Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 730: 150341, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39018965

ABSTRACT

Cardiomyocyte injury is closely related to various myocardial diseases, and S-Allyl-L-cysteine (SAC) has been found to have myocardial protective effects, but its mechanism is currently unclear. Meanwhile, copper also has various physiological functions, and this study found that copper inhibited cell viability in a concentration and time-dependent manner, and was associated with multiple modes of death. Elesclomol plus CuCl2 (ES + Cu) significantly inhibited cell viability, and this effect could only be blocked by copper chelator TTM, indicating that "ES + Cu" induced cuproptosis in cardiomyocytes. SAC reduced the inhibitory effects of high concentration copper and "ES + Cu" on cell viability in a concentration and time-dependent manner, indicating that SAC plays a cardioprotective role under stress. Further mechanism study showed that high concentration of copper significantly induced cardiomyocyte apoptosis and increased the levels of LDH, MDA and ROS, while SAC inhibited the apoptosis and injury of cardiomyocytes induced by copper. "ES + Cu" significantly increased intracellular copper levels and decreased the expression of FDX1, LIAS, Lip-DLST and Lip-DLAT; FDX1 siRNA did not affect the expression of LIAS, but further reduced the expression of Lip-DLST and Lip-DLAT; SAC did not affect the expression of these genes, but enhanced the effect of "ES + Cu" in down-regulating these gene expression and restored intracellular copper levels. In addition, "ES + Cu" reduced ATP production, weakened the activity of mitochondrial complex I and III, inhibited cell viability, and increased the contents of injury markers LDH, MDA, CK-MB and cTnI, while SAC significantly improved mitochondrial function injury and cardiomyocyte injury induced by "ES + Cu". Therefore, SAC can inhibit apoptosis and cuproptosis to play a cardioprotective role.

2.
J Cardiovasc Pharmacol ; 78(6): 891-899, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34596622

ABSTRACT

ABSTRACT: Endothelial dysfunction participates in the pathogenesis of various cardiovascular disorders, and dysregulated angiogenesis involves the vascular endothelial growth factor (VEGF)-matrix metalloproteinases (MMP) system. Nicotinamide phosphoribosyltransferase (NAMPT) is known to enhance endothelial function and angiogenesis. The study found that NAMPT overexpression protected human coronary artery endothelial cells (HCAECs) from H2O2-induced injury through promoting cell viability, inhibiting cell apoptosis, enhancing cell motility, and promoting tube formation. Through analyses based on 2 Protein-Protein Interaction databases, Mentha and BioGrid, we identified CUL5 as a protein that may interact with NAMPT, which was then validated by Co-IP experiments. Through interacting with NAMPT, CUL5 inhibited NAMPT expression. In contrast to NAMPT, CUL5 overexpression further aggravated H2O2-induced HCAEC dysfunction. In the meantime, CUL5 overexpression reduced, whereas NAMPT overexpression increased the phosphorylation of p38 and Akt and the protein levels of VEGF and MMP2. More importantly, NAMPT overexpression partially reversed the effects of CUL5 overexpression on H2O2-stimulated HCAECs and the MAPK/phosphatidylinositol 3-kinase-Akt/VEGF/MMP signaling. In conclusion, CUL5 interacts with NAMPT in H2O2-stimulated HCAECs, suppressing cell viability, promoting cell apoptosis, and inhibiting cell mobility and tube formation. NAMPT overexpression protects against H2O2-induced HCAEC dysfunction by promoting cell viability, inhibiting cell apoptosis, and enhancing cell mobility and tube formation.


Subject(s)
Cell Proliferation , Cullin Proteins/metabolism , Cytokines/metabolism , Endothelial Cells/enzymology , Neovascularization, Physiologic , Nicotinamide Phosphoribosyltransferase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis , Cell Proliferation/drug effects , Cells, Cultured , Cullin Proteins/genetics , Cytokines/genetics , Endothelial Cells/drug effects , Endothelial Cells/pathology , Humans , Hydrogen Peroxide/toxicity , Neovascularization, Physiologic/drug effects , Nicotinamide Phosphoribosyltransferase/genetics , Phosphorylation , Proteolysis , Signal Transduction
4.
J Cell Biochem ; 120(9): 14518-14530, 2019 09.
Article in English | MEDLINE | ID: mdl-31026096

ABSTRACT

Endothelial progenitor cells (EPCs) have been reported to replace the damaged endothelial cells to repair the injured or dead endothelium. However, EPC senescence might lead to the failure in EPC function. Thus, developing an in-depth understanding of the mechanism of EPC senescence might provide novel strategies for related vascular disorders' treatments. Herein, nicotinamide phosphoribosyltransferase (NAMPT) overexpression could increase cell proliferation and suppress cell senescence in EPCs. miR-223 directly bound to the 3'-untranslated region of NAMPT to inhibit its expression, therefore modulating EPC proliferation and senescence through NAMPT and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling. Long noncoding RNA (lncRNA) GAS5 sponges miR-223, consequently downregulating miR-223 expression. GAS5 knockdown inhibited EPC proliferation and promoted senescence. GAS5 might serve as a competing endogenous RNA for miR-223 to counteract miR-223-mediated suppression on NAMPT, thus regulating EPC proliferation and senescence via the PI3K/AKT signaling pathway. In summary, our findings provide a solid experimental basis for understanding the role and mechanism of lncRNA GAS5/miR-223/NAMPT axis in EPC proliferation and senescence.


Subject(s)
Cytokines/genetics , Endothelial Progenitor Cells/cytology , MicroRNAs/genetics , Nicotinamide Phosphoribosyltransferase/genetics , RNA, Long Noncoding/genetics , 3' Untranslated Regions , Cell Line , Cell Proliferation , Cellular Senescence , Endothelial Progenitor Cells/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
5.
Food Chem Toxicol ; 123: 374-384, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30428381

ABSTRACT

Ochratoxin A (OTA) and citrinin (CTN) are important mycotoxins, which often coexist in food and feed stuff. In this study, individual and combinative cytotoxicity of OTA and CTN were tested in human embryonic kidney (HEK) 293 cells via MTT assay, and synergistic cytotoxic effects were found following co-treatment with OTA and CTN, manifested by significant accumulation of HEK293 cells in S and G2/M stages. Transcriptomic and sRNA sequencing were performed to explore molecular signatures mediating individual or combinative cytotoxicity. A total of 378 miRNAs were identified, among which 66 miRNAs targeting thousands of genes were differentially expressed in response to different treatments, and 120 differentially expressed genes (DEGs) were regulated by either individual or combinative treatments. Correlations between two representative miRNAs (hsa-miR-1-3p and hsa-miR-122-5p), and their target genes, programmed cell death 10 (PDCD10) and cyclin G1 (CCNG1), associated with apoptotic signaling and cell cycle were analyzed by luciferase assay system. Further, their expression patterns were validated by quantitative real-time PCR and western blot analysis, suggesting that both miRNA-target interactions might account for the mycotoxin-induced cell death. Taken together, these findings provide molecular evidences for synergistic cytotoxic effects of exposure to single and mixture of OTA and CTN in HEK293 cells.


Subject(s)
Citrinin/toxicity , Ochratoxins/toxicity , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle/drug effects , Cell Survival/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , HEK293 Cells , Humans , Kidney/cytology , Kidney/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
6.
Biochem Biophys Res Commun ; 478(3): 1382-8, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27569277

ABSTRACT

The importance of endothelial progenitor cells (EPCs) in cardiovascular diseases has been demonstrated by numerous studies. Previous studies have shown that Nicotinamide phosphoribosyltransferase (NAMPT) plays a role in EPC development by regulating Sirtuin 1 (SIRT1), but the specific mechanism has not yet been elucidated. After stimulating EPCs with NAMPT, expression of SIRT1 and SIRT1 antisense long non-coding RNA (AS lncRNA) was upregulated. Upon transfection of an SIRT1 AS lncRNA overexpression vector into EPCs, SIRT1 expression was upregulated. Upon transfection of a small interfering RNA (siRNA) that targets SIRT1 AS lncRNA along with NAMPT, SIRT1 AS lncRNA was downregulated and NAMPT-induced SIRT1 expression was reduced. We used software analyses and a dual-luciferase reporter assay to demonstrate that microRNA (miR)-22 regulated SIRT1 and SIRT1 AS lncRNA. Our data suggest that SIRT1 AS lncRNA relieves miR-22-induced SIRT1 downregulation by competitively sponging miR-22. By measuring EPC senescence, proliferation, and migration, we found that NAMPT inhibited EPC senescence through an SIRT1 AS lncRNA/miR-22/SIRT1 pathway and promoted EPC proliferation and migration. These findings provide a new theoretical basis for the prevention and treatment of atherosclerosis (AS) and other cardiovascular diseases.


Subject(s)
Cell Movement/drug effects , Cellular Senescence/drug effects , Endothelial Progenitor Cells/metabolism , MicroRNAs/metabolism , Nicotinamide Phosphoribosyltransferase/pharmacology , RNA, Long Noncoding/metabolism , Signal Transduction/drug effects , Sirtuin 1/metabolism , Animals , Base Sequence , Cell Proliferation/drug effects , Endothelial Progenitor Cells/drug effects , Mice , RNA, Long Noncoding/genetics , Signal Transduction/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
7.
Int J Mol Med ; 38(2): 643-9, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27277186

ABSTRACT

Endothelial progenitor cells (EPCs) play an important role in aging-associated senescence, thereby potentially contributing to vascular pathologies. Visfatin, identified as a new adipocytokine, is closely associated with the senescence of human cells. However, the effects of visfatin on the oxidized low-density lipoprotein (ox-LDL)-induced senescence of EPCs has not yet been explored, to the best of our knowledge. For this purpose, in the present study, we examined the effects of visfatin in ox-LDL-stimulated EPCs as well as the underlying mechanism responsible for these effects. We found that visfatin attenuated the ox-LDL-induced senescence of EPCs by repressing ß-galactosidase expression and recovering telomerase activity. Western blot analysis confirmed that visfatin induced a dose-dependent increase in sirtuin 1 (SIRT1) expression in EPCs and ox-LDL exposure decreased SIRT1 expression. Silencing SIRT1 abolished the inhibition of EPC senescence and the suppression of p53 expression induced by visfatin. Moreover, visfatin attenuated the inhibition of phosphorylation of Akt, phosphoinositide-3-kinase (PI3K) and extracellular signal-regulated kinase (ERK) induced by ox-LDL. Taken together, these findings suggest that the treatment of EPCs with visfatin markedly attenuates the ox-LDL-induced senescence of EPCs by upregulating SIRT1 expression through the PI3K/Akt/ERK pathway.


Subject(s)
Cellular Senescence/drug effects , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Lipoproteins, LDL/pharmacology , Nicotinamide Phosphoribosyltransferase/metabolism , Signal Transduction/drug effects , Sirtuin 1/genetics , Up-Regulation/drug effects , Endothelial Progenitor Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Pharmacology ; 97(1-2): 63-71, 2016.
Article in English | MEDLINE | ID: mdl-26641856

ABSTRACT

A disintegrin and metalloproteinase (ADAM) 17, constitutively expressed in alveolar epithelium, is the pivotal shedding enzyme mediating acute lung inflammation. On the other hand, angiotensin (Ang)-(1-7)/Mas signaling has been shown to improve acute respiratory distress syndrome and protect alveolar epithelial cells from apoptosis. In this study, we explored the effect of Ang-(1-7)/Mas signaling on the expression and activity of ADAM17 and assessed its impact on apoptosis in lipopolysaccharide (LPS)-treated human alveolar epithelial cells. LPS markedly induced the shedding activity of ADAM17 in alveolar epithelial cells, which was blocked by selective c-Jun N-terminal kinase (JNK) inhibitor SP600125. Ang-(1-7) concentration-dependently inhibited LPS-induced ADAM17 shedding activity, which was abolished by selective Mas blocker A779 and Mas shRNA. LPS and Ang-(1-7) showed no significant effect on the expression of ADAM17. Overexpression of ADAM17 synergized with LPS on increasing the shedding activity of ADAM17 and apoptosis in alveolar epithelial cells, counteracting the inhibitory effects of Ang-(1-7). In addition, LPS significantly increased the JNK activity in alveolar epithelial cells; Ang-(1-7) concentration-dependently inhibited LPS-induced JNK activity, which was abolished by A779 and Mas shRNA. In conclusion, this study suggests that Ang-(1-7)/Mas signaling inhibits LPS-induced alveolar epithelial cell apoptosis by inhibiting LPS-induced shedding activity of ADAM17, likely by a JNK-dependent mechanism.


Subject(s)
ADAM Proteins/biosynthesis , Angiotensin I/metabolism , Apoptosis/drug effects , Epithelial Cells/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Pulmonary Alveoli/metabolism , Receptors, G-Protein-Coupled/metabolism , ADAM17 Protein , Anthracenes/pharmacology , Humans , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 4/antagonists & inhibitors , Proto-Oncogene Mas , RNA, Small Interfering , Signal Transduction , Transfection
9.
Int J Mol Med ; 36(3): 890-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26178576

ABSTRACT

Pre-B cell colony-enhancing factor (PBEF) has been shown to have a variety of biological functions. Studies have proven that PBEF plays a functional role in acute lung injury (ALI). Therefore, in this study, we aimed to confirm the importance of PBEF in ALI. The effects of PBEF overexpression on the apoptosis of human pulmonary microvascular endothelial cells (HPMECs) were analyzed by flow cytometry, and the results indicated that PBEF promoted the apoptosis of HPMECs, which aggravated the development of ALI. Comparative experiments involving increasing and decreasing PBEF expression demonstrated that PBEF promoted the expression of inflammatory factors, such as interleukin (IL)­1ß, IL­6 and IL­8 in the HPMECs , thus intensifying the inflammatory response. PBEF also inhibited the expression of aquaporin 1 (AQP1), which caused a dysfunction and imbalance in water transport. Moreover, we also found that tumor necrosis factor (TNF)­α promoted the expression of PBEF in the HPMECs. After blocking the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) pathways, we found that PBEF regulated the expression of inflammatory factors and AQP1, mainly through the MAPK pathways. Taken together, these results demonstrate that the increase in intracellular PBEF expression promoted the apoptosis of HPMECs and the expression of inflammatory factors and thus enhanced the inflammatory response and inhibited the expression of AQP1, which resulted in abnormal water transport, diminishing the regulatory effects of AQP1 on water transport.


Subject(s)
Apoptosis , Aquaporin 1/immunology , Cytokines/immunology , Inflammation Mediators/immunology , Lung/blood supply , MAP Kinase Signaling System , Microvessels/immunology , Nicotinamide Phosphoribosyltransferase/immunology , Cell Line , Cytokines/genetics , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Interleukins/immunology , Microvessels/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Up-Regulation
10.
Metabolism ; 63(8): 1012-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24930994

ABSTRACT

OBJECTIVE: Current whole genome-wide association study has identified the association of JAZF1 with type 2 diabetes; its close relation with glucose and lipid metabolism has also been revealed. However, to date, JAZF1 remains a relatively new gene with unknown function. MATERIALS/METHODS: We constructed JAZF1 overexpression vector and synthesized JAZF1 siRNA, then transfected them into 3T3-L1 adipocytes, investigated the relationship between the regulations of JAZF1, visfatin, and other adipokines, researched the specific function of JAZF1 in glucose and lipid metabolism. RESULTS: This study found that the expression of JAZF1 was gradually but significantly upregulated during the induced differentiation of 3T3-L1 preadipocytes, and that the trend of its expression was consistent with that of visfatin. Further studies indicated that JAZF1 promoted the expressions of visfatin, PPARα, and PPARß/δ in adipocytes but simultaneously inhibited the expressions of TAK1 and PPARγ. Luciferase reporter assay revealed that JAZF1 activated the transcription of visfatin, but ChIP assay results indicated that JAZF1 did not directly bind to visfatin PPRE. Our results also showed that the JAZF1 overexpression-increased visfatin expression was abolished by the addition of PPARα antagonist GW 6471 and PPARß/δ antagonist GSK 3787 respectively. And these results were further confirmed by the experiment with PPARα and PPARß/δ siRNAs. Meanwhile, we also found that JAZF1 inhibited the lipid accumulation during the differentiation of 3T3-L1 into mature adipocyte. CONCLUSIONS: Our results indicate that JAZF1 might firstly upregulated the expression of PPARα and PPARß/δ, which in turn activated the transcription of visfatin. JAZF1 plays an important role in lipid metabolism and may thus provide a potential tool for the treatment of obesity and lipid metabolism disorders among other diseases.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/physiology , Nicotinamide Phosphoribosyltransferase/metabolism , Nuclear Proteins/physiology , Peroxisome Proliferator-Activated Receptors/metabolism , Signal Transduction , 3T3-L1 Cells , Animals , Base Sequence , Chromatin Immunoprecipitation , Co-Repressor Proteins , DNA Primers , DNA-Binding Proteins , Fluorescence , Lipid Metabolism , Mice , Polymerase Chain Reaction
11.
J Diabetes ; 6(4): 279-89, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24606011

ABSTRACT

MicroRNAs (miRNAs) are important endogenous regulators in eukaryotic gene expression and a broad range of biological processes. MiRNA-related genetic variations have been proved to be associated with human diseases, such as type 2 diabetes mellitus (T2DM). Polymorphisms in miRNA genes (primary miRNAs, precursor miRNAs, mature miRNAs, and miRNA regulatory regions) may be involved in the development of T2DM by changing the expression and structure of miRNAs and target gene expression. Genetic polymorphisms of the 3'-untranslated region (UTR) in miRNA target genes may destroy putative miRNA binding sites or create new miRNA binding sites, which affects the binding of UTRs with miRNAs, finally resulting in susceptibility to and development of T2DM. Therefore, focusing on studies into genetic polymorphisms in miRNAs or miRNA binding sites will help our understanding of the pathophysiology of T2DM development and lead to better health management. Herein, we review the association of genetic polymorphisms in miRNA and miRNA targets genes with T2DM development.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation , MicroRNAs/genetics , Polymorphism, Genetic/genetics , RNA, Messenger/genetics , Animals , Binding Sites , Diabetes Mellitus, Type 2/metabolism , Humans , MicroRNAs/metabolism , RNA, Messenger/metabolism
12.
Biochem Biophys Res Commun ; 445(3): 673-80, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24583129

ABSTRACT

JAZF1 is a newly identified gene with unknown functions. A recent genome-wide association study showed that JAZF1 is associated with type 2 diabetes and is highly expressed in liver and adipose tissue. Studies have demonstrated that JAZF1 is the co-repressor for nuclear orphan receptor TAK1, whereas most nuclear orphan receptor family members are involved in the regulation of lipid metabolism. Therefore, JAZF1 could be closely related to glycolipid metabolism. In this study, JAZF1 was significantly upregulated during the induced differentiation process of 3T3-L1 preadipocytes. The overexpression of JAZF1 inhibited lipid accumulation in differentiated mature 3T3-L1 adipocytes and significantly inhibited the expression of SREBPl, ACC, and FAS, which were important in lipid synthesis, while upregulating the expression of key enzyme hormone-sensitive lipase in lipoclasis. Moreover, SREBPl exhibited an inhibitory function on the expression of JAZF1. SREBP1 reversed the inhibitory action on lipid accumulation of JAZF1. SREBP1 and JAZF1 were observed to regulate each other in adipocytes. Therefore, JAZF1 could regulate the expression of particular genes related to lipid metabolism and inhibit lipid accumulation in adipocytes. This result suggests that JAZF1 may be a potential target for the treatment of diseases, such as obesity and lipid metabolism disorders.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Developmental , Nuclear Proteins/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipogenesis , Animals , Carrier Proteins/genetics , Co-Repressor Proteins , DNA-Binding Proteins , Glucose/metabolism , Lipid Metabolism , Lipids/genetics , Mice , Nuclear Proteins/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Up-Regulation
13.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 37(4): 332-7, 2012 Apr.
Article in Chinese | MEDLINE | ID: mdl-22561562

ABSTRACT

OBJECTIVE: To determine whether central venous-to-arterial carbon dioxide tension difference (Pcv-aCO(2)) could still be used as a goal of fluid resuscitation in septic patients who already had ScvO2 greater than 70% after early resuscitation. METHODS: A prospective observational study was performed on 56 septic patients admitted to the Intensive Care Unit (ICU) in a single University Hospital, who already had ScvO2 greater than 70% after early resuscitation. They were divided into two groups, based on whether the patients' initial Pcv-aCO2 was less than 6 mmHg (low gap group) or greater than or equal to 6 mmHg (high gap group). The following data were collected at 0, 12, and 24 hours (T(0), T(12), T(24)) after study inclusion: hemodynamic indices [mean blood pressure (MAP), heart rate (HR), cardiac output (CO), central venous pressure (CVP)], perfusion-related indexes [ScvO(2), Pcv-aCO2, serum lactate (Lac), Lac clearance rate], organ function- related indices [oxygenation index (PaO2/FiO(2)), serum creatinine (SCr), creatine kinase (CK-MB)], APACHE II score, SOFA score, and 24 hours amounts of fluid infusion. RESULTS: Twenty patients (42.9%) with initial Pcv-aCO(2) ≥ 6 mmHg were included in the high gap group and another thirty-two patients were included in the low gap group. At T12 and T24, ScvO(2) and CO were significantly higher, and Lac and SCr were significantly lower in low gap patients than high gap patients (P<0.05). At T(12) and T(24), Lac clearance rate was significantly higher (P<0.05), and 24-hours amounts of fluid infusion was significantly less [(3449.47 ± 695.41) mL vs (4070.66 ± 757.43) mL, P= 0.002] for the low gap group than for the high gap group, as well as the descrease of SOFA score at T(24) (P<0.05). There was no significant difference of APACHE II score between the 2 groups (P<0.05). CO and Pcv-aCO(2) values were inversely correlated (P< 0.05). CONCLUSION: Septic patients targeting only ScvO(2) may still have inappropriate tissue perfusion, especially when Pcv-aCO2 ≥6 mmHg, which indicates insufficient resuscitation. When ScvO(2) > 70% has achieved after early resuscitation, Pcv-aCO2 can still be used as a goal of fluid resuscitation in septic patients .


Subject(s)
Carbon Dioxide/blood , Hemodynamics , Sepsis/blood , Sepsis/therapy , Vena Cava, Superior , Adult , Blood Gas Analysis , Central Venous Pressure , Female , Fluid Therapy , Humans , Male , Middle Aged , Prospective Studies , Sepsis/physiopathology , Severity of Illness Index
14.
Mol Biol Rep ; 37(7): 3051-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19823951

ABSTRACT

The aim of this study is to investigate whether silencing of Fas could have an influence on type II alveolar epithelial cell (AEC) apoptosis and inflammatory cytokine production, which prevents alveolar healing after acute lung injury (ALI). Rat primary type II AECs were isolated by elastase cell dispersion and IgG panning. The cells were transfected with Fas-specific small interfering RNA (siRNA) followed by treatment with lipopolysaccharide (LPS), Fas ligand (FasL) or both. The effects of siRNA-mediated silencing of Fas on LPS-induced apoptosis and cytokine release were then assessed. Notably, LPS, either alone or together with FasL, significantly stimulated type II AEC apoptosis and the release of tumor necrosis factor-alpha (TNF-α) and monocyte chemoattractant protein 1 (MCP-1) (P < 0.05 versus the control without treatment). Moreover, the effects exerted by both LPS and FasL were considerably counteracted by pretreatment with Fas-siRNA (P < 0.05 versus treatment with LPS and FasL). In conclusion, inhibition of Fas can diminish LPS-induced apoptosis and inflammatory cytokine production in type II AECs, and Fas specific siRNAs may have therapeutic potentials for intervention of ALI/ARDS.


Subject(s)
Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Apoptosis/drug effects , Chemokine CCL2/metabolism , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/antagonists & inhibitors , Alveolar Epithelial Cells/drug effects , Animals , Gene Silencing/drug effects , RNA, Small Interfering/metabolism , Rats , fas Receptor/metabolism
15.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 31(2): 241-4, 2006 Apr.
Article in Chinese | MEDLINE | ID: mdl-16706124

ABSTRACT

OBJECTIVE: To investigate the effect of lidocaine on LPS induced apoptosis of cultured adult rat alveolar Type II (AT-II) cells. METHODS: Cultured cells were exposed to LPS and lidocaine for 24 hours. Apoptosis and necrosis rates of cells were detected by flow cytometry and electron microscope. The activity of lactic dehydrogenase (LDH) was analyzed by using LDH kits. RESULTS: LPS induced the AT-II cell injuries by increasing not only the necrosis and apoptosis rates but also the LDH release of cultured AT-II in vitro. Lidocaine decreased the necrosis and apoptosis rates of AT-II cells. CONCLUSION: Lidocaine can directly inhibit the apoptosis and necrosis induced by LPS in cultured AT-II cells.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/pathology , Lidocaine/pharmacology , Pulmonary Alveoli/pathology , Animals , Cells, Cultured , Flow Cytometry , Lactate Dehydrogenases/metabolism , Lipopolysaccharides , Necrosis , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...