antibody. Our findings identified a mechanism that H. pylori abrogate CtsC to impair neutrophil activation and to ensure persistence in gastric mucosa. Efforts to enable and boost this neutrophil activation pathway by active CtsC may therefore become valuable strategies in treating H. pylori infection.-Liu, Y. G., Teng, Y. S., Cheng, P., Kong, H., Lv, Y. P., Mao, F. Y., Wu, X. L., Hao, C. J., Chen, W., Yang, S. M., Zhang, J. Y., Peng, L. S., Wang, T. T., Han, B., Ma, Q., Zou, Q. M., Zhuang, Y. Abrogation of cathepsin C by Helicobacter pylori impairs neutrophil activation to promote gastric infection.


Asunto(s)
Catepsina C/metabolismo , Mucosa Gástrica/microbiología , Helicobacter pylori/patogenicidad , Activación Neutrófila/fisiología , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Infecciones por Helicobacter/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Fagocitosis/fisiología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Neural Plast ; 2020: 6235948, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32617095

RESUMEN

The objective of this study was to explore the molecular mechanisms of acute noise-induced hearing loss and recovery of steady-state noise-induced hearing loss using miniature pigs. We used miniature pigs exposed to white noise at 120 dB (A) as a model. Auditory brainstem response (ABR) measurements were made before noise exposure, 1 day and 7 days after noise exposure. Proteomic Isobaric Tags for Relative and Absolute Quantification (iTRAQ) was used to observe changes in proteins of the miniature pig inner ear following noise exposure. Western blot and immunofluorescence were performed for further quantitative and qualitative analysis of proteomic changes. The average ABR-click threshold of miniature pigs before noise exposure, 1 day and 7 days after noise exposure, were 39.4 dB SPL, 67.1 dB SPL, and 50.8 dB SPL, respectively. In total, 2,158 proteins were identified using iTRAQ. Both gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) database analyses showed that immune and metabolic pathways were prominently involved during the impairment stage of acute hearing loss. During the recovery stage of acute hearing loss, most differentially expressed proteins were related to cholesterol metabolism. Western blot and immunofluorescence showed accumulation of reactive oxygen species and nuclear translocation of NF-κB (p65) in the hair cells of miniature pig inner ears during the acute hearing loss stage after noise exposure. Nuclear translocation of NF-κB (p65) may be associated with overexpression of downstream inflammatory factors. Apolipoprotein (Apo) A1 and Apo E were significantly upregulated during the recovery stage of hearing loss and may be related to activation of cholesterol metabolic pathways. This is the first study to use proteomics analysis to analyze the molecular mechanisms of acute noise-induced hearing loss and its recovery in a large animal model (miniature pigs). Our results showed that activation of metabolic, inflammatory, and innate immunity pathways may be involved in acute noise-induced hearing loss, while cholesterol metabolic pathways may play an important role in recovery of hearing ability following noise-induced hearing loss.


Asunto(s)
Colesterol/metabolismo , Cóclea/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Pérdida Auditiva Provocada por Ruido/metabolismo , Redes y Vías Metabólicas/fisiología , Recuperación de la Función/fisiología , Estimulación Acústica , Animales , Cóclea/fisiopatología , Bases de Datos Factuales , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva Provocada por Ruido/fisiopatología , Proteómica , Porcinos , Porcinos Enanos
9.
Cell Commun Signal ; 17(1): 63, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186051

RESUMEN

Human telomerase reverse transcriptase (hTERT) is the core subunit of human telomerase and plays important roles in human cancers. Aberrant expression of hTERT is closely associated with tumorigenesis, cancer cell stemness maintaining, cell proliferation, apoptosis inhibition, senescence evasion and metastasis. The molecular basis of hTERT regulation is highly complicated and consists of various layers. A deep and full-scale comprehension of the regulatory mechanisms of hTERT is pivotal in understanding the pathogenesis and searching for therapeutic approaches. In this review, we summarize the recent advances regarding the diverse regulatory mechanisms of hTERT, including the transcriptional (promoter mutation, promoter region methylation and histone acetylation), post-transcriptional (mRNA alternative splicing and non-coding RNAs) and post-translational levels (phosphorylation and ubiquitination), which may provide novel perspectives for further translational diagnosis or therapeutic strategies targeting hTERT.


Asunto(s)
Telomerasa/metabolismo , Humanos , Mutación , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional , Telomerasa/genética
11.
BMC Neurosci ; 19(1): 28, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29716524

RESUMEN

BACKGROUND: Auditory function and cochlear morphology have previously been described in a porcine model with spontaneous WS2-like phenotype. In the present study, cochlear histopathology was further investigated in the inner ear of the developing spontaneous deafness pig. RESULTS: We found that the stria vascularis transformed into a complex tri-laminar tissue at embryonic 85 days (E85) in normal pigs, but not in the MITF-/- pigs. As the neural crest (NC) of cochlea was derived by melanocytes. MITF mutation caused failure of development of melanocytes which caused a subsequent collapse of cochlear duct and deficits of the epithelium after E100. Furthermore, the spiral ganglion neurons of cochlea in the MITF-/- pigs began to degenerate at postnatal 30 days (P30). Thus, our histopathological results indicated that the malformation of the stria vascularis was a primary defect in MITF-/- induced WT pigs which was resulted from the loss of NC-derived melanocytes. Subsequently, the cochleae underwent secondary degeneration of the vestibular organs. As the degeneration of spiral ganglion neurons happened after P30, it suggests that WS patients should be considered as candidates for cochlear implant. CONCLUSIONS: Our porcine model of MITF-M mutation may provide a crucial animal model for cochlear implant, cell therapy in patients with congenital hereditary hearing loss.


Asunto(s)
Cóclea/patología , Sordera/patología , Oído Interno/patología , Animales , Implantes Cocleares , Oído Interno/crecimiento & desarrollo , Melanocitos/patología , Porcinos , Vestíbulo del Laberinto/patología
12.
Cell Commun Signal ; 16(1): 57, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30208972

RESUMEN

FOXM1 (forkhead box protein M1) is a critical proliferation-associated transcription factor that is widely spatiotemporally expressed during the cell cycle. It is closely involved with the processes of cell proliferation, self-renewal, and tumorigenesis. In most human cancers, FOXM1 is overexpressed, and this indicates a poor prognosis for cancer patients. FOXM1 maintains cancer hallmarks by regulating the expression of target genes at the transcriptional level. Due to its potential role as molecular target in cancer therapy, FOXM1 was named the Molecule of the Year in 2010. However, the mechanism of FOXM1 dysregulation remains indistinct. A comprehensive understanding of FOXM1 regulation will provide novel insight for cancer and other diseases in which FOXM1 plays a major role. Here, we summarize the transcriptional regulation, post-transcriptional regulation and post-translational modifications of FOXM1, which will provide extremely important implications for novel strategies targeting FOXM1.


Asunto(s)
Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Neoplasias/metabolismo , Animales , Proteína Forkhead Box M1/antagonistas & inhibidores , Humanos , Ratones , Neoplasias/genética , Neoplasias/patología , Procesamiento Proteico-Postraduccional , Transcripción Genética , Microambiente Tumoral
13.
Cell Commun Signal ; 16(1): 31, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29903018

RESUMEN

The MiT/TFE transcription factors play a pivotal role in the regulation of autophagy and lysosomal biogenesis. The subcellular localization and activity of MiT/TFE proteins are primarily regulated through phosphorylation. And the phosphorylated protein is retained in the cytoplasm and subsequently translocates to the nucleus upon dephosphorylation, where it stimulates the expression of hundreds of genes, leading to lysosomal biogenesis and autophagy induction. The transcription factor-mediated lysosome-to-nucleus signaling can be directly controlled by several signaling molecules involved in the mTORC1, PKC, and AKT pathways. MiT/TFE family members have attracted much attention owing to their intracellular clearance of pathogenic factors in numerous diseases. Recently, multiple studies have also revealed the MiT/TFE proteins as master regulators of cellular metabolic reprogramming, converging on autophagic and lysosomal function and playing a critical role in cancer, suggesting that novel therapeutic strategies could be based on the modulation of MiT/TFE family member activity. Here, we present an overview of the latest research on MiT/TFE transcriptional factors and their potential mechanisms in cancer.


Asunto(s)
Factor de Transcripción Asociado a Microftalmía/metabolismo , Animales , Autofagosomas/metabolismo , Autofagia , Humanos , Lisosomas/metabolismo , Transducción de Señal
14.
Neural Plast ; 2018: 7513217, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29849567

RESUMEN

Glucose is a fundamental source of energy for mammalian cells; however, whether glucose is taken up through the lateral walls of cochlear outer hair cells (OHCs) is unknown. The OHC lateral wall is complex, composed of a plasma membrane, cortical lattice, and subsurface cisternae. This study assessed the uptake of glucose by OHCs using live-cell microscopy and examined the distribution of glucose transporter isoforms by immunohistochemistry. We found that glucose transporter-4 was mostly expressed on the lateral wall of OHCs. Glucose crossed the lateral walls of OHCs via glucose transporters-4 mainly, and this process could be modulated. These results suggest that the lateral walls are involved in modulating energy transport into OHCs.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Glucosa/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Animales , Membrana Celular/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Cobayas
15.
Gut ; 66(1): 31-42, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26370108

RESUMEN

BACKGROUND AND AIMS: Human telomerase reverse transcriptase (hTERT) plays an important role in cancer invasion, but the relevant mechanism is not well known. This study aims to investigate the role and mechanism of hTERT in gastric cancer metastasis. DESIGN: Proteomics analysis, qPCR and western blotting were used to screen for hTERT-regulated candidate molecules in gastric cancer invasion. Chromatin immunoprecipitation (ChIP) qPCR was performed to identify the binding sites of hTERT at the regulatory region of the integrin ß1 (ITGB1) gene. ChIP assays were further applied to elucidate the transcription factors that bound to the regulatory region. The interactions between hTERT and the transcription factors were tested by co-immunoprecipitation (Co-IP) and glutathione S-transferase (GST) pull-down experiments. Moreover, the revealed pathway was verified in tumour-bearing nude mice and human gastric cancer tissues. RESULTS: ITGB1 was identified as a downstream gene of hTERT, and there were two hTERT-binding regions within this gene. hTERT alleviated the binding of forkhead box O3 (FOXO3a) to FOXO3a binding element (+9972∼+9978), but it enhanced the binding of forkhead box M1 (FOXM1) to FOXM1 binding element (-1104∼-1109) in ITGB1 gene. Importantly, FOXO3a played a major role in hTERT-induced ITGB1 expression, and the hTERT/murine double minute 2 (MDM2) complex promoted the ubiquitin-mediated degradation of FOXO3a. Moreover, hTERT increased ITGB1 expression in xenograft gastric cancer, and the level of hTERT was positively correlated with that of ITGB1 in human gastric cancer tissues. CONCLUSIONS: The hTERT/MDM2-FOXO3a-ITGB1 pathway markedly contributes to hTERT-promoted gastric cancer invasion, suggesting that this pathway might be a novel target for the prevention and treatment of gastric cancer metastasis.


Asunto(s)
Proteína Forkhead Box O3/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Telomerasa/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Femenino , Proteína Forkhead Box M1/metabolismo , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Metástasis Linfática , Ratones , Ratones Desnudos , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal , Neoplasias Gástricas/genética , Tasa de Supervivencia , Telomerasa/genética , Ubiquitinación , Regulación hacia Arriba
18.
Ear Hear ; 38(6): 647-652, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28471842

RESUMEN

Hearing loss (HL) is a common sensory impairment in humans, with significant economic and social impacts. With nearly 20% of the world's population, China has focused on economic development and health awareness to improve the care for its hearing-impaired population. Recently, the Chinese government has initiated national programs such as the China Disabled Persons Federation to fund prevention, treatment, and rehabilitation of hearing impairment. Newborn hearing screening and auditory rehabilitation programs in China have expanded exponentially with government support. While facing many challenges and overcoming obstacles, cochlear implantation (CI) programs in China have also experienced considerable growth. This review discusses the implementation of CI programs for HL in China and presents current HL data including epidemiology, newborn hearing screening, and determination of genetic etiologies. Sharing the experience in Chinese auditory rehabilitation and CI programs will shine a light on the developmental pathway of healthcare infrastructure to meet emerging needs of the hearing-impaired population in other developing countries.


Asunto(s)
Implantación Coclear , Corrección de Deficiencia Auditiva , Pérdida Auditiva/rehabilitación , China/epidemiología , Implantes Cocleares , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/epidemiología , Pruebas Auditivas , Humanos , Recién Nacido , Tamizaje Neonatal , Desarrollo de Programa
19.
Surg Endosc ; 31(1): 462-468, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27126625

RESUMEN

BACKGROUND AND AIM: It is usually difficult to obtain a good view of the dissection plane during esophageal endoscopic submucosal dissection (ESD). Therefore, the aim of this study was to investigate the efficacy and safety of clip traction in ESD for the treatment of early esophageal carcinoma (EEC) or precancerous lesions. METHODS: This is a case-matched comparative study. We selected 100 EEC patients who had undergone ESD. Fifty cases underwent ESD without clip traction (non-clip group), and 50 cases underwent ESD with clip traction (clip group). The patient-related variables, dissection time, data regarding muscularis propria injury, etc. were statistically analyzed. RESULTS: ESD was successful in all cases without complication. There were no significant differences between the two groups with respect to age, gender, the longitudinal diameter of the lesions, etc. Wide visual field exposure of the submucosal tissue below the lesion was obtained by applying clip traction. The dissection time of ESD was shorter in the clip group than in the non-clip group [22.02 (6.77) min vs 26.48 (12.56); P = 0.018] when the extent of lesion was less than half of the circumference of the esophagus; otherwise, there was no difference between the two groups (P = 0.252). Moreover, the muscularis propria injuries in the clip group were obviously less than the non-clip group (10 vs 30 %, P = 0.007). CONCLUSION: Clip traction can decrease the rate of muscularis propria injury and shorten the dissection time. It is recommended as a safe and effective auxiliary procedure for the treatment of esophageal ESD.


Asunto(s)
Resección Endoscópica de la Mucosa/métodos , Neoplasias Esofágicas/cirugía , Lesiones Precancerosas/cirugía , Instrumentos Quirúrgicos , Tracción/métodos , Anciano , Neoplasias Esofágicas/patología , Esofagoscopía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Membrana Mucosa/lesiones , Tempo Operativo , Lesiones Precancerosas/patología , Tracción/instrumentación , Resultado del Tratamiento
20.
Reprod Fertil Dev ; 29(3): 585-593, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28442065

RESUMEN

The purpose of this study was to examine the morphological and functional development of the lateral wall of the scala media of the cochlea in miniature pigs; light and transmission electron microscopy and electrophysiology were used for this purpose. We showed that the lateral wall of the scala media of the cochlea appears at embryonic Day 21 (E21) when the cochlear duct begins to form. From E28 to E49, the lateral wall can be distinguished according to its position along the cochlea. At E56, cells in the lateral wall begin to differentiate into three different types. At E70, three cell types, marginal, intermediate and basal, can be clearly distinguished. At E91, the stria vascularis is adult-like and the organ of Corti is also morphologically mature. The average endocochlear potential measured from the second turn of the cochlea (at E98, postnatal Day 1 (P1), P13 and P30) was 71.4±2.5 (n=7), 78.8±1.5 (n=10), 77.3±2.3 (n=10) and 78.0±2.1 mV (n=10), respectively. Our results suggest that in miniature pigs the stria vascularis develops during the embryonic period, concurrent with maturation of the organ of Corti. The magnitude of the endocochlear potential reached its mature level when the stria vascularis was morphologically adult-like at E98. These findings provide a morphological and functional basis for future animal studies using the miniature pig model concerning the pathogenesis of various inner-ear diseases.


Asunto(s)
Cóclea/embriología , Organogénesis/fisiología , Estría Vascular/citología , Animales , Cóclea/citología , Porcinos , Porcinos Enanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA