Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Publication year range
1.
Front Public Health ; 10: 888089, 2022.
Article in English | MEDLINE | ID: mdl-35812501

ABSTRACT

Background: Urinary tract infection (UTI) is a common complication in pediatric urological surgery patients and is associated with long-term sequelae, including subsequent recurrent infections and renal scarring. In this study, we aimed to explore the risk factors for UTI in pediatric urological surgery patients and construct a predictive model for UTI. Materials and Methods: A total of 2,235 pediatric patients who underwent urological surgery at a tertiary hospital between February 2019 and January 2020 were included. A multivariate logistic regression model was applied to identify the predictive factors, and a predictive model was constructed using a receiver operating characteristic curve. A multifactorial predictive model was used to categorize the risk of UTI based on the weight of the evidence. Results: A total of 341 patients with UTI were identified, which corresponded to a prevalence of 15.26% in pediatric urological surgery patients. Multivariate analysis identified six significant risk factors for UTI, including age <12.0 months, upper urinary tract disease, not using an indwelling drainage tube, hospital stay ≥10 days, administration of two or more types of antibiotics, and stent implantation. A combination of the aforementioned factors produced an area under the curve value of 88.37% for preventing UTI in pediatric urological surgery patients. A multifactorial predictive model was created based on the combination of these factors. Conclusions: The constructed multifactorial model could predict UTI risk in pediatric urological surgery patients with a relatively high predictive value.


Subject(s)
Urinary Tract Infections , Child , Humans , Infant , Prevalence , ROC Curve , Risk Factors , Urinary Tract Infections/complications , Urinary Tract Infections/etiology
2.
Front Pharmacol ; 13: 819826, 2022.
Article in English | MEDLINE | ID: mdl-35645824

ABSTRACT

Ilex rotunda Thunb (IR) is a traditional Chinese medicine used for the clinical treatment of gastric ulcers and duodenal ulcers; however, the effect of IR on ulcerative colitis (UC) and its underlying mechanism remains unclear. This study investigated the therapeutic effect of IR on UC mice induced by dextran sulfate sodium (DSS) as well as the potential underlying mechanism. The main components of IR were analyzed by ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry. Then we established a model of UC mice by administering 2.0% DSS for 7 days followed by 2 weeks of tap water for three cycles and administered IR. On day 56, the disease activity index (DAI), colon length, pathological changes, and inflammatory response of the colon tissue of mice were assessed. The oxidative stress and apoptosis of colon tissue were detected, and the integrity of the intestinal mucosal barrier was evaluated to assess the effect of IR. Furthermore, the relationship between oncostatin M (OSM) and its receptor (OSMR) in addition to the IR treatment of UC were evaluated using a mouse model and Caco2 cell model. The results showed that IR significantly alleviated the symptoms of UC including rescuing the shortened colon length; reducing DAI scores, serum myeloperoxidase and lipopolysaccharide levels, pathological damage, inflammatory cell infiltration and mRNA levels of interleukin one beta, tumor necrosis factor alpha, and interleukin six in colon tissue; alleviating oxidative stress and apoptosis by decreasing kelch-like ECH-associated protein 1 expression and increasing nuclear factor-erythroid factor 2-related factor 2 and heme oxygenase-1 protein expression; and promoting the regeneration of epithelial cells. IR also promoted the restoration of the intestinal mucosal barrier and modulated the OSM/OSMR pathway to alleviate UC. It was found that IR exerted therapeutic effects on UC by restoring the intestinal mucosal barrier and regulating the OSM/OSMR pathway.

3.
Front Cell Dev Biol ; 9: 682574, 2021.
Article in English | MEDLINE | ID: mdl-34409030

ABSTRACT

The inflammatory response of endothelial cells accelerates various vascular diseases. MicroRNAs (miRNAs) participate in diverse cellular processes during inflammation. In the present study, we found that miR-302a is an effective suppressor of vascular inflammation in endothelial cells. It was revealed that miR-302a exhibited a lower level in a lipopolysaccharide (LPS)-induced mouse model and in patients with vascular inflammatory disease. Genetic haploinsufficiency of miR-302 aggravated the LPS-induced vascular inflammatory response in mice, and overexpression of miR-302a attenuated vascular inflammation in mice. Furthermore, overexpression of miR-302a inhibited the synthesis and secretion of adhesion factors in endothelial cells, and suppressed the adhesion of monocytes to endothelium. In the study of molecular mechanism, we found that miR-302a relieved vascular inflammation mainly by regulating the nuclear factor kappa-B (NF-κB) pathway in endothelial cells. The results showed that interleukin-1 receptor-associated kinase4 (IRAK4) and zinc finger protein 91 (ZFP91) were the binding targets of miR-302a. MiR-302a prevented the nuclear translocation of NF-κB by inhibiting phosphorylation of IκB kinase complex ß (IKKß) and inhibitors of κBα (IκBα) via targeting IRAK4. In addition, miR-302a downregulated the expression of NF-κB by directly binding with ZFP91. These findings indicate that miR-302a negatively regulates inflammatory responses in the endothelium via the NF-κB pathway and it may be a novel target for relieving vascular inflammation.

4.
Cell Death Dis ; 9(6): 610, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29795190

ABSTRACT

The development of nephrotoxicity largely limits the clinical use of chemotherapy. MiRNAs are able to target various genes and involved in the regulation of diverse cellular processes, including cell apoptosis and death. Our study showed that miR-181a expression was significantly increased after 5-fluorouracil (5-FU) treatment in renal mesangial cells and kidney tissue, which was associated with decreased baculoviral inhibition of apoptosis protein repeat-containing 6 (BIRC6) expression and increased apoptotic rate. Enforced miR-181a expression enhanced 5-FU-induced p53-dependent mitochondrial apoptosis, including declined Bcl-2/Bax ratio, loss of mitochondrial membrane potential, cytochrome c release, and caspase-9 and caspase-3 activation. However, inhibition of miR-181a was associated with reduced p53-mediated mitochondrial apoptosis induced by 5-FU. Moreover, miR-181a increased BIRC6 downstream gene p53 protein expression and transcriptional activity by reducing ubiquitin-mediated protein degradation. We found that miR-181a directly targeted 3'-UTR of BIRC6 mRNA and negatively regulated BIRC6 expression. In vivo study, knockdown of miR-181a with adeno-associated virus harboring miR-181a-tough decoy attenuated 5-FU-induced renal cell apoptosis, inflammation and kidney injury. In conclusion, these results demonstrate that miR-181a increases p53 protein expression and transcriptional activity by targeting BIRC6 and promotes 5-FU-induced apoptosis in mesangial cells. Inhibition of miR-181a ameliorates 5-FU-induced nephrotoxicity, suggesting that miR-181a may be a novel therapeutic target for nephrotoxicity treatment during chemotherapy.


Subject(s)
Apoptosis , Fluorouracil/adverse effects , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney/pathology , Mesangial Cells/pathology , MicroRNAs/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , HCT116 Cells , Humans , Inflammation/pathology , Inhibitor of Apoptosis Proteins/metabolism , Mesangial Cells/drug effects , Mice, Inbred C57BL , MicroRNAs/genetics , Mitochondria/metabolism , Signal Transduction , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
5.
Biochem Biophys Res Commun ; 495(2): 1864-1870, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29225169

ABSTRACT

Orai1-dependent Ca2+ entry plays an essential role in inflammatory response through regulating T cell and macrophage activation and neutrophil infiltration. However, whether Orai1 Ca2+ entry contributes to endothelial activation, one of the early steps of vascular inflammation, remains elusive. In the present study, we observed that knockdown of Orai1 reduced, whereas overexpression of Orai1 potentiated, TNFα-induced expression of adhesion molecules such as ICAM-1 and VCAM-1 in HUVECs, and subsequently blocked adhesion of monocyte to HUVECs. In vivo, Orai1 downregulation attenuated TNFα-induced ICAM-1 and VCAM-1 expression in mouse aorta and the levels of pro-inflammatory cytokines in the serum. In addition, Orai1 knockdown also dramatically decreased the expression of pro-inflammatory cytokines and neutrophil infiltration in the lung after TNFα treatment, and thus protected lung tissue injury. Notably, among all isoforms of nuclear factor of activated T cells (NFATs), TNFα only triggered NFATc4 nuclear accumulation in HUVECs. Knockdown of Orai1 or inhibition of calcineurin prevented TNFα-induced NFATc4 nuclear translocation and reduced ICAM-1 and VCAM-1 expression in HUVECs. Overexpression of NFATc4 further enhanced ICAM-1 and VCAM-1 expression induced by TNFα. Our study demonstrates that Orai1-Ca2+-calcineurin-NFATc4 signaling is an essential inflammatory pathway required for TNFα-induced endothelial cell activation and vascular inflammation. Therefore, Orai1 may be a potential therapeutic target for treatment of inflammatory diseases.


Subject(s)
Aortitis/immunology , Calcineurin/immunology , Calcium/immunology , Cell Adhesion Molecules/immunology , Endothelium, Vascular/immunology , NFATC Transcription Factors/immunology , ORAI1 Protein/immunology , Animals , Aortitis/pathology , Cells, Cultured , Down-Regulation/immunology , Humans , Inflammation Mediators/immunology , Metabolic Networks and Pathways/immunology , Mice , Mice, Inbred C57BL
6.
J Cell Mol Med ; 21(5): 904-915, 2017 05.
Article in English | MEDLINE | ID: mdl-27878958

ABSTRACT

Increasing evidence supports that activation of store-operated Ca2+ entry (SOCE) is implicated in the chemoresistance of cancer cells subjected to chemotherapy. However, the molecular mechanisms underlying chemoresistance are not well understood. In this study, we aim to investigate whether 5-FU induces hepatocarcinoma cell death through regulating Ca2+ -dependent autophagy. [Ca2+ ]i was measured using fura2/AM dye. Protein expression was determined by Western blotting and immunohistochemistry. We found that 5-fluorouracil (5-FU) induced autophagic cell death in HepG2 hepatocarcinoma cells by inhibiting PI3K/AKT/mTOR pathway. Orai1 expression was obviously elevated in hepatocarcinoma tissues. 5-FU treatment decreased SOCE and Orai1 expressions, but had no effects on Stim1 and TRPC1 expressions. Knockdown of Orai1 or pharmacological inhibition of SOCE enhanced 5-FU-induced inhibition of PI3K/AKT/mTOR pathway and potentiated 5-FU-activated autophagic cell death. On the contrary, ectopic overexpression of Orai1 antagonizes 5-FU-induced autophagy and cell death. Our findings provide convincing evidence to show that Orai1 expression is increased in hepatocarcinoma tissues. 5-FU can induce autophagic cell death in HepG2 hepatocarcinoma cells through inhibition of SOCE via decreasing Orai1 expression. These findings suggest that Orai1 expression is a predictor of 5-FU sensitivity for hepatocarcinoma treatment and blockade of Orai1-mediated Ca2+ entry may be a promising strategy to sensitize hepatocarcinoma cells to 5-FU treatment.


Subject(s)
Calcium/metabolism , Carcinoma, Hepatocellular/metabolism , Fluorouracil/pharmacology , Liver Neoplasms/metabolism , ORAI1 Protein/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Carcinoma, Hepatocellular/pathology , Down-Regulation/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
7.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-666569

ABSTRACT

OBJECTIVE To determine the characterization, anti-tumor efficacy and pharmacokinetics of bufalin- loaded PEGylated liposomes compared with bufalin entity. METHODS Bufalin- loaded PEGylated liposomes and bufalin- loaded liposomes were prepared reproducibly with homogeneous particle size by the combination of thin film evaporation method and high pressure homogenization method. The particle size and zeta potential of the liposomes were determined by dynamic light scattering technique. The direct imaging of morphology of liposomes was charactered by transmission electron microscope. The content of bufalin in liposomes was analysed by HPLC method. The entrapment efficiency and the particle size was applied to assess the stability profile, after storage at 4℃ on day 0, 7, 15, 30 and 90. The in-vitro release behaviours of bufalin from liposomes were conducted using dialysis bag technique at 37℃. In-vitro cytotoxicity studies were carried out using MTT〔3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide〕assay on several kinds of tumor cell lines including SW620, PC-3, MDA-MB-231, A549, U251, U87 and HepG2. In-vivo pharmacokinetic study of bufalin liposomes was evaluated by HPLC method. RESULTS Their mean particle sizes were 127.6 nm and 155.0 nm, mean zeta potentials were 2.24 mV and - 18.5 mV, entrapment efficiencies were 76.31% and 78.40% , respectively. In- vitro release profile revealed that the release of bufalin in bufalin- loaded PEGylated liposomes was slower than that of bufalin-loaded liposomes. The cytotoxicity of blank liposomes has been found within acceptable range, whereas bufalin-loaded PEGylated liposomes showed enhanced cytotoxicity to U251 cells compared with bufalin entity. In-vivo pharmacokinetics indicated that bufalin-loaded PEGylated liposomes could extend eliminate half-life time of bufalin in plasma in rats. CONCLUSION The results suggested that bufalin-loaded PEGylated liposomes improved the solubility and increased the drug concentration in plasma.

8.
Circ J ; 80(4): 1024-33, 2016.
Article in English | MEDLINE | ID: mdl-26911455

ABSTRACT

BACKGROUND: Previous work has demonstrated that the volume-regulated chloride channel is activated during foam cell formation, and inhibition of chloride movement prevents intracellular lipid accumulation. However, the mechanism explaining how chloride movement promotes foam cell formation is not clear. METHODS AND RESULTS: Foam cell formation was determined by Oil Red O staining. Western blotting and co-immunoprecipitation were used to examine protein expression and protein-protein interaction. [Cl(-)]iwas measured using 6-methoxy-N-ethylquinolinium iodide dye. The results showed that [Cl(-)]iwas decreased in monocytes/macrophages from patients with hypercholesterolemia and from apoE(-/-)mice fed with a high-fat diet. Lowering [Cl(-)]iupregulated scavenger receptor A (SR-A) expression, increased the binding and uptake of oxLDL, enhanced pro-inflammatory cytokine production and subsequently accelerated foam cell formation in macrophages from humans and mice. In addition, low Cl(-)solution stimulated the activation of JNK and p38 mitogen-activated protein kinases. Inhibition of JNK and p38 blocked Cl(-)reduced medium-induced SR-A expression and lipid accumulation. In contrast, reduction of [Cl(-)]ipromoted the interaction of SR-A with caveolin-1, thus facilitating caveolin-1-dependent SR-A endocytosis. Moreover, disruption of caveolae attenuated SR-A internalization, JNK and p38 activation, and ultimately prevented SR-A expression and foam cell formation stimulated by low Cl(-)medium. CONCLUSIONS: This data provide strong evidence that reduction of [Cl(-)]iis a critical contributor to intracellular lipid accumulation, suggesting that modulation of [Cl(-)]iis a novel avenue to prevent foam cell formation and atherosclerosis.


Subject(s)
Chlorides/metabolism , Foam Cells/metabolism , Hypercholesterolemia/metabolism , Animals , Apolipoproteins E/deficiency , Caveolin 1/genetics , Caveolin 1/metabolism , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/genetics , Foam Cells/pathology , Hypercholesterolemia/chemically induced , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Arterioscler Thromb Vasc Biol ; 36(4): 618-28, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26916730

ABSTRACT

OBJECTIVE: To determine the role of orai1 store-operated Ca(2+) entry in foam cell formation and atherogenesis. APPROACH AND RESULTS: Acute administration of oxidized low-density lipoprotein (oxLDL) activates an orai1-dependent Ca(2+) entry in macrophages. Chelation of intracellular Ca(2+), inhibition of orai1 store-operated Ca(2+) entry, or knockdown of orai1 dramatically inhibited oxLDL-induced upregulation of scavenger receptor A, uptake of modified LDL, and foam cell formation. Orai1-dependent Ca(2+) entry induces scavenger receptor A expression and foam cell formation through activation of calcineurin but not calmodulin kinase II. Activation of nuclear factor of activated T cells is not involved in calcineurin signaling to foam cell formation. However, oxLDL dephosohorylates and activates apoptosis signal-regulating kinase 1 in macrophages. Orai1 knockdown prevents oxLDL-induced apoptosis signal-regulating kinase 1 activation. Knockdown of apoptosis signal-regulating kinase 1, or inhibition of its downstream effectors, JNK and p38 mitogen-activated protein kinase, reduces scavenger receptor A expression and foam cell formation. Notably, orai1 expression is increased in atherosclerotic plaques of apolipoprotein E(-/-) mice fed with high-cholesterol diet. Knockdown of orai1 with adenovirus harboring orai1 siRNA or inhibition of orai1 Ca(2+) entry with SKF96365 for 4 weeks dramatically inhibits atherosclerotic plaque development in high-cholesterol diet feeding apolipoprotein E(-/-) mice. In addition, inhibition of orai1 Ca(2+) entry prevents macrophage apoptosis in atherosclerotic plaque. Moreover, the expression of inflammatory genes in atherosclerotic lesions and the infiltration of myeloid cells into the aortic sinus plaques are decreased after blocking orai1 signaling. CONCLUSIONS: Orai1-dependent Ca(2+) entry promotes atherogenesis possibly by promoting foam cell formation and vascular inflammation, rendering orai1 Ca(2+) channel a potential therapeutic target against atherosclerosis.


Subject(s)
Anticholesteremic Agents/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium/metabolism , Cholesterol/metabolism , Foam Cells/drug effects , Macrophages, Peritoneal/drug effects , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Calcineurin/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Chelating Agents/pharmacology , Calcium Signaling/drug effects , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Foam Cells/metabolism , Foam Cells/pathology , Humans , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipoproteins, LDL/pharmacology , MAP Kinase Kinase Kinase 5/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Mice, Knockout , ORAI1 Protein , Plaque, Atherosclerotic , RNA Interference , Scavenger Receptors, Class A/metabolism , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...