Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.034
Filter
1.
Eur Rev Med Pharmacol Sci ; 28(18): 4277-4289, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39359199

ABSTRACT

OBJECTIVE: Ferroptosis of neurons is a significant cause of brain injury following intracerebral hemorrhage (ICH). As an iron-containing compound in hemoglobin, heme contributes to nerve injury post-ICH. Melatonin has been shown to mitigate the effects of ICH, yet its specific functions remain largely elusive. In this study, we aimed to explore the roles and mechanisms of melatonin in heme-induced ferroptosis subsequent to ICH. MATERIALS AND METHODS: C57BL/6 mice were intracranially injected with heme and then treated with melatonin. Behavior tests [modified neurological severity score (mNSS), forelimb placing, and corner turn tests], H&E staining, Nissl staining, and Prussian blue staining were used to evaluate mouse brain tissue injury. In vitro, HT-22 cells were stimulated with heme and cell viability was determined by crystal violet staining. The iron contents were determined in heme-treated brains and cells, and the levels of 4-hydroxynonenal (4-HNE) and malonaldehyde (MDA) were assessed by ELISA. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to investigate the mRNA levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Immunoblotting was used to analyze the protein expression of glutathione peroxidase 4 (GPX4), solute carrier family 7 member 11 (SLC7A11), Nrf2, and HO-1. Finally, small interfering RNA (siRNA) was used to knock down Nrf2 in HT-22 cells. RESULTS: Melatonin treatment alleviated heme-induced injuries to neural function, as indicated by improved behavior in the mice. Moreover, melatonin decreased cell death and iron concentrations, increased MDA and 4-HNE levels, and reversed the decreases in GPX4, SLC7A11, Nrf2, and HO-1 induced by heme in vitro and in vivo. These results indicated that melatonin could improve the ferroptosis induced by heme. In addition, we found that Nrf2 knockdown attenuated the therapeutic effect of melatonin on neuronal ferroptosis induced by heme. CONCLUSIONS: In general, melatonin alleviates heme-induced ferroptosis by activating the Nrf2/HO-1 pathway, which implies that melatonin is a promising treatment for ferroptosis in ICH.


Subject(s)
Ferroptosis , Heme Oxygenase-1 , Heme , Melatonin , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Neurons , Animals , Ferroptosis/drug effects , Melatonin/pharmacology , NF-E2-Related Factor 2/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Heme Oxygenase-1/metabolism , Heme/metabolism , Male , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Signal Transduction/drug effects , Disease Models, Animal , Membrane Proteins
2.
Acta Cir Bras ; 39: e396124, 2024.
Article in English | MEDLINE | ID: mdl-39356932

ABSTRACT

PURPOSE: To examine whether isoflurane preconditioning (IsoP) has a protective effect against renal ischemia/reperfusion injury (I/RI) in diabetic conditions and to further clarify the underlying mechanisms. METHODS: Control and streptozotocin-induced diabetic rats were randomly assigned to five groups, as follows: normal sham, normal I/R, diabetic sham, diabetic I/R, and diabetic I/R + isoflurane. Renal I/RI was induced by clamping renal pedicle for 45 min followed by reperfusion for 24 h. IsoP was achieved by exposing the rats to 2% isoflurane for 30 min before vascular occlusion. Kidneys and blood were collected after reperfusion for further analysis. Renal histology, blood urea nitrogen, serum creatinine, oxidative stress, inflammatory cytokines, and renal cell apoptosis were assessed. Furthermore, the expression of brahma related gene 1 (Brg1), nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and nuclear factor-κB (NF-κB) were determined. RESULTS: Compared with control, diabetic rats undergoing I/R presented more severe renal injury, oxidative stress, inflammatory reaction, and apoptosis with the impairment of Brg1/Nrf2/HO-1 signaling. All these alterations were significantly attenuated by pretreatment with isoflurane. CONCLUSIONS: These findings suggest that isoflurane could alleviate renal I/RI in diabetes, possibly through improving Brg1/Nrf2/HO-1 signaling.


Subject(s)
Apoptosis , Diabetes Mellitus, Experimental , Ischemic Preconditioning , Isoflurane , NF-E2-Related Factor 2 , Oxidative Stress , Random Allocation , Reperfusion Injury , Signal Transduction , Transcription Factors , Animals , Isoflurane/pharmacology , Reperfusion Injury/prevention & control , Diabetes Mellitus, Experimental/complications , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Male , Ischemic Preconditioning/methods , Oxidative Stress/drug effects , Apoptosis/drug effects , DNA Helicases/metabolism , Kidney/drug effects , Kidney/blood supply , Kidney/pathology , Nuclear Proteins/metabolism , Heme Oxygenase-1/metabolism , Anesthetics, Inhalation/pharmacology , Rats , Rats, Sprague-Dawley , NF-kappa B/metabolism
3.
J Immunol Res ; 2024: 8273732, 2024.
Article in English | MEDLINE | ID: mdl-39359694

ABSTRACT

Oxidative stress is crucial in ulcerative colitis (UC) and colitis-associated colorectal cancer (CAC). Intestinal epithelial cells (IECs) are an important component of the intestinal barrier. In previous studies, we have demonstrated that suppressing microRNA-222-3p (miR-222-3p) can protect against oxidative stress in IECs, which ameliorates colonic injuries in UC mice and prevents the conversion of UC to CAC. In this case, we hope to explore whether moxibustion can alleviate UC and CAC by inhibiting miR-222-3p based on mouse models of UC and CAC. After herb-partitioned moxibustion (HPM) intervention, the disease activity index (DAI) and colon macroscopic damage index (CMDI) were significantly reduced in UC mice, and the number and volume of intestinal tumors were decreased considerably in CAC mice. Meanwhile, we found that HPM suppressed miR-222-3p expression and upregulated the mRNA and protein expression of Brahma-related gene 1 (BRG1), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), while inhibiting Kelch-like ECH-associated protein 1 (Keap1) expression in IECs of UC and CAC mice. With changes in reactive oxygen species (ROS), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), and inflammatory cytokines interleukin (IL)-1ß and tumor necrosis factor (TNF)-α), we verified that HPM protects against oxidative stress and inflammation in IECs of UC and CAC mice. The effect of HPM was inhibited in miR-222-3p overexpression mice, further demonstrating that the protective effect of HPM on UC and CAC mice was through inhibiting miR-222-3p. In summary, HPM regulates the BRG1/Nrf2/HO-1 pathway by inhibiting miR-222-3p to attenuate oxidative stress in IECs in UC and CAC.


Subject(s)
Colitis, Ulcerative , Disease Models, Animal , Heme Oxygenase-1 , MicroRNAs , Moxibustion , NF-E2-Related Factor 2 , Oxidative Stress , Signal Transduction , Transcription Factors , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Colitis, Ulcerative/therapy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/genetics , Mice , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , DNA Helicases/metabolism , DNA Helicases/genetics , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Humans
4.
Biomed Res ; 45(5): 197-207, 2024.
Article in English | MEDLINE | ID: mdl-39370298

ABSTRACT

Our study explored the therapeutic effect and the mechanism of quercetin against hypoxia/reoxygenation (H/R)-induced injury in human coronary artery endothelial cells (CAECs). Quercetin was selected as a potential component for the BuShenKangShuaiPian formula (BSKSP) treatment via the Network pharmacology analysis. Cell viability and reactive oxygen species (ROS) production were measured by CCK8 assay and immunofluorescence, respectively. The expression of Bax, Bcl-2, Cle-caspase-3, cytochrome c (Cyt-C), NF-E2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) protein was quantified by western blotting. The superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA) activity, mtDNA copy number, and ATP production were measured via corresponding kits. Quercetin was selected from the BSKSP for its high degree value (Degree value: 22). Besides, quercetin protected CAECs against H/R-induced cytotoxicity and apoptosis. The H/R-induced increased ROS level, ATP production, Cyt-C release, and decreased mtDNA copy number were removed by the quercetin. Moreover, quercetin upregulated the Nrf2/ HO-1 axis, SOD, and CAT activity, and downregulated MDA levels in H/R treated CAECs, while knockdown Nrf2 reversed the protection of quercetin against H/R-induced oxidative stress, mitochondrial damage, and apoptosis. Quercetin protects CAECs against H/R-induced mitochondrial apoptosis via the Nrf2/HO-1 axis, which innovatively suggests the therapeutic potential of quercetin for coronary heart disease (CHD) treatment.


Subject(s)
Apoptosis , Coronary Vessels , Endothelial Cells , Heme Oxygenase-1 , Mitochondria , NF-E2-Related Factor 2 , Quercetin , Reactive Oxygen Species , Signal Transduction , Humans , Quercetin/pharmacology , Apoptosis/drug effects , NF-E2-Related Factor 2/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Reactive Oxygen Species/metabolism , Coronary Vessels/cytology , Coronary Vessels/metabolism , Coronary Vessels/drug effects , Signal Transduction/drug effects , Cell Survival/drug effects , Oxidative Stress/drug effects , Cell Hypoxia/drug effects
5.
J Med Virol ; 96(10): e29945, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39370874

ABSTRACT

Borna disease virus 1 (BoDV-1) is a neurotropic RNA virus that has been linked to fatal BoDV-1 encephalitis (BVE) in humans. Ferroptosis represents a newly recognized kind of programmed cell death that marked by iron overload and lipid peroxidation. Various viral infections are closely related to ferroptosis. However, the link between BoDV-1 infection and ferroptosis, as well as its role in BVE pathogenesis, remains inadequately understood. Herein, we used primary rat cortical neurons, human microglial HMC3 cells, and Sprague‒Dawley rats as models. BoDV-1 infection induced ferroptosis, as ferroptosis characteristics were detected (iron overload, reactive oxygen species buildup, decreased antioxidant capacity, lipid peroxidation, and mitochondrial damage). Analysis via qRT-PCR and Western blot demonstrated that BoDV-1-induced ferroptosis was mediated through Nrf2/HO-1/SLC7a11/GPX4 antioxidant pathway suppression. Nrf2 downregulation was due to BoDV-1 infection promoting Nrf2 ubiquitination and degradation. Following BoDV-1-induced ferroptosis, the PTGS2/PGE2 signaling pathway was activated, and various intracellular lipid peroxidation products and damage-associated molecular patterns were released, contributing to BVE occurrence and progression. More importantly, inhibiting ferroptosis or the ubiquitin‒proteasome system effectively alleviated BVE. Collectively, these findings demonstrate the interaction between BoDV-1 infection and ferroptosis and reveal BoDV-1-induced ferroptosis as an underlying pathogenic mechanism of BVE.


Subject(s)
Borna Disease , Borna disease virus , Ferroptosis , Lipid Peroxidation , NF-E2-Related Factor 2 , Neurons , Rats, Sprague-Dawley , Borna disease virus/physiology , Animals , Rats , Humans , Neurons/virology , Neurons/pathology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Borna Disease/virology , Borna Disease/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Microglia/virology , Microglia/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Cell Line , Encephalitis/virology , Encephalitis/pathology , Cells, Cultured
6.
Front Immunol ; 15: 1457010, 2024.
Article in English | MEDLINE | ID: mdl-39380993

ABSTRACT

Nrf2 is a master transcriptional regulator of a number of genes involved in the adaptive response to oxidative stress. Among the genes upregulated by Nrf2, heme oxygenase-1 (HO-1) has received significant attention, given that the products of HO-1-induced heme catabolism have well established antioxidant and anti-inflammatory properties. This is evidenced in numerous models of inflammatory and autoimmune disease whereby induction of HO-1 expression or administration of tolerable amounts of HO-1 reaction products can ameliorate disease symptoms. Unsurprisingly, Nrf2 and HO-1 are now considered viable drug targets for a number of conditions. In recent years, the term 'inflammaging' has been used to describe the low-grade chronic inflammation observed in aging/aged cells. Increased oxidative stress is also a key factor associated with aging and there is convincing evidence that Nrf2, not only declines with age, but that Nrf2 and HO-1 can reduce cellular senescence and the senescence-associated secretory phenotype (SASP) which is now considered an underlying driver of age-related inflammatory disease. In this review, we describe the role of oxidative stress in 'inflammaging' and highlight the potential anti-aging properties of the Nrf2-HO-1 system. We also highlight established and newly emerging Nrf2 activators and their therapeutic application in age-related disease.


Subject(s)
Aging , Heme Oxygenase-1 , Inflammation , NF-E2-Related Factor 2 , Oxidative Stress , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Humans , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Inflammation/metabolism , Inflammation/immunology , Animals , Aging/immunology , Cellular Senescence , Signal Transduction
7.
Neurotherapeutics ; 21(5): e00444, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39353831

ABSTRACT

This study aims to investigate whether butylphthalide can inhibit ferroptosis and ameliorate cerebral ischaemia-reperfusion (I/R) injury in rats by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) / heme oxygenase-1 (HO-1) signalling pathway, known for its antioxidative and cytoprotective properties. Middle cerebral artery occlusion reperfusion (MCAO/R) rat models were established. Male rats were randomly divided into five groups: a sham-operated group (sham), MCAO/R group, MCAO/R â€‹+ â€‹ML385 (Nrf2-specific inhibitor) group, MCAO/R â€‹+ â€‹NBP (butylphthalide) group and MCAO/R â€‹+ â€‹ML385 â€‹+ â€‹NBP group. The effect of butylphthalide on cerebral I/R injury was evaluated using neurological deficit scores. The expression levels of Nrf2, HO-1, glutathione peroxidase 4 (GPX4), acyl-CoA synthetase long-chain family member 4 (ACSL4) and transferrin receptor 1 (TfR1) protein were detected using Western blot. Moreover, the expression levels of GPX4, HO-1 and TfR1 mRNA were determined through real-time fluorescence quantitative reverse transcription polymerase chain reaction. The distribution of Nrf2, HO-1, GPX4 and TfR1 was detected using immunohistochemical staining. The levels of iron and related lipid peroxidation indexes, such as reduced glutathione, reactive oxygen species, malondialdehyde and nitric oxide, were measured using a kit. The changes in mitochondria were observed through transmission electron microscopy. Butylphthalide treatment significantly improved neurological dysfunction, reduced cerebral infarction volume and mitigated histopathological damage in MCAO/R rats. It induced the nuclear translocation of Nrf2 and upregulated HO-1 expression, which was attenuated by ML385. Butylphthalide also attenuated lipid peroxidation, iron accumulation and mitochondrial damage induced by MCAO/R. The expression of GPX4, ACSL4 and TfR1 proteins, as well as their mRNA levels, was modulated through butylphthalide treatment, with improvements observed in mitochondrial morphology. Butylphthalide exerts neuroprotective effects by attenuating neurological dysfunction and ferroptosis in MCAO/R rats through the activation of the Nrf2/HO-1 pathway and inhibition of lipid peroxidation and iron accumulation.


Subject(s)
Benzofurans , Ferroptosis , NF-E2-Related Factor 2 , Rats, Sprague-Dawley , Reperfusion Injury , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Male , Benzofurans/pharmacology , Benzofurans/therapeutic use , Ferroptosis/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Rats , Signal Transduction/drug effects , Heme Oxygenase-1/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Heme Oxygenase (Decyclizing)/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/metabolism
8.
Sci Rep ; 14(1): 23527, 2024 10 09.
Article in English | MEDLINE | ID: mdl-39384968

ABSTRACT

This study investigated the effects of heat shock protein 22 (HSP22) against doxorubicin (DOX)-induced kidney injury. Mice were randomly assigned to four groups: CON, ad-HSP22, DOX, and ad-HSP22 + DOX. Adeno-associated virus carrying the HSP22 gene (ad-HSP22) was administered via tail vein injection for four weeks, followed by intraperitoneal simulation with DOX (20 mg/kg) for another five days. Upon euthanasia, ELISA, histological staining (H&E, IHC, DHE, and TUNEL), and western blot analyses were employed to assess relevant markers. Serum biomarkers of kidney injury, SCr, and BUN, were upregulated after DOX administration but normalized with HSP22 overexpression. Pathological changes induced by DOX were also reversed by HSP22 overexpression in H&E, IHC, DHE, and TUNEL stains. DOX-induced upregulation of NOX-2 and NOX-4 and downregulation of SOD-1 and SOD-2 were reversed by HSP22 overexpression. Similarly, DOX-induced increases in Bax and decrease in Bcl-2 were attenuated by HSP22 overexpression. The study further demonstrated that the Nrf2/HO-1 signaling pathway was activated by HSP22 overexpression. In vitro experiments corroborated the findings from in vivo experiments. In conclusion, HSP22 alleviates DOX-induced kidney injury by suppressing oxidative stress and apoptosis, primarily through the activation of the Nrf2/HO-1 signaling pathway. These results suggest HSP22 as a potential therapeutic target for DOX-induced kidney injury.


Subject(s)
Apoptosis , Doxorubicin , Heat-Shock Proteins , Oxidative Stress , Animals , Doxorubicin/adverse effects , Oxidative Stress/drug effects , Apoptosis/drug effects , Mice , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Male , Kidney/metabolism , Kidney/drug effects , Kidney/pathology , Molecular Chaperones/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Signal Transduction/drug effects , Superoxide Dismutase/metabolism
9.
Carbohydr Polym ; 345: 122597, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39227128

ABSTRACT

Fucoxanthin (Fx) has garnered significant interest due to its exceptional biological properties. However, its efficacy in enhancing food quality and human health is contingent upon the solubility of the compound in water and its physicochemical stability. Therefore, nanocarriers must be developed to enhance the stability and biocompatibility of Fx. In this study, oxidized paramylon and Fx self-assembled nanoparticles (Fx-OEP) were prepared via the anti-solvent method, with a loading rate of 82.47 % for Fx. The Fx-OEP exhibited robust storage and photostability. In vitro simulated digestion assays demonstrated that Fx-OEP effectively protected Fx from premature gastric release, while achieving a release efficiency of 72.17 % in the intestinal phase. Fx-OEP has the capacity to scavenge a range of reactive oxygen species (ROS) induced by cellular oxidative stress. Treatment with Fx-OEP resulted in a significant reduction in ROS accumulation in insulin-resistant HepG2 cells, which was attributed to the activation of the nuclear factor E2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) pathway. This, in turn, activated insulin receptor substrate 1/glucose transporter type 4 (IRS1/GLUT4), promoting cellular glucose absorption and utilization. These findings indicate the potential of self-assembled nanoparticles based on oxidized paramylon as a new type of nanocarrier for delivering hydrophobic substances.


Subject(s)
Insulin Resistance , Nanoparticles , Xanthophylls , Humans , Xanthophylls/pharmacology , Xanthophylls/chemistry , Nanoparticles/chemistry , Hep G2 Cells , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Drug Carriers/chemistry , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Heme Oxygenase-1/metabolism , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins/metabolism , Drug Liberation , Glucans/chemistry , Glucans/pharmacology
10.
Mol Biol Rep ; 51(1): 976, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259343

ABSTRACT

OBJECTIVE: Bidirectional influences between senescence and inflammation are newly discovered. This study aimed to clarify the roles and mechanism of Porphyromonas gingivalis (P. gingivalis) in exacerbating senescence in human gingival fibroblasts (HGFs). DESIGN: Subgingival plaque and gingivae were collected from twenty-four periodontitis patients and eighteen periodontally healthy subjects. Quantities of P. gingivalis in subgingival plaque were explored using real-time PCR and the expressions of p53, p21 and SIRT6 in gingivae were detected by IHC. Moreover, senescence in HGFs was induced by P. gingivalis lipopolysaccharide (LPS) and the expressions of senescence-related ß-galactosidase (SA-ß-gal), p53, p21 and senescence-associated secretory phenotype (IL-6 and IL-8) with or without treatment by SIRT6 activator UBCS039 were explored by IHC, western blot and ELISA, respectively. In addition, the levels of SIRT6, Nrf2, HO-1 and reactive oxygen species (ROS) were examined by western blot and flow cytometry. RESULTS: Quantities of P. gingivalis in subgingival plaque and semi-quantitative scores of p53 and p21 in gingivae of periodontitis patients were increased compared with healthy controls (p < 0.05), while SIRT6 score in periodontitis patients was decreased (p < 0.001). Quantities of P. gingivalis were positively correlated with p53 and p21 scores (0.6 < r < 0.9, p < 0.01), and negatively correlated with SIRT6 score (-0.9 < r<-0.6, p < 0.01). Moreover, P. gingivalis LPS increased the levels of SA-ß-gal, p53, p21, IL-6, IL-8 and ROS and decreased the levels of SIRT6, Nrf2 and HO-1 in HGFs, which was rescued by UBCS039 (p < 0.05). CONCLUSIONS: P. gingivalis LPS could induce senescence of HGFs, which could be reversed by SIRT6 via Nrf2-HO-1 signaling pathway.


Subject(s)
Cellular Senescence , Fibroblasts , Gingiva , NF-E2-Related Factor 2 , Porphyromonas gingivalis , Reactive Oxygen Species , Sirtuins , Humans , Porphyromonas gingivalis/pathogenicity , Gingiva/microbiology , Gingiva/metabolism , Fibroblasts/metabolism , Sirtuins/metabolism , Sirtuins/genetics , Male , Female , Adult , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species/metabolism , Lipopolysaccharides/pharmacology , Periodontitis/microbiology , Periodontitis/metabolism , Tumor Suppressor Protein p53/metabolism , Middle Aged , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Interleukin-6/metabolism , Interleukin-8/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics
11.
J Orthop Surg Res ; 19(1): 531, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218922

ABSTRACT

BACKGROUND: Bone loss caused by microgravity exposure presents a serious threat to the health of astronauts, but existing treatment strategies have specific restrictions. This research aimed to investigate whether salidroside (SAL) can mitigate microgravity-induced bone loss and its underlying mechanism. METHODS: In this research, we used hindlimb unloading (HLU) and the Rotary Cell Culture System (RCCS) to imitate microgravity in vivo and in vitro. RESULTS: The results showed that salidroside primarily enhances bone density, microstructure, and biomechanical properties by stimulating bone formation and suppressing bone resorption, thereby preserving bone mass in HLU rats. In MC3T3-E1 cells cultured under simulated microgravity in rotary wall vessel bioreactors, the expression of osteogenic genes significantly increased after salidroside administration, indicating that salidroside can promote osteoblast differentiation under microgravity conditions. Furthermore, the Nrf2 inhibitor ML385 diminished the therapeutic impact of salidroside on microgravity-induced bone loss. Overall, this research provides the first evidence that salidroside can mitigate bone loss induced by microgravity exposure through stimulating the Nrf2/HO-1 pathway. CONCLUSION: These findings indicate that salidroside has great potential for treating space-related bone loss in astronauts and suggest that Nrf2/HO-1 is a viable target for counteracting microgravity-induced bone damage.


Subject(s)
Glucosides , NF-E2-Related Factor 2 , Phenols , Weightlessness Simulation , Glucosides/pharmacology , Glucosides/therapeutic use , Animals , Phenols/pharmacology , Phenols/therapeutic use , NF-E2-Related Factor 2/metabolism , Mice , Weightlessness Simulation/adverse effects , Rats , Male , Heme Oxygenase-1/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Weightlessness/adverse effects , Osteogenesis/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Hindlimb Suspension , Bone Resorption/prevention & control , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Density/drug effects , Membrane Proteins
12.
Int J Nanomedicine ; 19: 8779-8796, 2024.
Article in English | MEDLINE | ID: mdl-39220192

ABSTRACT

Purpose: Atopic dermatitis (AD) is a chronic inflammatory skin condition that can affect individuals of all ages. Recent research has shown that oxidative stress plays a crucial role in the development of AD. Therefore, inhibiting oxidative stress may be an effective therapeutic approach for AD. Nano-molybdenum is a promising material for use as an antioxidant. We aimed to evaluate the therapeutic effects and preliminary mechanisms of molybdenum nanoparticles (Mo NPs) by using a murine model of chemically induced AD-like disease. Methods: HaCaT cells, a spontaneously immortalized human keratinocyte cell line, were stimulated by tumor necrosis factor-alpha /interferon-gamma after pre-treatment with Mo NPs. Reactive oxygen species levels, production of inflammatory factors, and activation of the nuclear factor kappa-B and the nuclear factor erythroid 2-related factor pathways were then evaluated. Mo NPs was topically applied to treat a murine model of AD-like disease induced by MC903, a vitamin D3 analog. Dermatitis scores, pruritus scores, transepidermal water loss and body weight were evaluated. AD-related inflammatory factors and chemokines were evaluated. Activation of the nuclear factor kappa-B and nuclear factor erythroid 2-related factor / heme oxygenase-1 pathways was assessed. Results: Our data showed that the topical application of Mo NPs dispersion could significantly alleviate AD skin lesions and itching and promote skin barrier repair. Further mechanistic experiments revealed that Mo NPs could inhibit the excessive activation of the nuclear factor kappa-B pathway, promote the expression of nuclear factor erythroid 2-related factor and heme oxygenase-1 proteins, and suppress oxidative stress reactions. Additionally, they inhibited the expression of thymic stromal lymphopoietin, inflammatory factors, and chemokines, thereby alleviating skin inflammation. Conclusion: Mo NPs present a promising alternative treatment option for patients with AD as they could address three pivotal mechanisms in the pathogenesis of AD concurrently.


Subject(s)
Dermatitis, Atopic , Heme Oxygenase-1 , Metal Nanoparticles , Molybdenum , NF-E2-Related Factor 2 , NF-kappa B , Reactive Oxygen Species , Signal Transduction , Animals , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/chemically induced , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Heme Oxygenase-1/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Molybdenum/pharmacology , Molybdenum/chemistry , Humans , Mice , Metal Nanoparticles/chemistry , Disease Models, Animal , Oxidative Stress/drug effects , HaCaT Cells , Antioxidants/pharmacology , Mice, Inbred BALB C , Nanoparticles/chemistry , Cell Line , Skin/drug effects , Skin/metabolism , Membrane Proteins
13.
CNS Neurosci Ther ; 30(9): e70030, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39233353

ABSTRACT

BACKGROUND: Ischemic stroke leads a primary cause of mortality in human diseases, with a high disability rate worldwide. This study aims to investigate the function of ß-1,4-galactosyltransferase 1 (B4galt1) in mouse brain ischemia/reperfusion (I/R) injury. METHODS: Recombinant human B4galt1 (rh-B4galt1) was intranasally administered to the mice model of middle cerebral artery occlusion (MCAO)/reperfusion. In this study, the impact of rh-B4galt1 on cerebral injury assessed using multiple methods, including the neurological disability status scale, 2,3,5-triphenyltetrazolium chloride (TTC), Nissl and TUNEL staining. This study utilized laser speckle Doppler flowmeter to monitor the cerebral blood flow. Western blotting was performed to assess the protein expression levels, and fluorescence-labeled dihydroethidium method was performed to determine the superoxide anion generation. Assay kits were used for the measurement of iron, malondialdehyde (MDA) and glutathione (GSH) levels. RESULTS: We demonstrated that rh-B4galt1 markedly improved neurological function, reduced cerebral infarct volume and preserved the completeness of blood-brain barrier (BBB) for preventing damage. These findings further illustrated that rh-B4galt1 alleviated oxidative stress, lipid peroxidation, as well as iron deposition induced by I/R. The vital role of ferroptosis was proved in brain injury. Furthermore, the rh-B4galt1 could increase the levels of TAZ, Nrf2 and HO-1 after I/R. And TAZ-siRNA and ML385 reversed the neuroprotective effects of rh-B4galt1. CONCLUSIONS: The results indicated that rh-B4galt1 implements neuroprotective effects by modulating ferroptosis, primarily via upregulating TAZ/Nrf2/HO-1 pathway. Thus, B4galt1 could be seen as a promising novel objective for ischemic stroke therapy.


Subject(s)
Brain Ischemia , Ferroptosis , Galactosyltransferases , Heme Oxygenase-1 , NF-E2-Related Factor 2 , Signal Transduction , Animals , Humans , Male , Mice , Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Ferroptosis/drug effects , Ferroptosis/physiology , Galactosyltransferases/metabolism , Heme Oxygenase-1/metabolism , Infarction, Middle Cerebral Artery , Membrane Proteins , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/physiology , Signal Transduction/drug effects , Transcription Factors/metabolism
14.
Behav Neurol ; 2024: 5698119, 2024.
Article in English | MEDLINE | ID: mdl-39233848

ABSTRACT

Objective: The objective of the study is to investigate whether quercetin ameliorates Alzheimer's disease (AD)-like pathology in APP/PS1 double transgenic mice and its hypothesized mechanism, contributing to the comprehension of AD pathogenesis. Methods: A total of 30 APP/PS1 transgenic mice were randomized into model group (APP/PS1), quercetin group (APP/PS1+Q), and donepezil hydrochloride group (APP/PS1+DON). Simultaneously, there were 10 C57 mice of the same age served as a control group. Three months posttreatment, the effects of quercetin on AD mice were evaluated using the Morris water maze (MWM) test, Y maze experiment, immunohistochemistry, immunofluorescence, and western blotting. Results: Results from the water maze and Y maze indicated that quercetin significantly improved cognitive impairment in APP/PS1 transgenic AD mice. Additionally, serum enzyme-linked immunosorbent assay (ELISA) results demonstrated that quercetin elevated MDA, superoxide dismutase (SOD), CAT, GSH, acetylcholine (ACh), and acetylcholinesterase (AChE) levels in AD mice. Hematoxylin-eosin (HE) staining, Nissl staining, and hippocampal tissue thioflavine staining revealed that quercetin reduced neuronal damage and Aß protein accumulation in AD mice. Western blot validated protein expression in the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2)/HO-1 pathway associated with oxidative stress and apoptosis, confirming quercetin's potential molecular mechanism of enhancing AD mouse cognition. Furthermore, western blot findings indicate that quercetin significantly alters protein expression in the Keap1/Nrf2/HO-1 pathway. Moreover, molecular docking analysis suggests that Keap1, NQO1, HO-1, caspase-3, Bcl-2, and Bax proteins in the Keap1/Nrf2/HO-1 pathway may be potential regulatory targets of quercetin. These findings will provide a molecular basis for quercetin's clinical application in AD treatment. Conclusion: Quercetin can improve cognitive impairment and AD-like pathology in APP/PS1 double transgenic mice, potentially related to quercetin's activation of the Keap1/Nrf2/HO-1 pathway and reduction of cell apoptosis.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Apoptosis , Brain , Cognitive Dysfunction , Disease Models, Animal , Heme Oxygenase-1 , Kelch-Like ECH-Associated Protein 1 , Mice, Transgenic , NF-E2-Related Factor 2 , Oxidative Stress , Quercetin , Animals , Quercetin/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Mice , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Heme Oxygenase-1/metabolism , Apoptosis/drug effects , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Signal Transduction/drug effects , Presenilin-1/genetics , Presenilin-1/metabolism , Male , Mice, Inbred C57BL , Membrane Proteins/metabolism , Membrane Proteins/genetics , Antioxidants/pharmacology , Antioxidants/metabolism
15.
Ann Clin Lab Sci ; 54(4): 446-451, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39293832

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) is a common chronic autoimmune inflammatory disease. The pathogenesis of RA is complex, and RA lacks effective therapeutic drugs. Heme oxygenase 1 (HO-1) is found to be reduced in RA. However, the role of HO-1 in RA and related mechanisms have not been elucidated. METHODS: RA rat model was established. The expression of HO-1 was upregulated by hemin. The increase weight rate, the degree of toe swelling, and the arthritis index were analyzed to evaluate the therapeutic effect of HO-1 on RA. In vitro RAW264.7 inflammatory cell model was established using 5 ng/mL IL-1. SnPP or hemin were used to inhibit or upregulate HO-1 expression. Tetrazolium salt colorimetric assay (MTT) was selected to test cell proliferation. ELISA was used to determine the concentrations of cellular inflammatory factors IL-1 and IL-6. Reactive oxygen species (ROS) activity was assessed. Western blot was performed to analyze NF-[Formula: see text]B and MMP-3 expressions. RESULTS: The expression of HO-1 was decreased in RA rats, and hemin increased HO-1 level in arthritic rats, which elevated the increase weight rate and decreased toe swelling degree and arthritis index (P<0.05). Hemin significantly upregulated HO-1 expression, inhibited inflammatory cell proliferation, decreased IL-1 and IL-6 expressions, declined ROS level, restrained NF-[Formula: see text]B expression, and enhanced MMP-3 expression in Raw264.7 cells induced by LPS (P<0.05). SnPP obviously inhibited the expression of HO-1, promoted cell proliferation, elevated IL-1 and IL-6 secretions, increased ROS level, promoted NF-[Formula: see text]B expression, and decreased MMP-3 level compared with LPS group (P<0.05). CONCLUSION: Upregulation of HO-1 can improve arthritis symptoms by reducing ROS expression, inhibiting NF-[Formula: see text]B signaling pathway, elevating MMP-3 expression, attenuating inflammatory factor secretion, and suppressing inflammatory cell proliferation.


Subject(s)
Arthritis, Rheumatoid , Heme Oxygenase-1 , Hemin , Reactive Oxygen Species , Animals , Mice , Rats , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Heme Oxygenase (Decyclizing) , Heme Oxygenase-1/metabolism , Hemin/pharmacology , Inflammation/pathology , Inflammation/metabolism , Interleukin-1/metabolism , Interleukin-6/metabolism , Matrix Metalloproteinase 3/metabolism , NF-kappa B/metabolism , Protoporphyrins/pharmacology , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
16.
Mol Biol Rep ; 51(1): 1015, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39325219

ABSTRACT

BACKGROUND: The adverse effects of radiotherapy (RT) primarily occur through oxidative stress, and attempts are being made to mitigate these effects. L-Carnitine (L-Car) involved in physiological functions, possesses antioxidant and tissue-protective properties. The goal of this investigation is to appraise the radioprotective efficacy of L-Car supplementation. METHODS AND RESULTS: The groups were established by dividing thirty-two rats as: control, RT (10 Gy), RT + L-Car (200 mg/kg/d), L-Car. Upon completion of the experiment, the livers were harvested for histopathological, immunostaining [tumor necrosis factor-alpha (TNF-α), Caspase-3], spectrophotometric [total oxidant status (TOS), total antioxidant status (TAS), oxidative stress index (OSI)], and mRNA expression [(Nuclear factor erythroid 2-related factor 2 (Nrf2), Kelch-like ECH-associated protein 1 (Keap-1), Heme Oxygenase (HO-1), Transforming growth factor beta 1 (TGF-ß1)] analyses. In the damage group, decreased Keap-1, Nrf2, HO-1, and TAS values, along with increased histopathological findings, alanine transferase, aspartate transferase, TNF-α, Caspase-3, TOS, OSI, TGF-ß1 levels were found. All findings were improved with L-Car treatment. CONCLUSIONS: Considering these findings, it can be inferred that L-Car exhibits tissue-protective effects against organ damage predominantly induced by RT-related oxidative stress. Additionally, it has prevented the development of inflammation, apoptosis, and fibrosis. Therefore, L-Car may be considered as a supplement to reduce complications associated with RT.


Subject(s)
Antioxidants , Carnitine , Dietary Supplements , Liver , NF-E2-Related Factor 2 , Oxidative Stress , Animals , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Carnitine/pharmacology , Rats , Liver/drug effects , Liver/metabolism , Liver/radiation effects , Liver/pathology , NF-E2-Related Factor 2/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Male , Radiation-Protective Agents/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Caspase 3/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Heme Oxygenase-1/metabolism , Rats, Wistar , Apoptosis/drug effects , Apoptosis/radiation effects
17.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39337313

ABSTRACT

Itaconic acid (IA) is recognized for its potential application in treating intestinal diseases owing to the anti-inflammatory and antioxidant properties. Perfluorooctanoic acid (PFOA) can accumulate in animals and result in oxidative and inflammatory damages to multi-tissue and organ, particularly in the intestinal tract. This study aimed to explore whether IA could mitigate intestinal damage induced by PFOA exposure in laying hens and elucidate its potential underlying mechanisms. The results showed that IA improved the antioxidant capacity of laying hens and alleviated the oxidative damage induced by PFOA, as evidenced by the elevated activities of T-SOD, GSH-Px, and CAT, and the decreased MDA content in both the jejunum and serum. Furthermore, IA improved the intestinal morphological and structural integrity, notably attenuating PFOA-induced villus shedding, length reduction, and microvillus thinning. IA also upregulated the mRNA expression of ZO-1, Occludin, Claudin-1, and Mucin-2 in the jejunum, thereby restoring intestinal barrier function. Compared with the PF group, IA supplementation downregulated the gene expression of Keap1 and upregulated the HO-1, NQO1, SOD1, and GPX1 expression in the jejunum. Meanwhile, the PF + IA group exhibited lower expressions of inflammation-related genes (NF-κB, IL-1ß, IFN-γ, TNF-α, and IL-6) compared to the PF group. Moreover, IA reversed the PFOA-induced imbalance in gut microbiota by reducing the harmful bacteria such as Escherichia-Shigella, Clostridium innocuum, and Ruminococcus torques, while increasing the abundance of beneficial bacteria like Lactobacillus. Correlation analysis further revealed a significant association between gut microbes, inflammatory factors, and the Keap1/Nrf2/HO-1 pathway expression. In conclusion, dietary IA supplementation could alleviate the oxidative and inflammatory damage caused by PFOA exposure in the intestinal tract by reshaping the intestinal microbiota, modulating the Keap1/Nrf2/HO-1 pathway and reducing oxidative stress and inflammatory response, thereby promoting intestinal homeostasis.


Subject(s)
Caprylates , Fluorocarbons , Gastrointestinal Microbiome , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Oxidative Stress , Animals , Gastrointestinal Microbiome/drug effects , Oxidative Stress/drug effects , Caprylates/pharmacology , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Signal Transduction/drug effects , Antioxidants/pharmacology , Antioxidants/metabolism , Chickens , Heme Oxygenase-1/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Female , Intestines/drug effects , Intestines/microbiology , Intestines/pathology
18.
Redox Biol ; 76: 103342, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39265498

ABSTRACT

BACKGROUND: Disruption of the blood-brain barrier (BBB) is a major contributor to hemorrhagic transformation (HT) in patients with acute ischemic stroke (AIS) following intravenous thrombolysis (IVT). However, the clinical therapies aimed at BBB protection after IVT remain limited. METHODS: One hundred patients with AIS who underwent IVT were enrolled (42 with HT and 58 without HT 24 h after IVT). Based on the cytokine chip, the serum levels of several AIS-related proteins, including LCN2, ferritin, matrix metalloproteinase-3, vascular endothelial-derived growth factor, and X-linked inhibitor of apoptosis, were detected upon admission, and their associations with HT were analyzed. After finding that LCN2 was related to HT in patients with IVT, we clarified whether the modulation of LCN2 influenced BBB dysfunction and HT after thrombolysis and investigated the potential mechanism. RESULTS: In patients with AIS following IVT, logistic regression analysis showed that baseline serum LCN2 (p = 0.023) and ferritin (p = 0.046) levels were independently associated with HT. A positive correlation between serum LCN2 and ferritin levels was identified in patients with HT. In experimental studies, recombinant LCN2 (rLCN2) significantly aggravated BBB dysfunction and HT in the thromboembolic stroke rats after thrombolysis, whereas LCN2 inhibition by ZINC006440089 exerted opposite effects. Further mechanistic studies showed that, LCN2 promoted endothelial cell ferroptosis, accompanied by the induction of high mobility group box 1 (HMGB1) and the inhibition of nuclear translocation of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) proteins. Ferroptosis inhibitor ferrostatin-1 (fer-1) significantly restricted the LCN2-mediated BBB disruption. Transfection of LCN2 and HMGB1 siRNA inhibited the endothelial cell ferroptosis, and this effects was reversed by Nrf2 siRNA. CONCLUSION: LCN2 aggravated BBB disruption after thrombolysis by promoting endothelial cell ferroptosis via regulating the HMGB1/Nrf2/HO-1 pathway, this may provide a promising therapeutic target for the prevention of HT after IVT.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Ferroptosis , HMGB1 Protein , Lipocalin-2 , NF-E2-Related Factor 2 , NF-E2-Related Factor 2/metabolism , Humans , Animals , Male , Rats , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , HMGB1 Protein/metabolism , Ferroptosis/drug effects , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Female , Lipocalin-2/metabolism , Signal Transduction/drug effects , Aged , Middle Aged , Thrombolytic Therapy , Ischemic Stroke/metabolism , Ischemic Stroke/drug therapy , Ischemic Stroke/pathology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics
19.
Int J Mol Sci ; 25(18)2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39337472

ABSTRACT

In pancreatic cancer, the tumor microenvironment (TME) accounts for up to 90% of the tumor mass. Pancreatitis, characterized by the increased infiltration of macrophages into the pancreas, is a known risk factor for pancreatic cancer. The NRF2 (nuclear factor erythroid 2-related factor 2) transcription factor regulates responses to oxidative stress and can promote cancer and chemoresistance. NRF2 also attenuates inflammation through the regulation of macrophage-specific genes. Heme oxygenase 1 (HO-1) is expressed by anti-inflammatory macrophages to degrade heme, and its expression is dependent on NRF2 translocation to the nucleus. In macrophages stimulated with conditioned media from pancreatic cancer cells, HO-1 protein levels increased, which correlated with higher NRF2 expression in the nuclear fraction. Significant differences in macrophage infiltration and HO-1 expression were detected in LSL-KrasG12D/+; Pdx-1-Cre (KC) mice, Nrf2 whole-body knockout (KO) mice and wildtype mice with pancreatitis. Since epigenetic modulation is a mechanism used by tumors to regulate the TME, using small molecules as epigenetic modulators to activate immune recognition is therapeutically desirable. When the bromodomain inhibitor I-BET-762 was used to treat macrophages or mice with pancreatitis, high levels of HO-1 were reduced. This study shows that bromodomain inhibitors can be used to prevent physiological responses to inflammation that promote tumorigenesis.


Subject(s)
Heme Oxygenase-1 , Macrophages , NF-E2-Related Factor 2 , Pancreatic Neoplasms , Transcription Factors , Animals , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Mice , Macrophages/metabolism , Macrophages/drug effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Pancreas/metabolism , Pancreas/pathology , Pancreas/drug effects , Humans , Pancreatitis/metabolism , Pancreatitis/drug therapy , Pancreatitis/genetics , Mice, Knockout , Tumor Microenvironment/drug effects , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Mice, Inbred C57BL , Bromodomain Containing Proteins , Membrane Proteins , Nuclear Proteins
20.
Clin Exp Pharmacol Physiol ; 51(11): e13922, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39348933

ABSTRACT

Neuropathic pain arises from impairments or malfunctions within the nervous system, resulting in atypical transmission and interpretation of pain signals. In the present study, we examined the neuroprotective effects of agomelatine (AGM) and agomelatine-loaded nanostructured lipid carriers (AGM-NLCs) in neuropathic animal models induced by chronic constriction injury (CCI) of the sciatic nerve. Male Sprague Dawley rats were divided into seven experimental groups to compare the effects of AGM and AGM-NLCs, which were administered at 20 mg/kg for 14 consecutive days after CCI. Our finding demonstrated that CCI triggered the onset of analgesia in these animals, corroborated by mechanical allodynia and thermal hyperalgesia. Furthermore, CCI induced an elevation in inflammatory mediators such as interleukin (IL)-1ß and inducible nitric oxide synthase (iNOS), and downregulated heme oxygenase-1 (HO-1) and nuclear factor E2-related factor (Nrf2). Treatment with AGM and AGM-NLCs reversed inflammatory cascades and elevated antioxidant enzyme levels, leading to a reduction in paw withdrawal latency and threshold in rats. To further investigate the effect of AGM and AGM-NLCs, all-trans retinoic acid (ATRA) was administered, which antagonizes Nrf2. ATRA substantially downregulated Nrf2 expression and exacerbated thermal hyperalgesia, whereas Nrf2 and HO-1 expressions were significantly upregulated after AGM-NLCs administration. Overall, the results demonstrated that AGM-NLCs offer promising antinociceptive and anti-inflammatory properties in alleviating neuropathic pain symptoms, which can be attributed to improved drug delivery and therapeutic outcomes compared with AGM alone.


Subject(s)
Acetamides , Drug Carriers , Lipids , NF-E2-Related Factor 2 , Nanostructures , Neuralgia , Rats, Sprague-Dawley , Signal Transduction , Animals , Neuralgia/drug therapy , Neuralgia/metabolism , Male , NF-E2-Related Factor 2/metabolism , Rats , Acetamides/pharmacology , Acetamides/administration & dosage , Signal Transduction/drug effects , Nanostructures/chemistry , Drug Carriers/chemistry , Hyperalgesia/drug therapy , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Naphthalenes
SELECTION OF CITATIONS
SEARCH DETAIL