Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.327
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Int J Biol Sci ; 20(12): 4888-4907, 2024.
Article in English | MEDLINE | ID: mdl-39309448

ABSTRACT

Reactive oxygen species are involved in the pathogenesis of cancers and metabolic diseases, including diabetes, obesity, and fatty liver disease. Thus, inhibiting the generation of free radicals is a promising strategy to control the onset of metabolic diseases and cancer progression. Various synthetic drugs and natural product-derived compounds that exhibit antioxidant activity have been reported to have a protective effect against a range of metabolic diseases and cancer. This review highlights the development and aggravation of cancer and metabolic diseases due to the imbalance between pro-oxidants and endogenous antioxidant molecules. In addition, we discuss the function of proteins that regulate the production of reactive oxygen species as a strategy to treat metabolic diseases. In particular, we summarize the role of proteins such as nuclear factor-like 2, Sestrin, and heme oxygenase-1, which regulate the expression of various antioxidant genes in metabolic diseases and cancer. We have included recent literature to discuss the latest research on identifying novel signals of antioxidant genes that can control metabolic diseases and cancer.


Subject(s)
Antioxidants , Heme Oxygenase-1 , Metabolic Diseases , NF-E2-Related Factor 2 , Neoplasms , Humans , Metabolic Diseases/metabolism , Metabolic Diseases/genetics , Neoplasms/metabolism , Neoplasms/genetics , Antioxidants/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Animals , Reactive Oxygen Species/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Oxidative Stress
8.
Wei Sheng Yan Jiu ; 53(5): 771-777, 2024 Sep.
Article in Chinese | MEDLINE | ID: mdl-39308109

ABSTRACT

OBJECTIVE: To probe into the protective effect of different dose of secoisolariciresinol diglucoside(SDG) on brain of offspring of mice anainst oxidative damage and inflammatory reaction induced by maternal exposure to trans fatty acids(TFA) during gestation, and observe the the changes of regulating Nrf2/Keap1 pathway in the course. METHODS: 30 healthy female mice(C57BL/6) were divided into 5 groups randomly, they are respectively control group, TFA-exposed group, and three SDG-intervention groups(low-(TFA+LSDG), medium-(TFA+MSDG) and high-(TFA+HSDG)). The pregnancy mice of control group and TFA group were treated with distilled water and 60 mg/kg·d TFA by gavage, in the same time, the mice of three SDG-intervention groups were treated with 60 mg/kg·d TFA by gavage and fed with feed included SDG(10, 20 and 30 mg/kg). The treatment to pregnancy mice continued to birth of offspring. After 21 days of lactation, the offspring were killed under anesthesia and the experiment was ended. The coefficient of brain was calculated. The levels of superoxide dismutase(SOD), glutathione peroxidase(GSH-Px), malondialdehyde(MDA), tumor necrosis factor-α(TNF-α), interferon-γ(IFN-γ) and amyloid-ß(Aß)of brain were detected. RT-PCR and Western Blot was used to detected gene expression and protein levels of nuclear factor erythroid-2 related factor 2(Nrf2), kelch-like ECH-associated protein 1(Keap1), quinone oxidoreductase 1(NQO1) and hemeoxygenase-l(HO-1). RESULTS: Compared with control group, the brain coefficient and Aß1-40 of offspring of TFA-group had no significant changes(P>0.05), the activity of SOD and GSH-Px reduced, the content of MDA, IFN-γ, TNF-α and Aß1-42 increased, the level of mRNA and protein expression of Nrf2, NQO1 and HO-1 decreased and the level of mRNA and protein expression of Keap1 increase because of the exposion to TFA during gestation and all the differences were statistically significant(P<0.05). Compared with TFA-group, the brain coefficient, Aß1-40 and the level of NQO1 mRNA of offspring of three SDG-intervention groups had no significant changes(P>0.05), the activity of SOD(the middle and high dose SDG intervention groups) and GSH-Px(three SDG-intervention groups) increased, the content of MDA(the middle and high dose SDG intervention groups), IFN-γ(the middle and high dose SDG intervention groups), TNF-α(three SDG-intervention groups) and Aß1-42(the middle and high dose SDG intervention groups) decreased, the mRNA expression of Nrf2 and HO-1(the middle and high dose SDG intervention groups) was up-regulated, the mRNA expression of Keap1(the middle and high dose SDG intervention group) decreased, proteic expression of Nrf2, NQO1 and HO-1 of three SDG-intervention groups increase and the level of protein of Keap1 decreased because of the intervention of SDG during gestation(P<0.05). CONCLUSION: These result suggest that maternal TFA exposure during gestation can result in oxidative stress and inflammation to brain of offspring in a way. SDG can protect brain of mice of offspring from TFA-induced oxidative injury by up-regulating the expression of mRNA and protein of Nrf2, down-regulating the expression of Keap1, accelerating expression of protein of NQO1 and HO-1 which are antioxidant protein lying downstream of pathway of Nrf2/Keap1.


Subject(s)
Brain , Butylene Glycols , Glucosides , Kelch-Like ECH-Associated Protein 1 , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Oxidative Stress , Trans Fatty Acids , Animals , Female , Mice , Glucosides/pharmacology , Pregnancy , NF-E2-Related Factor 2/metabolism , Brain/metabolism , Brain/drug effects , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Oxidative Stress/drug effects , Butylene Glycols/pharmacology , Trans Fatty Acids/adverse effects , Prenatal Exposure Delayed Effects/metabolism , Inflammation/metabolism , Inflammation/chemically induced , Maternal Exposure/adverse effects , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Malondialdehyde/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics
9.
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
10.
J Neuroimmune Pharmacol ; 19(1): 50, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39312021

ABSTRACT

Depression is a global psychiatric illness that imposes a substantial economic burden. Unfortunately, traditional antidepressants induce many side effects which limit patient compliance thus, exploring alternative therapies with fewer adverse effects became urgent. This study aimed to investigate the effect of trimetazidine (TMZ); a well-known anti-ischemic drug in lipopolysaccharide (LPS) mouse model of depression focusing on its ability to regulate toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) as well as nuclear factor erythroid 2 related factor 2 (Nrf2)/ heme oxygenase-1 (HO-1) signaling pathways. Male Swiss albino mice were injected with LPS (500 µg/kg, i.p) every other day alone or parallel with oral doses of either escitalopram (Esc) (10 mg/kg/day) or TMZ (20 mg/kg/day) for 14 days. Treatment with TMZ attenuated LPS-induced animals' despair with reduced immobility time inforced swimming test. TMZ also diminished LPS- induced neuro-inflammation via inhibition of TLR4/NF-κB pathway contrary to Nrf2/HO-1 cascade activation with consequent increase in reduced glutathione (GSH) and HO-1 levels whereas the pro-inflammatory cytokines; tumor necrosis factor-α (TNF-α) and interleukin (IL)-1ß were evidently reduced. Besides, TMZ replenished brain serotonin levels via serotonin transporter (SERT) inhibition. Thus, TMZ hindered LPS-induced neuro-inflammation, oxidative stress, serotonin deficiency besides its anti-apoptotic effect which was reflected by decreased caspase-3 level. Neuroprotective effects of TMZ were confirmed by the histological photomicrographs which showed prominent neuronal survival. Here we showed that TMZ is an affluent nominee for depression management via targeting TLR4/NF-κB and Nrf2/HO-1 pathways. Future research addressing TMZ-antidepressant activity in humans is mandatory to enroll it as a novel therapeutic strategy for depression.


Subject(s)
Depression , Lipopolysaccharides , NF-E2-Related Factor 2 , NF-kappa B , Toll-Like Receptor 4 , Trimetazidine , Animals , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Male , Mice , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Lipopolysaccharides/toxicity , Depression/drug therapy , Depression/chemically induced , Depression/metabolism , Trimetazidine/pharmacology , Trimetazidine/therapeutic use , Signal Transduction/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Heme Oxygenase-1/metabolism , Membrane Proteins
11.
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
12.
J Nanobiotechnology ; 22(1): 531, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218878

ABSTRACT

Ferroptosis, triggered by iron overload and excessive lipid peroxidation, plays a pivotal role in the progression of DOX-induced cardiomyopathy (DIC), and thus limits the use of doxorubicin (DOX) in clinic. Here, we further showed that cardiac ferroptosis induced by DOX in mice was attributed to up-regulation of Hmox1, as knockdown of Hmox1 effectively inhibited cardiomyocyte ferroptosis. To targeted delivery of siRNA into cardiomyocytes, siRNA-encapsulated exosomes were injected followed by ultrasound microbubble targeted destruction (UTMD) in the heart region. UTMD greatly facilitated exosome delivery into heart. Consistently, UTMD assisted exosomal delivery of siHomox1 nearly blocked the ferroptosis and the subsequent cardiotoxicity induced by doxorubicin. In summary, our findings reveal that the upregulation of HMOX1 induces ferroptosis in cardiomyocytes and UTMD-assisted exosomal delivery of siHmox1 can be used as a potential therapeutic strategy for DIC.


Subject(s)
Doxorubicin , Exosomes , Ferroptosis , Heme Oxygenase-1 , Microbubbles , Myocytes, Cardiac , RNA, Small Interfering , Ferroptosis/drug effects , Animals , Doxorubicin/pharmacology , Exosomes/metabolism , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Heme Oxygenase-1/metabolism , RNA, Small Interfering/pharmacology , Mice, Inbred C57BL , Male , Drug Delivery Systems , Cardiomyopathies/metabolism , Membrane Proteins
13.
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
14.
Int J Mol Sci ; 25(17)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39273143

ABSTRACT

Prostate cancer (PC) is a significant cause of mortality in men worldwide, hence the need for a comprehensive understanding of the molecular mechanisms underlying its progression and resistance to treatment. Heme oxygenase-1 (HO-1), an inducible enzyme involved in heme catabolism, has emerged as a critical player in cancer biology, including PC. This review explores the multifaceted role of HO-1 in PC, encompassing its function, regulation, and implications in cancer therapy. HO-1 influences cell proliferation, anti-apoptotic pathways, angiogenesis, and the tumor microenvironment, thereby influencing tumor growth and metastasis. HO-1 has also been associated with therapy resistance, affecting response to standard treatments. Moreover, HO-1 plays a significant role in immune modulation, affecting the tumor immune microenvironment and potentially influencing therapy outcomes. Understanding the intricate balance of HO-1 in PC is vital for developing effective therapeutic strategies. This review further explores the potential of targeting HO-1 as a therapeutic approach, highlighting challenges and opportunities. Additionally, clinical implications are discussed, focusing on the prognostic value of HO-1 expression and the development of novel combined therapies to augment PC sensitivity to standard treatment strategies. Ultimately, unraveling the complexities of HO-1 in PC biology will provide critical insights into personalized treatment approaches for PC patients.


Subject(s)
Heme Oxygenase-1 , Prostatic Neoplasms , Tumor Microenvironment , Humans , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Prostatic Neoplasms/genetics , Male , Gene Expression Regulation, Neoplastic , Animals , Cell Proliferation
15.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167496, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39237046

ABSTRACT

Liver ischemia-reperfusion (I/R) injury is a detrimental complication of organ transplantation, shock, and sepsis. However, the available drugs to mitigate I/R injury remain limited. Jujuboside A (JuA) is renowned for its antioxidant, anti-inflammatory, and anti-apoptotic properties; nevertheless, its potential in liver I/R injury remains unknown. Thus, this study aimed to explore the role and underlying mechanisms of JuA in liver I/R injury. Mouse models of I/R and AML12 cell models of hypoxia/reoxygenation (H/R) were constructed. Haematoxylin and eosin staining, serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) detection, and cell viability analysis were used to assess liver injury. To evaluate oxidative stress, inflammation, apoptosis, and mitochondrial damage, immunofluorescence staining, transmission electron microscopy analysis, enzyme-linked immunosorbent assay, and flow cytometry were conducted. Moreover, molecular docking techniques and western blot were employed to identify downstream target molecules and pathways affected by JuA. The results showed that JuA pretreatment effectively attenuated liver necrosis and ALT and AST level elevations induced by I/R while enhancing AML12 cell viability following H/R. Furthermore, JuA pretreatment suppressed oxidative stress triggered by I/R and H/R, thereby inhibiting the level of pro-inflammatory factors and NLRP3 inflammasome activation. Notably, JuA pretreatment alleviated mitochondrial damage and apoptosis. Mechanistically, JuA pretreatment resulted in the activation of the AKT/NRF2/HO-1 signalling pathways, whereas MK2206, the inhibitor of AKT, partially reversed the hepatoprotective effects of JuA during liver I/R. Collectively, our findings illustrated that JuA mitigated oxidative stress, inflammation, apoptosis, and mitochondrial damage by facilitating the AKT/NRF2/HO-1 signalling pathway, thereby alleviating liver I/R injury.


Subject(s)
Apoptosis , Liver , NF-E2-Related Factor 2 , Proto-Oncogene Proteins c-akt , Reperfusion Injury , Signal Transduction , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/drug therapy , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice , Signal Transduction/drug effects , Male , Liver/pathology , Liver/metabolism , Liver/drug effects , Apoptosis/drug effects , Oxidative Stress/drug effects , Mice, Inbred C57BL , Heme Oxygenase-1/metabolism , Cell Line , Membrane Proteins/metabolism , Heme Oxygenase (Decyclizing)/metabolism
16.
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
17.
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
18.
Int J Mol Sci ; 25(18)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39337609

ABSTRACT

Age-related macular degeneration (AMD) is a common disease contributing to vision loss in the elderly. All-trans-retinal (atRAL) is a retinoid in the retina, and its abnormal accumulation exhibits toxicity to the retina and promotes oxidative stress-induced photoreceptor degeneration, which plays a crucial role in AMD progression. Crocin is a natural product extracted from saffron, which displays significant antioxidant and anti-inflammatory effects. The present study elucidates the protective effects of crocin on photoreceptor cell damage by atRAL and its potential mechanisms. The results revealed that crocin significantly attenuated cytotoxicity by repressing oxidative stress, mitochondrial injury, and DNA damage in atRAL-loaded photoreceptor cells. Moreover, crocin visibly inhibited DNA damage-induced apoptosis and gasdermin E (GSDME)-mediated pyroptosis in photoreceptor cells after exposure to atRAL. It was also observed that crocin distinctly prevented an increase in Fe2+ levels and lipid peroxidation caused by atRAL via suppressing the Kelch-like ECH-associated protein 1 (KEAP1)/nuclear factor-erythroid 2-related factor 2 (NRF2)/heme oxygenase-1 (HO-1) signaling pathway, thereby ameliorating photoreceptor cell ferroptosis. In short, these findings provide new insights that crocin mitigates atRAL-induced toxicity to photoreceptor cells by inhibiting oxidative stress, apoptosis, pyroptosis, and ferroptosis.


Subject(s)
Carotenoids , Oxidative Stress , Retinaldehyde , Animals , Mice , Carotenoids/pharmacology , Oxidative Stress/drug effects , Retinaldehyde/metabolism , Cell Line , NF-E2-Related Factor 2/metabolism , Apoptosis/drug effects , Signal Transduction/drug effects , Antioxidants/pharmacology , Heme Oxygenase-1/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Ferroptosis/drug effects , DNA Damage/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Macular Degeneration/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Protective Agents/pharmacology , Lipid Peroxidation/drug effects , Pyroptosis/drug effects
19.
Int Immunopharmacol ; 142(Pt A): 113065, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39243557

ABSTRACT

BACKGROUND: Alveolar epithelial cell (AEC) ferroptosis contributes to the progression of acute lung injury (ALI). Esketamine (ESK) is a new clinical sedative, anesthetic, and analgesic drug that has attracted substantial attention in mental health research because of its antidepressant effects. However, the effects of ESK on ferroptosis-mediated ALI remain unclear. OBJECTIVE: This study aimed to explore the protective effect of ESK on AEC ferroptosis in ALI and its potential molecular mechanism in vivo and in vitro. METHODS: The antiferroptotic and anti-inflammatory effects of ESK were assessed in a mouse model of lipopolysaccharide (LPS)-induced ALI. In vitro, the epithelial cell lines MLE-12 and A549 were used to examine the underlying mechanism by which ESK regulates inflammation and ferroptosis. RESULTS: ESK protected mice against LPS-induced ALI, significantly attenuated pathological changes in the lungs and decreased inflammation and ferroptosis. In vitro, ESK inhibited LPS-induced inflammation and ferroptosis in MLE-12 and A549 cells. Moreover, ferroptosis mediated inflammation in LPS-induced ALI in vivo and in vitro, and ESK decreased the LPS-induced inflammatory response by suppressing ferroptosis. ESK promoted the HIF-1α/HO-1 pathway in LPS-treated AECs and in the lung tissues of mice with LPS-induced ALI. Moreover, pretreatment with ESK and the HIF-1α stabilizer dimethyloxaloylglycine (DMOG) substantially attenuated lung injury and prevented changes in ferroptosis-related biochemical indicators, including glutathione (GSH) depletion, malondialdehyde (MDA) production and glutathione peroxidase 4 (GPX4) downregulation, in untreated LPS-induced mice but not in LPS-induced mice treated with the HO-1 inhibitor zinc protoporphyrin (ZNPP). Similar effects were observed in vitro in HO-1 siRNA-transfected A549 cells after LPS incubation but not in control siRNA-transfected cells. CONCLUSION: ESK can inhibit ferroptosis-mediated lipid peroxidation by increasing the expression of HIF-1α/HO-1 pathway, highlighting the potential of ESK to treat LPS-induced ALI.


Subject(s)
Acute Lung Injury , Ferroptosis , Heme Oxygenase-1 , Hypoxia-Inducible Factor 1, alpha Subunit , Ketamine , Lipopolysaccharides , Mice, Inbred C57BL , Signal Transduction , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Ferroptosis/drug effects , Ketamine/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Heme Oxygenase-1/metabolism , Mice , Male , A549 Cells , Signal Transduction/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Line , Disease Models, Animal , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Lung/pathology , Lung/drug effects , Lung/immunology , Membrane Proteins
20.
Redox Biol ; 76: 103345, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39255694

ABSTRACT

Plaque rupture with consequent thrombosis is the leading cause of acute cardiovascular events, during which macrophage death is a hallmark. Ferroptosis is a pivotal intermediate link between early and advanced atherosclerosis. Existing evidence indicates the involvement of macrophage ferroptosis in plaque vulnerability; however, the exact mechanism remains elusive. The aim of this study was to explore key ferroptosis-related genes (FRGs) involved in plaque progression and the underlying molecular mechanisms involved. The expression landscape of FRGs was obtained from atherosclerosis-related GEO datasets. Molecular mechanism studies of ferroptosis were performed using bone marrow-derived macrophages (BMDMs) and macrophage-derived foam cells (MDFCs). Bioinformatics analysis and immunohistochemistry revealed that macrophage haem oxygenase-1 (HMOX1) is the key FRG involved in plaque destabilization. Hypoxic conditions induced a significant increase in Hmox1 expression in MDFCs but not in macrophages. In addition, the beneficial or deleterious effects of Hmox1 were dependent on the degree of Hmox1 induction. Hmox1 overexpression drove inflammatory responses and ferroptotic oxidative stress in MDFCs and aggravated the plaque burden in atherosclerotic model mice. Further mechanistic investigations demonstrated that hypoxia-mediated degradation of egl-9 family hypoxia-inducible factor 3 (Egln3) stabilized Hif1a, which subsequently promoted Hmox1 transcription. Our findings suggest that high Hmox1 expression under hypoxia is deleterious to MDFC viability and plaque stability, providing a reference for the management of acute cardiovascular events.


Subject(s)
Ferroptosis , Foam Cells , Heme Oxygenase-1 , Plaque, Atherosclerotic , Ferroptosis/genetics , Animals , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Mice , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/genetics , Foam Cells/metabolism , Foam Cells/pathology , Macrophages/metabolism , Disease Models, Animal , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Humans , Oxidative Stress , Male , Gene Expression Regulation , Membrane Proteins
SELECTION OF CITATIONS
SEARCH DETAIL