Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 433
Filter
1.
Chem Biol Interact ; 402: 111211, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39197814

ABSTRACT

There is a pressing medical need for improved treatments in skin fibrosis including keloids and hypertrophic scars (HTS). This study aimed to characterize the role of phosphodiesterase 4 (PDE4), specifically PDE4B in fibrotic skin remodeling in vitro and in vivo. In vitro, effects of PDE4A-D (Roflumilast) or PDE4B (siRNA) inhibition on TGFß1-induced myofibroblast differentiation and dedifferentiation were studied in normal (NHDF) and keloid (KF) human dermal fibroblasts. In vivo, the role of PDE4 on HOCl-induced skin fibrosis in mice was addressed in preventive and therapeutic protocols. PDE4B (mRNA, protein) was increased in Keloid > HTS compared to healthy skin and in TGFß-stimulated NHDF and KF. In Keloid > HTS, collagen Iα1, αSMA, TGFß1 and NOX4 mRNA were all elevated compared to healthy skin confirming skin fibrosis. In vitro, inhibition of PDE4A-D and PDE4B similarly prevented TGFß1-induced Smad3 and ERK1/2 phosphorylation and myofibroblast differentiation, elevated NOX4 protein and proliferation in NHDF. PDE4A-D inhibition enabled myofibroblast dedifferentiation and curbed TGFß1-induced reactive oxygen species and fibroblast senescence. In KF PDE4A-D inhibition restrained TGFß1-induced Smad3 and ERK1/2 phosphorylation, myofibroblast differentiation and senescence. Mechanistically, PDE4A-D inhibition rescued from TGFß1-induced loss in PPM1A, a Smad3 phosphatase. In vivo, PDE4 inhibition mitigated HOCl-induced skin fibrosis in mice in preventive and therapeutic protocols. The current study provides novel evidence evolving rationale for PDE4 inhibitors in skin fibrosis (including keloids and HTS) and delivered evidence for a functional role of PDE4B in this fibrotic condition.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Fibroblasts , Fibrosis , Keloid , Phosphodiesterase 4 Inhibitors , Skin , Transforming Growth Factor beta1 , Keloid/pathology , Keloid/metabolism , Humans , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Mice , Phosphodiesterase 4 Inhibitors/pharmacology , Transforming Growth Factor beta1/metabolism , Skin/pathology , Skin/metabolism , Skin/drug effects , Cell Differentiation/drug effects , Male , Cells, Cultured , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/antagonists & inhibitors , NADPH Oxidase 4/genetics , Hypochlorous Acid/metabolism , Reactive Oxygen Species/metabolism , Smad3 Protein/metabolism , Cell Proliferation/drug effects , Female
3.
Clin Transl Sci ; 17(8): e13910, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39143889

ABSTRACT

Ferroptosis is a novel, iron-dependent regulatory cell death mainly caused by an imbalance between the production and degradation of intracellular reactive oxygen species (ROS). Recently, ferroptosis induction has been considered a potential therapeutic approach for hepatocellular carcinoma (HCC). Fibroblast growth factor 21 (FGF21) is a new modulator of ferroptosis; however, the regulatory role of FGF21 in HCC ferroptosis has not been investigated. In this study, we explored the role of FGF21 and its underlying molecular mechanism in the ferroptotic death of HCC cells. We identified Major vault protein (MVP) as a target of FGF21 and revealed that knockdown of MVP inhibited the lipid peroxidation levels of HCC cells by decreasing NADPH oxidase 4 (NOX4, a major source of ROS) transcription, thereby attenuating the effect of FGF21-mediated ferroptosis. On the other hand, MVP overexpression showed the opposite results. Mechanistically, MVP binds to IRF1 and thus interferes with the interaction between IRF1 and the YAP1 promoter, leading to an increase in NOX4 transcription. Importantly, forced expression of IRF1 or downregulation of YAP1 partially reversed the effect of MVP overexpression on HCC ferroptosis. Furthermore, the results in xenograft tumor models suggested that overexpression of MVP can efficiently increase the level of lipid peroxidation in vivo. Taken together, these results provide new insights into the regulatory mechanism of ferroptosis in HCC.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Lipid Peroxidation , Liver Neoplasms , NADPH Oxidase 4 , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Humans , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Animals , Mice , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Reactive Oxygen Species/metabolism , Male
4.
Sci Rep ; 14(1): 18175, 2024 08 06.
Article in English | MEDLINE | ID: mdl-39107431

ABSTRACT

Cytoglobin (CYGB) is a member of the oxygen-binding globin superfamily. In this study we generated stable CYGB overexpressing A375 melanoma cells and performed RNA-sequencing to comprehensively explore the CYGB-dependent transcriptome. Our findings reveal that ectopic expression of CYGB dysregulated multiple cancer-associated genes, including the mTORC1 and AKT/mTOR signaling pathways, which are frequently overactivated in tumors. Moreover, several cancer-associated pathways, such as epithelial-mesenchymal transition (EMT) mediated by CSPG4, were downregulated upon CYGB overexpression. Intriguingly, ectopic expression suggested anti-inflammatory potential of CYGB, as exemplified by downregulation of key inflammasome-associated genes, including NLRP1, CASP1 and CD74, which play pivotal roles in cytokine regulation and inflammasome activation. Consistent with established globin functions, CYGB appears to be involved in redox homeostasis. Furthermore, our study indicates CYGB's association to DNA repair mechanisms and its regulation of NOX4, reinforcing its functional versatility. Additionally, multiple significantly enriched pathways in CYGB overexpressing cells were consistently dysregulated in opposite direction in CYGB depleted cells. Collectively, our RNA-sequencing based investigations illustrate the diverse functions of CYGB in melanoma cells, pointing to its putative roles in cellular protection against oxidative stress, inflammation, and cancer-associated pathways. These findings pave the way for further research into the physiological role of CYGB and its potential as a candidate therapeutic target in melanoma.


Subject(s)
Cytoglobin , Gene Expression Regulation, Neoplastic , Inflammation , Melanoma , Oxidative Stress , Transcriptome , Cytoglobin/metabolism , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Inflammation/genetics , Inflammation/metabolism , Cell Line, Tumor , Signal Transduction , Epithelial-Mesenchymal Transition/genetics , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Gene Expression Profiling
5.
Cell Commun Signal ; 22(1): 393, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118129

ABSTRACT

BACKGROUND: Disruptions in intracellular pH (pHi) homeostasis, causing deviations from the physiological range, can damage renal epithelial cells. However, the existence of an adaptive mechanism to restore pHi to normalcy remains unclear. Early research identified H+ as a critical mediator of ischemic preconditioning (IPC), leading to the concept of acidic preconditioning (AP). This concept proposes that short-term, repetitive acidic stimulation can enhance a cell's capacity to withstand subsequent adverse stress. While AP has demonstrated protective effects in various ischemia-reperfusion (I/R) injury models, its application in kidney injury remains largely unexplored. METHODS: An AP model was established in human kidney (HK2) cells by treating them with an acidic medium for 12 h, followed by a recovery period with a normal medium for 6 h. To induce hypoxia/reoxygenation (H/R) injury, HK2 cells were subjected to hypoxia for 24 h and reoxygenation for 1 h. In vivo, a mouse model of IPC was established by clamping the bilateral renal pedicles for 15 min, followed by reperfusion for 4 days. Conversely, the I/R model involved clamping the bilateral renal pedicles for 35 min and reperfusion for 24 h. Western blotting was employed to evaluate the expression levels of cleaved caspase 3, cleaved caspase 9, NHE1, KIM1, FAK, and NOX4. A pH-sensitive fluorescent probe was used to measure pHi, while a Hemin/CNF microelectrode monitored kidney tissue pH. Immunofluorescence staining was performed to visualize the localization of NHE1, NOX4, and FAK, along with the actin cytoskeleton structure in HK2 cells. Cell adhesion and scratch assays were conducted to assess cell motility. RESULTS: Our findings demonstrated that AP could effectively mitigate H/R injury in HK2 cells. This protective effect and the maintenance of pHi homeostasis by AP involved the upregulation of Na+/H+ exchanger 1 (NHE1) expression and activity. The activity of NHE1 was regulated by dynamic changes in pHi-dependent phosphorylation of Focal Adhesion Kinase (FAK) at Y397. This process was associated with NOX4-mediated reactive oxygen species (ROS) production. Furthermore, AP induced the co-localization of FAK, NOX4, and NHE1 in focal adhesions, promoting cytoskeletal remodeling and enhancing cell adhesion and migration capabilities. CONCLUSIONS: This study provides compelling evidence that AP maintains pHi homeostasis and promotes cytoskeletal remodeling through FAK/NOX4/NHE1 signaling. This signaling pathway ultimately contributes to alleviated H/R injury in HK2 cells.


Subject(s)
Reperfusion Injury , Sodium-Hydrogen Exchanger 1 , Animals , Humans , Male , Mice , Acids/metabolism , Cell Line , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Hydrogen-Ion Concentration , Ischemic Preconditioning , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Phosphorylation , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sodium-Hydrogen Exchanger 1/metabolism , Sodium-Hydrogen Exchanger 1/genetics
6.
BMC Cardiovasc Disord ; 24(1): 409, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103770

ABSTRACT

BACKGROUND: This study evaluated the effects of concurrent isolated training (T) or training combined with the antioxidant N-acetylcysteine (NAC) on cardiac remodeling and oxidative stress in spontaneously hypertensive rats (SHR). METHODS: Six-month-old male SHR were divided into sedentary (S, n = 12), concurrent training (T, n = 13), sedentary supplemented with NAC (SNAC, n = 13), and concurrent training with NAC supplementation (TNAC, n = 14) groups. T and TNAC rats were trained three times a week on a treadmill and ladder; NAC supplemented groups received 120 mg/kg/day NAC in rat chow for eight weeks. Myocardial antioxidant enzyme activity and lipid hydroperoxide concentration were assessed by spectrophotometry. Gene expression of NADPH oxidase subunits Nox2, Nox4, p22 phox, and p47 phox was evaluated by real time RT-PCR. Statistical analysis was performed using ANOVA and Bonferroni or Kruskal-Wallis and Dunn. RESULTS: Echocardiogram showed concentric remodeling in TNAC, characterized by increased relative wall thickness (S 0.40 ± 0.04; T 0.39 ± 0.03; SNAC 0.40 ± 0.04; TNAC 0.43 ± 0.04 *; * p < 0.05 vs T and SNAC) and diastolic posterior wall thickness (S 1.50 ± 0.12; T 1.52 ± 0.10; SNAC 1.56 ± 0.12; TNAC 1.62 ± 0.14 * mm; * p < 0.05 vs T), with improved contractile function (posterior wall shortening velocity: S 39.4 ± 5.01; T 36.4 ± 2.96; SNAC 39.7 ± 3.44; TNAC 41.6 ± 3.57 * mm/s; * p < 0.05 vs T). Myocardial lipid hydroperoxide concentration was lower in NAC treated groups (S 210 ± 48; T 182 ± 43; SNAC 159 ± 33 *; TNAC 110 ± 23 *# nmol/g tissue; * p < 0.05 vs S, # p < 0.05 vs T and SNAC). Nox 2 and p22 phox expression was higher and p47 phox lower in T than S [S 1.37 (0.66-1.66); T 0.78 (0.61-1.04) *; SNAC 1.07 (1.01-1.38); TNAC 1.06 (1.01-1.15) arbitrary units; * p < 0.05 vs S]. NADPH oxidase subunits did not differ between TNAC, SNAC, and S groups. CONCLUSION: N-acetylcysteine supplementation alone reduces oxidative stress in untreated spontaneously hypertensive rats. The combination of N-acetylcysteine and concurrent exercise further decreases oxidative stress. However, the lower oxidative stress does not translate into improved cardiac remodeling and function in untreated spontaneously hypertensive rats.


Subject(s)
Acetylcysteine , Hypertension , NADPH Oxidases , Oxidative Stress , Rats, Inbred SHR , Ventricular Remodeling , Animals , Male , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Ventricular Remodeling/drug effects , Hypertension/physiopathology , Hypertension/drug therapy , Hypertension/metabolism , NADPH Oxidases/metabolism , NADPH Oxidases/genetics , Rats , Antioxidants/pharmacology , Physical Conditioning, Animal , Disease Models, Animal , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Myocardium/metabolism , Myocardium/pathology , Lipid Peroxides/metabolism , Ventricular Function, Left/drug effects , Dietary Supplements , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Hypertrophy, Left Ventricular/metabolism
7.
Free Radic Biol Med ; 223: 296-305, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39069268

ABSTRACT

Striated muscle cells, encompassing cardiac myocytes and skeletal muscle fibers, are fundamental to athletic performance, facilitating blood circulation and coordinated movement through contraction. Despite their distinct functional roles, these muscle types exhibit similarities in cytoarchitecture, protein expression, and excitation-contraction coupling. Both muscle types also undergo molecular remodeling in energy metabolism and cell size in response to acute and repeated exercise stimuli to enhance exercise performance. Reactive oxygen species (ROS) produced by NADPH oxidase (NOX) isoforms 2 and 4 have emerged as signaling molecules that regulate exercise adaptations. This review systematically compares NOX2 and NOX4 expression, regulation, and roles in cardiac and skeletal muscle responses across exercise modalities. We highlight the many gaps in our knowledge and opportunities to let future skeletal muscle research into NOX-dependent mechanisms be inspired by cardiac muscle studies and vice versa. Understanding these processes could enhance the development of exercise routines to optimize human performance and health strategies that capitalize on the advantages of physical activity.


Subject(s)
Adaptation, Physiological , Exercise , Muscle, Skeletal , Myocardium , NADPH Oxidase 2 , Reactive Oxygen Species , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Muscle, Skeletal/enzymology , Myocardium/metabolism , Myocardium/enzymology , Exercise/physiology , Animals , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Reactive Oxygen Species/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Signal Transduction
8.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999931

ABSTRACT

Aging is associated with a decline in the functionality of various cell types, including dermal fibroblasts, which play a crucial role in maintaining skin homeostasis and wound healing. Chronic inflammation and increased reactive oxygen species (ROS) production are hallmark features of aging, contributing to impaired wound healing. MicroRNA-146a (miR-146a) has been implicated as a critical regulator of inflammation and oxidative stress in different cell types, yet its role in aged dermal fibroblasts and its potential relevance to wound healing remains poorly understood. We hypothesize that miR-146a is differentially expressed in aged dermal fibroblasts and that overexpression of miR-146a will decrease aging-induced inflammatory responses and ROS production. Primary dermal fibroblasts were isolated from the skin of 17-week-old (young) and 88-week-old (aged) mice. Overexpression of miR-146a was achieved through miR-146a mimic transfection. ROS were detected using a reliable fluorogenic marker, 2,7-dichlorofluorescin diacetate. Real-time PCR was used to quantify relative gene expression. Our investigation revealed a significant reduction in miR-146a expression in aged dermal fibroblasts compared to their younger counterparts. Moreover, aged dermal fibroblasts exhibited heightened levels of inflammatory responses and increased ROS production. Importantly, the overexpression of miR-146a through miR-146a mimic transfection led to a substantial reduction in inflammatory responses through modulation of the NF-kB pathway in aged dermal fibroblasts. Additionally, the overexpression of miR-146a led to a substantial decrease in ROS production, achieved through the downregulation of NOX4 expression in aged dermal fibroblasts. These findings underscore the pivotal role of miR-146a in mitigating both inflammatory responses and ROS production in aged dermal fibroblasts, highlighting its potential as a therapeutic target for addressing age-related skin wound healing.


Subject(s)
Fibroblasts , Inflammation , MicroRNAs , Reactive Oxygen Species , MicroRNAs/genetics , MicroRNAs/metabolism , Fibroblasts/metabolism , Reactive Oxygen Species/metabolism , Animals , Mice , Inflammation/metabolism , Inflammation/genetics , Inflammation/pathology , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Skin/metabolism , Skin/pathology , Skin/cytology , NF-kappa B/metabolism , Cells, Cultured , Aging/metabolism , Aging/genetics , Oxidative Stress
9.
Int Immunopharmacol ; 139: 112688, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39029227

ABSTRACT

Chondrocytes, known for their metabolic adaptability in response to varying stimuli, play a significant role in osteoarthritis (OA) progression. Glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the pentose phosphate pathway, has recently been found to upregulate in OA chondrocyte. However, the exact role of G6PD in temporomandibular joint osteoarthritis (TMJOA) and its effect on chondrocyte function remains unclear. In present study, we induced OA-like conditions in the rat temporomandibular joint via occlusal disharmony (OD), noting a marked increase in G6PD expression in the condylar cartilage. Our data show that G6PD knockdown in mandibular condylar chondrocytes (MCCs) reduces the expression of catabolic enzymes (e.g., MMP3, MMP13) and inflammatory cytokines (e.g., IL6) induced by IL-1ß. G6PD knockdown also mitigates IL-1ß-induced upregulation of ERK, JNK, and p38 phosphorylation and reduces reactive oxygen species (ROS) levels by decreasing the nicotinamide adenine dinucleotide phosphate (NADPH) and NADPH oxidases 4 (NOX4) mRNA expression. In summary, G6PD appears to regulate the inflammatory state of condylar chondrocytes via the NOX-ROS-MAPK axis, highlighting its potential as a therapeutic target for TMJOA.


Subject(s)
Chondrocytes , Glucosephosphate Dehydrogenase , NADPH Oxidase 4 , Osteoarthritis , Reactive Oxygen Species , Animals , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Chondrocytes/drug effects , Chondrocytes/metabolism , Osteoarthritis/metabolism , Reactive Oxygen Species/metabolism , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase/genetics , Male , Rats , Rats, Sprague-Dawley , Interleukin-1beta/metabolism , Temporomandibular Joint/pathology , Temporomandibular Joint/metabolism , Cells, Cultured , Cytokines/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , MAP Kinase Signaling System/drug effects , Inflammation/metabolism , Signal Transduction , Disease Models, Animal , Humans
10.
J Agric Food Chem ; 72(31): 17444-17454, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39074384

ABSTRACT

Acute kidney injury (AKI), if not well controlled, may progress to chronic kidney disease (CKD). Diosgenin is a natural phytosteroid sapogenin from plants. This study aimed to investigate the mechanistic effects of diosgenin on AKI and AKI related development of CKD. The mouse model of ischemia/reperfusion (I/R)-induced AKI was used, and its progressive changes were followed. Human renal proximal tubular epithelial cells were used, and hypoxia stimulation was applied to mimic the in vivo I/R. Diosgenin, given after renal injury, preserved kidney function, as evidenced by a reduction in serum levels of BUN, creatinine, and UACR in both acute and chronic phases of AKI. Diosgenin alleviated I/R-induced tubular injury and prevented macrophage infiltration and renal fibrosis in AKI mice. Furthermore, diosgenin also mitigated the development of CKD from AKI with reduced renal expression of inflammatory, fibrotic, and epithelial-mesenchymal transition markers. In human renal tubular epithelial cells, diosgenin downregulated the hypoxia-induced oxidative stress and cellular damages that were dependent on the NOX4/p65 signaling pathways. Taken together, diosgenin treatment reduced I/R-induced AKI and ameliorated the progression to CKD from AKI probably by modifying the NOX4/p65 signaling pathways.


Subject(s)
Acute Kidney Injury , Diosgenin , Mice, Inbred C57BL , NADPH Oxidase 4 , Renal Insufficiency, Chronic , Signal Transduction , Diosgenin/pharmacology , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Animals , Humans , Mice , Signal Transduction/drug effects , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Male , Oxidative Stress/drug effects , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Disease Progression , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , Cell Line
11.
Front Immunol ; 15: 1410832, 2024.
Article in English | MEDLINE | ID: mdl-38975335

ABSTRACT

Introduction: Aging increases the risk of atherosclerotic vascular disease and its complications. Macrophages are pivotal in the pathogenesis of vascular aging, driving inflammation and atherosclerosis progression. NOX4 (NADPH oxidase 4) expression increases with age, correlating with mitochondrial dysfunction, inflammation, and atherosclerosis. We hypothesized that the NOX4-dependent mitochondrial oxidative stress promotes aging-associated atherosclerosis progression by causing metabolic dysfunction and inflammatory phenotype switch in macrophages. Methods: We studied atherosclerotic lesion morphology and macrophage phenotype in young (5-month-old) and aged (16-month-old) Nox4 -/-/Apoe -/- and Apoe -/- mice fed Western diet. Results: Young Nox4-/-/Apoe-/- and Apoe-/- mice had comparable aortic and brachiocephalic artery atherosclerotic lesion cross-sectional areas. Aged mice showed significantly increased lesion area compared with young mice. Aged Nox4-/-/Apoe-/- had significantly lower lesion areas than Apoe-/- mice. Compared with Apoe-/- mice, atherosclerotic lesions in aged Nox4-/-/Apoe-/- showed reduced cellular and mitochondrial ROS and oxidative DNA damage, lower necrotic core area, higher collagen content, and decreased inflammatory cytokine expression. Immunofluorescence and flow cytometry analysis revealed that aged Apoe-/- mice had a higher percentage of classically activated pro-inflammatory macrophages (CD38+CD80+) in the lesions. Aged Nox4-/-/Apoe-/- mice had a significantly higher proportion of alternatively activated pro-resolving macrophages (EGR2+/CD163+CD206+) in the lesions, with an increased CD38+/EGR2+ cell ratio compared with Apoe-/- mice. Mitochondrial respiration assessment revealed impaired oxidative phosphorylation and increased glycolytic ATP production in macrophages from aged Apoe-/- mice. In contrast, macrophages from Nox4-/-/Apoe-/- mice were less glycolytic and more aerobic, with preserved basal and maximal respiration and mitochondrial ATP production. Macrophages from Nox4-/-/Apoe-/- mice also had lower mitochondrial ROS levels and reduced IL1ß secretion; flow cytometry analysis showed fewer CD38+ cells after IFNγ+LPS treatment and more EGR2+ cells after IL4 treatment than in Apoe-/- macrophages. In aged Apoe-/- mice, inhibition of NOX4 activity using GKT137831 significantly reduced macrophage mitochondrial ROS and improved mitochondrial function, resulting in decreased CD68+CD80+ and increased CD163+CD206+ lesion macrophage proportion and attenuated atherosclerosis. Discussion: Our findings suggest that increased NOX4 in aging drives macrophage mitochondrial dysfunction, glycolytic metabolic switch, and pro-inflammatory phenotype, advancing atherosclerosis. Inhibiting NOX4 or mitochondrial dysfunction could alleviate vascular inflammation and atherosclerosis, preserving plaque integrity.


Subject(s)
Aging , Atherosclerosis , Macrophages , Mitochondria , NADPH Oxidase 4 , Phenotype , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/etiology , Atherosclerosis/immunology , Mitochondria/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Aging/immunology , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Disease Progression , Mice, Knockout , Oxidative Stress , Inflammation/immunology , Inflammation/metabolism , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Male , Disease Models, Animal , Apolipoproteins E/genetics , Apolipoproteins E/deficiency , Mice, Knockout, ApoE , Metabolic Reprogramming
12.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000202

ABSTRACT

The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) protein plays an essential role in the cisplatin (CDDP)-induced generation of reactive oxygen species (ROS). In this study, we evaluated the suitability of ultrasound-mediated lysozyme microbubble (USMB) cavitation to enhance NOX4 siRNA transfection in vitro and ex vivo. Lysozyme-shelled microbubbles (LyzMBs) were constructed and designed for siNOX4 loading as siNOX4/LyzMBs. We investigated different siNOX4-based cell transfection approaches, including naked siNOX4, LyzMB-mixed siNOX4, and siNOX4-loaded LyzMBs, and compared their silencing effects in CDDP-treated HEI-OC1 cells and mouse organ of Corti explants. Transfection efficiencies were evaluated by quantifying the cellular uptake of cyanine 3 (Cy3) fluorescein-labeled siRNA. In vitro experiments showed that the high transfection efficacy (48.18%) of siNOX4 to HEI-OC1 cells mediated by US and siNOX4-loaded LyzMBs significantly inhibited CDDP-induced ROS generation to almost the basal level. The ex vivo CDDP-treated organ of Corti explants of mice showed an even more robust silencing effect of the NOX4 gene in the siNOX4/LyzMB groups treated with US sonication than without US sonication, with a marked abolition of CDDP-induced ROS generation and cytotoxicity. Loading of siNOX4 on LyzMBs can stabilize siNOX4 and prevent its degradation, thereby enhancing the transfection and silencing effects when combined with US sonication. This USMB-derived therapy modality for alleviating CDDP-induced ototoxicity may be suitable for future clinical applications.


Subject(s)
Cisplatin , Hair Cells, Auditory , Microbubbles , Muramidase , NADPH Oxidase 4 , Ototoxicity , Reactive Oxygen Species , Cisplatin/pharmacology , Animals , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Mice , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Reactive Oxygen Species/metabolism , Ototoxicity/genetics , Muramidase/genetics , RNA, Small Interfering/genetics , Ultrasonic Waves , Gene Knockdown Techniques , Cell Line
13.
J Vet Sci ; 25(4): e49, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38910308

ABSTRACT

IMPORTANCE: Endochondral ossification plays an important role in skeletal development. Recent studies have suggested a link between increased intracellular reactive oxygen species (ROS) and skeletal disorders. Moreover, previous studies have revealed that increasing the levels of myeloperoxidase (MPO) and osteopontin (OPN) while inhibiting NADPH oxidase 4 (NOX4) can enhance bone growth. This investigation provides further evidence by showing a direct link between NOX4 and MPO, OPN in bone function. OBJECTIVE: This study investigates NOX4, an enzyme producing hydrogen peroxide, in endochondral ossification and bone remodeling. NOX4's role in osteoblast formation and osteogenic signaling pathways is explored. METHODS: Using NOX4-deficient (NOX4-/-) and ovariectomized (OVX) mice, we identify NOX4's potential mediators in bone maturation. RESULTS: NOX4-/- mice displayed significant differences in bone mass and structure. Compared to the normal Control and OVX groups. Hematoxylin and eosin staining showed NOX4-/- mice had the highest trabecular bone volume, while OVX had the lowest. Proteomic analysis revealed significantly elevated MPO and OPN levels in bone marrow-derived cells in NOX4-/- mice. Immunohistochemistry confirmed increased MPO, OPN, and collagen II (COLII) near the epiphyseal plate. Collagen and chondrogenesis analysis supported enhanced bone development in NOX4-/- mice. CONCLUSIONS AND RELEVANCE: Our results emphasize NOX4's significance in bone morphology, mesenchymal stem cell proteomics, immunohistochemistry, collagen levels, and chondrogenesis. NOX4 deficiency enhances bone development and endochondral ossification, potentially through increased MPO, OPN, and COLII expression. These findings suggest therapeutic implications for skeletal disorders.


Subject(s)
NADPH Oxidase 4 , Osteogenesis , Osteopontin , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Osteopontin/metabolism , Osteopontin/genetics , Ovariectomy , Peroxidase/metabolism , Peroxidase/genetics , Collagen/metabolism
14.
J Alzheimers Dis ; 100(3): 799-808, 2024.
Article in English | MEDLINE | ID: mdl-38943386

ABSTRACT

Background: Ferroptosis is extremely relevant to the progression of neurodegenerative pathologies such as Alzheimer's disease (AD). Ubiquitin-specific proteases (USP) can affect the NADPH oxidase family. Objective: Our study aimed to elucidate the potential role and molecular basis of a certain USP19 in reducing ferroptosis and mitochondrial injury in AD cells by targeting NOX4 stability. Methods: The deubiquitinase USP family gene USP19, which affects the stability of NOX4 protein, was first screened. The cell model of AD was constructed after interfering with SH-SY5Y cells by Aß1-40, and then SH-SY5Y cells were infected with lentiviral vectors to knock down USP19 and overexpress NOX4, respectively. Finally, the groups were tested for cell viability, changes in cellular mitochondrial membrane potential, lipid reactive oxygen species, intracellular iron metabolism, and NOX4, Mf1, Mf2, and Drp1 protein expression. Results: 5 µmol/L Aß1-40 intervened in SH-SY5Y cells for 24 h to construct a cell model of AD. Knockdown of USP19 decreased the expression of NOX4 protein, promoted the expression of mitochondrial fusion proteins Mnf1 and Mnf2, and inhibited the expression of the splitting protein Drp1. Furthermore, USP19 knockdown decreased mitochondrial membrane potential, SOD, MDA, intracellular iron content and increased GSH/GSSG ratio in SH-SY5Y cells. Our study revealed that NOX4 protein interacts with USP19 and knockdown of USP19 enhanced ubiquitination to maintain NOX4 protein stability. Conclusions: USP19 attenuates mitochondrial damage in SH-SY5Y cells by targeting NOX4 protein with Aß1-40.


Subject(s)
Amyloid beta-Peptides , Ferroptosis , Mitochondria , NADPH Oxidase 4 , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Ferroptosis/physiology , Ferroptosis/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Peptide Fragments/metabolism , Reactive Oxygen Species/metabolism
15.
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
16.
Cell Death Dis ; 15(6): 398, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844470

ABSTRACT

In chronic kidney disease (CKD), renal fibrosis is an unavoidable result of various manifestations. However, its pathogenesis is not yet fully understood. Here, we revealed the novel role of Homeobox D10 (HOXD10) in CKD-related fibrosis. HOXD10 expression was downregulated in CKD-related in vitro and in vivo fibrosis models. UUO model mice were administered adeno-associated virus (AAV) containing HOXD10, and HOXD10 overexpression plasmids were introduced into human proximal tubular epithelial cells induced by TGF-ß1. The levels of iron, reactive oxygen species (ROS), lipid ROS, the oxidized glutathione/total glutathione (GSSG/GSH) ratio, malonaldehyde (MDA), and superoxide dismutase (SOD) were determined using respective assay kits. Treatment with AAV-HOXD10 significantly attenuated fibrosis and renal dysfunction in UUO model mice by inhibiting NOX4 transcription, ferroptosis pathway activation, and oxidative stress. High levels of NOX4 transcription, ferroptosis pathway activation and profibrotic gene expression induced by TGF-ß1/erastin (a ferroptosis agonist) were abrogated by HOXD10 overexpression in HK-2 cells. Moreover, bisulfite sequencing PCR result determined that HOXD10 showed a hypermethylated level in TGF-ß1-treated HK-2 cells. The binding of HOXD10 to the NOX4 promoter was confirmed by chromatin immunoprecipitation (ChIP) analysis and dual-luciferase reporter assays. Targeting HOXD10 may represent an innovative therapeutic strategy for fibrosis treatment in CKD.


Subject(s)
Ferroptosis , Fibrosis , Homeodomain Proteins , NADPH Oxidase 4 , Renal Insufficiency, Chronic , Ferroptosis/genetics , Animals , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Male , Mice, Inbred C57BL , Disease Models, Animal , Transcription Factors/metabolism , Transcription Factors/genetics , Kidney/pathology , Kidney/metabolism , Transforming Growth Factor beta1/metabolism , Reactive Oxygen Species/metabolism , Oxidative Stress , Cell Line
17.
Open Biol ; 14(6): 230427, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862020

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is a monogenic cardiac disorder commonly induced by sarcomere gene mutations. However, the mechanism for HCM is not well defined. Here, we generated transgenic MYH7 R453C and MYH6 R453C piglets and found both developed typical cardiac hypertrophy. Unexpectedly, we found serious fibrosis and cardiomyocyte loss in the ventricular of MYH7 R453C, not MYH6 R453C piglets, similar to HCM patients. Then, RNA-seq analysis and western blotting identified the activation of ERK1/2 and PI3K-Akt pathways in MYH7 R453C. Moreover, we observed an increased expression of fetal genes and an excess of reactive oxygen species (ROS) in MYH7 R453C piglet models, which was produced by Nox4 and subsequently induced inflammatory response. Additionally, the phosphorylation levels of Smad2/3, ERK1/2 and NF-kB p65 proteins were elevated in cardiomyocytes with the MYH7 R453C mutation. Furthermore, epigallocatechin gallate, a natural bioactive compound, could be used as a drug to reduce cell death by adjusting significant downregulation of the protein expression of Bax and upregulated Bcl-2 levels in the H9C2 models with MYH7 R453C mutation. In conclusion, our study illustrated that TGF-ß/Smad2/3, ERK1/2 and Nox4/ROS pathways have synergistic effects on cardiac remodelling and inflammation in MYH7 R453C mutation.


Subject(s)
Myosin Heavy Chains , NADPH Oxidase 4 , NF-kappa B , Reactive Oxygen Species , Signal Transduction , Transforming Growth Factor beta , Animals , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Transforming Growth Factor beta/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Swine , Myocytes, Cardiac/metabolism , Humans , Cardiac Myosins/metabolism , Cardiac Myosins/genetics , Disease Models, Animal , MAP Kinase Signaling System , Animals, Genetically Modified , Smad2 Protein/metabolism , Smad2 Protein/genetics , Mutation , Smad3 Protein/metabolism , Smad3 Protein/genetics , Ventricular Remodeling , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Rats
18.
J Mol Cell Cardiol ; 193: 100-112, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38851627

ABSTRACT

Nicotine, a key constituent of tobacco/electronic cigarettes causes cardiovascular injury and mortality. Nicotine is known to induce oxidative stress and mitochondrial dysfunction in cardiomyocytes leading to cell death. However, the underlying mechanisms remain unclear. Pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP) is a member of metal-dependent protein phosphatase (PPM) family and is known to dephosphorylate several AGC family kinases and thereby regulate a diverse set of cellular functions including cell growth, survival, and death. Our lab has previously demonstrated that PHLPP1 removal reduced cardiomyocyte death and cardiac dysfunction following injury. Here, we present a novel finding that nicotine exposure significantly increased PHLPP1 protein expression in the adolescent rodent heart. Building upon our in vivo finding, we determined the mechanism of PHLPP1 expression in cardiomyocytes. Nicotine significantly increased PHLPP1 protein expression without altering PHLPP2 in cardiomyocytes. In cardiomyocytes, nicotine significantly increased NADPH oxidase 4 (NOX4), which coincided with increased reactive oxygen species (ROS) and increased cardiomyocyte apoptosis which were dependent on PHLPP1 expression. PHLPP1 expression was both necessary and sufficient for nicotine induced mitochondrial dysfunction. Mechanistically, nicotine activated extracellular signal-regulated protein kinases (ERK1/2) and subsequent eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) to increase PHLPP1 protein expression. Inhibition of protein synthesis with cycloheximide (CHX) and 4EGI-1 abolished nicotine induced PHLPP1 protein expression. Moreover, inhibition of ERK1/2 activity by U0126 significantly blocked nicotine induced PHLPP1 expression. Overall, this study reveals a novel mechanism by which nicotine regulates PHLPP1 expression through ERK-4E-BP1 signaling axis to drive cardiomyocyte injury.


Subject(s)
Myocytes, Cardiac , Nicotine , Oxidative Stress , Phosphoprotein Phosphatases , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/genetics , Nicotine/pharmacology , Nicotine/adverse effects , Oxidative Stress/drug effects , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Rats , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , MAP Kinase Signaling System/drug effects , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Rats, Sprague-Dawley , Mice , Extracellular Signal-Regulated MAP Kinases/metabolism , Male
19.
Am J Physiol Endocrinol Metab ; 327(1): E1-E12, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38690939

ABSTRACT

High levels of testosterone (Testo) are associated with cardiovascular risk by increasing reactive oxygen species (ROS) formation. NADPH oxidases (NOX) are the major source of ROS in the vasculature of cardiovascular diseases. NOX4 is a unique isotype, which produces hydrogen peroxide (H2O2), and its participation in cardiovascular biology is controversial. So far, it is unclear whether NOX4 protects from Testo-induced endothelial injury. Thus, we hypothesized that supraphysiological levels of Testo induce endothelial NOX4 expression to attenuate endothelial injury. Human mesenteric vascular endothelial cells (HMECs) and human umbilical vein endothelial cells (HUVEC) were treated with Testo (10-7 M) with or without a NOX4 inhibitor [GLX351322 (10-4 M)] or NOX4 siRNA. In vivo, 10-wk-old C57Bl/6J male mice were treated with Testo (10 mg/kg) for 30 days to study endothelial function. Testo increased mRNA and protein levels of NOX4 in HMECs and HUVECs. Testo increased superoxide anion (O2-) and H2O2 production, which were abolished by NOX1 and NOX4 inhibition, respectively. Testo also attenuated bradykinin-induced NO production, which was further impaired by NOX4 inhibition. In vivo, Testo decreased H2O2 production in aortic segments and triggered endothelial dysfunction [decreased relaxation to acetylcholine (ACh)], which was further impaired by GLX351322 and by a superoxide dismutase and catalase mimetic (EUK134). Finally, Testo led to a dysregulated endothelial cell migration, which was exacerbated by GLX351322. These data indicate that supraphysiological levels of Testo increase the endothelial expression and activity of NOX4 to counterbalance the deleterious effects caused by Testo in endothelial function.NEW & NOTEWORTHY By inducing ROS formation, high levels of testosterone play a major role in the pathogenesis of cardiovascular disease. NOXs are the major sources of ROS in the vasculature of cardiovascular diseases. Herein, we describe a novel compensatory mechanism by showing that NOX4 is a protective oxidant enzyme and counterbalances the deleterious effects of testosterone in endothelial cells by modulating hydrogen peroxide formation.


Subject(s)
Cell Movement , Endothelium, Vascular , Human Umbilical Vein Endothelial Cells , Hydrogen Peroxide , Mice, Inbred C57BL , NADPH Oxidase 4 , Testosterone , Animals , Humans , Male , Mice , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Reactive Oxygen Species/metabolism , Testosterone/pharmacology , Testosterone/metabolism
20.
Biochem Pharmacol ; 225: 116328, 2024 07.
Article in English | MEDLINE | ID: mdl-38815628

ABSTRACT

Early stages of diabetes are characterized by elevations of insulin and glucose concentrations. Both factors stimulate reactive oxygen species (ROS) production, leading to impairments in podocyte function and disruption of the glomerular filtration barrier. Podocytes were recently shown to be an important source of αKlotho (αKL) expression. Low blood Klotho concentrations are also associated with an increase in albuminuria, especially in patients with diabetes. We investigated whether ADAM10, which is known to cleave αKL, is activated in glomeruli and podocytes under diabetic conditions and the potential mechanisms by which ADAM10 mediates ROS production and disturbances of the glomerular filtration barrier. In cultured human podocytes, high glucose increased ADAM10 expression, shedding, and activity, NADPH oxidase activity, ROS production, and albumin permeability. These effects of glucose were inhibited when cells were pretreated with an ADAM10 inhibitor or transfected with short-hairpin ADAM10 (shADAM10) or after the addition soluble Klotho. We also observed increases in ADAM10 activity, NOX4 expression, NADPH oxidase activity, and ROS production in αKL-depleted podocytes. This was accompanied by an increase in albumin permeability in shKL-expressing podocytes. The protein expression and activity of ADAM10 also increased in isolated glomeruli and urine samples from diabetic rats. Altogether, these results reveal a new mechanism by which hyperglycemia in diabetes increases albumin permeability through ADAM10 activation and an increase in oxidative stress via NOX4 enzyme activation. Moreover, αKlotho downregulates ADAM10 activity and supports redox balance, consequently protecting the slit diaphragm of podocyteσ under hyperglycemic conditions.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Diabetes Mellitus, Experimental , Glucuronidase , Klotho Proteins , Membrane Proteins , Podocytes , Reactive Oxygen Species , Podocytes/metabolism , Podocytes/drug effects , Klotho Proteins/metabolism , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Reactive Oxygen Species/metabolism , Humans , Animals , Glucuronidase/metabolism , Glucuronidase/genetics , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Rats , Male , Diabetes Mellitus, Experimental/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidases/metabolism , Cells, Cultured , Glucose/metabolism , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL