Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Chem Biol Drug Des ; 103(4): e14520, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570710

ABSTRACT

Quercetin, a bioactive natural compound renowned for its potent anti-inflammatory, antioxidant, and antiviral properties, has exhibited therapeutic potential in various diseases. Given that bronchopulmonary dysplasia (BPD) development is closely linked to inflammation and oxidative stress, and quercetin, a robust antioxidant known to activate NRF2 and influence the ferroptosis pathway, offers promise for a wide range of age groups. Nonetheless, the specific role of quercetin in BPD remains largely unexplored. This study aims to uncover the target role of quercetin in BPD through a combination of network pharmacology, molecular docking, computer analyses, and experimental evaluations.


Subject(s)
Bronchopulmonary Dysplasia , Ferroptosis , Hyperoxia , Animals , Infant, Newborn , Humans , Bronchopulmonary Dysplasia/drug therapy , Bronchopulmonary Dysplasia/metabolism , Hyperoxia/drug therapy , Hyperoxia/metabolism , Quercetin/pharmacology , Quercetin/therapeutic use , Molecular Docking Simulation , Cyclooxygenase 2 , Animals, Newborn , Antioxidants , Network Pharmacology
2.
Peptides ; 170: 171109, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37804931

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia (BPD) is a common pulmonary injury among premature infants, which is often caused by hyperoxia exposure. Irisin is a novel hormone-like myokine derived mainly from skeletal muscles as well as adipose tissues. Many studies have indicated that Irisin exert a variety of properties against hyperoxia-induced inflammation and oxidative stress (OS). We aimed to evaluate the effects of irisin on hyperoxia-induced lung injury explore the underlying mechanisms. METHODS: BPD model was established after exposing newborn mouse to 85% oxygen. BPD mouse received continuous intraperitoneal injection of irisin at a dose of 25 µg/kg/day. Lung tissues were collected for histological examination at 7 and 14 days after birth. The alveolarization and alveolar vascularization of each animal was assessed. Levels of oxidative stress indicators, and the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) in lung tissues were detected at 14 days after birth. RESULTS: Hyperoxia exposure induced a markedly alveolar simplification and a disrupted alveolar angiogenesis, which was ameliorated by irisin treatment. The hyperoxia-induced increase in these oxidative stress indicators was significantly reversed by irisin treatment. The Nrf2/HO-1 pathway is inducted in the hyperoxia-induced BPD mouse model, which is further activated by irisin treatment. CONCLUSION: Our results demonstrated the beneficial effects of irisin in reducing the OS, enhancing alveolarization, and promoting vascular development through activation of Nrf2/HO-1 axis in a hyperoxia-induced experimental model of BPD.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Lung Injury , Animals , Mice , Animals, Newborn , Bronchopulmonary Dysplasia/drug therapy , Bronchopulmonary Dysplasia/metabolism , Disease Models, Animal , Fibronectins/metabolism , Heme Oxygenase-1/metabolism , Hyperoxia/drug therapy , Hyperoxia/metabolism , Lung/metabolism , Lung Injury/metabolism , NF-E2-Related Factor 2/metabolism
3.
J Breath Res ; 17(3)2023 06 12.
Article in English | MEDLINE | ID: mdl-37207635

ABSTRACT

Prolonged exposure to hyperbaric hyperoxia can lead to pulmonary oxygen toxicity (PO2tox). PO2tox is a mission limiting factor for special operations forces divers using closed-circuit rebreathing apparatus and a potential side effect for patients undergoing hyperbaric oxygen (HBO) treatment. In this study, we aim to determine if there is a specific breath profile of compounds in exhaled breath condensate (EBC) that is indicative of the early stages of pulmonary hyperoxic stress/PO2tox. Using a double-blind, randomized 'sham' controlled, cross-over design 14 U.S. Navy trained diver volunteers breathed two different gas mixtures at an ambient pressure of 2 ATA (33 fsw, 10 msw) for 6.5 h. One test gas consisted of 100% O2(HBO) and the other was a gas mixture containing 30.6% O2with the balance N2(Nitrox). The high O2stress dive (HBO) and low O2stress dive (Nitrox) were separated by at least seven days and were conducted dry and at rest inside a hyperbaric chamber. EBC samples were taken immediately before and after each dive and subsequently underwent a targeted and untargeted metabolomics analysis using liquid chromatography coupled to mass spectrometry (LC-MS). Following the HBO dive, 10 out of 14 subjects reported symptoms of the early stages of PO2tox and one subject terminated the dive early due to severe symptoms of PO2tox. No symptoms of PO2tox were reported following the nitrox dive. A partial least-squares discriminant analysis of the normalized (relative to pre-dive) untargeted data gave good classification abilities between the HBO and nitrox EBC with an AUC of 0.99 (±2%) and sensitivity and specificity of 0.93 (±10%) and 0.94 (±10%), respectively. The resulting classifications identified specific biomarkers that included human metabolites and lipids and their derivatives from different metabolic pathways that may explain metabolomic changes resulting from prolonged HBO exposure.


Subject(s)
Hyperbaric Oxygenation , Hyperoxia , Humans , Breath Tests , Hyperbaric Oxygenation/adverse effects , Hyperoxia/drug therapy , Nitrogen/therapeutic use , Oxygen , Cross-Over Studies
4.
Int J Mol Sci ; 24(9)2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37175511

ABSTRACT

The risk of oxidative stress is unavoidable in preterm infants and increases the risk of neonatal morbidities. Premature infants often require sedation and analgesia, and the commonly used opioids and benzodiazepines are associated with adverse effects. Impairment of cerebellar functions during cognitive development could be a crucial factor in neurodevelopmental disorders of prematurity. Recent studies have focused on dexmedetomidine (DEX), which has been associated with potential neuroprotective properties and is used as an off-label application in neonatal units. Wistar rats (P6) were exposed to 80% hyperoxia for 24 h and received as pretreatment a single dose of DEX (5µg/kg, i.p.). Analyses in the immature rat cerebellum immediately after hyperoxia (P7) and after recovery to room air (P9, P11, and P14) included examinations for cell death and inflammatory and oxidative responses. Acute exposure to high oxygen concentrations caused a significant oxidative stress response, with a return to normal levels by P14. A marked reduction of hyperoxia-mediated damage was demonstrated after DEX pretreatment. DEX produced a much earlier recovery than in controls, confirming a neuroprotective effect of DEX on alterations elicited by oxygen stress on the developing cerebellum.


Subject(s)
Dexmedetomidine , Hyperoxia , Infant, Newborn , Animals , Rats , Humans , Hyperoxia/complications , Hyperoxia/drug therapy , Rats, Wistar , Animals, Newborn , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Infant, Premature , Apoptosis , Oxidative Stress , Oxygen/pharmacology , Interneurons
5.
Biomaterials ; 293: 121943, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36527790

ABSTRACT

Recent trends in the design of regenerative materials include the development of bioactive matrices to harness the innate healing ability of the body using various biophysicochemical stimuli (defined as in situ tissue regeneration). Among these, hyperoxia (>21% pO2) is a well-known therapeutic factor for promoting tissue regeneration, such as immune cell recruitment, cell proliferation, angiogenesis, and fibroblast differentiation into myofibroblast. Although various strategies to induce hyperoxia are reported, developing advanced hyperoxia-inducing biomaterials for tissue regeneration is still challenging. In this study, a catalase-immobilized syringe (defined as an Oxyringe) via calcium peroxide-mediated surface modification is developed as a new type of oxygen-supplying system. Hyperoxia-inducible hydrogels are fabricated utilizing Oxyringe. This hydrogel plays a role as a physical barrier for hemostasis. In addition, hyperoxic matrices induce transient hyperoxia in vivo (up to 46.0% pO2). Interestingly, the hydrogel-induced hyperoxia boost the initial macrophage recruitment and rapid inflammation resolution. Furthermore, hyperoxic oxygen release of hydrogels facilitates neovascularization and cell proliferation involved in the proliferation phase, expediting tissue maturation related to the remodeling phase in wound healing. In summary, Oxyringe has excellent potential as an advanced oxygen-supplying platform to create hyperoxia-inducing hydrogels for in situ tissue regeneration.


Subject(s)
Hyperoxia , Humans , Hyperoxia/drug therapy , Hydrogels/pharmacology , Syringes , Oxygen , Wound Healing
6.
Eur J Ophthalmol ; 33(4): NP66-NP69, 2023 Jul.
Article in English | MEDLINE | ID: mdl-35593072

ABSTRACT

PURPOSE: To study the long-term anatomic and physiologic effects of nocturnal normobaric hyperoxia (NNBH) in a patient with treatment-resistant diabetic macular edema (DME). METHODS: A 64-year-old diabetic man with bilateral DME requiring regular anti-VEGF treatments in both eyes was started on 5 LPM (40% FiO2) NNBH treatment 6-h per night. Visual acuity, OCT measurements of retinal thickness and volume, as well as the number of injections given in each eye were retrospectively examined one year prior and prospectively after initiation of NNBH, as well as before and after a planned 1-month discontinuation of NNBH. RESULTS: The patient received 12 anti-VEGF injections in the year prior to beginning NNBH treatment (4 OD; 8 OS) and did not require any injections after commencing NNBH treatment. Visual acuity improved and stabilized to 20/20 and macular edema rapidly resolved in both eyes following initiation of NNBH. After a planned 1-month NNBH vacation, DME recurred but quickly resolved once NNBH treatment was restarted. CONCLUSION: This model case demonstrates that a 6-h NNBH regimen can be successful in treating DME and improving vision, without the need for intravitreal injections. NNBH is a more acceptable treatment regimen compared to 24-h continuous oxygen delivery and may provide a less invasive alternate method for treating DME in patients with diabetes. Further study is warranted.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Hyperoxia , Macular Edema , Male , Humans , Middle Aged , Macular Edema/diagnosis , Macular Edema/etiology , Macular Edema/therapy , Diabetic Retinopathy/complications , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/therapy , Retrospective Studies , Hyperoxia/drug therapy , Intravitreal Injections , Angiogenesis Inhibitors/therapeutic use , Tomography, Optical Coherence , Diabetes Mellitus/drug therapy
7.
Redox Rep ; 27(1): 270-278, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36357965

ABSTRACT

Objectives: Caffeine has been shown to reduce the incidence of bronchopulmonary dysplasia (BPD). To investigate the protective mechanism of caffeine in a hyperoxia-based cell model of BPD in vitro.Methods: Type II alveolar epithelial cells (AECs II) were isolated and randomly divided into 6 groups: the normal, hyperoxia, caffeine (50 µM caffeine), antagonist (5 µM ZM241385), agonist (5 µM CGS21680), and DMSO groups. Transfection with siRNA against adenosine A2A receptor (siA2AR) was performed in AECs II.Results: Caffeine alone or in combination with adenosine A2A receptor (A2AR) antagonist inhibited apoptosis, promoted proliferation and reduced oxidative stress (OS). The cyclic adenosine monophosphate (cAMP), protein kinase A (PKA) mRNA, A2AR mRNA and the protein levels of A2AR, phospho-Src, phospho-ERK1/2, phospho-P38 and cleaved caspase-3 were decreased in the caffeine and antagonist groups compared with that in the hyperoxia group. However, the effects of caffeine above were weakened by the A2AR agonist. Knockdown of A2AR showed similar results to caffeine.Discussion: Caffeine can reduce apoptosis, promote proliferation, and alleviate OS in hyperoxia-induced AECs II injury by inhibiting the A2AR/cAMP/PKA/Src/ERK1/2/p38MAPK signaling pathway. Caffeine and A2AR may serve as a promising therapeutic target for BPD in prematurity.


Subject(s)
Hyperoxia , Lung Injury , Infant, Newborn , Humans , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/pharmacology , Caffeine/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/pharmacology , MAP Kinase Signaling System , Hyperoxia/complications , Hyperoxia/drug therapy , Signal Transduction , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Oxidative Stress , RNA, Messenger/metabolism , RNA, Messenger/pharmacology
8.
Microbiol Spectr ; 10(5): e0128221, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36094220

ABSTRACT

The ring-stage survival assay was utilized to assess the impact of physiological hyperoxic stress on dihydroartemisinin (DHA) tolerance for a panel of Plasmodium falciparum strains with and without Kelch13 mutations. Strains without naturally acquired Kelch13 mutations or the postulated genetic background associated with delayed parasite clearance time demonstrated reduced proliferation under hyperoxic conditions in the subsequent proliferation cycle. Dihydroartemisinin tolerance in three isolates with naturally acquired Kelch13 mutations but not two genetically manipulated laboratory strains was modulated by in vitro hyperoxic stress exposure of early-ring-stage parasites in the cycle before drug exposure. Reduced parasite tolerance to additional derivatives, including artemisinin, artesunate, and OZ277, was observed within the second proliferation cycle. OZ439 and epoxomicin completely prevented parasite survival under both hyperoxia and normoxic in vitro culture conditions, highlighting the unique relationship between DHA tolerance and Kelch13 mutation-associated genetic background. IMPORTANCE Artemisinin-based combination therapy (ACT) for treating malaria is under intense scrutiny following treatment failures in the Greater Mekong subregion of Asia. This is further compounded by the potential for extensive loss of life if treatment failures extend to the African continent. Although Plasmodium falciparum has become resistant to all antimalarial drugs, artemisinin "resistance" does not present in the same way as resistance to other antimalarial drugs. Instead, a partial resistance or tolerance is demonstrated, associated with the parasite's genetic profile and linked to a molecular marker referred to as K13. It is suggested that parasites may have adapted to drug treatment, as well as the presence of underlying population health issues such as hemoglobinopathies, and/or environmental pressures, resulting in parasite tolerance to ACT. Understanding parasite evolution and control of artemisinin tolerance will provide innovative approaches to mitigate the development of artemisinin tolerance and thereby artemisinin-based drug treatment failure and loss of life globally to malaria infections.


Subject(s)
Antimalarials , Artemisinins , Hyperoxia , Malaria, Falciparum , Parasites , Animals , Plasmodium falciparum/genetics , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artesunate/pharmacology , Artesunate/therapeutic use , Hyperoxia/drug therapy , Drug Resistance/genetics , Protozoan Proteins/genetics , Protozoan Proteins/pharmacology , Protozoan Proteins/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use , Mutation , Drug Tolerance , Malaria, Falciparum/drug therapy
9.
Exp Lung Res ; 48(4-6): 187-197, 2022.
Article in English | MEDLINE | ID: mdl-35924334

ABSTRACT

Background: Insulin-like growth factor-1 (IGF-1), a member of the insulin family, has a high degree of homology with insulin and exhibits anti-inflammatory and anti-oxidative stress properties. However, the potential protective effect of IGF-1 on hyperoxia-induced lung injury remains unknown. In this study, we aimed to explore the effects and mechanism of action of IGF-1 in hyperoxia-induced lung injury in neonatal rats. Materials and Methods: Hematoxylin-eosin staining was used to observe pathological changes in lung tissue; transmission electron microscopy was used to examine the ultrastructure, and ELISA was used to detect the level of pro-inflammatory cytokines in bronchoalveolar lavage fluid. Further, malondialdehyde, glutathione, and superoxide dismutase activities in lung tissue were evaluated. TUNEL staining was used to detect cell apoptosis, and western blot analysis was used to detect the expression of Bax, Bcl-2, Caspase-3, p-PERK, p-eIF2α, ATF4, and CHOP in the lung tissue. Moreover, the wet/dry weight ratio of lung tissue was determined. Results: Intraperitoneal injection of IGF-1 effectively reduced lung tissue damage induced by hyperoxia; production of inflammatory cells and release of pro-inflammatory cytokines, oxidative stress, and cell apoptosis. Further, IGF-1 down-regulated the expression of ATF4, CHOP, and Bax/Bcl-2, and inhibited the phosphorylation of PERK and eIF2α. Conclusion: The results suggest that IGF-1 reduces hyperoxia-induced lung inflammation and oxidative stress in neonatal rats through the PERK/eIF2α/ATF4/CHOP signaling pathway and inhibits cell apoptosis.


Subject(s)
Hyperoxia , Insulins , Lung Injury , Pneumonia , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/pharmacology , Animals , Apoptosis , Cytokines/metabolism , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/pharmacology , Hyperoxia/complications , Hyperoxia/drug therapy , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Insulins/metabolism , Insulins/pharmacology , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Signal Transduction , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/pharmacology , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/pharmacology , eIF-2 Kinase/metabolism , eIF-2 Kinase/pharmacology
10.
Oxid Med Cell Longev ; 2022: 8336070, 2022.
Article in English | MEDLINE | ID: mdl-35509841

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly found in premature infants. Excessive inflammation and oxidative stress contribute to BPD occurrence and development. Simvastatin, as an inhibitor of HMG-CoA reductase, has been reported to have antioxidative and anti-inflammatory effects. However, its effect and possible mechanisms in hyperoxia-induced lung injury are rarely reported. In this study, in vivo and in vitro experiments were conducted to investigate whether simvastatin could ameliorate hyperoxia-induced lung injury and explore its potential mechanism. For the in vivo study, simvastatin could improve alveolar development after hyperoxic lung injury and reduce hyperoxic stress and inflammation. The in vitro study revealed that simvastatin can reduce inflammation in A549 cells after high-oxygen exposure. Simvastatin suppressed NLRP3 inflammasome activation and played anti-inflammatory and antioxidant roles by increasing KLF2 (Krüppel-like factor 2) expression. In vitro experiments also revealed that these effects of simvastatin were partially reversed by KLF2 shRNA, indicating that KLF2 was involved in simvastatin effects. In summary, our findings indicate that simvastatin could downregulate NLRP3 inflammasome activation and attenuate lung injury in hyperoxia-induced bronchopulmonary dysplasia via KLF2-mediated mechanism.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Lung Injury , Animals , Animals, Newborn , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Bronchopulmonary Dysplasia/genetics , Humans , Hyperoxia/complications , Hyperoxia/drug therapy , Hyperoxia/genetics , Infant, Newborn , Inflammasomes/metabolism , Inflammation/metabolism , Kruppel-Like Transcription Factors/metabolism , Lung/metabolism , Lung Injury/drug therapy , Lung Injury/etiology , Lung Injury/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Simvastatin/pharmacology , Simvastatin/therapeutic use , Transcription Factors/metabolism
11.
Environ Toxicol ; 37(6): 1472-1482, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35212449

ABSTRACT

Hyperoxia, is often used in preterm supportive care, leading to high oxygen exposure in neonates. Coenzyme Q10 (CoQ10) is a free radical scavenger that has been studied in older children but never be investigated for its role in preterm care. We hypothesize that the administration of exogenous CoQ10 would raise serum concentrations of CoQ10 and mitigate the adverse effects of hyperoxia on the organs by reducing oxygen-free radicals and inflammation. The aim of this study was to evaluate the effects of oxidative stress, inflammatory response, and survival in neonatal rats after CoQ10 treatment. Neonatal rats delivered from four pregnant Wistar rats were randomly divided into four groups: (a) control, (b) CoQ10, (c) hyperoxia (O2 group), and (d) treatment (CoQ10 + O2 ) groups. The dose of CoQ10 injected was 30 mg/kg. The CoQ9, CoQ10, cytokines, oxidative stress, and antioxidant enzyme activity were measured. Tissue samples were histologically examined and mortality was monitored for 16 days. The level of CoQ9 significantly increased in the liver, kidney, and plasma, while the level of CoQ10 significantly increased in most organ tissues in the CoQ10 + O2 group. Additionally, CoQ10 decrease oxidative stress in the liver, increase antioxidant enzyme activity in the heart, kidney, and brain, and reverse an inclined level of hematopoietic growth factors. However, CoQ10 had no effect on inflammation, organ damage, or mortality. Therefore, the use of CoQ10 in potential adjuvant therapy for neonatal hyperoxia requires further research.


Subject(s)
Antioxidants , Hyperoxia , Animals , Animals, Newborn , Antioxidants/metabolism , Female , Hyperoxia/drug therapy , Inflammation/metabolism , Oxidative Stress , Oxygen , Pregnancy , Rats , Rats, Wistar , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
12.
Pediatr Res ; 92(2): 436-444, 2022 08.
Article in English | MEDLINE | ID: mdl-34725500

ABSTRACT

BACKGROUND: We aimed to compare the effect of dexamethasone (Dex), hydrocortisone (Hc), and methylprednisolone (Mpz) at equivalent doses on somatic growth, lung healing, and neurotoxicity in a hyperoxic rat model. We hypothesized that Mpz and Hc would be superior to Dex with less neurotoxicity by exerting similar therapeutic efficacy on the injured lung. METHODS: Neonatal rats were randomized to control, bronchopulmonary dysplasia (BPD), Dex, Hc, and Mpz groups. All drugs were administered daily following day 15 over 7 days. Histopathological and immunohistochemical analyses of the lung and brain were performed on day 22. RESULTS: All types had much the same impact on lung repair. Oxidative markers in the lung were similar in the steroid groups. While nuclear factor erythroid 2-related factor and heat-shock protein 70 dropped following steroid treatment, no difference was noted in other biochemical markers in the brain between the study groups. Apoptotic activity and neuron loss in the parietal cortex and hippocampus were noted utmost in Dex, but alike in other BPD groups. CONCLUSIONS: Mpz does not appear to be superior to Dex or Hc in terms of pulmonary outcomes and oxidative damage in the brain, but safer than Dex regarding apoptotic neuron loss. IMPACT: This is the first study that compared the pulmonary efficacy and neurotoxic effects of Dex, Hc, and Mpz simultaneously in an established BPD model. This study adds to the literature on the importance of possible antioxidant and protective effects of glucocorticoid therapy in an oxidative stress-exposed brain. Mpz ended up with no more additional neuron loss or apoptosis risk by having interchangeable effects with others for the treatment of established BPD. Mpz and Hc seem safe as a rescue therapy in terms of adverse outcomes for established BPD in which lung and brain tissue is already impaired.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Lung Injury , Neurotoxicity Syndromes , Animals , Humans , Infant, Newborn , Rats , Animals, Newborn , Antioxidants , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/drug therapy , Dexamethasone , Glucocorticoids/therapeutic use , HSP70 Heat-Shock Proteins , Hydrocortisone , Hyperoxia/complications , Hyperoxia/drug therapy , Lung , Lung Injury/drug therapy , Methylprednisolone/therapeutic use
13.
J Integr Neurosci ; 20(1): 109-118, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33834697

ABSTRACT

Preterm birth causes neurological deficits. Previously, we demonstrated that fetal zone steroids reduce hyperoxia-mediated cell death in vitro. In immature oligodendrocytes (OLN-93 cells), dehydroepiandrosterone + 17ß-estradiol co-treatment had synergistic beneficial effects while signals were transduced through different receptors. In immature astrocytes (C6 cells), both hormones compete for the same receptor and no synergistic effects were observed. 17ß-estradiol and progesterone drastically decrease while fetal zone steroids, mainly dehydroepiandrosterone, remain persistently high within preterm infants until term. Substitution of 17ß-estradiol and progesterone does not improve neurological outcomes. We investigated the influence of dehydroepiandrosterone, 17ß-estradiol or dehydroepiandrosterone + 17ß-estradiol treatment in C6 or OLN-93 cells on steroid receptor availability and activation of intracellular signaling molecules in hyperoxic cell culture. We sought explanations of the observed synergistic effect in preliminary study. In C6 cells, the generated signaling of dehydroepiandrosterone + 17ß-estradiol treatment has no synergistic effects. The combined effect on this particular pathway does not potentiate cell survival. In OLN-93 cells, we observed significant differences in the early generated signaling of 17ß-estradiol + dehydroepiandrosterone treatment to either 17ß-estradiol dehydroepiandrosterone alone but never to both at the same time. The latter finding needs, therefore, further investigation to explain synergistic effects. Nevertheless, we add insight into the receptor and signaling cascade alterations induced by 17ß-estradiol, dehydroepiandrosterone or 17ß-estradiol + dehydroepiandrosterone treatment of C6 and OLN-93 cells in hyperoxia.


Subject(s)
Astrocytes/drug effects , Dehydroepiandrosterone/pharmacology , Estradiol/pharmacology , Hyperoxia/drug therapy , Infant, Premature, Diseases/drug therapy , Oligodendroglia/drug effects , Cells, Cultured , Drug Synergism , Drug Therapy, Combination , Humans
14.
Comb Chem High Throughput Screen ; 24(8): 1243-1250, 2021.
Article in English | MEDLINE | ID: mdl-32933455

ABSTRACT

BACKGROUND/AIM: This study aimed to ascertain the effects of astaxanthin on the lungs of rat pups with bronchopulmonary dysplasia (BPD) induced by hyperoxia and lipopolysaccharide (LPS). MATERIALS AND METHODS: Forty-two newborn Wistar rats, born to spontaneous pregnant rats, were divided into three groups: Hyperoxia (95% O2) + lipopolysaccharide (LPS) group, hyperoxia + LPS + astaxhantin group, and control: no treatment group (21% O2). Pups in the hyperoxia + LPS + astaxanthin group were given 100 mg/kg/day oral astaxanthin from the first day to the fifth day. Histopathologic and biochemical evaluations, including glutathione (GSH), total anti-oxidant status (TAS), total oxidant status (TOS), lipid hydroperoxide (LPO), 8-hydroxydeoxyguanosine (8-OHdG), advanced oxidation protein products (AOPP), myeloperoxidase (MPO), total thiol, tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1ß), and caspase-3 activities, were performed. RESULTS: Better survival rates and weight gain were demonstrated in the hyperoxia + LPS + astaxanthin group (p <0.001). In the histopathologic evaluation, the severity of lung damage was significantly reduced in the hyperoxia+LPS+astaxanthin group, as well as decreased apoptosis (ELISA for caspase-3) (p <0.001). The biochemical analyses of lung tissues showed that TAS, GSH, and Total thiol levels were significantly higher in the astaxanthin treated group compared to the hyperoxia + LPS group (p <0.05) while TOS, AOPP, LPO, 8-OHdG, MPO levels were significantly lower (p <0.001). In addition, unlike the hyperoxia + LPS group, TNF-α and IL-1ß levels in lung tissue were significantly lower in the astaxanthin-treated group (p <0.001). CONCLUSION: Astaxanthin was shown to reduce lung damage caused by inflammation and hyperoxia with its anti-inflammatory, anti-oxidant, anti-apoptotic properties, and to protect the lung from severe destruction.


Subject(s)
Hyperoxia , Lung Injury , Animals , Animals, Newborn , Disease Models, Animal , Female , Hyperoxia/complications , Hyperoxia/drug therapy , Hyperoxia/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Lung/metabolism , Lung/pathology , Lung Injury/drug therapy , Lung Injury/etiology , Lung Injury/metabolism , Pregnancy , Rats , Rats, Wistar , Xanthophylls
15.
Int J Mol Sci ; 21(18)2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32967238

ABSTRACT

Following traumatic brain injury (TBI), the time window during which secondary injuries develop provides a window for therapeutic interventions. During this time, many TBI victims undergo exposure to hyperoxia and anesthetics. We investigated the effects of genetic background on the interaction of oxygen and volatile general anesthetics with brain pathophysiology after closed-head TBI in the fruit fly Drosophila melanogaster. To test whether sevoflurane shares genetic risk factors for mortality with isoflurane and whether locomotion is affected similarly to mortality, we used a device that generates acceleration-deceleration forces to induce TBI in ten inbred fly lines. After TBI, we exposed flies to hyperoxia alone or in combination with isoflurane or sevoflurane and quantified mortality and locomotion 24 and 48 h after TBI. Modulation of TBI-induced mortality and locomotor impairment by hyperoxia with or without anesthetics varied among fly strains and among combinations of agents. Resistance to increased mortality from hyperoxic isoflurane predicted resistance to increased mortality from hyperoxic sevoflurane but did not predict the degree of locomotion impairment under any condition. These findings are important because they demonstrate that, in the context of TBI, genetic background determines the latent toxic potentials of oxygen and anesthetics.


Subject(s)
Anesthetics, Inhalation/pharmacology , Genetic Background , Head Injuries, Closed , Hyperoxia , Isoflurane/pharmacology , Sevoflurane/pharmacology , Animals , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Drosophila melanogaster , Head Injuries, Closed/drug therapy , Head Injuries, Closed/genetics , Head Injuries, Closed/metabolism , Head Injuries, Closed/pathology , Humans , Hyperoxia/drug therapy , Hyperoxia/genetics , Hyperoxia/metabolism , Hyperoxia/pathology , Oxygen Consumption/drug effects
16.
Physiol Rep ; 8(18): e14587, 2020 09.
Article in English | MEDLINE | ID: mdl-32959498

ABSTRACT

We investigated the hypothesis that exposure of lungs at the saccular stage of development to hyperoxia leads to persistent growth arrest and dysfunction of 5'AMP-activated protein kinase (AMPK), a key energy sensor in the cell. We exposed neonatal rat pups from postnatal day 1- day 10 (P1-P10) to ≥90% oxygen or control normoxia. Pups were euthanized at P4 or P10 or recovered in normoxia until euthanasia at P21. Half of the pups in each group received AMPK activator, metformin, or saline intraperitoneally from P1 to P10. Lung histology, morphometric analysis, immunofluorescence, and immunoblots were done for changes in lung structure at P10 and P21 and AMPK function at P4, P10, and P21. Phosphorylation of AMPK (p-AMPK) was decreased in lungs at P10 and P21 in hyperoxia-exposed pups. Metformin increased the levels of p-AMPK and PGC-1α, a downstream AMPK target which regulates mitochondrial biogenesis, at P4, P10, and P21 in hyperoxia pups. Lung ATP levels decreased during hyperoxia and were increased by metformin at P10 and P21. Radial alveolar count and alveolar septal tips were decreased and mean linear intercept increased in hyperoxia-exposed pups at P10 and the changes persisted at P21; these were improved by metformin. Lung capillary number was decreased in hyperoxia-exposed pups at P10 and P21 and was restored by metformin. Hyperoxia leads to impaired AMPK function, energy balance and alveolar simplification. The AMPK activator, metformin improves AMPK function and alveolar and vascular growth in this rat pup model of hyperoxia-induced lung injury.


Subject(s)
Antioxidants/therapeutic use , Hyperoxia/drug therapy , Hypoglycemic Agents/therapeutic use , Lung/metabolism , Metformin/therapeutic use , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Antioxidants/pharmacology , Female , Hypoglycemic Agents/pharmacology , Lung/drug effects , Lung/growth & development , Male , Metformin/pharmacology , Organelle Biogenesis , Oxygen/toxicity , PPAR gamma/genetics , PPAR gamma/metabolism , Rats , Rats, Sprague-Dawley
17.
Physiol Rep ; 8(16): e14555, 2020 08.
Article in English | MEDLINE | ID: mdl-32812392

ABSTRACT

BACKGROUND: This study was undertaken to test the hypothesis that the newly synthesized curcuminoids B2BrBC and C66 supplementation will overcome hyperoxia-induced tracheal hyperreactivity and impairment of relaxation of tracheal smooth muscle (TSM). MATERIALS AND METHODS: Rat pups (P5) were exposed to hyperoxia (>95% O2 ) or normoxia for 7 days. At P12, tracheal cylinders were used to study in vitro contractile responses induced by methacholine (10-8 -10-4 M) or relaxation induced by electrical field stimulation (5-60 V) in the presence/absence of B2BrBC or C66, or to study the direct relaxant effects elicited by both analogs. RESULTS: Hyperoxia significantly increased contraction and decreased relaxation of TSM compared to normoxia controls. Presence of B2BrBC or C66 normalized both contractile and relaxant responses altered by hyperoxia. Both, curcuminoids directly induced dose-dependent relaxation of preconstricted TSM. Supplementation of hyperoxic animals with B2BrBC or C66, significantly increased catalase activity. Lung TNF-α was significantly increased in hyperoxia-exposed animals. Both curcumin analogs attenuated increases in TNF-α in hyperoxic animals. CONCLUSION: We show that B2BrBC and C66 provide protection against adverse contractility and relaxant effect of hyperoxia on TSM, and whole lung inflammation. Both analogs induced direct relaxation of TSM. Through restoration of catalase activity in hyperoxia, we speculate that analogs are protective against hyperoxia-induced tracheal hyperreactivity by augmenting H2 O2 catabolism. Neonatal hyperoxia induces increased tracheal contractility, attenuates tracheal relaxation, diminishes lung antioxidant capacity, and increases lung inflammation, while monocarbonyl CUR analogs were protective of these adverse effects of hyperoxia. Analogs may be promising new therapies for neonatal hyperoxic airway and lung disease.


Subject(s)
Bronchial Hyperreactivity/drug therapy , Curcumin/analogs & derivatives , Hyperoxia/drug therapy , Muscle Relaxation , Muscle, Smooth/drug effects , Animals , Catalase/metabolism , Curcumin/pharmacology , Female , Lung/metabolism , Male , Muscle Contraction , Muscle, Smooth/physiology , Rats , Rats, Wistar , Trachea/cytology , Trachea/drug effects , Trachea/physiology , Tumor Necrosis Factor-alpha/metabolism
18.
Am J Physiol Lung Cell Mol Physiol ; 319(3): L497-L512, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32697651

ABSTRACT

Hyperoxia (HO)-induced lung injury contributes to bronchopulmonary dysplasia (BPD) in preterm newborns. Intractable wheezing seen in BPD survivors is associated with airway remodeling (AWRM). Sphingosine kinase 1 (SPHK1)/sphingosine-1-phosphate (S1P) signaling promotes HO-mediated neonatal BPD; however, its role in the sequela of AWRM is not known. We noted an increased concentration of S1P in tracheal aspirates of neonatal infants with severe BPD, and earlier, demonstrated that Sphk1-/- mice showed protection against HO-induced BPD. The role of SPHK1/S1P in promoting AWRM following exposure of neonates to HO was investigated in a murine model. Therapy using PF543, the specific SPHK1 inhibitor, during neonatal HO reduced alveolar simplification followed by reduced AWRM in adult mice. This was associated with reduced airway hyperreactivity to intravenous methacholine. Neonatal HO exposure was associated with increased expression of SPHK1 in lung tissue of adult mice, which was reduced with PF543 therapy in the neonatal stage. This was accompanied by amelioration of HO-induced reduction of E-cadherin in airway epithelium. This may be suggestive of arrested partial epithelial mesenchymal transition (EMT) induced by HO. In vitro studies using human primary airway epithelial cells (HAEpCs) showed that SPHK1 inhibition or deletion restored HO-induced reduction in E-cadherin and reduced formation of mitochondrial reactive oxygen species (mtROS). Blocking mtROS with MitoTempo attenuated HO-induced partial EMT of HAEpCs. These results collectively support a therapeutic role for PF543 in preventing HO-induced BPD in neonates and the long-term sequela of AWRM, thus conferring a long-term protection resulting in improved lung development and function.


Subject(s)
Airway Remodeling/drug effects , Bronchopulmonary Dysplasia/drug therapy , Hyperoxia/drug therapy , Methanol/analogs & derivatives , Pyrrolidines/pharmacology , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Disease Models, Animal , Hyperoxia/chemically induced , Lung/drug effects , Lung/metabolism , Methanol/pharmacology , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sulfones
19.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 36(1): 73-76, 2020 Jan 28.
Article in Chinese | MEDLINE | ID: mdl-32476376

ABSTRACT

OBJECTIVE: To find if edaravone can play a protective role in a mouse model of pulmonary oxygen toxicity and explore the intervention mechanism. METHODS: Thirty male C57BL/6 mice were randomly divided into 3 groups(Air +Vehicle, Hyperbaric oxygen(HBO) +Vehicle and HBO + Edaravone). Mice were either given edaravone (5 mg/(kg·d)) in sterilized water or a sterilized water vehicle for 3 days before oxygen exposure. Mice in HBO groups were exposed to 0.23 MPa hyperoxia (≥95% O2) for 6 h. Lung tissues were collected and the wet/dry ratio of lung were analyzed. For histologic analysis, lung sections were stained with hematoxylin and eosin (HE). Proinflammatory cytokine levels and antioxidant enzyme activities in lungs were determined by using ELISA kits. The expression levels of pro-apoptosis protein were determined with Western blot analysis. RESULTS: Edaravone treatment could significantly reduce lung permeability, decrease tissue pro-apoptosis protein (cleaved-caspase3) and inflammation (IL-1ß). However, edaravone treatment had no effect on antioxidant enzyme activities. CONCLUSION: These results showed that edaravone treatment had a protective role in pulmonary oxygen toxicity through curbing inflammation and apoptosis.


Subject(s)
Edaravone/therapeutic use , Hyperoxia/drug therapy , Oxygen/toxicity , Protective Agents/therapeutic use , Animals , Apoptosis , Inflammation , Lung , Male , Mice , Mice, Inbred C57BL , Random Allocation
20.
Med Sci Monit ; 26: e920467, 2020 May 21.
Article in English | MEDLINE | ID: mdl-32437336

ABSTRACT

BACKGROUND Hyperoxic acute lung injury (ALI) is a complication of ventilation in patients with respiratory failure. Nuclear factor erythroid-2-related factor 2 (Nrf2) has an important role in ALI. Kelch-like ECH-associated protein 1 (Keap1) binds to Nrf2. ZJ01 is a small molecule inhibitor of Keap1-Nrf2 protein-protein interaction (PPI) that can reduce Keap1-induced inhibition of Nrf2. This study aimed to investigate the effects of ZJ01 and the heme oxygenase-1 (HO-1) inhibitor, zinc protoporphyrin IX (ZnPP IX), in a mouse model of hyperoxic ALI. MATERIAL AND METHODS C57BL/6J mice included five study groups: the room air+vehicle-treated group; the room air+ZJ01 group; the hyperoxia+vehicle-treated group; the hyperoxia+ZJ01 group; and the hyperoxia+ZJ01+ZnPP IX group. ZJ01, ZnPP IX, or vehicle were given 1 h after the hyperoxia challenge. The lungs from the mice were harvested at 72 h following the hyperoxia challenge. RESULTS Hyperoxia exposure for 72 h increased the activity of myeloperoxidase, the lung water content, the levels of tumor necrosis factor-alpha (TNF-alpha), and matrix metalloprotease-9 (MMP-9) in the vehicle-treated mice. ZJ01 treatment reduced hyperoxia-induced inflammation and increased the activation of Nrf2 and HO-1 compared with the vehicle-treated mice. Histology of the lungs showed that ZJ01 treatment reduced the changes of hyperoxia-induced ALI. Pretreatment with ZnPP IX reversed the beneficial effect of ZJ01. CONCLUSIONS ZJ01, a Keap1-Nrf2 PPI inhibitor, reduced hyperoxic ALI in a mouse model through the Nrf2/HO-1 pathway.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Benzothiazoles/pharmacology , Kelch-Like ECH-Associated Protein 1/antagonists & inhibitors , NF-E2-Related Factor 2/antagonists & inhibitors , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/metabolism , Hyperoxia/drug therapy , Hyperoxia/metabolism , Hyperoxia/pathology , Kelch-Like ECH-Associated Protein 1/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Protein Interaction Domains and Motifs/drug effects , Protoporphyrins/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...