Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.485
Filter
1.
Stroke ; 55(7): 1904-1913, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38913800

ABSTRACT

BACKGROUND: The mitochondrial unfolded protein response (UPRmt) is an evolutionarily conserved mitochondrial response that is critical for maintaining mitochondrial and energetic homeostasis under cellular stress after tissue injury and disease. Here, we ask whether UPRmt may be a potential therapeutic target for ischemic stroke. METHODS: We performed the middle cerebral artery occlusion and oxygen-glucose deprivation models to mimic ischemic stroke in vivo and in vitro, respectively. Oligomycin and meclizine were used to trigger the UPRmt. We used 2,3,5-triphenyltetrazolium chloride staining, behavioral tests, and Nissl staining to evaluate cerebral injury in vivo. The Cell Counting Kit-8 assay and the Calcein AM Assay Kit were conducted to test cerebral injury in vitro. RESULTS: Inducing UPRmt with oligomycin protected neuronal cultures against oxygen-glucose deprivation. UPRmt could also be triggered with meclizine, and this Food and Drug Administration-approved drug also protected neurons against oxygen-glucose deprivation. Blocking UPRmt with siRNA against activating transcription factor 5 eliminated the neuroprotective effects of meclizine. In a mouse model of focal cerebral ischemia, pretreatment with meclizine was able to induce UPRmt in vivo, which reduced infarction and improved neurological outcomes. CONCLUSIONS: These findings suggest that the UPRmt is important in maintaining the survival of neurons facing ischemic/hypoxic stress. The UPRmt mechanism may provide a new therapeutic avenue for ischemic stroke.


Subject(s)
Brain Ischemia , Glucose , Mitochondria , Neurons , Unfolded Protein Response , Animals , Neurons/metabolism , Neurons/drug effects , Mice , Glucose/deficiency , Unfolded Protein Response/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Brain Ischemia/metabolism , Male , Infarction, Middle Cerebral Artery/metabolism , Oxygen/metabolism , Mice, Inbred C57BL , Cells, Cultured , Neuroprotective Agents/pharmacology
2.
Biochem Biophys Res Commun ; 724: 150216, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38851140

ABSTRACT

Death-associated protein kinase (DAPK) 1 is a critical mediator for neuronal cell death in cerebral ischemia, but its role in blood-brain barrier (BBB) disruption is incompletely understood. Here, we found that endothelial-specific deletion of Dapk1 using Tie2 Cre protected the brain of Dapk1fl/fl mice against middle cerebral artery occlusion (MCAO), characterized by mitigated Evans blue dye (EBD) extravasation, reduced infarct size and improved behavior. In vitro experiments also indicated that DAPK1 deletion inhibited oxygen-glucose deprivation (OGD)-induced tight junction alteration between cerebral endothelial cells (CECs). Mechanistically, we revealed that DAPK1-DAPK3 interaction activated cytosolic phospholipase A2 (cPLA2) in OGD-stimulated CECs. Our results thus suggest that inhibition of endothelial DAPK1 specifically prevents BBB damage after stroke.


Subject(s)
Blood-Brain Barrier , Death-Associated Protein Kinases , Endothelial Cells , Animals , Death-Associated Protein Kinases/metabolism , Death-Associated Protein Kinases/genetics , Death-Associated Protein Kinases/deficiency , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Mice , Endothelial Cells/metabolism , Endothelial Cells/pathology , Male , Gene Deletion , Stroke/metabolism , Stroke/pathology , Stroke/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/genetics , Mice, Inbred C57BL , Mice, Knockout , Glucose/metabolism , Glucose/deficiency , Tight Junctions/metabolism
3.
BMC Neurol ; 24(1): 204, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879468

ABSTRACT

Hypoxia can cause a variety of diseases, including ischemic stroke and neurodegenerative diseases. Within a certain range of partial pressure of oxygen, cells can respond to changes in oxygen. Changes in oxygen concentration beyond a threshold will cause damage or even necrosis of tissues and organs, especially for the central nervous system. Therefore, it is very important to find appropriate measures to alleviate damage. MiRNAs can participate in the regulation of hypoxic responses in various types of cells. MiRNAs are involved in regulating hypoxic responses in many types of tissues by activating the hypoxia-inducible factor (HIF) to affect angiogenesis, glycolysis and other biological processes. By analyzing differentially expressed miRNAs in hypoxia and hypoxia-related studies, as well as the HT22 neuronal cell line under hypoxic stress, we found that the expression of miR-18a was changed in these models. MiR-18a could regulate glucose metabolism in HT22 cells under hypoxic stress by directly regulating the 3'UTR of the Hif1a gene. As a small molecule, miRNAs are easy to be designed into small nucleic acid drugs, so this study can provide a theoretical basis for the research and treatment of nervous system diseases caused by hypoxia.


Subject(s)
Glucose , Hippocampus , Hypoxia-Inducible Factor 1, alpha Subunit , MicroRNAs , Neurons , Animals , Humans , Mice , Cell Hypoxia/physiology , Cell Line , Glucose/metabolism , Glucose/deficiency , Hippocampus/metabolism , Hippocampus/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MicroRNAs/metabolism , MicroRNAs/genetics , Neurons/metabolism
4.
Biomed Pharmacother ; 176: 116778, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38788601

ABSTRACT

The incidence of cerebral infarction triggered by abnormal glucose tolerance has increased; however, the relationship between glucose concentration in the brain and the detailed mechanism of post ischemic cell death remains unclear. Nicotinamide phosphoribosyltransferase (NAMPT), an adipocytokine, is the rate-limiting enzyme for NAD+ synthesis in the salvage pathway. Although NAMPT activation prevents neuronal injury, the relationship between NAMPT activity, glucose metabolism disorders, and cerebral ischemia-induced neuronal cell death is unknown. In this study, we determined changes in NAMPT on cerebral ischemic injuries with diabetes using a db/db mouse model of type 2 diabetes and then identified the underlying mechanisms using Neuro2a cells. The expression of inflammatory cytokine mRNAs was increased in db/db and db/+ middle cerebral artery occlusion and reperfusion (MCAO/R) mice. Although NeuN-positive cells were decreased after MCAO/R, the number of NAMPT and NeuN double-positive cells in NeuN-positive neuronal cells increased in db/db MCAO/R mice. Next, the role of NAMPT in Neuro2a cells under conditions of high glucose (HGC) and oxygen-glucose deprivation (OGD), which mimics diabetes-complicated cerebral infarction, was examined. Treatment with P7C3-A20, a NAMPT activator, suppressed the decrease in cell viability caused by HGC/OGD; however, there were no significant differences in the levels of cleaved caspase-3 and Bax proteins. Moreover, increased FoxO3a and LC3-II levels after HGC/OGD were inhibited by P7C3-A20 treatment. Our findings indicate that NAMPT activation is associated with neuronal survival under ischemic conditions with abnormal glucose tolerance through the regulation of FoxO3a/LC3.


Subject(s)
Brain Ischemia , Cell Survival , Forkhead Box Protein O3 , Glucose , Neurons , Nicotinamide Phosphoribosyltransferase , Signal Transduction , Animals , Nicotinamide Phosphoribosyltransferase/metabolism , Forkhead Box Protein O3/metabolism , Glucose/metabolism , Glucose/deficiency , Neurons/metabolism , Neurons/pathology , Neurons/drug effects , Male , Mice , Cell Survival/drug effects , Signal Transduction/drug effects , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cytokines/metabolism , Mice, Inbred C57BL , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/metabolism , Cell Line, Tumor , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications
5.
Tissue Cell ; 88: 102401, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749116

ABSTRACT

Myocardial infarction (MI) is a kind of cardiovascular diseases with high morbidity and mortality. Poricoic acid A (PAA) is the main active substance in Poria cocos, which has been discovered to exhibit an ameliorative role in the progression of many diseases. However, no report has been focused on the regulatory effects of PAA on MI progression. In this study, at first, oxygen glucose deprivation (OGD) treatment was performed in human cardiac microvascular endothelial cells (HCMECs) to mimic MI cell model. Our findings demonstrated that cell proliferation was reduced post OGD treatment, but which was reversed by PAA treatment. Moreover, PAA suppressed cell apoptosis in OGD-triggered HCMEC cells. Next, it revealed that PAA induced autophagy in OGD-treated HCMEC cells through enhancing LC3-II/LC3-I level and reducing P62 level. In addition, PAA strengthened the angiogenesis ability and migration ability in OGD-induced HCMEC cells. Lastly, it was uncovered that PAA modulated the AMPK/mTOR signaling pathway through affecting the p-mTOR/mTOR and p-AMPK/AMPK levels. In conclusion, PAA can promote angiogenesis and myocardial regeneration after MI by inducing autophagy through modulating the AMPK/mTOR pathway. This work suggested that PAA may be a potential and useful drug for MI treatment.


Subject(s)
Autophagy , Myocardial Infarction , Neovascularization, Physiologic , Signal Transduction , Autophagy/drug effects , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Humans , Neovascularization, Physiologic/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Regeneration/drug effects , Myocardium/pathology , Myocardium/metabolism , Cell Proliferation/drug effects , Glucose/deficiency , Glucose/metabolism , Cell Movement/drug effects , AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Angiogenesis
6.
J Biol Chem ; 300(5): 107270, 2024 May.
Article in English | MEDLINE | ID: mdl-38599381

ABSTRACT

Higher demand for nutrients including glucose is characteristic of cancer. "Starving cancer" has been pursued to curb tumor progression. An intriguing regime is to inhibit glucose transporter GLUT1 in cancer cells. In addition, during cancer progression, cancer cells may suffer from insufficient glucose supply. Yet, cancer cells can somehow tolerate glucose starvation. Uncovering the underlying mechanisms shall shed insight into cancer progression and benefit cancer therapy. TFE3 is a transcription factor known to activate autophagic genes. Physiological TFE3 activity is regulated by phosphorylation-triggered translocation responsive to nutrient status. We recently reported TFE3 constitutively localizes to the cell nucleus and promotes cell proliferation in kidney cancer even under nutrient replete condition. It remains unclear whether and how TFE3 responds to glucose starvation. In this study, we show TFE3 promotes kidney cancer cell resistance to glucose starvation by exposing cells to physiologically relevant glucose concentration. We find glucose starvation triggers TFE3 protein stabilization through increasing its O-GlcNAcylation. Furthermore, through an unbiased functional genomic study, we identify SLC36A1, a lysosomal amino acid transporter, as a TFE3 target gene sensitive to TFE3 protein level. We find SLC36A1 is overexpressed in kidney cancer, which promotes mTOR activity and kidney cancer cell proliferation. Importantly, SLC36A1 level is induced by glucose starvation through TFE3, which enhances cellular resistance to glucose starvation. Suppressing TFE3 or SLC36A1 significantly increases cellular sensitivity to GLUT1 inhibitor in kidney cancer cells. Collectively, we uncover a functional TFE3-SLC36A1 axis that responds to glucose starvation and enhances starvation tolerance in kidney cancer.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Glucose , Kidney Neoplasms , Humans , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glucose/deficiency , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Amino Acid Transport Systems , Symporters
7.
Am J Physiol Cell Physiol ; 326(6): C1611-C1624, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38646789

ABSTRACT

The influence of SGLT-1 on perivascular preadipocytes (PVPACs) and vascular remodeling is not well understood. This study aimed to elucidate the role and mechanism of SGLT-1-mediated PVPACs bioactivity. PVPACs were cultured in vitro and applied ex vivo to the carotid arteries of mice using a lentivirus-based thermosensitive in situ gel (TISG). The groups were treated with Lv-SGLT1 (lentiviral vector, overexpression), Lv-siSGLT1 (RNA interference, knockdown), or specific signaling pathway inhibitors. Assays were conducted to assess changes in cell proliferation, apoptosis, glucose uptake, adipogenic differentiation, and vascular remodeling in the PVPACs. Protein expression was analyzed by Western blotting, immunocytochemistry, and/or immunohistochemistry. The methyl thiazolyl tetrazolium (MTT) assay and Hoechst 33342 staining indicated that SGLT-1 overexpression significantly promoted PVPACs proliferation and inhibited apoptosis in vitro. Conversely, SGLT-1 knockdown exerted the opposite effect. Oil Red O staining revealed that SGLT-1 overexpression facilitated adipogenic differentiation, while its inhibition mitigated these effects. 3H-labeled glucose uptake experiments demonstrated that SGLT-1 overexpression enhanced glucose uptake by PVPACs, whereas RNA interference-mediated SGLT-1 inhibition had no significant effect on glucose uptake. Moreover, RT-qPCR, Western blotting, and immunofluorescence analyses revealed that SGLT-1 overexpression upregulated FABP4 and VEGF-A levels and activated the Akt/mTOR/p70S6K signaling pathway, whereas SGLT-1 knockdown produced the opposite effects. In vivo studies corroborated these findings and indicated that SGLT-1 overexpression facilitated carotid artery remodeling. Our study demonstrates that SGLT-1 activation of the Akt/mTOR/p70S6K signaling pathway promotes PVPACs proliferation, adipogenesis, glucose uptake, glucolipid metabolism, and vascular remodeling.NEW & NOTEWORTHY SGLT-1 is expressed in PVPACs and can affect preadipocyte glucolipid metabolism and vascular remodeling. SGLT-1 promotes the biofunctions of PVPACs mediated by Akt/mTOR/p70S6K signaling pathway. Compared with caudal vein or intraperitoneal injection, the external application of lentivirus-based thermal gel around the carotid artery is an innovative attempt at vascular remodeling model, it may effectively avoid the transfection of lentiviral vector into the whole body of mice and the adverse effect on experimental results.


Subject(s)
Adipocytes , Cell Proliferation , Proto-Oncogene Proteins c-akt , Ribosomal Protein S6 Kinases, 70-kDa , Signal Transduction , Sodium-Glucose Transporter 1 , TOR Serine-Threonine Kinases , Animals , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Mice , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Adipocytes/metabolism , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 1/genetics , Male , Adipogenesis/physiology , Mice, Inbred C57BL , Vascular Remodeling , Cells, Cultured , Apoptosis , Cell Differentiation , Glucose/metabolism , Glucose/deficiency
8.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 142-147, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38650139

ABSTRACT

The diagnostic biomarkers associated with ischemic stroke (IS) that may have clinical utility remain elucidated. Thus, the potential functional lncRNAs in IS were explored. The Gene Expression Omnibus database provided the transcriptome profile of IS for download. WGCNA analysis and integrated bioinformatics were used to find genes that were differentially expressed (DEGs). The Starbase database created the lncRNA-based ceRNA network. In order to investigate the molecular mechanism and involved pathways of DEGs in IS, functional enrichment analysis was carried out. Using qRT-PCR, lncRNAs identified as putative IS biomarkers were confirmed to be expressed in a permanent middle cerebral artery occlusion (MCAO) model. Using the annexin V/PI apoptosis test, the amount of apoptosis in oxygen-glucose deprivation (OGD) cells was measured. A total of 1600 common differentially expressed - protein-coding RNA (DE-pcRNAs) and 26 DE-lncRNAs were identified. The results of enrichment analysis indicate that the cytokine may be regulated by common DE-pcRNAs and are vital in the progress of IS. A lncRNAs-mediated ceRNA network including lncRNAs AU020206, Brip1os, F630028O10Rik and 9530082P21Rik was constructed. The expression of these lncRNAs was significantly increased in MCAO model. Knockdown of lncRNA AU020206 inhibited microglia apoptosis in OGD cell model. We constructed a lncRNAs-mediated ceRNA network and found that lncRNA AU020206 inhibited microglia apoptosis in OGD cell model. These findings provided further evidence for the diagnosis and a novel avenue for targeted therapy of IS.


Subject(s)
Apoptosis , Ischemic Stroke , Microglia , RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Apoptosis/genetics , Apoptosis/drug effects , Ischemic Stroke/genetics , Ischemic Stroke/pathology , Ischemic Stroke/metabolism , Animals , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Gene Knockdown Techniques , Male , Gene Regulatory Networks , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Glucose/metabolism , Glucose/deficiency , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation/drug effects , Transcriptome/genetics , Disease Models, Animal
9.
Neurochem Res ; 49(7): 1762-1781, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38551797

ABSTRACT

Lactate has received attention as a potential therapeutic intervention for brain diseases, particularly those including energy deficit, exacerbated inflammation, and disrupted redox status, such as cerebral ischemia. However, lactate roles in metabolic or signaling pathways in neural cells remain elusive in the hypoxic and ischemic contexts. Here, we tested the effects of lactate on the survival of a microglial (BV-2) and a neuronal (SH-SY5Y) cell lines during oxygen and glucose deprivation (OGD) or OGD followed by reoxygenation (OGD/R). Lactate signaling was studied by using 3,5-DHBA, an exogenous agonist of lactate receptor GPR81. Inhibition of lactate dehydrogenase (LDH) or monocarboxylate transporters (MCT), using oxamate or 4-CIN, respectively, was performed to evaluate the impact of lactate metabolization and transport on cell viability. The OGD lasted 6 h and the reoxygenation lasted 24 h following OGD (OGD/R). Cell viability, extracellular lactate concentrations, microglial intracellular pH and TNF-ɑ release, and neurite elongation were evaluated. Lactate or 3,5-DHBA treatment during OGD increased microglial survival during reoxygenation. Inhibition of lactate metabolism and transport impaired microglial and neuronal viability. OGD led to intracellular acidification in BV-2 cells, and reoxygenation increased the release of TNF-ɑ, which was reverted by lactate and 3,5-DHBA treatment. Our results suggest that lactate plays a dual role in OGD, acting as a metabolic and a signaling molecule in BV-2 and SH-SY5Y cells. Lactate metabolism and transport are vital for cell survival during OGD. Moreover, lactate treatment and GPR81 activation during OGD promote long-term adaptations that potentially protect cells against secondary cell death during reoxygenation.


Subject(s)
Cell Survival , Glucose , Lactic Acid , Microglia , Neurons , Oxygen , Microglia/metabolism , Microglia/drug effects , Glucose/metabolism , Glucose/deficiency , Humans , Neurons/metabolism , Neurons/drug effects , Oxygen/metabolism , Lactic Acid/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Animals , Mice , Neuroprotective Agents/pharmacology , Cell Hypoxia/physiology , Cell Hypoxia/drug effects , Tumor Necrosis Factor-alpha/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Line, Tumor , Cell Line , Monocarboxylic Acid Transporters/metabolism
10.
J Neurosci ; 44(22)2024 May 29.
Article in English | MEDLINE | ID: mdl-38548341

ABSTRACT

The neurovascular unit (NVU) includes multiple different cell types, including neurons, astrocytes, endothelial cells, and pericytes, which respond to insults on very different time or dose scales. We defined differential vulnerability among these cell types, using response to two different insults: oxygen-glucose deprivation (OGD) and thrombin-mediated cytotoxicity. We found that neurons are most vulnerable, followed by endothelial cells and astrocytes. After temporary focal cerebral ischemia in male rats, we found significantly more injured neurons, compared with astrocytes in the ischemic area, consistent with differential vulnerability in vivo. We sought to illustrate different and shared mechanisms across all cell types during response to insult. We found that gene expression profiles in response to OGD differed among the cell types, with a paucity of gene responses shared by all types. All cell types activated genes relating to autophagy, apoptosis, and necroptosis, but the specific genes differed. Astrocytes and endothelial cells also activated pathways connected to DNA repair and antiapoptosis. Taken together, the data support the concept of differential vulnerability in the NVU and suggest that different elements of the unit will evolve from salvageable to irretrievable on different time scales while residing in the same brain region and receiving the same (ischemic) blood flow. Future work will focus on the mechanisms of these differences. These data suggest future stroke therapy development should target different elements of the NVU differently.


Subject(s)
Astrocytes , Endothelial Cells , Neurons , Rats, Sprague-Dawley , Animals , Male , Rats , Astrocytes/metabolism , Astrocytes/pathology , Endothelial Cells/metabolism , Neurons/metabolism , Brain/metabolism , Brain/pathology , Glucose/deficiency , Glucose/metabolism , Brain Ischemia/pathology , Brain Ischemia/metabolism , Brain Ischemia/genetics , Pericytes/metabolism , Pericytes/pathology , Neurovascular Coupling/physiology
11.
J Neurochem ; 168(5): 899-909, 2024 May.
Article in English | MEDLINE | ID: mdl-38299375

ABSTRACT

Cofilactin rods (CARs), which are 1:1 aggregates of cofilin-1 and actin, lead to neurite loss in ischemic stroke and other disorders. The biochemical pathways driving CAR formation are well-established, but how these pathways are engaged under ischemic conditions is less clear. Brain ischemia produces both ATP depletion and glutamate excitotoxicity, both of which have been shown to drive CAR formation in other settings. Here, we show that CARs are formed in cultured neurons exposed to ischemia-like conditions: oxygen-glucose deprivation (OGD), glutamate, or oxidative stress. Of these conditions, only OGD produced significant ATP depletion, showing that ATP depletion is not required for CAR formation. Moreover, the OGD-induced CAR formation was blocked by the glutamate receptor antagonists MK-801 and kynurenic acid; the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors GSK2795039 and apocynin; as well as an ROS scavenger. The findings identify a biochemical pathway leading from OGD to CAR formation in which the glutamate release induced by energy failure leads to activation of neuronal glutamate receptors, which in turn activates NADPH oxidase to generate oxidative stress and CARs.


Subject(s)
Energy Metabolism , Glutamic Acid , Neurons , Animals , Cells, Cultured , Neurons/metabolism , Neurons/drug effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glutamic Acid/metabolism , Rats , Adenosine Triphosphate/metabolism , Glucose/metabolism , Glucose/deficiency , Actins/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , NADPH Oxidases/metabolism , Acetophenones/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Dizocilpine Maleate/pharmacology , Kynurenic Acid/pharmacology , Kynurenic Acid/metabolism , Rats, Sprague-Dawley
12.
Inflammation ; 47(3): 1028-1040, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38236385

ABSTRACT

Hepatic ischemia-reperfusion (I/R) injury is still a major risk factor and unsolved problem in hepatic surgery. Methyltransferase-like 3 (METTL3), an important m6A-modified methylase, regulates inflammation and cellular stress response. In this study, we demonstrated the special role of METTL3 and its underlying mechanism in hepatic I/R injury. In the mouse model of hepatic I/R and in the oxygen-glucose deprivation and reoxygenation (OGD/R)-induced AML12 and NCTC 1469 cells, the expression of METTL3 was significantly upregulated. Inhibition of METTL3 in OGD/R-induced AML12 and NCTC 1469 cells both increased the cell viability, declined the cell apoptosis, and decreased the reactive oxygen species (ROS) and the release levels of interleukin-1ß (IL-1ß) and interleukin-18 (IL-18), diminishing NLRP3 and Caspase1-p20 expressions. Moreover, METTL3 positively modulated TXNIP expression in an m6A manner. TXNIP overexpression reversed the effects of METTL3 knockdown on OGD/R-induced injury in AML12 cells. Furthermore, inhibition of NLRP3 inflammasome activity contributed to the protective effects of TXNIP knockdown in OGD/R-induced AML12 cells. In conclusion, METTL3 knockdown alleviated OGD/R-induced hepatocyte injury, and the specific mechanism was associated with the inhibition of NLRP3 inflammasome activation, which was attributed to the reduction of TXNIP in an m6A-dependent manner.


Subject(s)
Glucose , Inflammasomes , Methyltransferases , NLR Family, Pyrin Domain-Containing 3 Protein , Oxygen , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Mice , Methyltransferases/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Glucose/metabolism , Glucose/deficiency , Oxygen/metabolism , Hepatocytes/metabolism , Carrier Proteins/metabolism , Cell Line , Mice, Inbred C57BL , Male , Thioredoxins/metabolism , Thioredoxins/genetics , Liver/metabolism , Liver/pathology , Reactive Oxygen Species/metabolism
13.
Mol Biotechnol ; 66(5): 1082-1094, 2024 May.
Article in English | MEDLINE | ID: mdl-38151617

ABSTRACT

Cerebral ischemia is a severe neurological disability related to neuronal apoptosis and cellular stress response. Circular RNAs (circRNAs) are emerging regulators of cerebral ischemia. Herein, this study proposed to probe the action of circ_0000115 in cerebral ischemia injury. The mouse neuroblastoma cells N2a and HT22 underwent oxygen-glucose deprivation (OGD) were used as a model of in vitro cerebral ischemia. Levels of genes and proteins were detected by qRT-PCR and western blotting. Cell proliferation and apoptosis were determined by EdU assay and flow cytometry. Western blotting was used to detect the protein level of pro-inflammatory factors. The oxidative stress injury was evaluated by detecting reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD) generation. Dual-luciferase reporter and RIP assays were used to confirm the target relationship between miR-1224-5p and circ_0000115 or nitric oxide synthase 3 (NOS3). OGD exposure decreased circ_0000115 and NOS3 expression, and increased miR-1224-5p in N2a and HT22 cells in a time-dependent manner. Circ_0000115 silencing attenuated OGD-induced apoptosis, oxidative stress and inflammation in N2a and HT22 cells. Mechanistically, circ_0000115 directly sponged miR-1224-5p, which targeted NOS3. Furthermore, rescue experiments showed that miR-1224-5p overexpression abolished the neuroprotective effect of circ_0000115 in N2a and HT22 cells under OGD treatment. Besides that, silencing of miR-1224-5p protected N2a and HT22 cells against OGD-evoked injury, which was counteracted by NOS3 knockdown. Circ_0000115 protects N2a and HT22 cells against OGD-evoked neuronal apoptosis, inflammation, and oxidative stress via the miR-1224-5p/NOS3 axis, providing an exciting view of the pathogenesis of cerebral ischemia.


Subject(s)
Apoptosis , Brain Ischemia , Inflammation , MicroRNAs , Neurons , Oxidative Stress , RNA, Circular , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , RNA, Circular/genetics , RNA, Circular/metabolism , Apoptosis/genetics , Mice , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Neurons/metabolism , Neurons/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Cell Line, Tumor , Glucose/metabolism , Glucose/deficiency , Reactive Oxygen Species/metabolism , Nitric Oxide Synthase Type III
14.
Int J Mol Sci ; 24(15)2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37569345

ABSTRACT

In previous work, we showed that cancer cells do not depend on glycolysis for ATP production, but they do on fatty acid oxidation. However, we found some cancer cells induced cell death after glucose deprivation along with a decrease of ATP production. We investigated the different response of glucose deprivation with two types of cancer cells including glucose insensitive cancer cells (GIC) which do not change ATP levels, and glucose sensitive cancer cells (GSC) which decrease ATP production in 24 h. Glucose deprivation-induced cell death in GSC by more than twofold after 12 h and by up to tenfold after 24 h accompanied by decreased ATP production to compare to the control (cultured in glucose). Glucose deprivation decreased the levels of metabolic intermediates of the pentose phosphate pathway (PPP) and the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) in both GSC and GIC. However, glucose deprivation increased reactive oxygen species (ROS) only in GSC, suggesting that GIC have a higher tolerance for decreased NADPH than GSC. The twofold higher ratio of reduced/oxidized glutathione (GSH/GSSG) in GIS than in GSC correlates closely with the twofold lower ROS levels under glucose starvation conditions. Treatment with N-acetylcysteine (NAC) as a precursor to the biologic antioxidant glutathione restored ATP production by 70% and reversed cell death caused by glucose deprivation in GSC. The present findings suggest that glucose deprivation-induced cancer cell death is not caused by decreased ATP levels, but rather triggered by a failure of ROS regulation by the antioxidant system. Conclusion is clear that glucose deprivation-induced cell death is independent from ATP depletion-induced cell death.


Subject(s)
Adenosine Triphosphate , Glucose , Neoplasms , Reactive Oxygen Species , Glucose/deficiency , Adenosine Triphosphate/metabolism , Pentose Phosphate Pathway , Reactive Oxygen Species/metabolism , NADP/metabolism , Glutathione/metabolism , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , PC-3 Cells , Humans , Neoplasms/metabolism , Neoplasms/pathology , Cell Death
15.
Nature ; 618(7963): 151-158, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37198494

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a lethal disease notoriously resistant to therapy1,2. This is mediated in part by a complex tumour microenvironment3, low vascularity4, and metabolic aberrations5,6. Although altered metabolism drives tumour progression, the spectrum of metabolites used as nutrients by PDA remains largely unknown. Here we identified uridine as a fuel for PDA in glucose-deprived conditions by assessing how more than 175 metabolites impacted metabolic activity in 21 pancreatic cell lines under nutrient restriction. Uridine utilization strongly correlated with the expression of uridine phosphorylase 1 (UPP1), which we demonstrate liberates uridine-derived ribose to fuel central carbon metabolism and thereby support redox balance, survival and proliferation in glucose-restricted PDA cells. In PDA, UPP1 is regulated by KRAS-MAPK signalling and is augmented by nutrient restriction. Consistently, tumours expressed high UPP1 compared with non-tumoural tissues, and UPP1 expression correlated with poor survival in cohorts of patients with PDA. Uridine is available in the tumour microenvironment, and we demonstrated that uridine-derived ribose is actively catabolized in tumours. Finally, UPP1 deletion restricted the ability of PDA cells to use uridine and blunted tumour growth in immunocompetent mouse models. Our data identify uridine utilization as an important compensatory metabolic process in nutrient-deprived PDA cells, suggesting a novel metabolic axis for PDA therapy.


Subject(s)
Glucose , Pancreatic Neoplasms , Ribose , Tumor Microenvironment , Uridine , Animals , Mice , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Ribose/metabolism , Uridine/chemistry , Glucose/deficiency , Cell Division , Cell Line, Tumor , MAP Kinase Signaling System , Uridine Phosphorylase/deficiency , Uridine Phosphorylase/genetics , Uridine Phosphorylase/metabolism , Humans
16.
Nat Commun ; 13(1): 572, 2022 01 31.
Article in English | MEDLINE | ID: mdl-35102143

ABSTRACT

Substrate inhibition of enzymes can be a major obstacle to the production of valuable chemicals in engineered microorganisms. Here, we show substrate inhibition of lycopene cyclase as the main limitation in carotenoid biosynthesis in Yarrowia lipolytica. To overcome this bottleneck, we exploit two independent approaches. Structure-guided protein engineering yields a variant, Y27R, characterized by complete loss of substrate inhibition without reduction of enzymatic activity. Alternatively, establishing a geranylgeranyl pyrophosphate synthase-mediated flux flow restrictor also prevents the onset of substrate inhibition by diverting metabolic flux away from the inhibitory metabolite while maintaining sufficient flux towards product formation. Both approaches result in high levels of near-exclusive ß-carotene production. Ultimately, we construct strains capable of producing 39.5 g/L ß-carotene at a productivity of 0.165 g/L/h in bioreactor fermentations (a 1441-fold improvement over the initial strain). Our findings provide effective approaches for removing substrate inhibition in engineering pathways for efficient synthesis of natural products.


Subject(s)
Lycopene/metabolism , Yarrowia/metabolism , Acetyl Coenzyme A/metabolism , Bioreactors , Carbon/metabolism , Cytosol/metabolism , Farnesyltranstransferase/metabolism , Fermentation , Glucose/deficiency , Intramolecular Lyases/metabolism , Lipid Metabolism , Lipids/biosynthesis , Lycopene/chemistry , Metabolic Flux Analysis , Protein Engineering , Substrate Specificity , Terpenes/metabolism
17.
Sci Rep ; 12(1): 1710, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35110605

ABSTRACT

The study is aimed at elucidating the effect of selenium nanoparticles (SeNPs) on the death of cells in the primary culture of mouse cerebral cortex during oxygen and glucose deprivation (OGD). A primary cell culture of the cerebral cortex containing neurons and astrocytes was subjected to OGD and reoxygenation to simulate cerebral ischemia-like conditions in vitro. To evaluate the neuroprotective effect of SeNPs, cortical astrocytes and neurons were incubated for 24 h with SeNPs, and then subjected to 2-h OGD, followed by 24-h reoxygenation. Vitality tests, fluorescence microscopy, and real-time PCR have shown that incubation of primary cultured neurons and astrocytes with SeNPs at concentrations of 2.5-10 µg/ml under physiological conditions has its own characteristics depending on the type of cells (astrocytes or neurons) and leads to a dose-dependent increase in apoptosis. At low concentration SeNPs (0.5 µg/ml), on the contrary, almost completely suppressed the processes of basic necrosis and apoptosis. Both high (5 µg/ml) and low (0.5 µg/ml) concentrations of SeNPs, added for 24 h to the cells of cerebral cortex, led to an increase in the expression level of genes Bcl-2, Bcl-xL, Socs3, while the expression of Bax was suppressed. Incubation of the cells with 0.5 µg/ml SeNPs led to a decrease in the expression of SelK and SelT. On the contrary, 5 µg/ml SeNPs caused an increase in the expression of SelK, SelN, SelT, SelP. In the ischemic model, after OGD/R, there was a significant death of brain cells by the type of necrosis and apoptosis. OGD/R also led to an increase in mRNA expression of the Bax, SelK, SelN, and SelT genes and suppression of the Bcl-2, Bcl-xL, Socs3, SelP genes. Pre-incubation of cell cultures with 0.5 and 2.5 µg/ml SeNPs led to almost complete inhibition of OGD/R-induced necrosis and greatly reduced apoptosis. Simultaneously with these processes we observed suppression of caspase-3 activation. We hypothesize that the mechanisms of the protective action of SeNPs involve the activation of signaling cascades recruiting nuclear factors Nrf2 and SOCS3/STAT3, as well as the activation of adaptive pathways of ESR signaling of stress arising during OGD and involving selenoproteins SelK and SelT, proteins of the Bcl-2 family ultimately leading to inactivation of caspase-3 and inhibition of apoptosis. Thus, our results demonstrate that SeNPs can act as neuroprotective agents in the treatment of ischemic brain injuries.


Subject(s)
Apoptosis/drug effects , Astrocytes/drug effects , Brain Ischemia/drug therapy , Cerebral Cortex/drug effects , Glucose/deficiency , Nanoparticles , Neurons/drug effects , Neuroprotective Agents/pharmacology , Selenium Compounds/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Hypoxia , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Coculture Techniques , Female , Male , Mice , Necrosis , Neurons/metabolism , Neurons/pathology , Primary Cell Culture , Selenoproteins/genetics , Selenoproteins/metabolism
18.
Int Immunopharmacol ; 104: 108464, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35021128

ABSTRACT

BACKGROUND: Emerging evidence has shown that circular RNAs (circRNAs) are involved in the pathogenesis of ischemic stroke (IS). Nonetheless, the function of circ_0000647 was not reported. METHODS: Oxygen-glucose deprivation and reperfusion (OGD/R)-treated SK-N-SH cells were used to mimic cerebral ischemia/reperfusion (I/R) conditions. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot were used to measure the levels of circ_0000647, microRNA-126-5p (miR-126-5p) and TNF receptor associated factor 3 (TRAF3). Cell Counting Kit-8 (CCK-8) assay, 5'-ethynyl-2'-deoxyuridine (EDU) assay and flow cytometry analysis were employed to assess cell proliferation and apoptosis. Enzyme-linked immunosorbent assay (ELISA) was conducted for the concentrations of IL-6 and TNF-α. Oxidative stress was assessed by determining malondialdehyde (MDA) level and superoxide dismutase (SOD) activity. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were adopted to estimate the relationships of circ_0000647, miR-126-5p and TRAF3. The morphology and size of exosomes were observed via transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) analysis. RESULTS: Circ_0000647 was elevated in OGD/R-treated SK-N-SH cells. OGD/R treatment suppressed the proliferation and promoted the apoptosis, inflammation and oxidative stress in SK-N-SH cells, while circ_0000647 knockdown reversed the effects. Circ_0000647 could sponge miR-126-5p, which directly targeted TRAF3. MiR-126-5p overexpression alleviated OGD/R-induced SK-N-SH cell damage and miR-126-5p inhibition reversed the effect of circ_0000647 knockdown on OGD/R-induced SK-N-SH cell damage. Moreover, TRAF3 elevation abated miR-126-5p-mediated effect on SK-N-SH cell injury. In addition, exosomal circ_0000647 level was increased in OGD/R-stimulated SK-N-SH cells. CONCLUSION: Circ_0000647 interference relieved OGD/R-induced SK-N-SH cell damage by altering miR-126-5p/TRAF3 axis.


Subject(s)
MicroRNAs , RNA, Circular , Reperfusion Injury/genetics , TNF Receptor-Associated Factor 3/genetics , Cell Hypoxia/genetics , Cell Line, Tumor , Glucose/deficiency , Humans , Interleukin-6/metabolism , Models, Biological , Oxygen , Reperfusion Injury/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Int Immunopharmacol ; 104: 108443, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35021129

ABSTRACT

AIM: Hepatic ischemia/reperfusion (I/R) injury is a significant pathological process that contributes to high morbidity and mortality rates, although the underlying mechanism is unknown. Recent studies have shown that transient receptor potential melastatin 2 (TRPM2) plays a critical role in organ I/R injury, but the exact mechanism is elusive. This study investigates the role and mechanism of TPRM2 in hepatic I/R injury and oxygen-glucosedeprivation/reoxygenation (OGD/R) induced hepatocyte injury. METHODS: We evaluated the effects of TRPM2 on hepatic I/R injury using a knockout mouse model of hepatic I/R. In a model of OGD/R in hepatocytes, we investigated the mechanism of TPRM2 in it using the autophagy agonist and inhibitor and an NLRP3 inhibitor. RESULTS: We discovered that knockout of TRPM2 protected against hepatic I/R accompanied by autophagy activation and NLRP3 inflammasome pathway inhibition. Furthermore, increasing autophagy attenuated OGD/R-induced cell injury and knockdown of TRPM2 alleviated the injury by activating autophagy. Additionally, we detected the expression of NLRP3 inflammasome pathway in the OGD/R-induced hepatocytes which had been treated with the autophagy agonist and inhibitor, and found that autophagy negatively regulated the NLRP3 inflammasome pathway. Moreover, we discovered that the administration of NLRP3-inhibitor INF39 increased cell viability and caused a decline in cell death in the OGD/R-treated hepatocytes. CONCLUSIONS: Downregulation of TRPM2 protected the liver against I/R injury and OGD/R induced injury, mediated by autophagy activation and inhibition of the NLRP3 inflammasome pathway, whereas autophagy negatively regulated the NLRP3 inflammasome pathway in this process.


Subject(s)
Liver Diseases/genetics , Reperfusion Injury/genetics , TRPM Cation Channels/genetics , Animals , Autophagy , Cell Hypoxia/genetics , Cell Line , Down-Regulation , Glucose/deficiency , Hepatocytes , Humans , Inflammasomes/metabolism , Liver Diseases/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/metabolism , Signal Transduction
20.
Sci Rep ; 12(1): 1230, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075260

ABSTRACT

Human gingival fibroblasts (HGnFs) maintain periodontal tissue homeostasis through active proliferation and migration. Clinically, it is considered that the wound-healing ability of the gingival tissue is maintained even in environments with insufficient supply of nutrients, such as glucose, immediately after periodontal surgery. However, the effects of such glucose-deficient environments on HGnFs remain unclear. This study aimed to investigate the effects of low-glucose environment on HGnFs homeostasis. We evaluated gingival wound healing by examining cell proliferation and migration and collagen synthesis in HGnFs cultured in 100, 50, 25, and 0 mg/dL glucose in vitro. The cellular stress levels were determined by measuring the lactate dehydrogenase (LDH) and reactive oxygen species (ROS) levels. The glucose metabolism of HGnFs in the low-glucose concentrations was studied by measuring glucose transporter type 1 (GLUT1) mRNA expression, glucose uptake assays, lactate and ATP productions. Molecular effects were examined with a focus on the LKB1-AMPK signaling pathway. Autophagy activity in glucose-deprived HGnFs was evaluated by measuring the levels of autophagy-related proteins. Low glucose levels increased cellular stress levels, autophagy activity, and enhanced glucose metabolism through the LKB1-AMPK signaling pathway, providing more ATPs to promote wound healing. Our results regarding glucose transfer suggest the rapid healing of gingival wounds.


Subject(s)
Autophagy , Fibroblasts/physiology , Gingiva/physiology , Glucose/deficiency , Wound Healing , AMP-Activated Protein Kinase Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Cells, Cultured , Glycolysis , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...