Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.538
Filter
1.
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
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.
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
4.
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
5.
Sci Rep ; 14(1): 23527, 2024 10 09.
Article in English | MEDLINE | ID: mdl-39384968

ABSTRACT

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


Subject(s)
Apoptosis , Doxorubicin , Heat-Shock Proteins , Oxidative Stress , Animals , Doxorubicin/adverse effects , Oxidative Stress/drug effects , Apoptosis/drug effects , Mice , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Male , Kidney/metabolism , Kidney/drug effects , Kidney/pathology , Molecular Chaperones/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Signal Transduction/drug effects , Superoxide Dismutase/metabolism
6.
Mol Biol Rep ; 51(1): 976, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259343

ABSTRACT

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


Subject(s)
Cellular Senescence , Fibroblasts , Gingiva , NF-E2-Related Factor 2 , Porphyromonas gingivalis , Reactive Oxygen Species , Sirtuins , Humans , Porphyromonas gingivalis/pathogenicity , Gingiva/microbiology , Gingiva/metabolism , Fibroblasts/metabolism , Sirtuins/metabolism , Sirtuins/genetics , Male , Female , Adult , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species/metabolism , Lipopolysaccharides/pharmacology , Periodontitis/microbiology , Periodontitis/metabolism , Tumor Suppressor Protein p53/metabolism , Middle Aged , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Interleukin-6/metabolism , Interleukin-8/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics
7.
Sci Rep ; 14(1): 21425, 2024 09 13.
Article in English | MEDLINE | ID: mdl-39271809

ABSTRACT

Oxidative stress in adipose tissue may alter the secretion pattern of adipocytokines and potentially promote atherosclerosis. However, the therapeutic role of hydrogen in adipose tissue under oxidative stress remains unclear. In this study, subcutaneous adipose tissue (SCAT) was collected from the mid-thoracic wounds of 12 patients who underwent open-heart surgery with a mid-thoracic incision. The adipose tissue was then immersed in a culture medium dissolved with hydrogen, which was generated using a hydrogen-generating device. The weight of the adipose tissue was measured before and after hydrogenation, and the tissue was immunostained for nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and superoxide dismutase (SOD), which are markers of oxidative stress. The immunostaining results showed that HO-1 and Nrf2 expression levels were significantly decreased in the hydrogenated group, whereas SOD expression levels increased, but did not attain statistical significance. Image analysis of adipose tissue revealed that a reduction in adipocyte size. Furthermore, hydrogenated adipose tissue showed a trend toward increased gene expression levels of adiponectin and decreased gene expression levels of chemerin, an adipocytokine involved in adipogenesis. These results demonstrated the therapeutic potential of hydrogen gas for oxidative stress in adipose tissue and for reducing adipocyte size.


Subject(s)
Adipose Tissue , Hydrogen , Oxidative Stress , Oxidative Stress/drug effects , Humans , Hydrogen/pharmacology , Hydrogen/metabolism , Male , Female , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Middle Aged , Superoxide Dismutase/metabolism , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Aged , Adiponectin/metabolism , Adiponectin/genetics , Adipocytes/metabolism , Adipocytes/drug effects , Subcutaneous Fat/metabolism , Subcutaneous Fat/drug effects , NF-E2-Related Factor 2
8.
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
9.
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
10.
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
11.
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
12.
ACS Infect Dis ; 10(10): 3516-3527, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39283729

ABSTRACT

Fungal keratitis (FK) is a blinding corneal infectious disease. The prognosis is frequently unfavorable due to fungal invasion and an excessive host inflammatory response. Licochalcone A (Lico A) exhibits a broad spectrum of pharmacological activities, encompassing antifungal, anti-inflammatory, antioxidation, and antitumor properties. However, the role of Lico A has not yet been studied in FK. In this study, we discovered that Lico A could disrupt Aspergillus fumigatus (A. fumigatus) biofilms, inhibit fungal growth and adhesion to host cells, induce alterations of hyphal morphology, and impair the cell membrane and cell wall integrity and mitochondrial structure of A. fumigatus. Lico A can alleviate the severity of FK in mice, reduce neutrophil infiltration and fungal load, and significantly decrease the pro-inflammatory cytokines in mouse corneas infected with A. fumigatus. In vitro, we also demonstrated that Lico A increased the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) around the nucleus in human corneal epithelial cells (HCECs) stimulated with A. fumigatus. We verified that the anti-inflammatory effect of Lico A is associated with the activation of the Nrf2/HO-1 axis. These results indicated that Lico A could provide a protective role in A. fumigatus keratitis through its anti-inflammatory and antifungal activities.


Subject(s)
Antifungal Agents , Aspergillosis , Aspergillus fumigatus , Chalcones , Heme Oxygenase-1 , Keratitis , NF-E2-Related Factor 2 , Signal Transduction , Aspergillus fumigatus/drug effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Animals , Keratitis/drug therapy , Keratitis/microbiology , Mice , Signal Transduction/drug effects , Humans , Aspergillosis/drug therapy , Aspergillosis/microbiology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Chalcones/pharmacology , Chalcones/chemistry , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Biofilms/drug effects , Disease Models, Animal , Cornea/microbiology , Cornea/drug effects , Female , Cytokines/metabolism
13.
Redox Biol ; 76: 103342, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39265498

ABSTRACT

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


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Ferroptosis , HMGB1 Protein , Lipocalin-2 , NF-E2-Related Factor 2 , NF-E2-Related Factor 2/metabolism , Humans , Animals , Male , Rats , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , HMGB1 Protein/metabolism , Ferroptosis/drug effects , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Female , Lipocalin-2/metabolism , Signal Transduction/drug effects , Aged , Middle Aged , Thrombolytic Therapy , Ischemic Stroke/metabolism , Ischemic Stroke/drug therapy , Ischemic Stroke/pathology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics
14.
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
16.
BMC Cardiovasc Disord ; 24(1): 452, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39192217

ABSTRACT

BACKGROUND: There is increasing evidence implicating hemoglobin/heme and their scavengers in oxidative stress-mediated pathologies, but information is limited in abdominal aortic aneurysm (AAA). METHODS AND RESULTS: In this case-control study, we assessed heme/heme-related markers in 142 men with AAA and 279 men with a normal aortic diameter consecutively recruited from an ultrasound screening program in Sweden. Enzyme-linked immunosorbent assays (ELISAs) were used to measure heme oxygenase-1 (HO-1) and hemopexin (Hpx) plasma levels, colorimetric assays for cell-free heme and whole blood hemoglobin (Hb) levels, and droplet digital PCR (ddPCR) and real-time PCR to determine haptoglobin (Hp) (pheno)type and genotype, respectively. Hpx and heme plasma levels at baseline were elevated, while HO-1 levels were lower in men with AAA (p < 0.001) and were significantly associated with AAA prevalence independently of potential confounders. A combination of heme and HO-1 showed the best diagnostic potential based on the area under the curve (AUC): 0.76, sensitivity: 80%, specificity: 48%. Additionally, when previously described inflammatory biomarker interleukin-6 (IL-6), was added to our model it significantly improved the diagnostic value (AUC: 0.87, sensitivity: 80%, specificity: 79%) compared to IL-6 alone (AUC: 0.73, sensitivity: 80%, specificity: 49%). Finally, Hb (positively) and Hpx (negatively) levels at baseline were associated with AAA growth rate (mm/year), and their combination showed the best prognostic value for discriminating fast and slow-growing AAA (AUC: 0.76, sensitivity: 80%, specificity: 62%). CONCLUSIONS: This study reports the distinct disruption of heme and related markers in both the development and progression of AAA, underscoring their potential in aiding risk stratification and therapeutic strategies.


Subject(s)
Aortic Aneurysm, Abdominal , Biomarkers , Haptoglobins , Heme Oxygenase-1 , Heme , Hemoglobins , Hemopexin , Predictive Value of Tests , Humans , Aortic Aneurysm, Abdominal/blood , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/diagnosis , Male , Biomarkers/blood , Heme Oxygenase-1/blood , Heme Oxygenase-1/genetics , Aged , Case-Control Studies , Haptoglobins/analysis , Middle Aged , Sweden/epidemiology , Hemoglobins/metabolism , Hemoglobins/analysis , Prognosis , Homeostasis , Interleukin-6/blood , Enzyme-Linked Immunosorbent Assay
18.
Poult Sci ; 103(10): 104154, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39137500

ABSTRACT

Lambda-cyhalothrin (LCT) is a common pyrethroid insecticide widely used for ectoparasite control and hygiene pest prevention in poultry and this study aimed to investigate the mechanisms of LCT-induced cardiac injury in chickens. Low, medium, and high-dose LCT exposure models in chickens were established and hematoxylin and eosin (H&E) staining, dihydroethidium (DHE) staining, TUNEL staining, immunofluorescence, biochemical analysis, and gene expression analysis were used to study the effects of LCT exposure on the chicken heart. The results showed that LCT exposure increased the serum levels of creatine kinase (CK) and lactate dehydrogenase (LDH), led to muscle fiber breakage and inflammatory cell infiltration and caused cardiac tissue damage. The DHE staining and biochemical analysis revealed that LCT exposure resulted in the excessive accumulation of ROS, decreased activities/levels of catalase (CAT), total superoxide dismutase (T-SOD), and glutathione (GSH), and increased levels of the oxidative damage marker malondialdehyde (MDA). The TUNEL staining indicated that LCT exposure increased apoptosis possibly through the elevated expression of pro-apoptotic genes in the mitochondrial pathway, the reduced expression of anti-apoptotic genes, the upregulation of pro-inflammatory factors and the downregulation of anti-inflammatory factors. Here, LCT exposure significantly inhibited the expression of genes in the Nrf2/HO-1 pathway and activated the expression of genes in the CYP450 enzyme system. Compared to the low-dose group, the high-dose LCT exposure group showed lower levels of apoptosis and inflammation, possibly related to the low oxidative stress levels mediated by the decreased expression of the CYP450 enzyme system. In conclusion, LCT exposure induces oxidative stress, apoptosis, and inflammation in chicken hearts, which may be associated with the inhibition of the Nrf2/HO-1 pathway and activation of the CYP450 enzyme system. This study provides a theoretical basis for the safer use of insecticides in poultry production.


Subject(s)
Avian Proteins , Chickens , Cytochrome P-450 Enzyme System , Insecticides , NF-E2-Related Factor 2 , Nitriles , Poultry Diseases , Pyrethrins , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Insecticides/toxicity , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/genetics , Poultry Diseases/chemically induced , Poultry Diseases/prevention & control , Avian Proteins/metabolism , Avian Proteins/genetics , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Dose-Response Relationship, Drug , Male , Heart Diseases/veterinary , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Oxidative Stress/drug effects , Signal Transduction/drug effects , Apoptosis/drug effects
19.
Invest Ophthalmol Vis Sci ; 65(10): 37, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39186260

ABSTRACT

Purpose: Metastatic uveal melanoma (UM) treatment is difficult, and effective treatments are urgently needed. We aimed to explore the role of heme oxygenase 1 (HO-1) in UM and provide new therapeutic strategies for UM. Methods: Bioinformatics was used to analyze the relationship between HMOX1 and immunity in UM and other tumors. Cell Counting Kit-8, Western blot, immunofluorescence staining, wound healing, and Transwell assays were used. A subcutaneous transplanted UM tumor model was used in mice to verify the therapeutic effect. Results: In UM, the expression level of HMOX1 was strongly correlated with the immune score and the infiltration level of various immune cells. ZnPP can inhibit the growth of UM cells, promote cell apoptosis, and block the cell cycle at G0/G1 phase in vitro. HO-1 knockout can effectively inhibit the proliferation of UM cells. ZnPP effectively inhibited the growth of UM and promoted the infiltration of CD8+ T cells in a subcutaneous tumor transplantation model. Conclusions: These results indicate that targeting HO-1 in UM has the potential for independent targeted immunotherapy or adjuvant immunotherapy.


Subject(s)
Apoptosis , CD8-Positive T-Lymphocytes , Cell Movement , Cell Proliferation , Heme Oxygenase-1 , Lymphocytes, Tumor-Infiltrating , Melanoma , Uveal Neoplasms , Uveal Neoplasms/pathology , Uveal Neoplasms/immunology , Uveal Neoplasms/metabolism , Animals , Melanoma/pathology , Melanoma/immunology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Mice , CD8-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Humans , Neoplasm Invasiveness , Blotting, Western , Protoporphyrins/pharmacology , Cell Line, Tumor , Mice, Inbred C57BL , Disease Models, Animal
20.
Free Radic Biol Med ; 223: 430-442, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39159887

ABSTRACT

Ferroptosis, a recently identified non-apoptotic form of cell death, is strongly associated with neurological diseases and has emerged as a potential therapeutic target. Nevertheless, the fundamental mechanisms are still predominantly unidentified. In the current investigation, sulfiredoxin-1 (SRXN1) has been identified as a crucial regulator that enhances the susceptibility to ferroptosis in HT-22 mouse hippocampal cells treated with erastin. Utilizing TMT-based proteomics, a significant increase in SRXN1 expression was observed in erastin-exposed HT-22 cells. Efficient amelioration of erastin-induced ferroptosis was achieved via the knockdown of SRXN1, which resulted in the reduction of intracellular Fe2+ levels and reactive oxygen species (ROS) in HT-22 cells. Notably, the activation of Heme Oxygenase-1 (HO-1) was found to be crucial for inducing SRXN1 expression in HT-22 cells upon treatment with erastin. SRXN1 increased intracellular ROS and Fe2+ levels by activating HO-1 expression, which promoted erastin-induced ferroptosis in HT-22 cells. Inhibiting SRXN1 or HO-1 alleviated erastin-induced autophagy in HT-22 cells. Additionally, upregulation of SRXN1 or HO-1 increased the susceptibility of HT-22 cells to ferroptosis, a process that was counteracted by the autophagy inhibitor 3-Methyladenine (3-MA). These results indicate that SRXN1 is a key regulator of ferroptosis, activating the HO-1 protein through cellular redox regulation, ferrous iron accumulation, and autophagy in HT-22 cells. These findings elucidate a novel molecular mechanism of erastin-induced ferroptosis sensitivity and suggest that SRXN1-HO-1-autophagy-dependent ferroptosis serves as a promising treatment approach for neurodegenerative diseases.


Subject(s)
Ferroptosis , Heme Oxygenase-1 , Hippocampus , Neurons , Oxidoreductases Acting on Sulfur Group Donors , Piperazines , Reactive Oxygen Species , Ferroptosis/drug effects , Ferroptosis/genetics , Animals , Mice , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Reactive Oxygen Species/metabolism , Piperazines/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/drug effects , Oxidoreductases Acting on Sulfur Group Donors/genetics , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Cell Line , Iron/metabolism , Gene Expression Regulation/drug effects , Membrane Proteins
SELECTION OF CITATIONS
SEARCH DETAIL