Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.471
Filter
1.
Mol Biol Rep ; 51(1): 703, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38822881

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is the leading cause of cancer morbidity and mortality worldwide, and new diagnostic markers are urgently needed. We aimed to investigate the mechanism by which hsa_circ_0096157 regulates autophagy and cisplatin (DDP) resistance in NSCLC. METHODS: A549 cells were treated with DDP (0 µg/mL or 3 µg/mL). Then, the autophagy activator rapamycin (200 nm) was applied to the A549/DDP cells. Moreover, hsa_circ_0096157 and Nrf2 were knocked down, and Nrf2 was overexpressed in A549/DDP cells. The expression of Hsa_circ_0096157, the Nrf2/ARE pathway-related factors Nrf2, HO-1, and NQO1, and the autophagy-related factors LC3, Beclin-1, and p62 was evaluated by qRT‒PCR or western blotting. Autophagosomes were detected through TEM. An MTS assay was utilized to measure cell proliferation. The associated miRNA levels were also tested by qRT‒PCR. RESULTS: DDP (3 µg/mL) promoted hsa_circ_0096157, LC3 II/I, and Beclin-1 expression and decreased p62 expression. Knocking down hsa_circ_0096157 resulted in the downregulation of LC3 II/I and Beclin-1 expression, upregulation of p62 expression, and decreased proliferation. Rapamycin reversed the effect of interfering with hsa_circ_0096157. Keap1 expression was lower, and Nrf2, HO-1, and NQO1 expression was greater in the A549/DDP group than in the A549 group. HO-1 expression was repressed after Nrf2 interference. In addition, activation of the Nrf2/ARE pathway promoted autophagy in A549/DDP cells. Moreover, hsa_circ_0096157 activated the Nrf2/ARE pathway. The silencing of hsa_circ_0096157 reduced Nrf2 expression by releasing miR-142-5p or miR-548n. Finally, we found that hsa_circ_0096157 promoted A549/DDP cell autophagy by activating the Nrf2/ARE pathway. CONCLUSION: Knockdown of hsa_circ_0096157 inhibits autophagy and DDP resistance in NSCLC cells by downregulating the Nrf2/ARE signaling pathway.


Subject(s)
Autophagy , Carcinoma, Non-Small-Cell Lung , Cisplatin , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Lung Neoplasms , NF-E2-Related Factor 2 , Signal Transduction , Humans , Cisplatin/pharmacology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Autophagy/drug effects , Autophagy/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , A549 Cells , Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Line, Tumor , Antioxidant Response Elements/genetics , Antineoplastic Agents/pharmacology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism
2.
Commun Biol ; 7(1): 621, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783088

ABSTRACT

Nuclear factor erythroid 2-related factor-2 (Nrf2) antioxidant signaling is involved in liver protection, but this generalization overlooks conflicting studies indicating that Nrf2 effects are not necessarily hepatoprotective. The role of Nrf2/heme oxygenase-1 (HO-1) in cholestatic liver injury (CLI) remains poorly defined. Here, we report that Nrf2/HO-1 activation exacerbates liver injury rather than exerting a protective effect in CLI. Inhibiting HO-1 or ameliorating bilirubin transport alleviates liver injury in CLI models. Nrf2 knockout confers hepatoprotection in CLI mice, whereas in non-CLI mice, Nrf2 knockout aggravates liver damage. In the CLI setting, oxidative stress activates Nrf2/HO-1, leads to bilirubin accumulation, and impairs mitochondrial function. High levels of bilirubin reciprocally upregulate the activation of Nrf2 and HO-1, while antioxidant and mitochondrial-targeted SOD2 overexpression attenuate bilirubin toxicity. The expression of Nrf2 and HO-1 is elevated in serum of patients with CLI. These results reveal an unrecognized function of Nrf2 signaling in exacerbating liver injury in cholestatic disease.


Subject(s)
Bilirubin , Cholestasis , Heme Oxygenase-1 , Mice, Knockout , NF-E2-Related Factor 2 , Oxidative Stress , Signal Transduction , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Animals , Mice , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Cholestasis/metabolism , Cholestasis/pathology , Cholestasis/genetics , Humans , Male , Bilirubin/metabolism , Bilirubin/blood , Mice, Inbred C57BL , Liver/metabolism , Liver/injuries , Liver/pathology , Disease Models, Animal , Membrane Proteins
3.
J Ovarian Res ; 17(1): 107, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762721

ABSTRACT

Abnormal granulosa cell (GC) death contributes to cyclophosphamide (CTX) induced primary ovarian insufficiency (POI). To investigate the contribution of GCs to POI, gene profiles of GCs exposed to CTX were assessed using RNA-Seq and bioinformatics analysis. The results showed the differentially expressed genes (DEGs) were enriched in the ferroptosis-related pathway, which is correlated with upregulated heme oxygenase 1 (HO-1) and downregulated glutathione peroxidase-4 (GPX4). Using CTX-induced cell culture (COV434 and KGN cells), the levels of iron, reactive oxygen species (ROS), lipid peroxide, mitochondrial superoxide, mitochondrial morphology and mitochondrial membrane potential (MMP) were detected by DCFDA, MitoSOX, C11-BODIPY, MitoTracker, Nonylacridine Orange (NAO), JC-1 and transmission electron microscopy respectively. The results showed iron overload and disrupted ROS, including cytoROS, mtROS and lipROS homeostasis, were associated with upregulation of HO-1 and could induce ferroptosis via mitochondrial dysfunction in CTX-induced GCs. Moreover, HO-1 inhibition could suppress ferroptosis induced GPX4 depletion. This implies a role for ROS in CTX-induced ferroptosis and highlights the effect of HO-1 modulators in improving CTX-induced ovarian damage, which may provide a theoretical basis for preventing or restoring GC and ovarian function in patients with POI.


Subject(s)
Cyclophosphamide , Ferroptosis , Granulosa Cells , Heme Oxygenase-1 , Mitochondria , Reactive Oxygen Species , Ferroptosis/drug effects , Female , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Cyclophosphamide/pharmacology , Cyclophosphamide/adverse effects , Reactive Oxygen Species/metabolism , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Membrane Potential, Mitochondrial/drug effects
4.
Bull Exp Biol Med ; 176(5): 562-566, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38724811

ABSTRACT

We studied the effect of an NO donor, nitrosyl iron complex with N-ethylthiourea, on Nrf2-dependent antioxidant system activation of tumor cells in vitro. The complex increased intracellular accumulation of Nrf2 transcription factor and induced its nuclear translocation. It was shown that both heme oxygenase-1 gene and protein expression increased significantly under the influence of the complex. Nrf2 activation was accompanied by a decrease in the intracellular accumulation of proinflammatory transcription factor NF-κB p65 subunit and expression of its target genes. The cytotoxic effect of N-ethylthiourea leads to induction of Nrf2/HO-1 antioxidant response and suppression of NF-κB-dependent processes in tumor cells.


Subject(s)
Heme Oxygenase-1 , Iron , NF-E2-Related Factor 2 , Thiourea , Humans , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Thiourea/analogs & derivatives , Thiourea/pharmacology , HeLa Cells , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Iron/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Nitrogen Oxides/metabolism , Nitrogen Oxides/pharmacology , Antioxidants/pharmacology
5.
Int Immunopharmacol ; 134: 112231, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38739977

ABSTRACT

The roles of immune cell infiltration and ferroptosis in the progression of proliferative diabetic retinopathy (PDR) remain unclear. To identify upregulated molecules associated with immune infiltration and ferroptosis in PDR, GSE60436 and GSE102485 datasets were downloaded from the Gene Expression Omnibus (GEO). Genes associated with immune cell infiltration were examined through Weighted Gene Co-expression Network Analysis (WGCNA) and CIBERSORT algorithm. Common differentially expressed genes (DEGs) were intersected with ferroptosis-associated and immune cell infiltration-related genes. Localization of cellular expression was confirmed by single-cell analysis of GSE165784 dataset. Findings were validated by qRT-PCR, ELISA, Western blotting, and immunofluorescence staining. As a result, the infiltration of M2 macrophages was significantly elevated in fibrovascular membrane samples from PDR patients than the retinas of control subjects. Analysis of DEGs, M2 macrophage-related genes and ferroptosis-related genes identified three hub intersecting genes, TP53, HMOX1 and PPARA. qRT-PCR showed that HMOX1 was significantly higher in the oxygen-induced retinopathy (OIR) mouse model retinas than in controls. Single-cell analysis confirmed that HMOX1 was located in M2 macrophages. ELISA and western blotting revealed elevated levels of HMOX1 in the vitreous humor of PDR patients and OIR retinas, and immunofluorescence staining showed that HMOX1 co-localized with M2 macrophages in the retinas of OIR mice. This study offers novel insights into the mechanisms associated with immune cell infiltration and ferroptosis in PDR. HMOX1 expression correlated with M2 macrophage infiltration and ferroptosis, which may play a crucial role in PDR pathogenesis.


Subject(s)
Diabetic Retinopathy , Ferroptosis , Heme Oxygenase-1 , Macrophages , Up-Regulation , Diabetic Retinopathy/genetics , Diabetic Retinopathy/immunology , Diabetic Retinopathy/pathology , Diabetic Retinopathy/metabolism , Animals , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Macrophages/immunology , Ferroptosis/genetics , Mice , Mice, Inbred C57BL , Retina/immunology , Retina/pathology , Retina/metabolism , Male , Disease Models, Animal , Membrane Proteins
6.
Mol Med Rep ; 30(1)2024 Jul.
Article in English | MEDLINE | ID: mdl-38785151

ABSTRACT

Periodontal disease is a common infectious disease that can lead to the loss of teeth. Hower how to effectively suppress the inflammation with medication is unclear. The aim of the present study was to investigate the anti­inflammatory effect of Oroxylin A in periodontitis and its potential role through heme oxygenase­1 (HO­1). Primary rat gingival fibroblasts (RGFs) were cultured using the tissue block method and identified by immunofluorescence. Following lipopolysaccharide (LPS) stimulation of RGFs, Oroxylin A was administered at 50, 100, 200 or 400 µg/ml. Reverse transcription­quantitative PCR was used to assess mRNA expression of cyclooxygenase (COX)­2, TNF­α, RANKL and osteoprotegerin (OPG). Western blotting was used to detect protein expression levels of COX ­2, TNF­α, RANKL and OPG. Following HO­1 knockdown, the same treatment was performed. The expression of COX­2 in rat gingival tissue was observed by immunohistochemistry. One­way analysis of variance and Student's t test were used for statistical analysis. Oroxylin A downregulated mRNA expression of COX­2, TNF­α, RANKL and OPG in LPS­induced RGFs. With increase of Oroxylin A dose, the expression of HO­1 was gradually upregulated. When HO­1 was knocked down, Oroxylin A did not downregulate the expression of COX­2, TNF­α, RANKL and OPG in LPS­induced RGFs. Immunohistochemical results showed that expression of COX­2 was downregulated by Oroxylin A, and the expression of TNF­α, RANKL and OPG were also downregulated. Oroxylin A decreased expression of inflammatory cytokines in LPS­induced RGFs and had a good inhibitory effect on periodontitis in rats.


Subject(s)
Cyclooxygenase 2 , Fibroblasts , Flavonoids , Periodontitis , RANK Ligand , Animals , Rats , Flavonoids/pharmacology , Periodontitis/metabolism , Periodontitis/drug therapy , Periodontitis/pathology , RANK Ligand/metabolism , RANK Ligand/genetics , Male , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Fibroblasts/metabolism , Fibroblasts/drug effects , Osteoprotegerin/metabolism , Osteoprotegerin/genetics , Lipopolysaccharides , Gingiva/metabolism , Gingiva/drug effects , Tumor Necrosis Factor-alpha/metabolism , Cytokines/metabolism , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Cells, Cultured , Rats, Sprague-Dawley
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 296-302, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38710513

ABSTRACT

Objective To evaluate the effects of heme oxygenase-1 (HO-1) gene deletion on immune cell composition and inflammatory injury in lung tissues of mice with lipopolysaccharide (LPS)-induced acute lung injury (ALI). Methods C57BL/6 wild-type (WT) mice and HO-1 conditional-knockout (HO-1-/-) mice on the same background were randomly divided into four groups (n=5 in every group): WT control group, LPS-treated WT group, HO-1-/- control group and LPS-treated HO-1-/- group. LPS-treated WT and HO-1-/- groups were injected with LPS (15 mg/kg) through the tail vein to establish ALI model, while WT control group and HO-1-/- control group were injected with an equivalent volume of normal saline through the tail vein, respectively. Twelve hours later, the mice were sacrificed and lung tissues from each group were collected for analysis. Histopathological alterations of lung tissues were assessed by HE staining. The levels of mRNA expression of tumor necrosis factor α (TNF-α), interleukin 1ß (IL-1ß), and IL-6 were determined by PCR. The percentages of neutrophils (CD45+CD11b+Ly6G+Ly6C-), total monocytes (CD45+CD11b+Ly6Chi), pro-inflammatory monocyte subsets (CD45+CD11b+Ly6ChiCCR2hi) and total macrophages (CD45+CD11b+F4/80+), M1 macrophage (CD45+CD11b+F4/80+CD86+), M2 macrophage (CD45+CD11b+F4/80+CD206+), total T cells (CD45+CD3+), CD3+CD4+ T cells, CD3+CD8+ T cells and myeloid suppressor cells (MDSCs, CD45+CD11b+Gr1+) were detected by flow cytometry. Results Compared with the corresponding control groups, HE staining exhibited increased inflammation in the lung tissues of both LPS-treated WT and HO-1-/- model mice; mRNA expression levels of TNF-α, IL-1ß and IL-6 were up-regulated; the proportions of neutrophils, total monocytes, pro-inflammatory monocyte subsets, MDSCs and total macrophages increased significantly. The percentage of CD3+, CD3+CD4+ and CD3+CD8+ T cells decreased significantly. Under resting-state, compared with WT control mice, the proportion of neutrophils, monocytes and pro-inflammatory monocyte subset increased in lung tissues of HO-1-/- control mice, while the proportion of CD3+ and CD3+CD8+ T cells decreased. Compared with LPS-treated WT mice, the mRNA expression levels of TNF-α and IL-1ß were up-regulated in lung tissues of LPS-treated HO-1-/- mice; the proportion of total monocytes, pro-inflammatory monocyte subsets, M1 macrophages and M1/M2 ratio increased greatly; the percentage of CD3+CD8+ T cells decreased significantly. Conclusion The deletion of HO-1 affects the function of the lung immune system and aggravates the inflammatory injury after LPS stimulation in ALI mice.


Subject(s)
Acute Lung Injury , Heme Oxygenase-1 , Lipopolysaccharides , Lung , Mice, Inbred C57BL , Mice, Knockout , Animals , Male , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation/genetics , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Lung/pathology , Lung/immunology , Lung/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Zhongguo Zhong Yao Za Zhi ; 49(9): 2308-2315, 2024 May.
Article in Chinese | MEDLINE | ID: mdl-38812131

ABSTRACT

This study aims to decipher the mechanism of tetramethylpyrazine(TMP) in regulating the migration of neural stem cells(NSCs) in the rat model of middle cerebral artery occlusion(MCAO) via the nuclear factor erythroid 2-related factor 2(Nrf2)/heme oxygenase 1(HO-1)/C-X-C motif chemokine receptor 4(CXCR4) pathway. SD rats were randomized into sham, MCAO(model), and tetramethylpyrazine(TMP, 20 mg·kg~(-1) and 40 mg·kg~(-1)) groups. The neurological impairment was assessed by the modified neurological severity score(mNSS). The immunofluorescence assay was employed to detect the cells stained with both 5-bromodeoxyuridine(BrdU) and doublecortin(DCX) in the brain tissue. The effect of TMP on the migration of C17.2 cells was observed. Western blot was employed to determine the protein levels of Nrf2, HO-1, p62, NAD(P)H quinone oxidoreductase 1(NQO1), stromal cell-derived factor 1(SDF-1), and CXCR4 in the brain tissue and C17.2 cells. The results showed that after 7 days and 21 days of mode-ling, the mNSS and BrdU~+/DCX~+ cells were increased, and the expression of Nrf2 and CXCR4 in the brain tissue was up-regulated. Compared with the model group, TMP(40 mg·kg~(-1)) reduced the mNSS, increased the number of BrdU~+/DCX~+ cells, and up-regulated the expression of Nrf2, CXCR4, and SDF-1. In addition, TMP promoted the migration of C17.2 cells and up-regulated the expression of p62, Nrf2, HO-1, and NQO1 in a time-and dose-dependent manner. The expression was the highest at the time point of 12 h in the TMP(50 µg·mL~(-1)) group(P<0.01). In conclusion, TMP activates the Nrf2/HO-1/CXCR4 pathway to promote the migration of NSCs to the ischemic area, thus exerting the therapeutic effect on the ischemia-reperfusion injury. This study provides experimental support for the application of TMP in ischemic stroke.


Subject(s)
Cell Movement , Heme Oxygenase-1 , NF-E2-Related Factor 2 , Neural Stem Cells , Pyrazines , Rats, Sprague-Dawley , Receptors, CXCR4 , Animals , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Pyrazines/pharmacology , Rats , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Cell Movement/drug effects , Male , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Doublecortin Protein , Signal Transduction/drug effects , Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , Humans
9.
J Am Heart Assoc ; 13(11): e033981, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38818928

ABSTRACT

BACKGROUND: Oxidative stress plays a principal role in the pathogenesis of white matter hyperintensities (WMHs). The induction of heme oxygenase-1 (HO-1) gene in the brain represents 1 of the pivotal mechanisms to counteract the noxious effects of reactive oxygen species, and the transcriptional modulation of HO-1 induction depends on the length of a GT-repeat (GT)n in the promoter region. We investigated whether the HO-1 gene (GT)n polymorphism is associated with the risk of WMHs. METHODS AND RESULTS: A total of 849 subjects from the memory clinic were consecutively enrolled, and the HO-1 (GT)n genotype was determined. WMHs were assessed with the Fazekas scale and further divided into periventricular WMHs and deep WMHs (DWMHs). Allelic HO-1 (GT)n polymorphisms were classified as short (≤24 (GT)n), median (25≤[GT]n<31), or long (31≤[GT]n). Multivariate logistic regression analysis was used to evaluate the effect of the HO-1 (GT)n variants on WMHs. The number of repetitions of the HO-1 gene (GT)n ranged from 15 to 39 with a bimodal distribution at lengths 23 and 30. The proportion of S/S genotypes was higher for moderate/severe DWMHs than none/mild DWMHs (22.22% versus 12.44%; P=0.001), but the association for periventricular WMHs was not statistically significant. Logistic regression suggested that the S/S genotype was significantly associated with moderate/severe DWMHs (S/S versus non-S/S: odds ratio, 2.001 [95% CI, 1.323-3.027]; P<0.001). The HO-1 gene (GT)n S/S genotype and aging synergistically contributed to the progression of DWMHs (relative excess risk attributable to interaction, 6.032 [95% CI, 0.149-11.915]). CONCLUSIONS: Short (GT)n variants in the HO-1 gene may confer susceptibility to rather than protection from DWMHs, but not periventricular WMHs. REGISTRATION: URL: https://www.chictr.org.cn; Unique identifier: ChiCTR2100045869.


Subject(s)
Genetic Predisposition to Disease , Heme Oxygenase-1 , Humans , Heme Oxygenase-1/genetics , Male , Female , Aged , Middle Aged , Polymorphism, Genetic , White Matter/diagnostic imaging , White Matter/pathology , Risk Factors , Magnetic Resonance Imaging , Promoter Regions, Genetic , Leukoencephalopathies/genetics , Leukoencephalopathies/diagnostic imaging , Phenotype
10.
Sheng Li Xue Bao ; 76(2): 215-223, 2024 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-38658371

ABSTRACT

This study aimed to investigate the effects of microtubule associated tumor suppressor 1 (MTUS1) on hemeoxygenase 1 (HMOX1) expression and hemin-induced apoptosis of vascular endothelial cells and its regulatory mechanism. RNA sequencing, RT-qPCR and Western blot were used to assess altered genes of hemin binding proteins, the expression of cAMP response element-binding protein (CREB) and nuclear respiratory factor 2 (NRF2), hemin-induced HMOX1 expression in MTUS1 knockdown human umbilical vein endothelial cells (HUVEC), and the effect of overexpression of CREB and NRF2 on HMOX1 expression in MTUS1 knockdown 293T cells. The effect of MTUS1 or HMOX1 knockdown on hemin-induced apoptosis in HUVEC, and the overexpression of NRF2 on hemin-induced apoptosis in MTUS1 knockdown 293T cells were assayed with CCK8 and Western blot. The results showed that MTUS1 was knocked down significantly in HUVEC by siRNA (P < 0.01), accompanied by decreased HMOX1 expression (P < 0.01). The increased HMOX1 expression induced by hemin was also inhibited by MTUS1 knockdown (P < 0.01). And the apoptosis of HUVEC induced by hemin was amplified by MTUS1 or HMOX1 knockdown (P < 0.01). Moreover the expression of CREB and NRF2 were both inhibited by MTUS1 knockdown in HUVEC (P < 0.01). The decreased HMOX1 regulated by MTUS1 knockdown could be rescued partly by overexpression of NRF2 (P < 0.01), however, not by overexpression of CREB. And the MTUS1 knockdown mediated decreased 293T cells viability induced by hemin could be partly rescued by NRF2 overexpression (P < 0.01). These results suggest that MTUS1 can inhibit hemin-induced apoptosis of HUVEC, and the mechanism maybe related to MTUS1/NRF2/HMOX1 pathway.


Subject(s)
Apoptosis , Heme Oxygenase-1 , Hemin , Human Umbilical Vein Endothelial Cells , NF-E2-Related Factor 2 , Humans , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Knockdown Techniques , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Hemin/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics
11.
Toxicol Ind Health ; 40(6): 312-322, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38590048

ABSTRACT

Previous epidemiologic research has shown that phthalate exposure in pregnant women is related to adverse birth outcomes in a sex-specific manner. However, the biological mechanism of phthalate exposure that causes these birth outcomes remains poorly defined. In this research, we investigated the association between phthalate exposure and placental oxidative stress in a large population-based cohort study, aiming to initially explore the relationship between phthalate exposure and gene expression in placental oxidative stress in a sex-specific manner. Quantitative PCR was performed to measure the expression of placental inflammatory mRNAs (HO-1, HIF1α, and GRP78) in 2469 placentae. The multiple linear regression models were used to investigate the associations between mRNA and urinary phthalate monoesters. Phthalate metabolites monomethyl phthalate (MMP) and mono-n-butyl phthalate (MBP) were positively correlated with higher HIF1α expression in placentae of male fetuses (p < .05). Mono-benzyl phthalate (MBzP) increased the expression of HO-1, HIF1α, and GRP78 in placentae of male fetuses, and mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP) up-regulated the expression of HIF1α and GRP78. Additionally, mono-(2-ethyl-5-oxohexyl) phthalate (MEOHP) was negatively correlated with HO-1, HIF1α, and GRP78 in placentae of female fetuses. Maternal phthalate exposure was associated with oxidative stress variations in placental tissues. The associations were closer in the placentas of male fetuses than in that of female ones. The placenta oxidative stress is worth further investigation as a potential mediator of maternal exposure-induced disease risk in children.


Subject(s)
Biomarkers , Endoplasmic Reticulum Chaperone BiP , Maternal Exposure , Oxidative Stress , Phthalic Acids , Placenta , Humans , Phthalic Acids/toxicity , Phthalic Acids/urine , Female , Oxidative Stress/drug effects , Pregnancy , Male , Placenta/drug effects , Placenta/metabolism , Biomarkers/urine , Prospective Studies , Adult , Maternal Exposure/adverse effects , Sex Factors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Cohort Studies
12.
Arch Oral Biol ; 163: 105965, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38593562

ABSTRACT

OBJECTIVE: Porphyromonas gingivalis (P. gingivalis) is a key etiological agent in periodontitis and functions as a facultative intracellular microorganism and involves many virulence factors. These virulence factors participate in multiple intracellular processes, like ferroptosis, the mechanistic underpinnings remain to be elucidated. Aim of this study was to investigate the effects of virulence factors on the host cells. DESIGN: Human umbilical vein endothelial cells (HUVECs) were treated with 4% paraformaldehyde-fixed P. gingivalis, and subsequent alterations in gene expression were profiled via RNA-seq. Further, the molecules associated with ferroptosis were quantitatively analyzed using qRT-PCR and Western blot. RESULTS: A total of 1125 differentially expressed genes (DEGs) were identified, encompassing 225 upregulated and 900 downregulated. Ferroptosis was conspicuously represented in the kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis, with notable upregulation of Heme oxygenase 1 (HMOX1), Ferritin light chain (FTL), and Solute carrier family 3 member 2 (SLC3A2) and downregulation of Scavenger receptor class A member 5 (SCARA5) and glutaminase (GLS). Random selection of DEGs for validation through qRT-PCR corroborated the RNA-Seq data (R2 = 0.93). Kelch like ECH associated protein 1 (Keap1) protein expression decreased after 4 and 8 h, while NFE2 like bZIP transcription factor 2 (Nrf2) and HMOX1 were elevated, with significant nuclear translocation of Nrf2. CONCLUSIONS: The virulence factors of P. gingivalis may potentially instigating ferroptosis through activation of the Keap1-Nrf2-HMOX1 signaling cascade, in conjunction with modulating the expression of other ferroptosis-associated elements. Further research is necessary to achieve a thorough comprehension of these complex molecular interactions.


Subject(s)
Ferroptosis , Human Umbilical Vein Endothelial Cells , Porphyromonas gingivalis , Virulence Factors , Porphyromonas gingivalis/pathogenicity , Porphyromonas gingivalis/genetics , Ferroptosis/genetics , Humans , Virulence Factors/genetics , Up-Regulation , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Blotting, Western , Down-Regulation , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism
13.
Cell Signal ; 119: 111165, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38583746

ABSTRACT

Emerging evidence suggests that long non-coding RNAs (lncRNAs) play significant roles in renal ischemia reperfusion (RIR) injury. However, the specific mechanisms by which lncRNAs regulate ferroptosis in renal tubular epithelial cells remain largely unknown. The objective of this study was to investigate the biological function of lncRNA heme oxygenase 1 (lnc-HMOX1) in RIR and its potential molecular mechanism. Our findings demonstrated that the expression of HMOX1-related lnc-HMOX1 was reduced in renal tubular epithelial cells treated with hypoxia-reoxygenation (HR). Furthermore, the over-expression of lnc-HMOX1 mitigated ferroptotic injury in renal tubular epithelial cells in vivo and in vitro. Mechanistically, lnc-HMOX1, as a competitive endogenous RNA (ceRNA), promoted the expression of HMOX1 by sponging miR-3587. Furthermore, the inhibition of HMOX1 effectively impeded the aforementioned effects exerted by lnc-HMOX1. Ultimately, the inhibitory or mimic action of miR-3587 reversed the promoting or refraining influence of silenced or over-expressed lnc-HMOX1 on ferroptotic injury during HR. In summary, our findings contribute to a comprehensive comprehension of the mechanism underlying ferroptotic injury mediated by lnc-HMOX1 during RIR. Significantly, we identified a novel lnc-HMOX1-miR-3587-HMOX1 axis, which holds promise as a potential therapeutic target for RIR injury.


Subject(s)
Ferroptosis , Heme Oxygenase-1 , MicroRNAs , RNA, Long Noncoding , Reperfusion Injury , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Ferroptosis/genetics , Mice , Male , Humans , Mice, Inbred C57BL , Kidney/pathology , Kidney/metabolism
14.
Arch Toxicol ; 98(6): 1781-1794, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38573338

ABSTRACT

Doxorubicin (DOX) is one of the most frequently used chemotherapeutic drugs belonging to the class of anthracyclines. However, the cardiotoxic effects of anthracyclines limit their clinical use. Recent studies have suggested that ferroptosis is the main underlying pathogenetic mechanism of DOX-induced cardiomyopathy (DIC). BTB-and-CNC homology 1 (Bach1) acts as a key role in the regulation of ferroptosis. However, the mechanistic role of Bach1 in DIC remains unclear. Therefore, this study aimed to investigate the underlying mechanistic role of Bach1 in DOX-induced cardiotoxicity using the DIC mice in vivo (DOX at cumulative dose of 20 mg/kg) and the DOX-treated H9c2 cardiomyocytes in vitro (1 µM). Our results show a marked upregulation in the expression of Bach1 in the cardiac tissues of the DOX-treated mice and the DOX-treated cardiomyocytes. However, Bach1-/- mice exhibited reduced lipid peroxidation and less severe cardiomyopathy after DOX treatment. Bach1 knockdown protected against DOX-induced ferroptosis in both in vivo and in vitro models. Ferrostatin-1 (Fer-1), a potent inhibitor of ferroptosis, significantly alleviated DOX-induced cardiac damage. However, the cardioprotective effects of Bach1 knockdown were reversed by pre-treatment with Zinc Protoporphyrin (ZnPP), a selective inhibitor of heme oxygenase-1(HO-1). Taken together, these findings demonstrated that Bach1 promoted oxidative stress and ferroptosis through suppressing the expression of HO-1. Therefore, Bach1 may present as a promising new therapeutic target for the prevention and early intervention of DOX-induced cardiotoxicity.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Cardiomyopathies , Doxorubicin , Ferroptosis , Heme Oxygenase-1 , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac , Oxidative Stress , Animals , Ferroptosis/drug effects , Doxorubicin/toxicity , Oxidative Stress/drug effects , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Male , Mice , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Cell Line , Rats , Cardiotoxicity , Antibiotics, Antineoplastic/toxicity , Lipid Peroxidation/drug effects , Protoporphyrins/pharmacology , Signal Transduction/drug effects , Cyclohexylamines , Membrane Proteins , Phenylenediamines
15.
Cancer Lett ; 590: 216826, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38574881

ABSTRACT

Ferroptosis, an iron-dependent regulated cell death caused by excessive lipid peroxide accumulation, has emerged as a promising therapeutic target in various cancers, including non-small cell lung cancer (NSCLC). In this study, we identified the long non-coding RNA RGMB-AS1 as a key regulator of ferroptosis in NSCLC. Mechanistically, RGMB-AS1 interacted with heme oxygenase 1 (HMOX1) and prevented its ubiquitination by the E3 ligase TRC8, leading to increased HMOX1 stability and enhanced ferroptosis. Additionally, RGMB-AS1 bound to the 82-87 amino acid region of N-alpha-acetyltransferase 10 (NAA10), stimulating its acetyltransferase activity and promoting the conversion of acetyl-CoA to HMG-CoA, further contributing to ferroptosis. The RGMB-AS1-HMOX1 and RGMB-AS1-NAA10 axes synergistically inhibited NSCLC growth both in vitro and in vivo. Clinically, low RGMB-AS1 expression was associated with advanced tumor stage and poor overall survival in NSCLC patients. Furthermore, adeno-associated virus-mediated RGMB-AS1 overexpression significantly suppressed tumor growth in mouse xenograft models. Our findings uncover a novel lncRNA-mediated regulatory mechanism of ferroptosis and highlight the potential of RGMB-AS1 as a prognostic biomarker and therapeutic target in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Ferroptosis , Heme Oxygenase-1 , Lung Neoplasms , RNA, Long Noncoding , Ubiquitination , Ferroptosis/genetics , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Mice , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Female , Male , Mice, Nude , A549 Cells , Xenograft Model Antitumor Assays , Cell Proliferation
16.
J Cell Biochem ; 125(5): e30563, 2024 May.
Article in English | MEDLINE | ID: mdl-38591551

ABSTRACT

High glucose (HG)-induced endothelial cell (EC) and smooth muscle cell (SMC) dysfunction is critical in diabetes-associated atherosclerosis. However, the roles of heme oxygenase-1 (HO-1), a stress-response protein, in hemodynamic force-generated shear stress and HG-induced metabolic stress remain unclear. This investigation examined the cellular effects and mechanisms of HO-1 under physiologically high shear stress (HSS) in HG-treated ECs and adjacent SMCs. We found that exposure of human aortic ECs to HSS significantly increased HO-1 expression; however, this upregulation appeared to be independent of adenosine monophosphate-activated protein kinase, a regulator of HO-1. Furthermore, HSS inhibited the expression of HG-induced intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and reactive oxygen species (ROS) production in ECs. In an EC/SMC co-culture, compared with static conditions, subjecting ECs close to SMCs to HSS and HG significantly suppressed SMC proliferation while increasing the expression of physiological contractile phenotype markers, such as α-smooth muscle actin and serum response factor. Moreover, HSS and HG decreased the expression of vimentin, an atherogenic synthetic phenotypic marker, in SMCs. Transfecting ECs with HO-1-specific small interfering (si)RNA reversed HSS inhibition on HG-induced inflammation and ROS production in ECs. Similarly, reversed HSS inhibition on HG-induced proliferation and synthetic phenotype formation were observed in co-cultured SMCs. Our findings provide insights into the mechanisms underlying EC-SMC interplay during HG-induced metabolic stress. Strategies to promote HSS in the vessel wall, such as continuous exercise, or the development of HO-1 analogs and mimics of the HSS effect, could provide an effective approach for preventing and treating diabetes-related atherosclerotic vascular complications.


Subject(s)
Endothelial Cells , Glucose , Heme Oxygenase-1 , Myocytes, Smooth Muscle , Reactive Oxygen Species , Stress, Mechanical , Humans , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Glucose/metabolism , Glucose/pharmacology , Myocytes, Smooth Muscle/metabolism , Reactive Oxygen Species/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Cell Proliferation , Coculture Techniques , Enzyme Activation , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Intercellular Adhesion Molecule-1/metabolism
17.
Ageing Res Rev ; 97: 102308, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615894

ABSTRACT

Aging entails the deterioration of the body's organs, including overall damages at both the genetic and cellular levels. The prevalence of age-related ocular disease such as macular degeneration, dry eye diseases, glaucoma and cataracts is increasing as the world's population ages, imposing a considerable economic burden on individuals and society. The development of age-related ocular disease is predominantly triggered by oxidative stress and chronic inflammatory reaction. Heme oxygenase-1 (HO-1) is a crucial antioxidant that mediates the degradative process of endogenous iron protoporphyrin heme. It catalyzes the rate-limiting step of the heme degradation reaction, and releases the metabolites such as carbon monoxide (CO), ferrous, and biliverdin (BV). The potent scavenging activity of these metabolites can help to defend against peroxides, peroxynitrite, hydroxyl, and superoxide radicals. Other than directly decomposing endogenous oxidizing substances (hemoglobin), HO-1 is also a critical regulator of inflammatory cells and tissue damage, exerting its anti-inflammation activity through regulating complex inflammatory networks. Therefore, promoting HO-1 expression may act as a promising therapeutic strategy for the age-related ocular disease. However, emerging evidences suggest that the overexpression of HO-1 significantly contributes to ferroptosis due to its dual nature. Surplus HO-1 leads to excessive Fe2+ and reactive oxygen species, thereby causing lipid peroxidation and ferroptosis. In this review, we elucidate the role of HO-1 in countering age-related disease, and summarize recent pharmacological trials that targeting HO-1 for disease management. Further refinements of the knowledge would position HO-1 as a novel therapeutic target for age-related ocular disease.


Subject(s)
Aging , Eye Diseases , Heme Oxygenase-1 , Humans , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Aging/metabolism , Aging/genetics , Eye Diseases/metabolism , Animals , Oxidative Stress/physiology
18.
Microbiol Res ; 284: 127727, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636241

ABSTRACT

Heme oxygenase HO-1 (HMOX) regulates cellular inflammation and apoptosis, but its role in regulation of autophagy in Mycoplasma bovis infection is unknown. The objective was to determine how the HO-1/CO- Protein kinase RNA-like endoplasmic reticulum kinase (PERK)-Ca2+- transcription factor EB (TFEB) signaling axis induces autophagy and regulates clearance of M. bovis by bovine mammary epithelial cells (bMECs). M. bovis inhibited autophagy and lysosomal biogenesis in bMECs and suppressed HO-1 protein and expression of related proteins, namely nuclear factor erythroid 2-related factor 2 (Nrf2) and Kelch-like ECH-associated protein 1 (keap1). Activation of HO-1 and its production of carbon monoxide (CO) were required for induction of autophagy and clearance of intracellular M. bovis. Furthermore, when HO-1 was deficient, CO sustained cellular autophagy. HO-1 activation increased intracellular calcium (Ca2+) and cytosolic localization activity of TFEB via PERK. Knockdown of PERK or chelation of intracellular Ca2+ inhibited HO-1-induced M. bovis autophagy and clearance. M. bovis infection affected nuclear localization of lysosomal TFEB in the MiT/TFE transcription factor subfamily, whereas activation of HO-1 mediated dephosphorylation and intranuclear localization of TFEB, promoting autophagy, lysosomal biogenesis and autophagic clearance of M. bovis. Nuclear translocation of TFEB in HO-1 was critical to induce M. bovis transport and survival of infected bMECs. Furthermore, the HO-1/CO-PERK-Ca2+-TFEB signaling axis induced autophagy and M. bovis clearance, providing a viable approach to treat persistent M. bovis infections.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Calcium , Cell Nucleus , Endoplasmic Reticulum , Epithelial Cells , Mammary Glands, Animal , Mycoplasma bovis , Animals , Cattle , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Calcium/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Endoplasmic Reticulum/metabolism , Mammary Glands, Animal/microbiology , Mammary Glands, Animal/metabolism , Cell Nucleus/metabolism , Female , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Mycoplasma Infections/microbiology , Mycoplasma Infections/veterinary , Mycoplasma Infections/metabolism , Lysosomes/metabolism , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Carbon Monoxide/metabolism , Signal Transduction , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics
19.
Int Immunopharmacol ; 132: 111994, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38581992

ABSTRACT

Acute liver failure (ALF) is a potentially fatal disorder characterized by extensive hepatocyte necrosis and rapid decline in liver function. Numerous factors, including oxidative stress, cell death, and inflammatory responses, are associated with its pathogenesis. Endotoxin tolerance (ET) refers to the phenomenon in which the body or cells exhibit low or no response to high-dose lipopolysaccharide (LPS) stimulation after pre-stimulation with low-dose LPS. However, the specific mechanism through which ET regulates LPS/D-galactosamine (D-GalN)-induced ALF remains unclear. An ALF mouse model was established by intraperitoneal injection of D-GalN (400 mg/kg) and LPS (10 mg/kg). A low dose of LPS (0.1 mg/kg/d) was continuously administered to mice for 5 d before modeling to assess the protective effect of ET. The data from this study showed that ET alleviated the inflammatory response in mice with LPS/D-GalN-induced ALF. ET inhibited LPS-induced oxidative damage and pyroptosis in macrophages in vitro. RNA sequencing analysis showed that the NF-κB/NLRP3 pathway was linked to the anti-inflammatory and antioxidative effects of ET. Furthermore, using western blot, RT-qPCR, and immunofluorescence, we verified that ET inhibited the NF-κB/NLRP3 pathway and triggered the Nrf2/HO-1 signaling pathway to attenuate oxidative stress and cell pyroptosis. Sirt1 knockdown reversed this protective effect. In summary, our research elucidates that ET prevents ALF advancement by upregulating Sirt1 levels, triggering the Nrf2/HO-1 signaling axis, and suppressing the NF-κB/NLRP3 signaling cascade to inhibit oxidative stress and cell pyroptosis. Our results provide a mechanistic explanation for the protective effect of ET against ALF.


Subject(s)
Galactosamine , Lipopolysaccharides , Liver Failure, Acute , Signal Transduction , Animals , Male , Mice , Disease Models, Animal , Endotoxins/toxicity , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Immune Tolerance/drug effects , Liver/drug effects , Liver/pathology , Liver/metabolism , Liver/immunology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/immunology , Liver Failure, Acute/metabolism , Liver Failure, Acute/drug therapy , Macrophages/drug effects , Macrophages/immunology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/drug effects , Pyroptosis/drug effects , Signal Transduction/drug effects , Sirtuin 1/metabolism , Sirtuin 1/genetics
20.
Ecotoxicol Environ Saf ; 277: 116314, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38642409

ABSTRACT

Fine particulate matter (PM2.5) has been extensively implicated in the pathogenesis of neurodevelopmental disorders, but the underlying mechanism remains unclear. Recent studies have revealed that PM2.5 plays a role in regulating iron metabolism and redox homeostasis in the brain, which is closely associated with ferroptosis. In this study, the role and underlying mechanism of ferroptosis in PM2.5-induced neurotoxicity were investigated in mice, primary hippocampal neurons, and HT22 cells. Our findings demonstrated that exposure to PM2.5 could induce abnormal behaviors, neuroinflammation, and neuronal loss in the hippocampus of mice. These effects may be attributed to ferroptosis induced by PM2.5 exposure in hippocampal neurons. RNA-seq analysis revealed that the upregulation of iron metabolism-related protein Heme Oxygenase 1 (HO-1) and the activation of mitophagy might play key roles in PM2.5-induced ferroptosis in HT22 cells. Subsequent in vitro experiments showed that PM2.5 exposure significantly upregulated HO-1 in primary hippocampal neurons and HT22 cells. Moreover, PM2.5 exposure activated mitophagy in HT22 cells, leading to the loss of mitochondrial membrane potential, alterations in the expression of autophagy-related proteins LC3, P62, and mTOR, as well as an increase in mitophagy-related protein PINK1 and PARKIN. As a heme-degradation enzyme, the upregulation of HO-1 promotes the release of excess iron, genetically inhibiting the upregulation of HO-1 in HT22 cells could prevent both PM2.5-induced mitophagy and ferroptosis. Furthermore, pharmacological inhibition of mitophagy in HT22 cells reduced levels of ferrous ions and lipid peroxides, thereby preventing ferroptosis. Collectively, this study demonstrates that HO-1 mediates PM2.5-induced mitophagy-dependent ferroptosis in hippocampal neurons, and inhibiting mitophagy or ferroptosis may be a key therapeutic target to ameliorate neurotoxicity following PM2.5 exposure.


Subject(s)
Ferroptosis , Heme Oxygenase-1 , Hippocampus , Mitophagy , Neurons , Particulate Matter , Up-Regulation , Animals , Particulate Matter/toxicity , Ferroptosis/drug effects , Mitophagy/drug effects , Hippocampus/drug effects , Hippocampus/pathology , Neurons/drug effects , Neurons/pathology , Mice , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Up-Regulation/drug effects , Male , Mice, Inbred C57BL , Air Pollutants/toxicity , Membrane Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...