Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Chin Med J (Engl) ; 134(19): 2333-2339, 2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34483253

ABSTRACT

BACKGROUND: A deep learning model (DLM) that enables non-invasive hypokalemia screening from an electrocardiogram (ECG) may improve the detection of this life-threatening condition. This study aimed to develop and evaluate the performance of a DLM for the detection of hypokalemia from the ECGs of emergency patients. METHODS: We used a total of 9908 ECG data from emergency patients who were admitted at the Second Affiliated Hospital of Nanchang University, Jiangxi, China, from September 2017 to October 2020. The DLM was trained using 12 ECG leads (lead I, II, III, aVR, aVL, aVF, and V1-6) to detect patients with serum potassium concentrations <3.5 mmol/L and was validated using retrospective data from the Jiangling branch of the Second Affiliated Hospital of Nanchang University. The blood draw was completed within 10 min before and after the ECG examination, and there was no new or ongoing infusion during this period. RESULTS: We used 6904 ECGs and 1726 ECGs as development and internal validation data sets, respectively. In addition, 1278 ECGs from the Jiangling branch of the Second Affiliated Hospital of Nanchang University were used as external validation data sets. Using 12 ECG leads (leads I, II, III, aVR, aVL, aVF, and V1-6), the area under the receiver operating characteristic curve (AUC) of the DLM was 0.80 (95% confidence interval [CI]: 0.77-0.82) for the internal validation data set. Using an optimal operating point yielded a sensitivity of 71.4% and a specificity of 77.1%. Using the same 12 ECG leads, the external validation data set resulted in an AUC for the DLM of 0.77 (95% CI: 0.75-0.79). Using an optimal operating point yielded a sensitivity of 70.0% and a specificity of 69.1%. CONCLUSIONS: In this study, using 12 ECG leads, a DLM detected hypokalemia in emergency patients with an AUC of 0.77 to 0.80. Artificial intelligence could be used to analyze an ECG to quickly screen for hypokalemia.


Subject(s)
Deep Learning , Hypokalemia , Artificial Intelligence , Electrocardiography , Humans , Hypokalemia/diagnosis , Retrospective Studies
2.
Sensors (Basel) ; 19(22)2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31731740

ABSTRACT

Due to the influence of altitude change on a driver's heart rate, it is difficult to estimate driving fatigue using heart rate variability (HRV) at a road segment with frequent and rapid altitude change. Accordingly, a novel method of driving fatigue estimation for driving at plateau area with frequent altitude changes is proposed to provide active safety monitoring in real time. A naturalistic driving experiment at Qinghai-Tibet highway was conducted to collect drivers' electrocardiogram data and eye movement data. The results of the eye movement-based method were selected to enhance the HRV-based driving fatigue degree estimation method. A correction factor was proposed to correct the HRV-based method at the plateau area so that the estimation can be made via common portable devices. The correction factors for both upslope and downslope segments were estimated using the field experiment data. The results on the estimation of revised driving fatigue degree can describe the driver's fatigue status accurately for all the road segments at the plateau area with altitudes from 3540 to 4767 m. The results can provide theoretical references for the design of the devices of active safety prevention.


Subject(s)
Altitude , Automobile Driving , Fatigue/diagnosis , Monitoring, Physiologic , Accidents, Traffic/prevention & control , Heart Rate/physiology , Humans , Reaction Time/physiology , Safety
3.
Onco Targets Ther ; 9: 2221-6, 2016.
Article in English | MEDLINE | ID: mdl-27143917

ABSTRACT

OBJECTIVE: To explore the expression and prognosis significance of BC200 in esophageal squamous cell carcinoma (ESCC) patients who received radical resection. METHODS: We used quantitative real-time polymerase chain reaction to detect the expression level of BC200 in cancer tissue and paired adjacent normal tissue samples from 70 ESCC patients who received radical surgical resection and analyzed the correlation of the relative expression level of BC200 with clinical-pathological features and prognosis. RESULTS: We found that the relative expression of BC200 was significantly higher in ESCC tissues compared with adjacent normal tissue samples (P=0.023). But the expression of BC200 were not related to clinical-pathological features, such as age, TNM stages, and histological grade (P>0.05). Kaplan-Meier analysis showed that high expression levels of BC200 were correlated with poor prognosis in ESCC patients. Patients with a high level of BC200 had a shorter disease-free survival and overall survival than those with low BC200 expression (P=0.034 and P=0.031, respectively). On multivariate analysis, the hazard ratio (HR) of BC200 expression was 2.17 (95% confidence interval [CI]=1.12-4.19, P=0.022) for disease-free survival and 2.24 (95% CI=1.12-4.49, P=0.023) for overall survival. CONCLUSION: Our results indicate that high expression of BC200 reflects poor prognosis and could serve as a novel predictive marker for ESCC patients who received radical resection.

4.
J Pharmacol Exp Ther ; 340(2): 422-32, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22085648

ABSTRACT

Phospho-nonsteroidal anti-inflammatory drugs (phospho-NSAIDs) are novel NSAID derivatives with improved anticancer activity and reduced side effects in preclinical models. Here, we studied the metabolism of phospho-NSAIDs by carboxylesterases and assessed the impact of carboxylesterases on the anticancer activity of phospho-NSAIDs in vitro and in vivo. The expression of human liver carboxylesterase (CES1) and intestinal carboxylesterase (CES2) in human embryonic kidney 293 cells resulted in the rapid intracellular hydrolysis of phospho-NSAIDs. Kinetic analysis revealed that CES1 is more active in the hydrolysis of phospho-sulindac, phospho-ibuprofen, phospho-naproxen, phospho-indomethacin, and phospho-tyrosol-indomethacin that possessed a bulky acyl moiety, whereas the phospho-aspirins are preferentially hydrolyzed by CES2. Carboxylesterase expression leads to a significant attenuation of the in vitro cytotoxicity of phospho-NSAIDs, suggesting that the integrity of the drug is critical for anticancer activity. Benzil and bis-p-nitrophenyl phosphate (BNPP), two carboxylesterase inhibitors, abrogated the effect of carboxylesterases and resensitized carboxylesterase-expressing cells to the potent cytotoxic effects of phospho-NSAIDs. In mice, coadministration of phospho-sulindac and BNPP partially protected the former from esterase-mediated hydrolysis, and this combination more effectively inhibited the growth of AGS human gastric xenografts in nude mice (57%) compared with phospho-sulindac alone (28%) (p = 0.037). Our results show that carboxylesterase mediates that metabolic inactivation of phospho-NSAIDs, and the inhibition of carboxylesterases improves the efficacy of phospho-NSAIDs in vitro and in vivo.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carboxylesterase/metabolism , Carboxylic Ester Hydrolases/metabolism , Organophosphates/metabolism , Organophosphates/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/blood , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspirin/analogs & derivatives , Aspirin/metabolism , Aspirin/pharmacology , Carboxylesterase/antagonists & inhibitors , Carboxylesterase/genetics , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/genetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Interactions/physiology , Drug Therapy, Combination/methods , Enzyme Inhibitors/pharmacology , Female , HEK293 Cells , Humans , Hydrolysis , Ibuprofen/analogs & derivatives , Ibuprofen/metabolism , Ibuprofen/pharmacology , Indomethacin/analogs & derivatives , Indomethacin/metabolism , Indomethacin/pharmacology , Inhibitory Concentration 50 , Kinetics , Mice , Mice, Inbred BALB C , Mice, Nude , Naproxen/analogs & derivatives , Naproxen/metabolism , Naproxen/pharmacology , Nitrophenols/pharmacology , Nitrophenols/therapeutic use , Organophosphates/blood , Organophosphates/therapeutic use , Organophosphorus Compounds/blood , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/pharmacology , Organophosphorus Compounds/therapeutic use , Phenylglyoxal/analogs & derivatives , Phenylglyoxal/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Sulindac/analogs & derivatives , Sulindac/blood , Sulindac/metabolism , Sulindac/pharmacology , Sulindac/therapeutic use
5.
Biochem Pharmacol ; 80(1): 22-30, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20206143

ABSTRACT

Artemisinin and its derivatives (ARTs) are effective antimalarial drugs and also possess profound anticancer activity. However, the mechanism accounted for its distinctive activity in tumor cells remains unelucidated. We computed Pair wise Pearson correlation coefficients to identify genes that show significant correlation with ARTs activity in NCI-55 cell lines using data obtained from studies with HG-U133A Affymetrix chip. We found c-myc is one of the genes that showed the highest positive correlation coefficients among the probe sets analyzed (r=0.585, P<0.001). Dihydroartemisinin (DHA), the main active metabolite of ARTs, induced significant apoptosis in HL-60 and HCT116 cells that express high levels of c-MYC. Stable knockdown of c-myc abrogated DHA-induced apoptosis in HCT116 cells. Conversely, forced expression of c-myc in NIH3T3 cells sensitized these cells to DHA-induced apoptosis. Interestingly, DHA irreversibly down-regulated the protein level of c-MYC in DHA-sensitive HCT116 cells, which is consistent to persistent G1 phase arrest induced by DHA. Further studies demonstrated that DHA accelerated the degradation of c-MYC protein and this process was blocked by pretreatment with the proteasome inhibitor MG-132 or GSK 3beta inhibitor LiCl in HCT116 cells. Taken together, ARTs might be useful in the treatment of c-MYC-overexpressing tumors. We also suggest that c-MYC may potentially be a biomarker candidate for prediction of the antitumor efficacies of ARTs.


Subject(s)
Antimalarials/pharmacology , Apoptosis/drug effects , Artemisinins/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Down-Regulation/drug effects , G1 Phase/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , HCT116 Cells , HL-60 Cells , Humans , Leupeptins/pharmacology , Lithium Chloride/pharmacology , Mice , NIH 3T3 Cells , Proto-Oncogene Proteins c-myc/antagonists & inhibitors
6.
Cancer Biol Ther ; 8(17): 1640-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19625781

ABSTRACT

Aberrant activation of the epidermal growth factor receptor (EGFR) is closely associated with malignant progression of tumors. EGFR inhibitors have been used successfully in clinic in the treatment of solid tumors. In the present study, we revealed that BB, a new synthetic quinonazoline derivative, was a potent EGFR inhibitor. BB selectively inhibited EGFR with a IC(50) value of 50 +/- 37 nM, at least 32-fold more potent than suppressed all other ten tested receptor tyrosine kinases including the same family member ErbB2 (IC(50) = 5.6 +/- 3.2 microM). BB effectively abrogated autophosphorylation of the EGF-stimulated EGFR and phosphorylation of its key downstream signaling molecules ERK and AKT in A549 cells. BB was shown to suppress EGF-stimulated proliferation of A549 cells with an apparently lower IC(50) value (0.33 +/- 0.07 microM) than that (2.7 +/- 0.4 microM) for the serum-stimulated cells. BB also inhibited the EGF-independent proliferation of a panel of tumor cells. In addition, BB exhibited anti-angiogenesis activity, as evidenced by antagonizing EGF-induced HMEC-1 migration in vitro, blocking HMEC-1 tube formation, and inhibiting microvessel sprouting from rat aortic rings. Most importantly, BB prominently inhibited in vivo tumorigenesis of NIH3T3 cells specifically driven by the activation-mutated EGFR genes. As reported, normal NIH3T3 cells lack tumorigenicity in nude mice. NIH3T3 cells transfected with the EGFR gene with activating mutation (A750P or L858R) produced rapidly growing xenografts in nude mice. BB, when given orally at 100 mg/kg consecutively for 2 w, prominently inhibited the growth of the xenografts and reduced the number of microvessels. Taken together, the data indicate that BB is a new selective EGFR inhibitor with potent antitumor activity, revealing its potential as a promising anticancer candidate.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , ErbB Receptors/metabolism , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , NIH 3T3 Cells , Rats , Transfection
7.
Acta Pharmacol Sin ; 30(7): 1039-45, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19483716

ABSTRACT

AIM: We aimed to investigate the potential modification of previously unrecognized surface glycoprotein(s) by alpha2,6-sialylation other than by integrins. METHODS: The expression of beta-galactoside alpha2,6-sialyltransferase (ST6Gal-I) in the colon cancer cell line HCT116 was reduced by siRNA. The adhesion and Boyden chamber assay were used to detect the variation in cell motility. alpha2,6-Sialylation proteins were detected with lectin affinity assay. The mRNA expression, protein expression and downstream signaling modulation with siRNA were detected using reverse transcription-polymerase chain reaction, flow cytometry analysis, and Western blot. RESULTS: In HCT116 cells, the knockdown of ST6Gal-I inhibited cell motility, but did not affect cell adhesion. This selectively altered cell migration was caused by the loss of alpha2,6-sialic acid structures on c-Met. Moreover, STAT3 was dephosphorylated at tyrosine 705 in ST6Gal-I-knockdown (ST6Gal-I-KD) HCT116 cells. CONCLUSION: c-Met is the substrate of ST6Gal-I. The hyposialylation of c-Met can abolish cell motility in ST6Gal-I-KD HCT116 cells.Acta Pharmacologica Sinica (2009) 30: 1039-1045; doi: 10.1038/aps.2009.84; published online 1 June 2009.


Subject(s)
Cell Movement/physiology , HCT116 Cells/physiology , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/metabolism , Sialic Acids/metabolism , Sialyltransferases , Animals , Cell Adhesion/physiology , Gene Knockdown Techniques , Humans , Proto-Oncogene Proteins c-met/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism , Sialic Acids/chemistry , Sialyltransferases/genetics , Sialyltransferases/metabolism , Tyrosine/metabolism , beta-D-Galactoside alpha 2-6-Sialyltransferase
8.
Int J Biochem Cell Biol ; 40(10): 2164-73, 2008.
Article in English | MEDLINE | ID: mdl-18387333

ABSTRACT

Human ADAM15 is unique among the A disintegrin and metalloprotease domain (ADAM) family because of the integrin binding motif Arg-Gly-Asp (RGD) within its disintegrin domain. Integrin alpha5beta1 has been reported to bind to ADAM15 in an RGD-dependent manner, but the biological significance of the interaction between ADAM15 and alpha5beta1 is unknown. To characterize the effects of ADAM15 on alpha5beta1-mediated cell adhesion and migration and elucidate the potential mechanism, CHO cells which express endogenous integrin alpha5beta1 were transfected with human ADAM15 cDNA. ADAM15 overexpression led to enhanced cell adhesion and decreased migration on fibronectin, which were suppressed by down-regulation of integrin alpha5. Overexpression of ADAM15 not only increased the cell surface expression of integrin alpha5 but also resulted in a more clustered staining of alpha5 on cell surface, while the beta1 subunit remained unchanged. Unexpectedly, results from immunoprecipitation and immunofluorescence indicated that ADAM15 and alpha5beta1 integrin did not interact directly in CHO cells. We found that ADAM15 expression decreased the phosphorylation of Erk1/2. Consistently, down-regulation of Erk1/2 phosphorylation by MEK inhibitor PD98059 or siRNA against Erk1/2 enhanced the expression of alpha5 on cell surface. By using a B16F10 pulmonary metastasis model, we revealed that overexpression of ADAM15 significantly reduced the number of metastatic nodules on the lung. Taken together, this study reveals for the first time that ADAM15 could drive alpha5 integrin expression on cell surface via down-regulation of phosphorylated Erk1/2. This presents a novel mechanism by which ADAM15 regulates cell-matrix adhesion and migration.


Subject(s)
ADAM Proteins/metabolism , Cell Membrane/enzymology , Cell Movement , Extracellular Signal-Regulated MAP Kinases/metabolism , Integrin alpha5beta1/metabolism , Membrane Proteins/metabolism , Animals , CHO Cells , Cell Adhesion , Cell Line, Tumor , Cricetinae , Cricetulus , Enzyme Activation , Fibronectins/metabolism , Humans , Lung Neoplasms/secondary , Melanoma/pathology , Mice , Transplantation, Heterologous
9.
Mol Pharmacol ; 72(3): 545-52, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17565003

ABSTRACT

Vascular endothelial growth factor (VEGF) signaling pathway is essential for tumor angiogenesis and has long been recognized as a promising target for cancer therapy. Current view holds that physical interaction between alpha(v)beta(3) integrin and kinase insert domain-containing receptor (KDR) is important in regulating angiogenesis and tumor development. We have reported previously that a new marine-derived compound, philinopside E (PE), exhibited the antiangiogenic activity via inhibition on KDR phosphorylation and downstream signaling. Herein, we have further demonstrated that PE specifically interacts with KDR extracellular domain, which is distinct from conventional small-molecule inhibitors targeting cytoplasmic kinase domain, to block its interaction with VEGF and the downstream signaling. We also noted that PE markedly suppresses alpha(v)beta(3) integrin-driven downstream signaling as a result of disturbance of the physical interaction between KDR and alpha(v)beta(3) integrin in HMECs, followed by disruption of the actin cytoskeleton organization and decreased cell adhesion to vitronectin. All of these findings substantiate PE to be an unrecognized therapeutic class in tumor angiogenesis and, more importantly, help appeal the interest of the therapeutic potential in angiogenesis and cancer development via targeting integrin-KDR interaction in the future.


Subject(s)
Glycosides/chemistry , Glycosides/pharmacology , Integrin alphaVbeta3/antagonists & inhibitors , Saponins/chemistry , Sea Cucumbers/chemistry , Sulfuric Acid Esters/chemistry , Triterpenes/chemistry , Triterpenes/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Cell Adhesion/drug effects , Cell Line , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Glycosides/isolation & purification , Humans , Molecular Structure , Triterpenes/isolation & purification , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/chemistry
10.
Cancer Biol Ther ; 5(8): 978-85, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16855382

ABSTRACT

C75, a well-known fatty acid synthase (FAS) inhibitor, has been shown to possess potent anti-cancer activity in vitro and in vivo. In this study, we reveal that C75 is a cell cycle arrest inducer and explore the potential mechanisms for this effect in hepatocellular carcinoma (HCC) cell lines with abundant FAS expression: HepG2 and SMMC7721 cells with wt-p53, and Hep3B cells with null p53. The results showed FAS protein expression and basal activity levels were higher in HepG2 cells than in the other two HCC cell lines. Treatment with C75 inhibited FAS activity within 30 min of administration and induced G(2) phase arrest accompanied by p53 overexpression in HepG2 and SMMC7721 cells. By contrast, C75 triggered G(1) phase arrest in Hep3B cells, and RNA interference targeting p53 did not attenuate C75-induced G(2) arrest in HepG2 cells. Similarly, p53 overexpression via p53 plasmid transfection did not affect C75-induced G(1) phase arrest in Hep3B cells. However, we observed a clear correlation between p38 MAPK activation triggered by C75 and the induction of cell cycle arrest in all three HCC cells. Furthermore, treatment with the p38 MAPK inhibitor SB203580 reduced p38 MAPK activity and cell cycle arrest, and also partially restored cyclin A, cyclin B1, cyclin D1 and p21 protein levels. Collectively, it was p38 MAPK but not p53 involved in C75-mediated tumor cell growth arrest in HCC cells.


Subject(s)
4-Butyrolactone/analogs & derivatives , Carcinoma, Hepatocellular/metabolism , Fatty Acid Synthases/antagonists & inhibitors , G2 Phase/drug effects , Liver Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , 4-Butyrolactone/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Cyclin A/metabolism , Cyclin B/metabolism , Cyclin B1 , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Flow Cytometry , Humans , Immunoprecipitation , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Plasmids/genetics , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , fas Receptor/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...