Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Biomed Tech (Berl) ; 68(6): 651-665, 2023 Dec 15.
Article En | MEDLINE | ID: mdl-37350466

Transthoracic impedance is one of the key factors affecting the success of defibrillation. Impedance compensation technique is used to adjust defibrillation parameters according to the transthoracic impedance of the defibrillator. In this paper, a combined impedance compensation strategy is proposed to address the shortcomings of existing compensation strategies. In order to evaluate the performance of the combined compensation strategy, this paper uses the prototype as the experimental machine, and uses two AED with representative impedance compensation strategies as the control machine, and the simulated defibrillation method is used for comparative testing. The results show that the combined impedance compensation has a more steadier distribution over the defibrillation energy and current: compared with the energy-based impedance compensation strategy, this strategy can significantly reduce the peak current (25 Ω: 27.8 vs. 54.7 A; 50 Ω: 20.7 vs. 32.3 A) and average current (25 Ω: 24.8 vs. 37.5 A) of defibrillation at low impedance, and compared with the current impedance compensation strategy, it can significantly reduce the defibrillation energy (150 Ω: 8.6 vs. 1.7 %, 175 Ω: 15.6 vs. 4.9 %, 200 Ω: 21.9 vs. 8.5 %) at high impedance. Impedance compensation is more precise and the current passing during defibrillation is steadier.


Defibrillators , Electric Countershock , Electric Impedance , Electric Countershock/methods , Cardiography, Impedance/methods
2.
Biomater Adv ; 135: 212741, 2022 Apr.
Article En | MEDLINE | ID: mdl-35929214

Surface functionalization to improve the blood compatibility is pivotal for the application of biomaterials. In this article, the surface of silicon was first functionalized with chemical groups, such as amino, quinone and phenol groups by the self-polymerization of dopamine, which were used to immobilize anticoagulant drugs hirudin. The detailed analysis and discussion about the grafting groups, morphology, wettability, the dynamic adsorption of proteins, the cytological property and the blood compatibility on the surfaces were carried on by the technology of contact angle, X-ray photoelectron spectroscopy, quartz crystal microbalance, endothelial cells culture and anticoagulant blood test in vivo. The surface with hirudin modification exhibited hydrophilic property and significantly inhibited the nonspecific adsorption of albumin, while it was more approachable to fibronectin. In vitro study displayed that the surface loaded with hirudin could promote the proliferation of endothelial cells. The evaluation of anticoagulant showed good anti-adhesion effect on platelets and the hemolysis rate decreased significantly to less than 0.4%. Activated partial thromboplastin time (APTT) of the silicon wafer loaded with hirudin can exceed 38 s, and the APTT prolongs as the hirudin concentration rises. This study suggested that such simple but effective surface functionalization technique, combining excellent anticoagulant activity together with reendothelialization potential due to the preferable fibronectin adsorption, provide great practical significance to the application of cardiovascular materials.


Fibronectins , Hirudins , Adsorption , Anticoagulants/pharmacology , Endothelial Cells , Fibronectins/pharmacology , Hirudins/chemistry , Silicon/chemistry
3.
Anal Chim Acta ; 1050: 132-138, 2019 Mar 07.
Article En | MEDLINE | ID: mdl-30661580

Loop-mediated isothermal amplification (LAMP) is a useful platform for nucleic acids detection in point-of-care (POC) situations, and development of single-step, close-tube LAMP reactions for specific detection of single nucleotide mutations (SNMs) remains a challenge. We develop a novel primer-activatable LAMP (PA-LAMP) strategy that enables highly specific and sensitive SNM detection using single-step, close-tube reactions. This strategy designs a terminal-blocked inner primer with a ribonucleotide insertion, which is cleaved and activated specifically to perfectly matched targets by ribonuclease (RNase) H2, to realize efficient amplification of mutant genes. It has shown dynamic responses of mutant target in a linear range from 220 aM to 22 pM with a lowest detectable concentration of 22 aM. It also demonstrates very high specificity in identifying the mutant in a large excess of the wild-type with a discrimination ratio as high as ∼10,000. It has been successfully applied to mutation detection of genomic DNA in tumor cells. The PA-LAMP strategy provides a useful, portable and affordable POC platform for highly sensitive and specific detection of genetic mutations in clinical applications.


DNA, Neoplasm/genetics , Nucleic Acid Amplification Techniques , Nucleotides/genetics , DNA, Neoplasm/isolation & purification , HT29 Cells , Humans , Mutation , Point-of-Care Systems , Tumor Cells, Cultured
4.
Analyst ; 143(18): 4422-4428, 2018 Sep 10.
Article En | MEDLINE | ID: mdl-30140810

In this work, a versatile point-of-care assay platform based on a microfluidic paper-based analytic device (µPAD) was developed for the simultaneous detection of multiple targets. The µPAD with a central zone and six test zones is fabricated by a simple and inexpensive wax printing method. A flower-like hybrid nanocomplex synthesized with specific dual enzymes and Cu3(PO4)2 inorganic nanocrystals is spotted in the test zones on the µPAD, followed by the introduction of assay targets. Using dual-enzyme inorganic hybrid nanoflowers in the µPAD as nanobiocatalysts, which preserve the activity and enhance the stability of the enzymes, based on the H2O2-mediated catalytic oxidizing chromogenic reaction produced by glucose/uric acid, the developed multiplex paper-based nanobiocatalytic system is demonstrated to enable simultaneous and sensitive detection of glucose and uric acid with a detection limit of 60 and 25 µM, respectively. More importantly, it has been successfully used for detecting glucose and uric acid levels in human whole blood samples. The developed multiplex paper-based nanobiocatalytic system features very easy fabrication and operation, low cost, and high sensitivity and has promising prospects for a clinical multianalyte point-of-care test.


Blood Glucose/analysis , Nanoparticles , Paper , Uric Acid/blood , Enzymes/chemistry , Humans , Hydrogen Peroxide , Microfluidic Analytical Techniques , Point-of-Care Testing
5.
Eur J Pharmacol ; 825: 10-18, 2018 Apr 15.
Article En | MEDLINE | ID: mdl-29454609

b-AP15, a potent and selective inhibitor of the ubiquitin-specific peptidase 14 (USP14), displays in vitro and in vivo antitumor abilities on some types of cancer cells. However, the mechanism underlying its action is not well elucidated. The purposes of the present study are to observe the potential impacts of b-AP15 on cell survival of hepatocellular carcinoma cells and to investigate whether and how this compound inhibits some survival-promoting signaling pathways. We found that b-AP15 significantly decreased cell viability and increased cell apoptosis in a dose-dependent manner in hepatocellular carcinoma cells, along with the perturbation of cell cycle and the decreased expressions of cell cycle-related proteins. We also demonstrated that the endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) were enhanced by b-AP15 supplementation. The inhibition of ER stress/UPR only partly attenuated the cytotoxicity of b-AP15 on hepatocellular carcinoma cells. In addition, b-AP15 treatment inhibited Wnt/ß-catenin and Notch1 signaling pathways, and suppressed phosphorylation of STAT3, Akt, and Erk1/2, which were not restored by the inhibition of ER stress/UPR. Furthermore, the expression levels of signaling molecules in Notch1 were reduced by specific inhibitor of Wnt/ß-catenin pathway. Notably, either Wnt or Notch1 signaling inhibitor mitigated phosphorylation of STAT3, Akt, and Erk1/2, and mimicked the cytotoxicity of b-AP15 on hepatocellular carcinoma cells. These results clearly indicate that b-AP15 induced cytotoxic response to hepatocellular carcinoma cells by augmenting ER stress/UPR and inhibiting Wnt/Notch1 signaling pathways. This new finding provides a novel mechanism by which b-AP15 produces its antitumor therapeutic effects.


Carcinoma, Hepatocellular/drug therapy , Cell Survival/drug effects , Deubiquitinating Enzymes/antagonists & inhibitors , Endoplasmic Reticulum Stress/drug effects , Liver Neoplasms/drug therapy , Piperidones/pharmacology , Receptor, Notch1/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Endoplasmic Reticulum/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Unfolded Protein Response/drug effects
6.
Can J Physiol Pharmacol ; 96(5): 521-526, 2018 May.
Article En | MEDLINE | ID: mdl-29394494

The proteasomal system is a promising target for cancer treatment. Quercetin (Que), a flavonoid compound with antitumor ability, displays the inhibitory effect on proteasome activity. However, the underlying molecular mechanisms are ill defined. The present study found that Que treatment significantly reduced the chymotrypsin-like protease activity of proteasome whereas the trypsin- and caspase-like protease activities remained unchanged in HepG2 cancer cells, along with activation of p38 MAPK and JNK and reduction of ERK1/2 phosphorylation. Que-reduced proteasome activity could not be reverted by inhibition of p38 MAPK and JNK signaling pathway. In addition, MEK1 overexpression or knockdown upregulated or downregulated the chymotrypsin-like protease activity of proteasome, respectively. Both Que and MEK1/ERK1/2 inhibitor attenuated the expression levels of proteasome ß subunits. These results indicate that Que-induced suppression of MEK1/ERK1/2 signaling and subsequent reduction of proteasome ß subunits is responsible for its inhibitory impacts on proteasome activity.


Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Chymotrypsin/metabolism , Liver Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Proteasome Endopeptidase Complex/metabolism , Quercetin/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Hep G2 Cells , Humans , MAP Kinase Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism
7.
Tumour Biol ; 39(3): 1010428317694317, 2017 Mar.
Article En | MEDLINE | ID: mdl-28349827

Staurosporine, which is an inhibitor of a broad spectrum of protein kinases, has shown cytotoxicity on several human cancer cells. However, the underlying mechanism is not well understood. In this study, we examined whether and how this compound has an inhibitory action on phosphatidylinositol 3-kinase (PI3K)/Akt pathway in vitro using HepG2 human hepatocellular carcinoma cell line. Cell viability and apoptosis were determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and terminal deoxyribonucleotidyl transferase-mediated dUTP-digoxigenin nick end labeling (TUNEL) assay, respectively. Glutathione S-transferase (GST) pull-down assay and co-immunoprecipitation were performed to detect protein-protein interactions. Small interfering RNA (siRNA) was used to silence the expression of targeted protein. We found that staurosporine significantly decreased cell viability and increased cell apoptosis in a concentration- and time-dependent manner in HepG2 cancer cells, along with the decreased expressions of PDK1 protein and Akt phosphorylation. Staurosporine was also found to enhance Omi/HtrA2 release from mitochondria. Furthermore, Omi/HtrA2 directly bound to PDK1. Pharmacological and genetic inhibition of Omi/HtrA2 restored protein levels of PDK1 and protected HepG2 cancer cells from staurosporine-induced cell death. In addition, staurosporine was found to activate autophagy. However, inhibition of autophagy exacerbated cell death under concomitant treatment with staurosporine. Taken together, our results indicate that staurosporine induced cytotoxicity response by inhibiting PI3K/Akt signaling pathway through Omi/HtrA2-mediated PDK1 degradation, and the process provides a novel mechanism by which staurosporine produces its therapeutic effects.


Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Mitochondrial Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Serine Endopeptidases/metabolism , Staurosporine/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Enzyme Inhibitors/pharmacology , Hep G2 Cells , High-Temperature Requirement A Serine Peptidase 2 , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Signal Transduction/drug effects
...