Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.548
Filter
1.
Genes (Basel) ; 15(6)2024 May 28.
Article in English | MEDLINE | ID: mdl-38927642

ABSTRACT

Chronic granulomatous disease (CGD) is an inherited immunodeficiency disease mainly caused by mutations in the X-linked CYBB gene that abrogate reactive oxygen species (ROS) production in phagocytes and microbial defense. Gene repair using the CRISPR/Cas9 system in hematopoietic stem and progenitor cells (HSPCs) is a promising technology for therapy for CGD. To support the establishment of efficient and safe gene therapies for CGD, we generated a mouse model harboring a patient-derived mutation in the CYBB gene. Our CybbC517del mouse line shows the hallmarks of CGD and provides a source for Cybb-deficient HSPCs that can be used to evaluate gene-therapy approaches in vitro and in vivo. In a setup using Cas9 RNPs and an AAV repair vector in HSPCs, we show that the mutation can be repaired in 19% of treated cells and that treatment restores ROS production by macrophages. In conclusion, our CybbC517del mouse line provides a new platform for refining and evaluating novel gene therapies and studying X-CGD pathophysiology.


Subject(s)
CRISPR-Cas Systems , Disease Models, Animal , Genetic Therapy , Granulomatous Disease, Chronic , NADPH Oxidase 2 , Granulomatous Disease, Chronic/therapy , Granulomatous Disease, Chronic/genetics , Animals , Genetic Therapy/methods , Mice , NADPH Oxidase 2/genetics , Reactive Oxygen Species/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Macrophages/metabolism , Mutation
2.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892109

ABSTRACT

Astronauts on exploratory missions will be exposed to galactic cosmic rays (GCR), which can induce neuroinflammation and oxidative stress (OS) and may increase the risk of neurodegenerative disease. As key regulators of inflammation and OS in the CNS, microglial cells may be involved in GCR-induced deficits, and therefore could be a target for neuroprotection. This study assessed the effects of exposure to helium (4He) and iron (56Fe) particles on inflammation and OS in microglia in vitro, to establish a model for testing countermeasure efficacy. Rat microglia were exposed to a single dose of 20 cGy (300 MeV/n) 4He or 2 Gy 56Fe (600 MeV/n), while the control cells were not exposed (0 cGy). Immediately following irradiation, fresh media was applied to the cells, and biomarkers of inflammation (cyclooxygenase-2 [COX-2], nitric oxide synthase [iNOS], phosphorylated IκB-α [pIκB-α], tumor necrosis factor-α [TNFα], and nitrite [NO2-]) and OS (NADPH oxidase [NOX2]) were assessed 24 h later using standard immunochemical techniques. Results showed that radiation did not increase levels of NO2- or protein levels of COX-2, iNOS, pIκB-α, TNFα, or NOX2 compared to non-irradiated control conditions in microglial cells (p > 0.05). Therefore, microglia in isolation may not be the primary cause of neuroinflammation and OS following exposures to helium or iron GCR particles.


Subject(s)
Biomarkers , Cosmic Radiation , Inflammation , Microglia , Oxidative Stress , Animals , Microglia/metabolism , Microglia/radiation effects , Cosmic Radiation/adverse effects , Oxidative Stress/radiation effects , Rats , Inflammation/metabolism , Inflammation/etiology , Biomarkers/metabolism , Nitric Oxide Synthase Type II/metabolism , Iron/metabolism , Cyclooxygenase 2/metabolism , Helium/pharmacology , Tumor Necrosis Factor-alpha/metabolism , NADPH Oxidase 2/metabolism
3.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38892302

ABSTRACT

Urban air pollution, a significant environmental hazard, is linked to adverse health outcomes and increased mortality across various diseases. This study investigates the neurotoxic effects of particulate matter (PM), specifically PM2.5 and PM10, by examining their role in inducing oxidative stress and subsequent neuronal cell death. We highlight the novel finding that PM increases mitochondrial ROS production via stimulating NOX4 activity, not through its expression level in Neuro-2A cells. Additionally, PMs provoke ROS production via increasing the expression and activity of NOX2 in SH-SY5Y human neuroblastoma cells, implying differential regulation of NOX proteins. This increase in mitochondrial ROS triggers the opening of the mitochondrial permeability transition pore (mPTP), leading to apoptosis through key mediators, including caspase3, BAX, and Bcl2. Notably, the voltage-dependent anion-selective channel 1 (VDAC1) increases at 1 µg/mL of PM2.5, while PM10 triggers an increase from 10 µg/mL. At the same concentration (100 µg/mL), PM2.5 causes 1.4 times higher ROS production and 2.4 times higher NOX4 activity than PM10. The cytotoxic effects induced by PMs were alleviated by NOX inhibitors GKT137831 and Apocynin. In SH-SY5Y cells, both PM types increase ROS and NOX2 levels, leading to cell death, which Apocynin rescues. Variability in NADPH oxidase sources underscores the complexity of PM-induced neurotoxicity. Our findings highlight NOX4-driven ROS and mitochondrial dysfunction, suggesting a potential therapeutic approach for mitigating PM-induced neurotoxicity.


Subject(s)
Apoptosis , Mitochondria , NADPH Oxidase 4 , Neurons , Particulate Matter , Reactive Oxygen Species , Particulate Matter/toxicity , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Cell Line, Tumor , Oxidative Stress/drug effects , Animals , Mice , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics
4.
J Am Heart Assoc ; 13(13): e033558, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38904226

ABSTRACT

BACKGROUND: The incidental use of statins during radiation therapy has been associated with a reduced long-term risk of developing atherosclerotic cardiovascular disease. We examined whether irradiation causes chronic vascular injury and whether short-term administration of statins during and after irradiation is sufficient to prevent chronic injury compared with long-term administration. METHODS AND RESULTS: C57Bl/6 mice were pretreated with pravastatin for 72 hours and then exposed to 12 Gy X-ray head-and-neck irradiation. Pravastatin was then administered either for an additional 24 hours or for 1 year. Carotid arteries were tested for vascular reactivity, altered gene expression, and collagen deposition 1 year after irradiation. Treatment with pravastatin for 24 hours after irradiation reduced the loss of endothelium-dependent vasorelaxation and protected against enhanced vasoconstriction. Expression of markers associated with inflammation (NFκB p65 [phospho-nuclear factor kappa B p65] and TNF-α [tumor necrosis factor alpha]) and with oxidative stress (NADPH oxidases 2 and 4) were lowered and subunits of the voltage and Ca2+ activated K+ BK channel (potassium calcium-activated channel subfamily M alpha 1 and potassium calcium-activated channel subfamily M regulatory beta subunit 1) in the carotid artery were modulated. Treatment with pravastatin for 1 year after irradiation completely reversed irradiation-induced changes. CONCLUSIONS: Short-term administration of pravastatin is sufficient to reduce chronic vascular injury at 1 year after irradiation. Long-term administration eliminates the effects of irradiation. These findings suggest that a prospective treatment strategy involving statins could be effective in patients undergoing radiation therapy. The optimal duration of treatment in humans has yet to be determined.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Mice, Inbred C57BL , Oxidative Stress , Pravastatin , Animals , Pravastatin/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Time Factors , Vasoconstriction/drug effects , Vasoconstriction/radiation effects , Vasodilation/drug effects , Vasodilation/radiation effects , Male , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Tumor Necrosis Factor-alpha/metabolism , Transcription Factor RelA/metabolism , NADPH Oxidases/metabolism , Mice , Radiation Injuries, Experimental/prevention & control , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/drug therapy , Drug Administration Schedule , Carotid Arteries/radiation effects , Carotid Arteries/drug effects , Chronic Disease , Disease Models, Animal , NADPH Oxidase 4
5.
Chem Biol Interact ; 398: 111091, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38825056

ABSTRACT

Investigation of acetaminophen (APAP)-induced liver damage recently indicated the significance of phagocytic NADPH oxidase (NOX)-derived reactive oxygen species (ROS) and ferroptosis in the liver. Here, we focused on phagocytosis by iron-containing erythrocyte-devouring splenic macrophages and explored upstream factors of known APAP hepatotoxic mechanisms in vivo. Splenectomy did not alter hepatic cytochrome P450 (CYP) 2E1 activity or hepatic glutathione (GSH) content. APAP injection into splenectomized mice almost completely suppressed increases in plasma alanine aminotransferase levels and centrilobular hepatic necrosis showing the spleen to be a critical tissue in APAP-induced liver damage. Hepatic GSH was recovered to approximately 50 % content at 8 h. In non-splenectomized mice, liver damage was dramatically suppressed by a sensitive redox probe (DCFH-DA), macrophage-depleting clodronate (CL), and a NOX2 inhibitor. APAP treatment resulted in markedly stronger fluorescence intensity from DCFH-DA due to excessive ROS around splenic macrophages, which was lost upon co-treatment with a CYP inhibitor and CL. Deformed erythrocytes disappeared in mice co-treated with DCFH-DA, CL, the NOX2 inhibitor, and the CYP inhibitor. Simultaneously, these four compounds significantly improved APAP-depleted GSH levels. The CYP inhibitor also prevented the formation of APAP-cell adducts in the blood and spleen. In the spleen, CL co-treatment markedly reduced the number of adducts. Splenic ferrous iron levels were significantly elevated by APAP. Therefore, we demonstrated that splenic macrophages devoured APAP metabolite-erythrocyte adducts and subsequently splenic macrophage-related ROS caused sustained hepatic GSH depletion and excessive erythrocyte deformation around 7 h. Our data indicate in vivo upstream factors of known APAP hepatotoxic mechanisms.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Erythrocytes , Glutathione , Liver , Macrophages , Reactive Oxygen Species , Spleen , Animals , Acetaminophen/toxicity , Reactive Oxygen Species/metabolism , Glutathione/metabolism , Macrophages/metabolism , Macrophages/drug effects , Mice , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Erythrocytes/metabolism , Erythrocytes/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Mice, Inbred C57BL , Splenectomy , Phagocytosis/drug effects , NADPH Oxidase 2/metabolism , Clodronic Acid/pharmacology
6.
J Neuroinflammation ; 21(1): 115, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698374

ABSTRACT

BACKGROUND: Macrophages play a pivotal role in the regulation of Japanese encephalitis (JE), a severe neuroinflammation in the central nervous system (CNS) following infection with JE virus (JEV). Macrophages are known for their heterogeneity, polarizing into M1 or M2 phenotypes in the context of various immunopathological diseases. A comprehensive understanding of macrophage polarization and its relevance to JE progression holds significant promise for advancing JE control and therapeutic strategies. METHODS: To elucidate the role of NADPH oxidase-derived reactive oxygen species (ROS) in JE progression, we assessed viral load, M1 macrophage accumulation, and cytokine production in WT and NADPH oxidase 2 (NOX2)-deficient mice using murine JE model. Additionally, we employed bone marrow (BM) cell-derived macrophages to delineate ROS-mediated regulation of macrophage polarization by ROS following JEV infection. RESULTS: NOX2-deficient mice exhibited increased resistance to JE progression rather than heightened susceptibility, driven by the regulation of macrophage polarization. These mice displayed reduced viral loads in peripheral lymphoid tissues and the CNS, along with diminished infiltration of inflammatory cells into the CNS, thereby resulting in attenuated neuroinflammation. Additionally, NOX2-deficient mice exhibited enhanced JEV-specific Th1 CD4 + and CD8 + T cell responses and increased accumulation of M1 macrophages producing IL-12p40 and iNOS in peripheral lymphoid and inflamed extraneural tissues. Mechanistic investigations revealed that NOX2-deficient macrophages displayed a more pronounced differentiation into M1 phenotypes in response to JEV infection, thereby leading to the suppression of viral replication. Importantly, the administration of H2O2 generated by NOX2 was shown to inhibit M1 macrophage polarization. Finally, oral administration of the ROS scavenger, butylated hydroxyanisole (BHA), bolstered resistance to JE progression and reduced viral loads in both extraneural tissues and the CNS, along with facilitated accumulation of M1 macrophages. CONCLUSION: In light of our results, it is suggested that ROS generated by NOX2 play a role in undermining the control of JEV replication within peripheral extraneural tissues, primarily by suppressing M1 macrophage polarization. Subsequently, this leads to an augmentation in the viral load invading the CNS, thereby facilitating JE progression. Hence, our findings ultimately underscore the significance of ROS-mediated macrophage polarization in the context of JE progression initiated JEV infection.


Subject(s)
Macrophages , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , Animals , Mice , Macrophages/metabolism , Macrophages/immunology , Macrophages/virology , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Encephalitis, Japanese/immunology , Reactive Oxygen Species/metabolism , Encephalitis Virus, Japanese , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/virology , Cell Polarity/drug effects , Cell Polarity/physiology
7.
PLoS One ; 19(5): e0289854, 2024.
Article in English | MEDLINE | ID: mdl-38771750

ABSTRACT

INTRODUCTION: Recent research suggests that endothelial activation plays a role in coronavirus disease 2019 (COVID-19) pathogenesis by promoting a pro-inflammatory state. However, the mechanism by which the endothelium is activated in COVID-19 remains unclear. OBJECTIVE: To investigate the mechanism by which COVID-19 activates the pulmonary endothelium and drives pro-inflammatory phenotypes. HYPOTHESIS: The "inflammatory load or burden" (cytokine storm) of the systemic circulation activates endothelial NADPH oxidase 2 (NOX2) which leads to the production of reactive oxygen species (ROS) by the pulmonary endothelium. Endothelial ROS subsequently activates pro-inflammatory pathways. METHODS: The inflammatory burden of COVID-19 on the endothelial network, was recreated in vitro, by exposing human pulmonary microvascular endothelial cells (HPMVEC) to media supplemented with serum from COVID-19 affected individuals (sera were acquired from patients with COVID-19 infection that eventually died. Sera was isolated from blood collected at admission to the Intensive Care Unit of the Hospital of the University of Pennsylvania). Endothelial activation, inflammation and cell death were assessed in HPMVEC treated with serum either from patients with COVID-19 or from healthy individuals. Activation was monitored by measuring NOX2 activation (Rac1 translocation) and ROS production; inflammation (or appearance of a pro-inflammatory phenotype) was monitored by measuring the induction of moieties such as intercellular adhesion molecule (ICAM-1), P-selectin and the NLRP3 inflammasome; cell death was measured via SYTOX™ Green assays. RESULTS: Endothelial activation (i.e., NOX2 activation and subsequent ROS production) and cell death were significantly higher in the COVID-19 model than in healthy samples. When HPMVEC were pre-treated with the novel peptide PIP-2, which blocks NOX2 activation (via inhibition of Ca2+-independent phospholipase A2, aiPLA2), significant abrogation of ROS was observed. Endothelial inflammation and cell death were also significantly blunted. CONCLUSIONS: The endothelium is activated during COVID-19 via cytokine storm-driven NOX2-ROS activation, which causes a pro-inflammatory phenotype. The concept of endothelial NOX2-ROS production as a unifying pathophysiological axis in COVID-19 raises the possibility of using PIP-2 to maintain vascular health.


Subject(s)
COVID-19 , Endothelial Cells , NADPH Oxidase 2 , Reactive Oxygen Species , SARS-CoV-2 , Signal Transduction , Humans , COVID-19/metabolism , Reactive Oxygen Species/metabolism , Endothelial Cells/metabolism , SARS-CoV-2/physiology , NADPH Oxidase 2/metabolism , Endothelium, Vascular/metabolism , Lung/pathology , Lung/metabolism , Lung/virology , Lung/blood supply , Peptides/metabolism , Intercellular Adhesion Molecule-1/metabolism
8.
G3 (Bethesda) ; 14(6)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38696730

ABSTRACT

Reactive oxygen species are important effectors and modifiers of the acute inflammatory response, recruiting phagocytes including neutrophils to sites of tissue injury. In turn, phagocytes such as neutrophils are both consumers and producers of reactive oxygen species. Phagocytes including neutrophils generate reactive oxygen species in an oxidative burst through the activity of a multimeric phagocytic nicotinamide adenine dinucleotide phosphate oxidase complex. Mutations in the NOX2/CYBB (previously gp91phox) nicotinamide adenine dinucleotide phosphate oxidase subunit are the commonest cause of chronic granulomatous disease, a disease characterized by infection susceptibility and an inflammatory phenotype. To model chronic granulomatous disease, we made a nox2/cybb zebrafish (Danio rerio) mutant and demonstrated it to have severely impaired myeloid cell reactive oxygen species production. Reduced early survival of nox2 mutant embryos indicated an essential requirement for nox2 during early development. In nox2/cybb zebrafish mutants, the dynamics of initial neutrophil recruitment to both mild and severe surgical tailfin wounds was normal, suggesting that excessive neutrophil recruitment at the initiation of inflammation is not the primary cause of the "sterile" inflammatory phenotype of chronic granulomatous disease patients. This nox2 zebrafish mutant adds to existing in vivo models for studying reactive oxygen species function in myeloid cells including neutrophils in development and disease.


Subject(s)
Mutation , Myeloid Cells , NADPH Oxidase 2 , Reactive Oxygen Species , Zebrafish , Animals , Reactive Oxygen Species/metabolism , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , Myeloid Cells/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Neutrophils/metabolism , Neutrophil Infiltration , Tail , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Granulomatous Disease, Chronic/genetics , Disease Models, Animal
9.
Proc Natl Acad Sci U S A ; 121(23): e2320388121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38805284

ABSTRACT

Essential for reactive oxygen species (EROS) protein is a recently identified molecular chaperone of NOX2 (gp91phox), the catalytic subunit of phagocyte NADPH oxidase. Deficiency in EROS is a recently identified cause for chronic granulomatous disease, a genetic disorder with recurrent bacterial and fungal infections. Here, we report a cryo-EM structure of the EROS-NOX2-p22phox heterotrimeric complex at an overall resolution of 3.56Å. EROS and p22phox are situated on the opposite sides of NOX2, and there is no direct contact between them. EROS associates with NOX2 through two antiparallel transmembrane (TM) α-helices and multiple ß-strands that form hydrogen bonds with the cytoplasmic domain of NOX2. EROS binding induces a 79° upward bend of TM2 and a 48° backward rotation of the lower part of TM6 in NOX2, resulting in an increase in the distance between the two hemes and a shift of the binding site for flavin adenine dinucleotide (FAD). These conformational changes are expected to compromise superoxide production by NOX2, suggesting that the EROS-bound NOX2 is in a protected state against activation. Phorbol myristate acetate, an activator of NOX2 in vitro, is able to induce dissociation of NOX2 from EROS with concurrent increase in FAD binding and superoxide production in a transfected COS-7 model. In differentiated neutrophil-like HL-60, the majority of NOX2 on the cell surface is dissociated with EROS. Further studies are required to delineate how EROS dissociates from NOX2 during its transport to cell surface, which may be a potential mechanism for regulation of NOX2 activation.


Subject(s)
Cryoelectron Microscopy , NADPH Oxidase 2 , NADPH Oxidases , Phagocytes , Humans , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , NADPH Oxidase 2/chemistry , Phagocytes/metabolism , NADPH Oxidases/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/chemistry , Protein Binding , Binding Sites , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/genetics , Models, Molecular , Reactive Oxygen Species/metabolism
10.
Redox Biol ; 73: 103214, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38805973

ABSTRACT

The chaperone protein EROS ("Essential for Reactive Oxygen Species") was recently discovered in phagocytes. EROS was shown to regulate the abundance of the ROS-producing enzyme NADPH oxidase isoform 2 (NOX2) and to control ROS-mediated cell killing. Reactive oxygen species are important not only in immune surveillance, but also modulate physiological signaling responses in multiple tissues. The roles of EROS have not been previously explored in the context of oxidant-modulated cell signaling. Here we show that EROS plays a key role in ROS-dependent signal transduction in vascular endothelial cells. We used siRNA-mediated knockdown and developed CRISPR/Cas9 knockout of EROS in human umbilical vein endothelial cells (HUVEC), both of which cause a significant decrease in the abundance of NOX2 protein, associated with a marked decrease in RAC1, a small G protein that activates NOX2. Loss of EROS also attenuates receptor-mediated hydrogen peroxide (H2O2) and Ca2+ signaling, disrupts cytoskeleton organization, decreases cell migration, and promotes cellular senescence. EROS knockdown blocks agonist-modulated eNOS phosphorylation and nitric oxide (NO●) generation. These effects of EROS knockdown are strikingly similar to the alterations in endothelial cell responses that we previously observed following RAC1 knockdown. Proteomic analyses following EROS or RAC1 knockdown in endothelial cells showed that reduced abundance of these two distinct proteins led to largely overlapping effects on endothelial biological processes, including oxidoreductase, protein phosphorylation, and endothelial nitric oxide synthase (eNOS) pathways. These studies demonstrate that EROS plays a central role in oxidant-modulated endothelial cell signaling by modulating NOX2 and RAC1.


Subject(s)
Human Umbilical Vein Endothelial Cells , NADPH Oxidase 2 , Oxidation-Reduction , Reactive Oxygen Species , Signal Transduction , rac1 GTP-Binding Protein , Humans , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Human Umbilical Vein Endothelial Cells/metabolism , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide/metabolism , Cell Movement , Phosphorylation , Cellular Senescence , Gene Knockdown Techniques
11.
Free Radic Biol Med ; 220: 56-66, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38697489

ABSTRACT

Apart from dopaminergic neurotoxicity, exposure to rotenone, a commonly used insecticide in agriculture, also adversely affects hippocampal and cortical neurons, resulting in cognitive impairments in mice. We recently established a role of microglia-mediated neuroinflammation in rotenone-elicited deficits of cognition, yet the mechanisms remain elusive. Here, we investigated the involvement of NADPH oxidase 2 (NOX2) catalytic subunit gp91phox in rotenone-induced cognitive deficits and the associated mechanisms. Our study demonstrated that rotenone exposure elevated expression of gp91phox and phosphorylation of the NOX2 cytosolic subunit p47phox, along with NADPH depletion in the hippocampus and cortex of mice, indicating NOX2 activation. Specific knockdown of gp91phox in microglia via adeno-associated virus delivery resulted in reduced microglial activation, proinflammatory gene expression and improved learning and memory capacity in rotenone-intoxicated mice. Genetic deletion of gp91phox also reversed rotenone-elicited cognitive dysfunction in mice. Furthermore, microglial gp91phox knockdown attenuated neuronal damage and synaptic loss in mice. This intervention also suppressed iron accumulation, disruption of iron-metabolism proteins and iron-dependent lipid peroxidation and restored the balance of ferroptosis-related parameters, including GPX4, SLC711, PTGS2, and ACSL4 in rotenone-lesioned mice. Intriguingly, pharmacological inhibition of ferroptosis with liproxstatin-1 conferred protection against rotenone-induced neurodegeneration and cognitive dysfunction in mice. In summary, our findings underscored the contribution of microglial gp91phox-dependent neuroinflammation and ferroptosis to learning and memory dysfunction in rotenone-lesioned mice. These results provided valuable insights into the pathogenesis of cognitive deficits associated with pesticide-induced Parkinsonism, suggesting potential therapeutic avenues for intervention.


Subject(s)
Ferroptosis , Memory Disorders , Microglia , NADPH Oxidase 2 , Neuroinflammatory Diseases , Rotenone , Animals , Mice , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Microglia/metabolism , Microglia/pathology , Microglia/drug effects , Rotenone/toxicity , Ferroptosis/drug effects , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/genetics , Memory Disorders/chemically induced , Memory Disorders/metabolism , Memory Disorders/genetics , Memory Disorders/pathology , Male , Mice, Inbred C57BL , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/drug effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Neurons/metabolism , Neurons/pathology , Neurons/drug effects , Mice, Knockout
12.
Free Radic Biol Med ; 220: 179-191, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38704053

ABSTRACT

Sepsis is a systemic inflammatory response syndrome caused by the invasion of pathogenic microorganisms. Despite major advances in diagnosis and technology, morbidity and mortality remain high. The level of neutrophil extracellular traps (NETs) is closely associated with the progression and prognosis of sepsis, suggesting the regulation of NET formation as a new strategy in sepsis treatment. Owing to its pleiotropic effects, atorvastatin, a clinical lipid-lowering drug, affects various aspects of sepsis-related inflammation and immune responses. To align closely with clinical practice, we combined it with imipenem for the treatment of sepsis. In this study, we used a cecum ligation and puncture-induced lung injury mouse model and employed techniques including western blot, immunofluorescence, and enzyme-linked immunosorbent assay to measure the levels of NETs and other sepsis-related lung injury indicators. Our findings indicate that atorvastatin effectively inhibited the formation of NETs. When combined with imipenem, it significantly alleviated lung injury, reduced systemic inflammation, and improved the 7-day survival rate of septic mice. Additionally, we explored the inhibitory mechanism of atorvastatin on NET formation in vitro, revealing its potential action through the ERK/NOX2 pathway. Therefore, atorvastatin is a potential immunomodulatory agent that may offer new treatment strategies for patients with sepsis in clinical settings.


Subject(s)
Atorvastatin , Disease Models, Animal , Extracellular Traps , Imipenem , NADPH Oxidase 2 , Sepsis , Animals , Atorvastatin/pharmacology , Extracellular Traps/drug effects , Extracellular Traps/metabolism , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/complications , Sepsis/pathology , Mice , Imipenem/pharmacology , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Lung Injury/drug therapy , Lung Injury/pathology , Lung Injury/metabolism , Male , MAP Kinase Signaling System/drug effects , Neutrophils/metabolism , Neutrophils/drug effects , Neutrophils/pathology , Signal Transduction/drug effects , Humans , Mice, Inbred C57BL , Drug Therapy, Combination
13.
J Neuroinflammation ; 21(1): 105, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649885

ABSTRACT

BACKGROUND: NADPH oxidase (NOX), a primary source of endothelial reactive oxygen species (ROS), is considered a key event in disrupting the integrity of the blood-retinal barrier. Abnormalities in neurovascular-coupled immune signaling herald the loss of ganglion cells in glaucoma. Persistent microglia-driven inflammation and cellular innate immune system dysregulation often lead to deteriorating retinal degeneration. However, the crosstalk between NOX and the retinal immune environment remains unresolved. Here, we investigate the interaction between oxidative stress and neuroinflammation in glaucoma by genetic defects of NOX2 or its regulation via gp91ds-tat. METHODS: Ex vivo cultures of retinal explants from wildtype C57BL/6J and Nox2 -/- mice were subjected to normal and high hydrostatic pressure (Pressure 60 mmHg) for 24 h. In vivo, high intraocular pressure (H-IOP) was induced in C57BL/6J mice for two weeks. Both Pressure 60 mmHg retinas and H-IOP mice were treated with either gp91ds-tat (a NOX2-specific inhibitor). Proteomic analysis was performed on control, H-IOP, and treatment with gp91ds-tat retinas to identify differentially expressed proteins (DEPs). The study also evaluated various glaucoma phenotypes, including IOP, retinal ganglion cell (RGC) functionality, and optic nerve (ON) degeneration. The superoxide (O2-) levels assay, blood-retinal barrier degradation, gliosis, neuroinflammation, enzyme-linked immunosorbent assay (ELISA), western blotting, and quantitative PCR were performed in this study. RESULTS: We found that NOX2-specific deletion or activity inhibition effectively attenuated retinal oxidative stress, immune dysregulation, the internal blood-retinal barrier (iBRB) injury, neurovascular unit (NVU) dysfunction, RGC loss, and ON axonal degeneration following H-IOP. Mechanistically, we unveiled for the first time that NOX2-dependent ROS-driven pro-inflammatory signaling, where NOX2/ROS induces endothelium-derived endothelin-1 (ET-1) overexpression, which activates the ERK1/2 signaling pathway and mediates the shift of microglia activation to a pro-inflammatory M1 phenotype, thereby triggering a neuroinflammatory outburst. CONCLUSIONS: Collectively, we demonstrate for the first time that NOX2 deletion or gp91ds-tat inhibition attenuates iBRB injury and NVU dysfunction to rescue glaucomatous RGC loss and ON axon degeneration, which is associated with inhibition of the ET-1/ERK1/2-transduced shift of microglial cell activation toward a pro-inflammatory M1 phenotype, highlighting NOX2 as a potential target for novel neuroprotective therapies in glaucoma management.


Subject(s)
Blood-Retinal Barrier , Intraocular Pressure , Mice, Inbred C57BL , NADPH Oxidase 2 , Neuroinflammatory Diseases , Animals , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Mice , Blood-Retinal Barrier/pathology , Blood-Retinal Barrier/metabolism , Intraocular Pressure/physiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Mice, Knockout , Cell Proliferation/physiology , MAP Kinase Signaling System/physiology , Neuroglia/metabolism , Neuroglia/pathology , Ocular Hypertension/pathology , Ocular Hypertension/metabolism , Glaucoma/pathology , Glaucoma/metabolism , Oxidative Stress/physiology
14.
Redox Biol ; 72: 103151, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593631

ABSTRACT

Salmonella infection entails a cascade of attacks and defence measures. After breaching the intestinal epithelial barrier, Salmonella is phagocytosed by macrophages, where the bacteria encounter multiple stresses, to which it employs relevant countermeasures. Our study shows that, in Salmonella, the polyamine spermidine activates a stress response mechanism by regulating critical antioxidant genes. Salmonella Typhimurium mutants for spermidine transport and synthesis cannot mount an antioxidative response, resulting in high intracellular ROS levels. These mutants are also compromised in their ability to be phagocytosed by macrophages. Furthermore, it regulates a novel enzyme in Salmonella, Glutathionyl-spermidine synthetase (GspSA), which prevents the oxidation of proteins in E. coli. Moreover, the spermidine mutants and the GspSA mutant show significantly reduced survival in the presence of hydrogen peroxide in vitro and reduced organ burden in the mouse model of Salmonella infection. Conversely, in macrophages isolated from gp91phox-/- mice, we observed a rescue in the attenuated fold proliferation previously observed upon infection. We found that Salmonella upregulates polyamine biosynthesis in the host through its effectors from SPI-1 and SPI-2, which addresses the attenuated proliferation observed in spermidine transport mutants. Thus, inhibition of this pathway in the host abrogates the proliferation of Salmonella Typhimurium in macrophages. From a therapeutic perspective, inhibiting host polyamine biosynthesis using an FDA-approved chemopreventive drug, D, L-α-difluoromethylornithine (DFMO), reduces Salmonella colonisation and tissue damage in the mouse model of infection while enhancing the survival of infected mice. Therefore, our work provides a mechanistic insight into the critical role of spermidine in stress resistance of Salmonella. It also reveals a bacterial strategy in modulating host metabolism to promote their intracellular survival and shows the potential of DFMO to curb Salmonella infection.


Subject(s)
Bacterial Proteins , Macrophages , Membrane Proteins , NADPH Oxidase 2 , Reactive Oxygen Species , Salmonella typhimurium , Spermidine , Animals , Salmonella typhimurium/metabolism , Salmonella typhimurium/drug effects , Spermidine/metabolism , Mice , Macrophages/microbiology , Macrophages/metabolism , Macrophages/drug effects , Reactive Oxygen Species/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Polyamines/metabolism , Phagocytosis/drug effects , Salmonella Infections/microbiology , Salmonella Infections/metabolism , NADPH Oxidases/metabolism , NADPH Oxidases/genetics , Host-Pathogen Interactions , Spermidine Synthase/metabolism , Spermidine Synthase/genetics , Oxidative Stress/drug effects
15.
Biochemistry ; 63(9): 1097-1106, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38669178

ABSTRACT

As a key component for NADPH oxidase 2 (NOX2) activation, the peripheral membrane protein p47phox translocates a cytosolic activating complex to the membrane through its PX domain. This study elucidates a potential regulatory mechanism of p47phox recruitment and NOX2 activation by inositol hexaphosphate (IP6). Through NMR, fluorescence polarization, and FRET experimental results, IP6 is shown to be capable of breaking the lipid binding and membrane anchoring events of p47phox-PX with low micromolar potency. Other phosphorylated inositol species such as IP5(1,3,4,5,6), IP4(1,3,4,5), and IP3(1,3,4) show weaker binding and no ability to inhibit lipid interactions in physiological concentration ranges. The low micromolar potency of IP6 inhibition of the p47phox membrane anchoring suggests that physiologically relevant concentrations of IP6 serve as regulators, as seen in other membrane anchoring domains. The PX domain of p47phox is known to be promiscuous to a variety of phosphatidylinositol phosphate (PIP) lipids, and this regulation may help target the domain only to the membranes most highly enriched with the highest affinity PIPs, such as the phagosomal membrane, while preventing aberrant binding to other membranes with high and heterogeneous PIP content, such as the plasma membrane. This study provides insight into a potential novel regulatory mechanism behind NOX2 activation and reveals a role for small-molecule regulation in this important NOX2 activator.


Subject(s)
NADPH Oxidases , Phytic Acid , Phytic Acid/metabolism , Phytic Acid/chemistry , NADPH Oxidases/metabolism , NADPH Oxidases/antagonists & inhibitors , Humans , Cell Membrane/metabolism , NADPH Oxidase 2/metabolism , Phosphatidylinositol Phosphates/metabolism
16.
Cancer Res Commun ; 4(4): 1135-1149, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38598844

ABSTRACT

Preclinical studies imply that surgery triggers inflammation that may entail tumor outgrowth and metastasis. The potential impact of surgery-induced inflammation in human pancreatic cancer is insufficiently explored. This study included 17 patients with periampullary cancer [pancreatic ductal adenocarcinoma (PDAC) n = 14, ampullary carcinoma n = 2, cholangiocarcinoma n = 1] undergoing major pancreatic cancer surgery with curative intent. We analyzed the potential impact of preoperative and postoperative immune phenotypes and function on postoperative survival with >30 months follow-up. The surgery entailed prompt expansion of monocytic myeloid-derived suppressor cells (M-MDSC) that generated NOX2-derived reactive oxygen species (ROS). Strong induction of immunosuppressive M-MDSC after surgery predicted poor postoperative survival and coincided with reduced functionality of circulating natural killer (NK) cells. The negative impact of surgery-induced M-MDSC on survival remained significant in separate analysis of patients with PDAC. M-MDSC-like cells isolated from patients after surgery significantly suppressed NK cell function ex vivo, which was reversed by inhibition of NOX2-derived ROS. High NOX2 subunit expression within resected tumors from patients with PDAC correlated with poor survival whereas high expression of markers of cytotoxic cells associated with longer survival. The surgery-induced myeloid inflammation was recapitulated in vivo in a murine model of NK cell-dependent metastasis. Surgical stress thus induced systemic accumulation of M-MDSC-like cells and promoted metastasis of NK cell-sensitive tumor cells. Genetic or pharmacologic suppression of NOX2 reduced surgery-induced inflammation and distant metastasis in this model. We propose that NOX2-derived ROS generated by surgery-induced M-MDSC may be targeted for improved outcome after pancreatic cancer surgery. SIGNIFICANCE: Pancreatic cancer surgery triggered pronounced accumulation of NOX2+ myeloid-derived suppressor cells that inhibited NK cell function and negatively prognosticated postoperative patient survival. We propose the targeting of M-MDSC as a conceivable strategy to reduce postoperative immunosuppression in pancreatic cancer.


Subject(s)
Myeloid-Derived Suppressor Cells , NADPH Oxidase 2 , Pancreatic Neoplasms , Reactive Oxygen Species , Female , Humans , Male , Carcinoma, Pancreatic Ductal/surgery , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/mortality , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/immunology , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Pancreatic Neoplasms/surgery , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/mortality , Postoperative Period , Reactive Oxygen Species/metabolism
17.
Pflugers Arch ; 476(6): 1007-1018, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38613695

ABSTRACT

Neutrophil granulocytes play a crucial role in host defense against invading pathogens and in inflammatory diseases. The aim of this study was to elucidate membrane potential dynamics during the initial phase of neutrophil activation and its relation to migration and production of reactive oxygen species (ROS). We performed ROS production measurements of neutrophils from healthy C57BL/6J mice after TNFα-priming and/or C5a stimulation. The actin cytoskeleton was visualized with fluorescence microscopy. Furthermore, we combined migration assays and measurements of membrane potential dynamics after stimulating unprimed and/or TNFα-primed neutrophils with C5a. We show that C5a has a concentration-dependent effect on ROS production and chemokinetic migration. Chemokinetic migration and chemotaxis are impaired at C5a concentrations that induce ROS production. The actin cytoskeleton of unstimulated and of ROS-producing neutrophils is not distributed in a polarized way. Inhibition of the phagocytic NADPH oxidase NOX2 with diphenyleneiodonium (DPI) leads to a polarized distribution of the actin cytoskeleton and rescues chemokinetic migration of primed and C5a-stimulated neutrophils. Moreover, C5a evokes a pronounced depolarization of the cell membrane potential by 86.6 ± 4.2 mV starting from a resting membrane potential of -74.3 ± 0.7 mV. The C5a-induced depolarization occurs almost instantaneously (within less than one minute) in contrast to the more gradually developing depolarization induced by PMA (lag time of 3-4 min). This initial depolarization is accompanied by a decrease of the migration velocity. Collectively, our results show that stimulation with C5a evokes parallel changes in membrane potential dynamics, neutrophil ROS production and motility. Notably, the amplitude of membrane potential dynamics is comparable to that of excitable cells.


Subject(s)
Complement C5a , Membrane Potentials , Mice, Inbred C57BL , Neutrophils , Reactive Oxygen Species , Animals , Neutrophils/metabolism , Neutrophils/drug effects , Neutrophils/physiology , Complement C5a/metabolism , Complement C5a/pharmacology , Reactive Oxygen Species/metabolism , Mice , Membrane Potentials/physiology , NADPH Oxidases/metabolism , Actin Cytoskeleton/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Movement/drug effects , Neutrophil Activation , NADPH Oxidase 2/metabolism
18.
Sci Rep ; 14(1): 6971, 2024 03 23.
Article in English | MEDLINE | ID: mdl-38521855

ABSTRACT

Doxorubicin has been used extensively as a potent anticancer agent, but its clinical use is limited by its cardiotoxicity. However, the underlying mechanisms remain to be fully elucidated. In this study, we tested whether NADPH oxidase 2 (Nox2) mediates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy, resulting in cardiac atrophy and dysfunction in doxorubicin-induced heart failure. Nox2 knockout (KO) and wild-type (WT) mice were randomly assigned to receive a single injection of doxorubicin (15 mg/kg, i.p.) or saline. WT doxorubicin mice exhibited the decreases in survival rate, left ventricular (LV) wall thickness and LV fractional shortening and the increase in the lung wet-to-dry weight ratio 1 week after the injections. These alterations were attenuated in Nox2 KO doxorubicin mice. In WT doxorubicin mice, myocardial oxidative stress was increased, myocardial noradrenergic nerve fibers were reduced, myocardial expression of PGP9.5, GAP43, tyrosine hydroxylase and norepinephrine transporter was decreased, and these changes were prevented in Nox2 KO doxorubicin mice. Myocyte autophagy was increased and myocyte size was decreased in WT doxorubicin mice, but not in Nox2 KO doxorubicin mice. Nox2 mediates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy-both of which contribute to cardiac atrophy and failure after doxorubicin treatment.


Subject(s)
Cardiomyopathies , Myocytes, Cardiac , NADPH Oxidase 2 , Animals , Mice , Autophagy , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Doxorubicin/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , Oxidative Stress , Sympathectomy
19.
Kidney Int ; 105(6): 1239-1253, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38431216

ABSTRACT

Intestinal microbiota and their metabolites affect systemic inflammation and kidney disease outcomes. Here, we investigated the key metabolites associated with the acute kidney injury (AKI)-to chronic kidney disease (CKD) transition and the effect of antibiotic-induced microbiota depletion (AIMD) on this transition. In 61 patients with AKI, 59 plasma metabolites were assessed to determine the risk of AKI-to-CKD transition. An AKI-to-CKD transition murine model was established four weeks after unilateral ischemia-reperfusion injury (IRI) to determine the effects of AIMD on the gut microbiome, metabolites, and pathological responses related to CKD transition. Human proximal tubular epithelial cells were challenged with CKD transition-related metabolites, and inhibitory effects of NADPH oxidase 2 (NOX2) signals were tested. Based on clinical metabolomics, plasma trimethylamine N-oxide (TMAO) was associated with a significantly increased risk for AKI-to-CKD transition [adjusted odds ratio 4.389 (95% confidence interval 1.106-17.416)]. In vivo, AIMD inhibited a unilateral IRI-induced increase in TMAO, along with a decrease in apoptosis, inflammation, and fibrosis. The expression of NOX2 and oxidative stress decreased after AIMD. In vitro, TMAO induced fibrosis with NOX2 activation and oxidative stress. NOX2 inhibition successfully attenuated apoptosis, inflammation, and fibrosis with suppression of G2/M arrest. NOX2 inhibition (in vivo) showed improvement in pathological changes with a decrease in oxidative stress without changes in TMAO levels. Thus, TMAO is a key metabolite associated with the AKI-to-CKD transition, and NOX2 activation was identified as a key regulator of TMAO-related AKI-to-CKD transition both in vivo and in vitro.


Subject(s)
Acute Kidney Injury , Anti-Bacterial Agents , Disease Models, Animal , Gastrointestinal Microbiome , Methylamines , NADPH Oxidase 2 , Oxidative Stress , Renal Insufficiency, Chronic , Acute Kidney Injury/chemically induced , Acute Kidney Injury/microbiology , Acute Kidney Injury/prevention & control , Acute Kidney Injury/pathology , Acute Kidney Injury/drug therapy , Methylamines/blood , Methylamines/metabolism , Animals , NADPH Oxidase 2/antagonists & inhibitors , NADPH Oxidase 2/metabolism , Humans , Male , Gastrointestinal Microbiome/drug effects , Renal Insufficiency, Chronic/microbiology , Renal Insufficiency, Chronic/complications , Middle Aged , Mice , Oxidative Stress/drug effects , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Mice, Inbred C57BL , Female , Reperfusion Injury/prevention & control , Aged , Apoptosis/drug effects , Disease Progression
20.
J Biol Chem ; 300(4): 107130, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38432630

ABSTRACT

The actin cytoskeleton and reactive oxygen species (ROS) both play crucial roles in various cellular processes. Previous research indicated a direct interaction between two key components of these systems: the WAVE1 subunit of the WAVE regulatory complex (WRC), which promotes actin polymerization and the p47phox subunit of the NADPH oxidase 2 complex (NOX2), which produces ROS. Here, using carefully characterized recombinant proteins, we find that activated p47phox uses its dual Src homology 3 domains to bind to multiple regions within the WAVE1 and Abi2 subunits of the WRC, without altering WRC's activity in promoting Arp2/3-mediated actin polymerization. Notably, contrary to previous findings, p47phox uses the same binding pocket to interact with both the WRC and the p22phox subunit of NOX2, albeit in a mutually exclusive manner. This observation suggests that when activated, p47phox may separately participate in two distinct processes: assembling into NOX2 to promote ROS production and engaging with WRC to regulate the actin cytoskeleton.


Subject(s)
NADPH Oxidase 2 , Wiskott-Aldrich Syndrome Protein Family , Humans , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actins/metabolism , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , NADPH Oxidases/metabolism , NADPH Oxidases/genetics , Protein Binding , Reactive Oxygen Species/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , Wiskott-Aldrich Syndrome Protein Family/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Binding Sites
SELECTION OF CITATIONS
SEARCH DETAIL
...