Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Braz J Cardiovasc Surg ; 39(3): e20210424, 2024 Apr 17.
Article En | MEDLINE | ID: mdl-38629941

OBJECTIVE: Intravenous non-volatile anaesthetics like propofol are commonly used in cardiac surgeries across several countries. Volatile anaesthetics like isoflurane may help in protecting the myocardium and minimize ischaemia-reperfusion injury. Hence, we did this review to compare the cardioprotective effect of isoflurane and propofol among patients undergoing coronary artery bypass grafting (CABG). METHODS: We conducted a search in the databases Medical Literature Analysis and Retrieval System Online (or MEDLINE), Embase, PubMed Central®, ScienceDirect, Google Scholar, and Cochrane Library from inception until April 2021. We carried out a meta-analysis with random-effects model and reported pooled risk ratio (RR) or standardized mean difference (SMD) with 95% confidence interval (CI) depending on the type of outcome. RESULTS: We analysed 13 studies including 808 participants. Almost all were low-quality studies. For cardiac index, the pooled SMD was 0.14 (95% CI: -0.22 to 0.50); for cardiac troponin I, pooled SMD was 0.10 (95% CI: -0.28 to 0.48). For mortality, the RR was 3.00 (95% CI: 0.32 to 28.43); for MI, pooled RR was 1.58 (95% CI: 0.59 to 4.20); and for inotropic drug use, pooled RR was 1.04 (95% CI: 0.90 to 1.21). For length of intensive care unit stay, the pooled SMD was 0.13 (95% CI: -0.29 to 0.55), while pooled SMD for mechanical ventilation time was -0.02 (95% CI: -0.54 to 0.51). CONCLUSION: Isoflurane did not have significant cardioprotective effect compared to propofol following CABG. Hence, the anaesthetists need to check some viable alternatives to manage these patients and reduce the rate of postoperative complications.


Anesthetics , Isoflurane , Propofol , Humans , Randomized Controlled Trials as Topic , Coronary Artery Bypass , Myocardium
2.
Stem Cells Int ; 2019: 7025473, 2019.
Article En | MEDLINE | ID: mdl-31611918

Mesenchymal stem cells have cannabinoid (CB) receptors type 1 and type 2 and can alleviate a variety of neuropathic pains, including chronic constriction injury (CCI). A selective CB2 receptor agonist is AM1241. In the present study, it was found that mice with CCI displayed a longer duration of mechanical and thermal analgesia when intrathecally (i.t.) injected with AM1241-treated mesenchymal stem cells, compared to those injected with untreated mesenchymal stem cells or AM1241 alone. Moreover, CCI-induced upregulation of the phosphorylated extracellular signal-regulated kinase (ERK) 1/2 (p-ERK1/2) was inhibited following i.t. injection of AM1241-treated mesenchymal stem cells and this inhibition was noticeably higher compared to injection with untreated mesenchymal stem cells. The expression of transforming growth factor-ß1 (TGF-ß1) was also analyzed in the dorsal root ganglion (DRGs) and spinal cord of CCI mice. In untreated CCI mice, expression of TGF-ß1 was increased, whereas pretreatment with AM1241-treated mesenchymal stem cells regulated the expression of TGF-ß1 on 10 days and 19 days after surgery. In addition, i.t. injection of exogenous TGF-ß1 slightly alleviated neuropathic pain whilst neutralization of TGF-ß1 potently blocked the effect of AM1241-treated mesenchymal stem cells on thermal hyperalgesia and mechanical allodynia of CCI mice. In an in vitro experiment, AM1241 could enhance the release of TGF-ß1 in the supernatant of BMSCs after lipopolysaccharide (LPS) simulation. Taken together, the findings of the current study show that i.t. administration of AM1241-treated mesenchymal stem cells has a positive effect on analgesia and that TGF-ß1 and p-ERK1/2 may be the molecular signaling pathway involved in this process.

3.
Cell Physiol Biochem ; 42(5): 1961-1972, 2017.
Article En | MEDLINE | ID: mdl-28793286

BACKGROUND/AIMS: Sepsis is a systemic inflammatory response during infection. There are limited therapeutic options for sepsis patients. Interleukin (IL)-33 has been reported recently with a beneficial effect in mouse sepsis. METHODS: In this study, we initiated a clinical study to measure serum levels of pro-inflammatory cytokines including IL-33 in sepsis patients. Next, we employed cecal ligation and puncture (CLP) to study the role of IL-33 during sepsis. To further dissect the molecular mechanism, we used in vivo knockout models and in vitro knockdown murine embryonic fibroblasts (MEFs) to investigate the cross-talk between IL-33 and IL-17 signaling, and to identify the potential downstream mediators. RESULTS: IL-33 and IL-17 were upregulated in both clinical and experimental sepsis. In CLP, IL-33 (-/-) mice showed higher mortality rate, and IL-33 treatment improved the survival rate. Elevated proinflammatory cytokines in sepsis were related to IL-17 from γδT cells. IL-33 treatment suppressed production of these cytokines by targeting IL-17 signaling both in vivo and in vitro. Finally, IL-33 was shown to inhibit the IL-17 pathway via activating suppressor of cytokine signaling (SOCS)-3. CONCLUSION: Collectively, the results suggest that IL-33 plays a negative regulatory role in sepsis progression by inhibiting IL-17 pathway through activating SOCS3. This finding would inspire a new therapeutic strategy for treating sepsis.


Interleukin-33/metabolism , Receptors, Interleukin-17/metabolism , Sepsis/diagnosis , Signal Transduction/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Systemic Inflammatory Response Syndrome/diagnosis , Animals , Case-Control Studies , Chemokine CXCL1/analysis , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Interleukin-17/analysis , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-33/analysis , Interleukin-33/genetics , Interleukin-6/analysis , Lentinula/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sepsis/mortality , Sepsis/pathology , Suppressor of Cytokine Signaling 3 Protein/antagonists & inhibitors , Suppressor of Cytokine Signaling 3 Protein/genetics , Transforming Growth Factor beta/deficiency , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation
4.
Inflammation ; 40(1): 285-294, 2017 Feb.
Article En | MEDLINE | ID: mdl-27878685

The objective of the study is to investigate the role and specific molecular mechanism of interleukin-33 (IL-33) acted on acute lung injury (ALI) induced by lipopolysaccharide (LPS). C57BL/6 mice intratracheally instilled LPS to induce ALI model. The mice were randomly divided into three groups: the sham operation group (Sham), ALI group (ALI), and pretreatment with IL-33 of ALI group (IL-33). By observing the survival rate, inflammatory cytokines in bronchoalveolar lavage fluid (BALF), myeloperoxidase (MPO) levels in lung tissue, lung histopathological examination, pulmonary capillary leakage, lung wet/dry (W/D) weight ratio, fibrosis levels in lung tissue, and associated pathways changes among the different groups, comparing to explore the role of IL-33 pretreatment on ALI mice and the possible molecular mechanisms. IL-33 pretreatment overall decreased the survival rate of ALI mice. IL-33 aggravated inflammation reaction showing as increasing the release of proinflammatory cytokines TNF-α and IL-6, increasing MPO levels in lung tissue, and aggravating lung pathology injury. In addition, IL-33 pretreatment further destroyed adherens junctions (AJs) by increasing the phosphorylation of VE-cadherin, resulting in the concomitantly pulmonary capillary barrier damage and pulmonary edema. During this process, mitogen-activated protein kinase (MAPK) pathways further activated. However, IL-33 pretreatment had no significant impact on collagen content of lung tissue. Our results indicated that IL-33 aggravated inflammatory reaction and increased microvascular permeability, but had little effect on pulmonary fibrosis, associated with the further activation of MAPK family proteins in the process. To sum up, IL-33 decreased survival rate and aggravated LPS-induced ALI.


Acute Lung Injury/drug therapy , Interleukin-33/pharmacology , Acute Lung Injury/chemically induced , Animals , Bronchoalveolar Lavage Fluid/chemistry , Capillary Permeability/drug effects , Cytokines/drug effects , Inflammation/chemically induced , Interleukin-33/administration & dosage , Lipopolysaccharides , Lung/drug effects , Lung/pathology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Survival Rate
5.
Adv Healthc Mater ; 5(10): 1213-21, 2016 05.
Article En | MEDLINE | ID: mdl-27028159

The long-term use of potent analgesics is often needed to treat chronic pain. However, it has been greatly hindered by their side effects such as addiction and withdrawal reactions. The study seeks to circumvent these drawbacks by taking advantage of a multifunctional delivery system based on nanoparticles to target on pathological neuroinflammation. A drug delivery system is designed and generated using mesoporous silica nanoparticles (MSNs) that are loaded with Δ9-THC (Δ9-tetrahydrocannabinol, a cannabinoid) and ARA290 (an erythropoietin-derived polypeptide), both of which possess analgesic and anti-inflammatory functions. The actions of such THC-MSN-ARA290 nanocomplexes depend on the enhanced permeability and retention of THC through nanosized carriers, and a redox-sensitive release of conjugated ARA290 peptide into the local inflammatory milieu. The biosafety and anti-inflammatory effects of the nanocomplexes are first evaluated in primary microglia in vitro, and further in a mouse model of chronic constriction injury. It is found that the nanocomplexes attenuate in vitro and in vivo inflammation, and achieve a sustained relief of neuropathic pain in injured animals induced by both thermal hyperalgesia and mechanical allodynia. Thus, a nanoparticle-based carrier system can be useful for the amelioration of chronic neuropathic pain through combinatorial drug delivery.


Analgesics/administration & dosage , Analgesics/chemistry , Neuralgia/drug therapy , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Cell Line , Dronabinol/administration & dosage , Dronabinol/chemistry , Drug Carriers/chemistry , Drug Delivery Systems/methods , Hyperalgesia/drug therapy , Inflammation/drug therapy , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Oxidation-Reduction , Pain Management/methods
6.
Oncotarget ; 7(6): 6436-47, 2016 Feb 09.
Article En | MEDLINE | ID: mdl-26824325

Chronic pain is commonly and closely correlated with inflammation. Both cannabinoid signaling and mesenchymal stem cells (MSCs) have been demonstrated to reduce inflammatory pain. Although cannabinoid signaling is essential for mesenchymal stem cell survival and differentiation, little is known about its role in modulatory effect of MSCs on inflammation and pain sensitivity. Here we showed that mouse bone-marrow derived MSCs (BM-MSCs) expressed both cannabinoid receptor type 1 and 2 (CB1 and CB2). CB2 expression level in BM-MSCs increased with their maturation. In addition, we found that tetrahydrocannabinol (THC) activated CB2 receptor and ERK signaling, consequently enhancing the modulation of MSCs on inflammation-associated cytokine release from lipopolysaccharides-stimulated microglia. Consistent with in vitro data, THC pretreatment enhanced the immunomodulatory effects of BM-MSC on thermal hyperalgesia and mechanical allodynia in chronic constriction injury model, by decreasing the release of pro-inflammation cytokines. Our study revealed the crucial role of THC in promoting the immunomodulatory effects of MSCs and proposed a new strategy to alleviate pain based on stem cells therapy.


Bone Marrow/immunology , Dronabinol/pharmacology , Immunomodulation , Mesenchymal Stem Cells/immunology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Animals , Apoptosis , Blotting, Western , Bone Marrow/drug effects , Bone Marrow/metabolism , Cannabinoid Receptor Agonists/pharmacology , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hyperalgesia/drug therapy , Hyperalgesia/immunology , Hyperalgesia/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Nerve/drug effects , Sciatic Nerve/immunology , Sciatic Nerve/metabolism , Signal Transduction , Transcriptional Activation , Up-Regulation
7.
Med Oncol ; 32(5): 145, 2015 May.
Article En | MEDLINE | ID: mdl-25814287

Wingless-type (Wnt) family of secreted glycoproteins is a group of signal molecules implicated in oncogenesis. Abnormal activation of Wnt signal pathway is associated with a variety of human cancers, including non-small cell lung cancer (NSCLC). Wnt antagonists, such as the secreted frizzled-related protein (SFRP) family, Wnt inhibitory factor-1 (WIF-1) and cerberus, inhibit Wnt signal pathway by directly binding to Wnt molecules. Norcantharidin (NCTD) is known to possess anticancer activity but less nephrotoxicity than cantharidin. In this study, we found that NCTD inhibited cell proliferation, induced apoptosis, arrested cell cycle and suppressed cell invasion/migration in vitro. Additionally, Wnt signal pathway transcription was also suppressed. NCTD treatment blocked cytoplasmic translocation of beta-catenin into the nucleus. Alterations of apoptosis-related proteins, such as Bax, cleaved caspase-3 (pro-apoptotic) and Bcl-2 (anti-apoptotic), had been detected. Furthermore, the expression levels of WIF-1 and SFRP1 were significantly increased in NCTD-treated groups compared with negative control (NC) groups. Abnormal methylation was observed in NC groups, while NCTD treatment promoted WIF-1 demethylation. The present study revealed that NCTD activated WIF-1 via promoter demethylation, inhibiting the canonical Wnt signal pathway in NSCLC, which may present a new therapeutic target in vivo.


Adaptor Proteins, Signal Transducing/genetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation/drug effects , Lung Neoplasms/genetics , Promoter Regions, Genetic/drug effects , Repressor Proteins/genetics , Wnt Signaling Pathway/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Caspase 3/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Methylation/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/drug therapy , Membrane Proteins/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Wnt Signaling Pathway/genetics , bcl-2-Associated X Protein/genetics , beta Catenin/genetics
...