Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Chem Biol Interact ; 395: 111010, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38679114

ABSTRACT

The incidence and mortality rate of myocardial infarction are increasing per year in China. The polarization of macrophages towards the classically activated macrophages (M1) phenotype is of utmost importance in the progression of inflammatory stress subsequent to myocardial infarction. Poly (ADP-ribose) polymerase 1(PARP1) is the ubiquitous and best characterized member of the PARP family, which has been reported to support macrophage polarization towards the pro-inflammatory phenotype. Yet, the role of PARP1 in myocardial ischemic injury remains to be elucidated. Here, we demonstrated that a myocardial infarction mouse model induced cardiac damage characterized by cardiac dysfunction and increased PARP1 expression in cardiac macrophages. Inhibition of PARP1 by the PJ34 inhibitors could effectively alleviate M1 macrophage polarization, reduce infarction size, decrease inflammation and rescue the cardiac function post-MI in mice. Mechanistically, the suppression of PARP1 increase NLRC5 gene expression, and thus inhibits the NF-κB pathway, thereby decreasing the production of inflammatory cytokines such as IL-1ß and TNF-α. Inhibition of NLRC5 promote infection by effectively abolishing the influence of this mechanism discussed above. Interestingly, inhibition of NLRC5 promotes cardiac macrophage polarization toward an M1 phenotype but without having major effects on M2 macrophages. Our results demonstrate that inhibition of PARP1 increased NLRC5 gene expression, thereby suppressing M1 polarization, improving cardiac function, decreasing infarct area and attenuating inflammatory injury. The aforementioned findings provide new insights into the proinflammatory mechanisms that drive macrophage polarization following myocardial infarction, thereby introducing novel potential targets for future therapeutic interventions in individuals affected by myocardial infarction.


Subject(s)
Intracellular Signaling Peptides and Proteins , Macrophages , Myocardial Infarction , NF-kappa B , Poly (ADP-Ribose) Polymerase-1 , Animals , Male , Mice , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/metabolism , Macrophages/drug effects , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , NF-kappa B/metabolism , Phenanthrenes/pharmacology , Phenanthrenes/therapeutic use , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Up-Regulation/drug effects
2.
Oxid Med Cell Longev ; 2019: 7192798, 2019.
Article in English | MEDLINE | ID: mdl-30911349

ABSTRACT

Bupivacaine, a typical local anesthetic, induces neurotoxicity via reactive oxygen species regulation of apoptosis. High glucose could enhance bupivacaine-induced neurotoxicity through regulating oxidative stress, but the mechanism of it is not clear. Mitochondrial calcium uniporter (MCU), a key channel for regulating the mitochondrial Ca2+ (mCa2+) influx, is closely related to oxidative stress via disruption of mCa2+ homeostasis. Whether MCU is involved in high glucose-sensitized bupivacaine-induced neurotoxicity remains unknown. In this study, human neuroblastoma (SH-SY5Y) cells were cultured with high glucose and/or bupivacaine, and the data showed that high glucose enhanced bupivacaine-induced MCU expression elevation, mCa2+ accumulation, and oxidative damage. Next, Ru360, an inhibitor of MCU, was employed to pretreated SH-SY5Y cells, and the results showed that it could decrease high glucose and bupivacaine-induced mCa2+ accumulation, oxidative stress, and apoptosis. Further, with the knockdown of MCU with a specific small interfering RNA (siRNA) in SH-SY5Y cells, we found that it also could inhibit high glucose and bupivacaine-induced mCa2+ accumulation, oxidative stress, and apoptosis. We propose that downregulation expression or activity inhibition of the MCU channel might be useful for restoring the mitochondrial function and combating high glucose and bupivacaine-induced neurotoxicity. In conclusion, our study demonstrated the crucial role of MCU in high glucose-mediated enhancement of bupivacaine-induced neurotoxicity, suggesting the possible use of this channel as a target for curing bupivacaine-induced neurotoxicity in diabetic patients.


Subject(s)
Bupivacaine/adverse effects , Calcium Channels/metabolism , Glucose/toxicity , Neurotoxins/toxicity , Oxidative Stress/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Apoptosis/drug effects , Calcium/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Ruthenium Compounds/pharmacology
3.
Neural Plast ; 2018: 9163521, 2018.
Article in English | MEDLINE | ID: mdl-29853850

ABSTRACT

Sustained activation of NLRP3 inflammasome is closely related to diabetes and stroke. However, it is unknown whether NLRP3 inflammasome plays an essential role in stroke in diabetes. We aim to investigate the effect and the potential mechanism of NLRP3 inflammasome in diabetic mice with cerebral ischemia-reperfusion injury. A type 2 diabetic mouse model was induced by a high-fat diet and streptozotocin (STZ). Diabetic mice received MCC950 (the specific molecule NLRP3 inhibitor) or vehicle 60 minutes before the middle cerebral artery occlusion (MCAO) and reperfusion. MCC950 reduced the neurological deficit score of 24 h after cerebral ischemia reperfusion and improved the 28-day survival rate of cerebral ischemia-reperfusion injury in diabetic mice. Furthermore, we found that the mRNA transcription levels of NLRP3, IL-1ß, and caspase-1 in the core ischemic area were remarkably amplified in diabetic mice with cerebral ischemia-reperfusion injury, whereas this phenomenon was obviously attenuated by MCC950 pretreatment. In conclusion, the NLRP3 inflammasome was involved in the complex diseases of diabetic stroke. MCC950, the NLRP3 specific inhibitor, ameliorated diabetic mice with cerebral ischemia-reperfusion injury and improved the 28-day survival rate during the recovery stage of ischemic stroke.


Subject(s)
Brain Ischemia/metabolism , Diabetes Mellitus, Type 2/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Reperfusion Injury/metabolism , Stroke/metabolism , Animals , Brain Ischemia/complications , Brain Ischemia/prevention & control , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/complications , Disease Models, Animal , Furans , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Indenes , Male , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/complications , Reperfusion Injury/prevention & control , Streptozocin , Stroke/complications , Sulfonamides , Sulfones/administration & dosage
4.
Sci Rep ; 7: 45316, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28338089

ABSTRACT

Local anaesthetics (LAs) may lead to neurological complications, but the underlying mechanism is still unclear. Many neurotoxicity research studies have examined different LAs, but none have comprehensively explored the distinct mechanisms of neurotoxicity caused by amide- (bupivacaine) and ester- (procaine) type LAs. Here, based on a CCK8 assay, LDH assay, Rhod-2-AM and JC-1 staining, 2',7'-dichlorohy-drofluorescein diacetate and dihydroethidium probes, an alkaline comet assay, and apoptosis assay, we show that both bupivacaine and procaine significantly induce mitochondrial calcium overload and a decline in the mitochondrial membrane potential as well as overproduction of ROS, DNA damage and apoptosis (P < 0.05). There were no significant differences in mitochondrial injury and apoptosis between the bupivacaine and procaine subgroups (P > 0.05). However, to our surprise, the superoxide anionic level after treatment with bupivacaine, which leads to more severe DNA damage, was higher than the level after treatment with procaine, while procaine produced more peroxidation than bupivacaine. Some of these results were also affirmed in dorsal root ganglia neurons of C57 mice. The differences in the superoxidation and peroxidation induced by these agents suggest that different types of LAs may cause neurotoxicity via different pathways. We can target more accurate treatment based on their different mechanisms of neurotoxicity.


Subject(s)
Anesthetics, Local/toxicity , Bupivacaine/toxicity , Neurons/drug effects , Procaine/toxicity , Amides/chemistry , Animals , Apoptosis/drug effects , Bupivacaine/chemistry , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , DNA Damage/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Lipid Peroxidation/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Procaine/chemistry , Reactive Oxygen Species
5.
Oxid Med Cell Longev ; 2015: 683197, 2015.
Article in English | MEDLINE | ID: mdl-26161242

ABSTRACT

Hyperglycemia can inhibit expression of the 8-oxoG-DNA glycosylase (OGG1) which is one of the key repair enzymes for DNA oxidative damage. The effect of hyperglycemia on OGG1 expression in response to local anesthetics-induced DNA damage is unknown. This study was designed to determine whether high glucose inhibits OGG1 expression and aggravates bupivacaine-induced DNA damage via reactive oxygen species (ROS). SH-SY5Y cells were cultured with or without 50 mM glucose for 8 days before they were treated with 1.5 mM bupivacaine for 24 h. OGG1 expression was measured by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. ROS was estimated using the redox-sensitive fluorescent dye DCFH-DA. DNA damage was investigated with immunostaining for 8-oxodG and comet assays. OGG1 expression was inhibited in cells exposed to high glucose with concomitant increase in ROS production and more severe DNA damage as compared to control culture conditions, and these changes were further exacerbated by bupivacaine. Treatment with the antioxidant N-acetyl-L-cysteine (NAC) prevented high glucose and bupivacaine mediated increase in ROS production and restored functional expression of OGG1, which lead to attenuated high glucose-mediated exacerbation of bupivacaine neurotoxicity. Our findings indicate that subjects with diabetes may experience more detrimental effects following bupivacaine use.


Subject(s)
Bupivacaine/toxicity , DNA Damage/drug effects , DNA Glycosylases/metabolism , Glucose/pharmacology , Oxidative Stress/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Acetylcysteine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Comet Assay , DNA Glycosylases/genetics , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Humans , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
6.
Oxid Med Cell Longev ; 2013: 159864, 2013.
Article in English | MEDLINE | ID: mdl-24228138

ABSTRACT

Local anesthetics are used routinely and effectively. However, many are also known to activate neurotoxic pathways. We tested the neuroprotective efficacy of ginkgolide B (GB), an active component of Ginkgo biloba, against ROS-mediated neurotoxicity caused by the local anesthetic bupivacaine. SH-SY5Y cells were treated with different concentrations of bupivacaine alone or following preincubation with GB. Pretreatment with GB increased SH-SY5Y cell viability and attenuated intracellular ROS accumulation, apoptosis, mitochondrial dysfunction, and ER stress. GB suppressed bupivacaine-induced mitochondrial depolarization and mitochondria complex I and III inhibition and increased cleaved caspase-3 and Htra2 expression, which was strongly indicative of activation of mitochondria-dependent apoptosis with concomitantly enhanced expressions of Grp78, caspase-12 mRNA, protein, and ER stress. GB also improved ultrastructural changes indicative of mitochondrial and ER damage induced by bupivacaine. These results implicate bupivacaine-induced ROS-dependent mitochondria, ER dysfunction, and apoptosis, which can be attenuated by GB through its antioxidant property.


Subject(s)
Apoptosis/drug effects , Bupivacaine/toxicity , Ginkgolides/pharmacology , Lactones/pharmacology , Neuroprotective Agents/pharmacology , Caspase 12/genetics , Caspase 12/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Shape/genetics , Cell Survival/drug effects , Cell Survival/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex III/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Enzyme Activation/drug effects , Flow Cytometry , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , High-Temperature Requirement A Serine Peptidase 2 , Humans , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Serine Endopeptidases/metabolism
7.
Neurochem Res ; 38(9): 1880-94, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23817845

ABSTRACT

Bupivacaine is one of the most toxic local anesthetics but the mechanisms underlying its neurotoxicity are still unclear. Intracellular nicotinamide adenine dinucleotide (NAD(+)) depletion has been demonstrated to play an essential role in neuronal injury. In the present study, we investigated whether intracellular NAD(+) depletion contributes to bupivacaine-induced neuronal injury and whether NAD(+) repletion attenuates the injury in SH-SY5Y cells. First, we evaluated the intracellular NAD(+) content after bupivacaine exposure. We also examined the cellular NAD(+) level after pretreatment with exogenous NAD(+). We next determined cell viability and the apoptosis rate after bupivacaine treatment in the presence or absence of NAD(+) incubation. Finally, cell injuries such as nuclear injury, reactive oxygen species (ROS) production, and mitochondrial depolarization were detected after bupivacaine treatment with or without NAD(+) pretreatment. Bupivacaine caused intracellular NAD(+) depletion in a time- and concentration-dependent manner. Cellular NAD(+) replenishment prevented cell death and apoptosis induced by bupivacaine. Importantly, exogenous NAD(+) attenuated bupivacaine-induced nuclear injury, ROS production, and mitochondrial depolarization. Our results suggest that NAD(+) depletion is necessary for bupivacaine-induced neuronal necrosis and apoptosis, and that NAD(+) repletion attenuates neurotoxicity resulting from bupivacaine-treatment.


Subject(s)
NAD/metabolism , Bupivacaine/pharmacology , Cell Line, Tumor , Flow Cytometry , Humans , Lidocaine/pharmacology , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL