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








Base de dados
Intervalo de ano de publicação
1.
Anesth Analg ; 137(6): 1257-1269, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37973132

RESUMO

BACKGROUND: Simple and rapid tools for screening high-risk patients for perioperative neurocognitive disorders (PNDs) are urgently needed to improve patient outcomes. We developed an online tool with machine-learning algorithms using routine variables based on multicenter data. METHODS: The entire dataset was composed of 49,768 surgical patients from 3 representative academic hospitals in China. Surgical patients older than 45 years, those undergoing general anesthesia, and those without a history of PND were enrolled. When the patient's discharge diagnosis was PND, the patient was in the PND group. Patients in the non-PND group were randomly extracted from the big data platform according to the surgical type, age, and source of data in the PND group with a ratio of 3:1. After data preprocessing and feature selection, general linear model (GLM), artificial neural network (ANN), and naive Bayes (NB) were used for model development and evaluation. Model performance was evaluated by the area under the receiver operating characteristic curve (ROCAUC), the area under the precision-recall curve (PRAUC), the Brier score, the index of prediction accuracy (IPA), sensitivity, specificity, etc. The model was also externally validated on the multiparameter intelligent monitoring in intensive care (MIMIC) Ⅳ database. Afterward, we developed an online visualization tool to preoperatively predict patients' risk of developing PND based on the models with the best performance. RESULTS: A total of 1051 patients (242 PND and 809 non-PND) and 2884 patients (6.2% patients with PND) were analyzed on multicenter data (model development, test [internal validation], external validation-1) and MIMIC Ⅳ dataset (external validation-2). The model performance based on GLM was much better than that based on ANN and NB. The best-performing GLM model on validation-1 dataset achieved ROCAUC (0.874; 95% confidence interval [CI], 0.833-0.915), PRAUC (0.685; 95% CI, 0.584-0.786), sensitivity (72.6%; 95% CI, 61.4%-81.5%), specificity (84.4%; 95% CI, 79.3%-88.4%), Brier score (0.131), and IPA (44.7%), and of which the ROCAUC (0.761, 95% CI, 0.712-0.809), the PRAUC (0.475, 95% CI, 0.370-0.581), Brier score (0.053), and IPA (76.8%) on validation-2 dataset. Afterward, we developed an online tool (https://pnd-predictive-model-dynnom.shinyapps.io/ DynNomapp/) with 10 routine variables for preoperatively screening high-risk patients. CONCLUSIONS: We developed a simple and rapid online tool to preoperatively screen patients' risk of PND using GLM based on multicenter data, which may help medical staff's decision-making regarding perioperative management strategies to improve patient outcomes.


Assuntos
Tomada de Decisão Clínica , Nomogramas , Humanos , Adulto , Teorema de Bayes , Algoritmos , Fatores de Risco , Estudos Retrospectivos
2.
Front Med (Lausanne) ; 10: 1151996, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37601798

RESUMO

Objective: Non-invasive methods for hemoglobin (Hb) monitoring can provide additional and relatively precise information between invasive measurements of Hb to help doctors' decision-making. We aimed to develop a new method for Hb monitoring based on mask R-CNN and MobileNetV3 with eye images as input. Methods: Surgical patients from our center were enrolled. After image acquisition and pre-processing, the eye images, the manually selected palpebral conjunctiva, and features extracted, respectively, from the two kinds of images were used as inputs. A combination of feature engineering and regression, solely MobileNetV3, and a combination of mask R-CNN and MobileNetV3 were applied for model development. The model's performance was evaluated using metrics such as R2, explained variance score (EVS), and mean absolute error (MAE). Results: A total of 1,065 original images were analyzed. The model's performance based on the combination of mask R-CNN and MobileNetV3 using the eye images achieved an R2, EVS, and MAE of 0.503 (95% CI, 0.499-0.507), 0.518 (95% CI, 0.515-0.522) and 1.6 g/dL (95% CI, 1.6-1.6 g/dL), which was similar to that based on MobileNetV3 using the manually selected palpebral conjunctiva images (R2: 0.509, EVS:0.516, MAE:1.6 g/dL). Conclusion: We developed a new and automatic method for Hb monitoring to help medical staffs' decision-making with high efficiency, especially in cases of disaster rescue, casualty transport, and so on.

3.
J Clin Transl Hepatol ; 11(5): 1150-1160, 2023 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-37577229

RESUMO

Background and Aims: The results of basic research implicate the vascular endothelial growth factor (VEGF) family as a potential target of hepatopulmonary syndrome (HPS). However, the negative results of anti-angiogenetic therapy in clinical studies have highlighted the need for markers for HPS. Therefore, we aimed to determine whether VEGF family members and their receptors can be potential biomarkers for HPS through clinical and experimental studies. Methods: Clinically, patients with chronic liver disease from two medical centers were enrolled and examined for HPS. Patients were divided into HPS, intrapulmonary vascular dilation [positive contrast-enhanced echocardiography (CEE) and normal oxygenation] and CEE-negative groups. Baseline information and perioperative clinical data were compared between HPS and non-HPS patients. Serum levels of VEGF family members and their receptors were measured. In parallel, HPS rats were established by common bile duct ligation. Liver, lung and serum samples were collected for the evaluation of pathophysiologic changes, as well as the expression levels of the above factors. Results: In HPS rats, all VEGF family members and their receptors underwent significant changes; however, only soluble VEGFR1 (sFlt-1) and the sFlt-1/ placental growth factor (PLGF) ratio were changed in almost the same manner as those in HPS patients. Furthermore, through feature selection and internal and external validation, sFlt-1 and the sFlt-1/PLGF ratio were identified as the most important variables to distinguish HPS from non-HPS patients. Conclusions: Our results from animal and human studies indicate that sFlt-1 and the sFlt-1/PLGF ratio in serum are potential markers for HPS.

5.
Med Sci Monit ; 27: e932348, 2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33762560

RESUMO

Retracted, due to breach of publishing guidelines, following the identification of non-original and manipulated figure images. Reference: Xiaokang Liu, Yu Li, Qiang Ma, Yanwei Wang, Ai Lin Song: Withaferin-A Inhibits Growth of Drug-Resistant Breast Carcinoma by Inducing Apoptosis and Autophagy, Endogenous Reactive Oxygen Species (ROS) Production, and Inhibition of Cell Migration and Nuclear Factor kappa B (Nf-kappaB)/Mammalian Target of Rapamycin (m-TOR) Signalling Pathway. Med Sci Monit 2019; 25:6855-6863. 10.12659/MSM.916931.

6.
Med Sci Monit ; 25: 6855-6863, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31512681

RESUMO

BACKGROUND The main purpose of this study was to assess in vitro and in vivo the anticancer effect of withaferin-A in human breast carcinoma cells (MDA-MB-231), and to assess its effects on autophagy, cell apoptosis, ROS production, cell migration and invasion, and Nf-kappaB/m-TOR signalling pathway. MATERIAL AND METHODS Proliferation of MDA-MB-231 cells at various doses of the drug was studied by CCK8 cell viability assay. Effects on cell apoptosis were studied by fluorescence microscopy in combination with flow cytometry and Western blot analysis. Effects on autophagy were evaluated by transmission electron microscopy and Western blot. Effects on cellular migration were examined in vitro by wound healing assay. RESULTS The results indicated that withaferin-A led to significant reduction of MDA-MB-231 cell viability. The anticancer action of withaferin-A was shown to be due to the stimulation of autophagy, which was accompanied by enhancement of LC3 expression. Withaferin-A prompted mitochondrial apoptosis, which was also associated with increased level of Bax and decreased Bcl-2 in MDA-MB-231 cells. It was also observed that withaferin-A has decreases cellular migration and invasion of the tested human breast cancer cells. The effects of withaferin-A were also investigated in vivo, and it was found that this molecule could inhibit the growth of tumor xenografts in tested mice. Withaferin-A led to suppression of the Nf-kappaB/m-TOR signalling pathway. CONCLUSIONS In brief, the withaferin-A molecule has great potential as an anticancer agent against drug-resistant breast cancer, and as such needs to be further studied in detail.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Vitanolídeos/farmacologia , Neoplasias da Mama/ultraestrutura , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Transdução de Sinais/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco , Vitanolídeos/química , Proteína X Associada a bcl-2/metabolismo
7.
Chin Med J (Engl) ; 132(9): 1071-1078, 2019 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-30896562

RESUMO

BACKGROUND: Colorectal cancer is the third most common cancer worldwide and still lack of effective therapy so far. Petasin, a natural product found in plants of the genus Petasites, has been reported to possess anticancer activity. The present study aimed to investigate the anticolon cancer activity of petasin both in vitro and in vivo. The molecular mechanism of petasin was also further explored. METHODS: Caco-2, LoVo, SW-620, and HT-29 cell lines were used to detect the inhibitory effect of petasin on colon cancer proliferation. Cell viability was determined using the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assay. Cell apoptosis was analyzed by flow cytometry. Hoechst 33258 staining was used to visualize morphological changes. Cell migration was assessed using a wound-healing migration assay, and cell invasion was investigated using Transwell chambers. Western blotting assays were employed to evaluate the expression levels of proteins in the protein kinase B/mammalian target of rapamycin (Akt/mTOR) signaling pathway. Finally, in vivo activity of petasin was evaluated using the SW-620 subcutaneous tumor model established in Balb/c nude mice. Twelve rats were randomly divided into control group and 10 mg/kg petasin group. The tumor volume was calculated every 7 days for 28 days. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was performed to assess the apoptotic effect of petasin. Differences between two groups were assessed by analysis of independent-sample t tests. RESULTS: Petasin significantly inhibited the proliferation of human colon carcinoma cell lines, induced apoptosis, and suppressed migration and invasion in SW-620 cells. Western blotting results showed that petasin decreased the phosphorylation of Akt (1.01 ±â€Š0.16 vs. 0.74 ±â€Š0.06, P = 0.042), mTOR (0.71 ±â€Š0.12 vs. 0.32 ±â€Š0.11, P = 0.013), and P70S6K (1.23 ±â€Š0.21 vs. 0.85 ±â€Š0.14, P = 0.008), elevated the expression of caspase-3 (0.41 ±â€Š0.09 vs. 0.74 ±â€Š0.12, P = 0.018) and caspase-9 (1.10 ±â€Š0.27 vs. 1.98 ±â€Š0.22, P = 0.009), decreased the Bcl-2 protein (2.75 ±â€Š0.47 vs. 1.51 ±â€Š0.36, P = 0.008), downregulated the expression of matrix metalloproteinase (MMP)-3 (1.51 ±â€Š0.31 vs. 0.82 ±â€Š0.11, P = 0.021) and MMP-9 (1.56 ±â€Š0.32 vs. 0.94 ±â€Š0.15, P = 0.039) in SW-620 cell. In vivo, 10 mg/kg petasin inhibited tumor growth in Balb/c nude mice (924.18 ±â€Š101.23 vs. 577.67 ±â€Š75.12 mm at day 28, P = 0.001) and induced apoptosis (3.6 ±â€Š0.7% vs. 36.0 ±â€Š4.9%, P = 0.001) in tumor tissues. CONCLUSIONS: Petasin inhibits the proliferation of colon cancer SW-620 cells via inactivating the Akt/mTOR pathway. Our findings suggest petasin as a potential candidate for colon cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sesquiterpenos/uso terapêutico , Serina-Treonina Quinases TOR/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células CACO-2 , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células HT29 , Humanos , Marcação In Situ das Extremidades Cortadas , Metaloproteinase 3 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética
8.
J BUON ; 24(6): 2442-2447, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31983118

RESUMO

PURPOSE: Xanthohumol is a prenylated flavonoid of plant origin and has been reported to exhibit a spectrum of pharmacological properties including anticancer effects. However, the anticancer properties of Xanthohumol have not been thoroughly evaluated against drug-resistant colon cancer cells. This study was undertaken to evaluate the anticancer effects of Xanthohumol against the human colon cancer cell line HT-29 and normal CDD-18Co cell line. METHODS: HT-29 cell viability was evaluated by cell counting kit-8 (CCK8) assay. Apoptotic effects were examined by fluorescence microscopy using DAPI staining and flow cytometry using annexin V/propidium iodide (PI) staining. Effects on cell cycle were studied by flow cytometry while western blot analysis was done to study effects on protein expressions. RESULTS: The results showed that Xanthohumol causes a dramatic decrease in the HT-29 cell viability with an IC50 of 10 µM. However, an IC50 >100 µM for Xanthohumol against the normal CDD-18Co cells suggested cancer cell specific activity. DAPI staining revealed nuclear fragmentation, suggesting xanthohumol induces apoptosis in HT-29 cells. Xanthohumol also caused activation of caspase-3 and 9 and increased the Bax/Bcl-2 ratio. Cell cycle analysis showed that this molecule caused arrest of the HT-29 cells at the G2/M phase of the cell cycle. The induction of G2/M cell cycle was also accompanied with depletion of the expression of cyclin B1. The effects of Xanthohumol were also investigated on the Ras/MEK/ERK signalling pathway which revealed that Xanthohumol also blocks the MEK/ERK signalling pathway in colon cancer cells in a concentration-dependent manner. CONCLUSIONS: Xanthohumol may prove an efficient lead molecule for the development of more potent anticancer agents through semi-synthetic approaches.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Neoplasias do Colo/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Flavonoides/farmacologia , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Propiofenonas/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células Tumorais Cultivadas , Proteínas ras/metabolismo
9.
Chin Med J (Engl) ; 124(4): 574-80, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21362284

RESUMO

BACKGROUND: Donor-pretreatment with ulinastatin may influence the liver graft during cold preservation. The aim of this research was to determine whether pretreatment of donor liver with Ulinastatin can attenuate cold preservation injury, and to explore the mechanism by which Ulinastatin affects the donor liver graft. METHODS: One hundred and forty-four Wistar rats were divided into the Ulinastatin treatment group (T group) pretreated with Ulinastatin 50 000 U/kg and control group (C group) treated with 0.9% normal saline via peritoneal injection prior to the anesthetization. After the abdominal cavity was opened and perfused with cold Ringer's lactate solution, the liver was harvested. The harvested liver was preserved in cold Ringer's lactate solution for 0, 2, 6, 24 hours, at which time the liver tissue was sampled for determination of dry weight and wet weight, Na(+)-K(+)-ATPase and Ca(2+)-ATPase activity, lactic acid dehydrogenase (LDH) activity, lactic acid and malondialdehyde levels. Light microscopy and electron microscopy were used to observe liver morphology. The liver cold-preservation solution was taken for measurement of aspartate aminotransferase (AST) and alanine transaminase (ALT) levels. Correlation between ATPase activity and lactic acid level was analyzed by SPSS 13.0 for Windows. RESULTS: The morphology in the T group had improved cell boundaries vs. the C group at each time point. Dry weight to wet weight in the T group was lower than in the C group at 6 hours (P < 0.05), but the difference was not significant at 24 hours. ALT levels in the T group were lower than that in the C group at 6 hours (P < 0.05) and 24 hours (P < 0.01). AST levels in the T group were lower than those in the C group at 2 hours (P < 0.05), 6 hours (P < 0.01) and 24 hours (P < 0.01). Na(+)-K(+)-ATPase activity in the T group was higher than in the C group and the mean difference between two groups was significant at 0 hour (P < 0.05) and 2 hours (P < 0.05). Ca(2+)-ATPase activity in the T group was higher than in the C group with the mean difference between two groups significant at 2 hours (P < 0.05). The T group had increased lactic acid levels at 0 hour (P < 0.01) and 2 hours (P < 0.05) compared with the C group, but there was no influence on the LDH activity at the same time. There were no obvious differences in the levels of malondialdehyde between the two groups at any time point. A linear correlation between Na(+)-K(+)-ATPase activity and lactic acid levels (r = 0.295, P < 0.05) was found. CONCLUSIONS: Donor-pretreatment with ulinastatin may protect the cells in a liver graft from ischemia injury during cold preservation; the mechanism may be due to its promotion for cell glycolysis and its preservation of ATPase activity.


Assuntos
Criopreservação/métodos , Glicoproteínas/farmacologia , Transplante de Fígado/métodos , Fígado/efeitos dos fármacos , Preservação de Órgãos/métodos , Animais , ATPases Transportadoras de Cálcio/metabolismo , Sobrevivência de Enxerto/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Fígado/metabolismo , Malondialdeído/metabolismo , Ratos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/metabolismo , Doadores de Tecidos
10.
World J Gastroenterol ; 12(25): 3977-82, 2006 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-16810743

RESUMO

AIM: To study the expression level and localization of insulin-like growth factor -I receptor (IGF-IR) in HepG2 cells and Chang liver cells, and to observe the effect of anti-IGF-IR monoclonal antibody (alphaIR3) on the growth of HepG2 cells. METHODS: The expression of IGF-IR in HepG2 cells and Chang liver cells was detected by immunohistochemistry. The influences of alphaIR3 on proliferation and apoptosis were examined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and electron microscopy, respectively. Flow cytometry (FCM) was applied for the analysis of cell cycle and apoptosis was observed under electron microscope. RESULTS: IGF-IR was located in the membranes of both HepG2 and Chang liver cell lines, and the expression level of IGF-IR was higher in HepG2 cells than in Chang liver cells. Treated with 0.1 microg/mL alphaIR3 for 48 h in vitro, the cell growth index (GI) of HepG2 cells was significantly higher than that of control (103.41% vs 100%, P<0.01). However, the alphaIR3 for 24 h at final concentration of 4.0 microg/mL made the GI of HepG2 cells lower than that of control (93.37% vs 100%, P<0.01). Compared with control, treated with alphaIR3 for 48 h at final concentrations ranging from 1.0 microg/mL to 4.0 microg/mL markedly reduced the GIs of HepG2 cells (97.63%, 97.16%, 95.13%, 92.53% vs 100%, P<0.05 or P<0.01), treated with alphaIR3 for 72 h at final concentrations ranging from 0.2 microg/mL to 4.0 microg/mL decreased the GIs of HepG2 cells obviously (95%, 91.63%, 90.77%, 89.84%, 88.51% vs 100%, P<0.01), and treated with alphaIR3 for 96 h at final concentrations ranging from 0.5 microg/mL to 4.0 microg/mL made GIs of HepG2 cells lower significantly (88.86%, 83.97%, 79.81%, 77.24%, 70.51% vs 100%, P<0.05 or P<0.01). Moreover, treated with alphaIR3 from 24 h to 96 h at final concentrations ranging from 0.2 microg/mL to 4.0 microg/mL reduced the GI of HepG2 cells from 97.63% to 70.51% in a dose- and time-dependent manner. Also, alphaIR3 treatment for 72 h at final concentration from 0.5 microg/mL to 2.0 microg/mL increased the proportion of G0/G1 phase cells (61.73%, 67.1%, 83.7%, 76.87% vs 44.47%, P<0.01) and significantly decreased that of S phase cells (28.63%, 25.13%, 15.63%, 23.13% vs 53.17%, P<0.01), in contrast to the proportion of G2/M phase cells. The apoptotic rates of HepG2 cells were increased more than that of control (7.83%, 16.13%, 21.1%, 37.73% vs 4.13%, P<0.01). CONCLUSION: The malignant cell phenotype of human hepatocarcinoma cell is related to overexpression of IGF-IR. The blockage of IGF-IR with alphaIR3 may contribute to the inhibition of proliferation and induction of apoptosis in HepG2 cells.


Assuntos
Anticorpos Monoclonais/farmacologia , Proliferação de Células/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Receptor IGF Tipo 1/metabolismo , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/ultraestrutura , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Hepatócitos/ultraestrutura , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/ultraestrutura , Receptor IGF Tipo 1/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA