Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Gastroenterol ; 58(5): 444-457, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36739585

RESUMEN

BACKGROUND: Amino acid transporters play an important role in supplying nutrition to cells and are associated with cell proliferation. L-type amino acid transporter 1 (LAT1) is highly expressed in many types of cancers and promotes tumor growth; however, how LAT1 affects tumor development is not fully understood. METHODS: To investigate the role of LAT1 in intestinal tumorigenesis, mice carrying LAT1 floxed alleles that also expressed Cre recombinase from the promoter of gene encoding Villin were crossed to an ApcMin/+ background (LAT1fl/fl; vil-cre; ApcMin/+), which were subject to analysis; organoids derived from those mice were also analyzed. RESULTS: This study showed that LAT1 was constitutively expressed in normal crypt base cells, and its conditional deletion in the intestinal epithelium resulted in fewer Paneth cells. LAT1 deletion reduced tumor size and number in the small intestine of ApcMin/+ mice. Organoids derived from LAT1-deleted ApcMin/+ intestinal crypts displayed fewer spherical organoids with reduced Wnt/ß-catenin target gene expression, suggesting a low tumor-initiation capacity. Wnt3 expression was decreased in the absence of LAT1 in the intestinal epithelium, suggesting that loss of Paneth cells due to LAT1 deficiency reduced the risk of tumor initiation by decreasing Wnt3 production. CONCLUSIONS: LAT1 affects intestinal tumor development in a cell-extrinsic manner through reduced Wnt3 expression in Paneth cells. Our findings may partly explain how nutrient availability can affect the risk of tumor development in the intestines.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon , Sistema de Transporte de Aminoácidos y+L , Neoplasias Intestinales , Células de Paneth , Animales , Ratones , Transformación Celular Neoplásica/genética , Mucosa Intestinal/patología , Neoplasias Intestinales/metabolismo , Intestino Delgado/patología , Intestinos , Células de Paneth/metabolismo , Células de Paneth/patología , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Sistema de Transporte de Aminoácidos y+L/metabolismo
3.
J Int Med Res ; 50(11): 3000605221134467, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36448965

RESUMEN

OBJECTIVE: To identify whether median nerve stimulation (MNS) may be a potential candidate for the treatment of consciousness disorders via a systematic review and meta-analysis. METHODS: PubMed, Cochrane Library, China National Knowledge Infrastructure, Chinese VIP Information, Wanfang, and SinoMed databases were searched. Risk of bias was assessed using the Cochrane Collaboration's tool. The Glasgow Coma Scale (GCS), Disability Rating Scale (DRS), electroencephalogram (EEG), days in the Intensive Care Unit (ICU), and cerebral blood flow measures were compared between the median nerve stimulation and control groups. The meta-analysis was conducted using Review Manager software. RESULTS: We identified 2244 studies, of which 23 (with data from 1856 patients) qualified for the analysis. MNS improved GCS scores (mean difference [MD] = 2.15), EEG scores (MD = 1.61), cerebral mean blood flow velocity (MD = 4.23), and cerebral systolic blood flow velocity (MD = 10.51). Furthermore, it decreased DRS scores (MD = -1.77) and days in the ICU (MD = -2.02). The effects of MNS on GCS scores increased with longer treatments (1 week, MD = 1.03; 1 month, MD = 2.35) and were better with right MNS (right, MD = 2.36; bilateral, MD = 1.72). CONCLUSIONS: MNS may promote recovery from consciousness disorders.


Asunto(s)
Trastornos de la Conciencia , Nervio Mediano , Humanos , Trastornos de la Conciencia/terapia , Estimulación Eléctrica , Electroencefalografía , Pueblo Asiatico
4.
Front Pharmacol ; 13: 965661, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36204225

RESUMEN

Dexmedetomidine (DEX) is a highly selective α2 receptor agonist that is routinely used in the clinic for sedation and anesthesia. Recently, an increasing number of studies have shown that DEX has a protective effect against brain injury caused by traumatic brain injury (TBI), subarachnoid hemorrhage (SAH), cerebral ischemia and ischemia-reperfusion (I/R), suggesting its potential as a neuroprotective agent. Here, we summarized the neuroprotective effects of DEX in several models of neurological damage and examined its mechanism based on the current literature. Ultimately, we found that the neuroprotective effect of DEX mainly involved inhibition of inflammatory reactions, reduction of apoptosis and autophagy, and protection of the blood-brain barrier and enhancement of stable cell structures in five way. Therefore, DEX can provide a crucial advantage in neurological recovery for patients with brain injury. The purpose of this study was to further clarify the neuroprotective mechanisms of DEX therefore suggesting its potential in the clinical management of the neurological injuries.

5.
J Food Sci ; 87(5): 2173-2184, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35411589

RESUMEN

Resistant starch (RS) has been reported to improve steatosis as well as obesity. Type 4 resistant starch (RS4), a chemically modified starch, is particularly hard to digest and suggesting higher efficacy. However, because the effects of RS4 on steatosis are not yet fully understood, the effects of RS4 on steatosis were examined using a murine high-fat diet model. Seven-week-old male mice were divided into three groups and fed a normal diet, a high-fat diet (HFD), or a high-fat diet with added RS (HFD + RS). Amylofiber SH® produced from acid-treated corn starch was used as the dietary RS. At 22 weeks old, hepatic steatosis and short chain fatty acid (SCFA) content and gut microbiota in cecum stool samples were analyzed. The ratio of body weight to 7 weeks was significantly suppressed in the HFD + RS group compared to the HFD group (132.2 ± 1.4% vs. 167.2 ± 3.9%, p = 0.0076). Macroscopic and microscopic steatosis was also suppressed in the HFD + RS group. Analysis of cecum stool samples revealed elevated SCFA levels in the HFD + RS group compared with the HFD group. Metagenome analysis revealed that Bifidobacterium (17.9 ± 1.9% vs. 3.6 ± 0.7%, p = 0.0019) and Lactobacillus (14.8 ± 3.4% vs. 0.72 ± 0.23%, p = 0.0045), which degrade RS to SCFA, were more prevalent in the HFD + RS group than the HFD group. In conclusion, RS4 suppressed steatosis, and increased Bifidobacterium and Lactobacillus, and SCFAs. RS4 may prevent steatosis by modulating the intestinal environment.


Asunto(s)
Dieta Alta en Grasa , Hígado Graso , Amilosa , Animales , Bifidobacterium/metabolismo , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos Volátiles/metabolismo , Masculino , Ratones , Almidón Resistente , Almidón/farmacología , Zea mays/química
6.
Med Princ Pract ; 31(1): 39-46, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34818236

RESUMEN

OBJECTIVE: G protein-coupled receptor 43 (GPR43), a receptor for short-chain fatty acids, plays a role in suppressing tumor growth; however, the detailed underlying mechanism needs to be comprehensively elucidated. In this study, we investigated the role of GPR43 in inhibiting tumor growth using ApcMin/+, a murine model of intestinal tumors. MATERIALS AND METHODS: Using GPR43-/- ApcMin/+ and GPR43+/- ApcMin/+ mice, the number of tumors was analyzed at the end of the experimental period. Immunohistochemistry, quantitative polymerase chain reaction, and Western blotting were performed to analyze cellular proliferation and proliferation-associated signal pathways. RESULTS: Our results revealed that GPR43 deficiency resulted in increased tumor numbers in ApcMin/+ mice. Ki67 was highly expressed in GPR43-/- mice (p > 0.05). Increased expression levels of proinflammatory cytokines, including interleukin-6 and tumor necrosis factor-α, and amino acid transporters were not observed in GPR43-deficient mice compared to GPR43-sufficient mice. Furthermore, GPR43-deficient tumor tissues showed enhanced mammalian target of rapamycin-mediated phosphorylated ribosomal protein S6 kinase beta-1 (p > 0.05) and phosphorylated eukaryotic translation initiation factor 4E-binding protein 1 (p > 0.05), but not Akt (protein kinase B) phosphorylation (p = 0.7088). CONCLUSION: Collectively, GPR43 affords protection against tumor growth at least partly through inhibition of the mammalian target of rapamycin complex 1 pathway.


Asunto(s)
Ácidos Grasos Volátiles , Neoplasias Intestinales , Receptores Acoplados a Proteínas G , Animales , Colon/patología , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/farmacología , Mucosa Intestinal , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Mamíferos/metabolismo , Ratones , Receptores Acoplados a Proteínas G/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
Kobe J Med Sci ; 66(4): E139-E148, 2021 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-33994517

RESUMEN

Daikenchuto (TU-100) is herbal medicine which predominantly contains ginger, Japanese pepper, and ginseng. We investigated whether TU-100 can affect the composition of gut flora and intestinal tumor development using ApcMin/+ mice, a murine model of intestinal tumor. Bacterial 16S rRNA sequencing and short-chain fatty acid analysis were performed on faecal samples. Tumor number and size were analysed. Any change in gene expression of the tumor tissues was assessed by real-time PCR. Principal coordinate analysis (PCoA) showed that the faecal microbiota cluster of TU-100-fed mice was different from the microbiota of control mice. However, no significant difference was observed in the concentration of short-chain fatty acids, tumor number, and gene expression levels between the two groups. Our data showed that TU-100 can affect the intestinal environment; however, it does not contribute in tumor progression or inhibition in our setting.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Medicina de Hierbas , Mucosa Intestinal/efectos de los fármacos , Neoplasias Intestinales/tratamiento farmacológico , Extractos Vegetales/farmacología , Animales , Heces , Microbioma Gastrointestinal/genética , Neoplasias Intestinales/patología , Ratones , Microbiota , Panax , ARN Ribosómico 16S , Reacción en Cadena en Tiempo Real de la Polimerasa , Zanthoxylum , Zingiberaceae
8.
Int J Ophthalmol ; 12(10): 1524-1530, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31637186

RESUMEN

AIM: To reveal the cytotoxicity and related mechanisms of gatifloxacin (GFX) to stromal fibroblasts (SFs) in vitro. METHODS: SFs were treated with GFX at different concentrations (0.009375%-0.3%), and their viability was detected by MTT method. The cell morphology was observed using light/transmission electron microscope. The plasma membrane permeability was measured by AO/EB double-staining. Then cell cycle, phosphatidylserine (PS) externalization, and mitochondrial transmembrane potential (MTP) were analyzed by flow cytometry. DNA damage was analyzed by electrophoresis and immunostaining. ELISA was used to evaluate the caspase-3/-8/-9 activation. Finally, Western blotting was applied for detecting the expressions of apoptosis-related proteins. RESULTS: Morphological changes and reduced viability of GFX-treated SFs demonstrated that GFX above 0.009375% had cytotoxicity to SFs with dependence of concentration and time. GFX-treating cells also showed G1 phase arrest, increased membrane permeability, PS externalization and DNA damage, which indicated that GFX induced apoptosis of SFs. Additionally, GFX could activate the caspase-8, caspase-9, and caspase-3, induce MTP disruption, downregulate B-cell leukemia-2 (Bcl-2) and B-cell leukemia-XL (Bcl-XL), and upregulate Bcl-2 assaciated X protein (Bax), Bcl-2-associated death promoter (Bad), Bcl-2 interacting domain (Bid) and cytoplasmic cytochrome C in SFs, suggesting that caspase-dependent extrinsic and intrinsic pathways were related to GFX-contributed apoptosis of SFs. CONCLUSION: The cytotoxicity of GFX induces apoptosis of SFs through triggering the caspase-dependent extrinsic and intrinsic pathways.

9.
J Toxicol Sci ; 40(4): 427-36, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26165639

RESUMEN

Proparacaine is a widely used topical anesthetic in ophthalmic optometry and surgery, and has been reported to have cytotoxic effects on rabbit corneal endothelial cells after prolonged and repeated usage. Since rabbit is an exceptive mammal whose corneal endothelial cells still maintaining proliferation abilities even in adulthood, whether proparacaine has cytotoxic effects on human corneal endothelial (HCE) cells need to be further verified. Our objectives in the present study were to investigate the cytotoxicity to HCE cells of proparacaine and its underlying mechanisms in vitro and verify the cytotoxicity using cat corneal endothelial (CCE) cells in an in vivo model of cat corneas. Cytotoxic evaluation results indicated that a dose- and time-dependent toxic response of HCE cells to proparacaine over 0.03125% was rated based on morphology and viability, and a toxic response of CCE cells to 0.5% (clinical applied dosage) proparacaine was also rated based on cell density and histology. Importantly, treatment with proparacaine resulted in significant elevation of plasma membrane permeability, cell cycle arrest at S phase, fragmentation of genomic DNA, formation of apoptotic bodies, and externalization of phosphatidylserine (PS) of HCE cells. Moreover, proparacaine demonstrated disrupting effects on mitochondrial transmembrane potential (MTP) of HCE cells and activating effects on caspase-3, -8 and -9. This study demonstrates that proparacaine has notable cytotoxicity to both HCE cells in vitro and CCE cells in vivo, and its dose- and time-dependent cytotoxicity to HCE cells is achieved by inducing apoptosis via a mitochondrion-mediated caspase-dependent pathway. These findings provide new insights into the cytotoxicity and apoptosis-inducing effect of local anesthetics which should be used with great caution in the eye clinic.


Asunto(s)
Anestésicos Locales/toxicidad , Células Endoteliales/efectos de los fármacos , Endotelio Corneal/efectos de los fármacos , Propoxicaína/toxicidad , Animales , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Gatos , Ciclo Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Fragmentación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Corneal/citología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fosfatidilserinas/metabolismo , Conejos , Fase S/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...