Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 12(6)2023 Jun 11.
Article in English | MEDLINE | ID: mdl-37371988

ABSTRACT

This study investigates the role of eugenol (EUG) on CS-induced acute lung injury (ALI) and how this compound is able to modulate macrophage activity. C57BL/6 mice were exposed to 12 cigarettes/day/5days and treated 15 min/day/5days with EUG. Rat alveolar macrophages (RAMs) were exposed to CSE (5%) and treated with EUG. In vivo, EUG reduced morphological changes inflammatory cells, oxidative stress markers, while, in vitro, it induced balance in the oxidative stress and reduced the pro-inflammatory cytokine release while increasing the anti-inflammatory one. These results suggest that eugenol reduced CS-induced ALI and acted as a modulator of macrophage activity.

2.
Antioxidants (Basel) ; 12(3)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36978796

ABSTRACT

Acute and chronic lung injuries are among the leading causes of mortality worldwide. Lung injury can affect several components of the respiratory system, including the airways, parenchyma, and pulmonary vasculature. Although acute and chronic lung injuries represent an enormous economic and clinical burden, currently available therapies primarily focus on alleviating disease symptoms rather than reversing and/or preventing lung pathology. Moreover, some supportive interventions, such as oxygen and mechanical ventilation, can lead to (further) deterioration of lung function and even the development of permanent injuries. Lastly, sepsis, which can originate extrapulmonary or in the respiratory system itself, contributes to many cases of lung-associated deaths. Considering these challenges, we aim to summarize molecular and cellular mechanisms, with a particular focus on airway inflammation and oxidative stress that lead to the characteristic pathophysiology of acute and chronic lung injuries. In addition, we will highlight the limitations of current therapeutic strategies and explore new antioxidant-based drug options that could potentially be effective in managing acute and chronic lung injuries.

3.
Antioxidants (Basel) ; 11(9)2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36139733

ABSTRACT

BACKGROUND: Gold nanoparticles (AuNPs) can inhibit pivotal pathological changes in experimental asthma, but their effect on steroid-insensitive asthma is unclear. The current study assessed the effectiveness of nebulized AuNPs in a murine model of glucocorticoid (GC)-resistant asthma. METHODS: A/J mice were sensitized and subjected to intranasal instillations of ovalbumin (OVA) once a week for nine weeks. Two weeks after starting allergen stimulations, mice were subjected to Budesonide or AuNP nebulization 1 h before stimuli. Analyses were carried out 24 h after the last provocation. RESULTS: We found that mice challenged with OVA had airway hyperreactivity, eosinophil, and neutrophil infiltrates in the lung, concomitantly with peribronchiolar fibrosis, mucus production, and pro-inflammatory cytokine generation compared to sham-challenged mice. These changes were inhibited in mice treated with AuNPs, but not Budesonide. In the GC-resistant asthmatic mice, oxidative stress was established, marked by a reduction in nuclear factor erythroid 2-related factor 2 (NRF2) levels and catalase activity, accompanied by elevated values of thiobarbituric acid reactive substances (TBARS), phosphoinositide 3-kinases δ (PI3Kδ) expression, as well as a reduction in the nuclear expression of histone deacetylase 2 (HDAC2) in the lung tissue, all of which sensitive to AuNPs but not Budesonide treatment. CONCLUSION: These findings suggest that AuNPs can improve GC-insensitive asthma by preserving HDAC2 and NRF2.

4.
Antioxidants (Basel) ; 11(7)2022 Jun 29.
Article in English | MEDLINE | ID: mdl-35883784

ABSTRACT

The use of annatto pigments has been evaluated as a therapeutic strategy in animal models of several health disorders. Beneficial effects were generally attributed to the inhibition of oxidative stress. Bixin is the main pigment present in annatto seeds and has emerged as an important scavenger of reactive oxygen (ROS) and nitrogen species (RNS). However, this carotenoid is highly hydrophobic, affecting its therapeutic applicability. Therefore, bixin represents an attractive target for nanotechnology to improve its pharmacokinetic parameters. In this study, we prepared bixin nanoparticles (npBX) and evaluated if they could prevent pulmonary inflammation and oxidative stress induced by cigarette smoke (CS). C57BL/6 mice were exposed to CS and treated daily (by gavage) with different concentrations of npBX (6, 12 and 18%) or blank nanoparticles (npBL, 18%). The negative control group was sham smoked and received 18% npBL. On day 6, the animals were euthanized, and bronchoalveolar lavage fluid (BALF), as well as lungs, were collected for analysis. CS exposure led to an increase in ROS and nitrite production, which was absent in animals treated with npBX. In addition, npBX treatment significantly reduced leukocyte numbers and TNF-α levels in the BALF of CS-exposed mice, and it strongly inhibited CS-induced increases in MDA and PNK in lung homogenates. Interestingly, npBX protective effects against oxidative stress seemed not to act via Nrf2 activation in the CS + npBX 18% group. In conclusion, npBX prevented oxidative stress and acute lung inflammation in a murine model of CS-induced acute lung inflammation.

5.
Phytother Res ; 36(2): 1032-1042, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35028976

ABSTRACT

Obesity is a chronic condition involving inflammation and oxidative stress that commonly predisposes affected individuals to develop metabolic disorders. We hypothesize that Ilex paraguariensis (IP) can modulate oxidative stress and inflammation underpinning metabolic disorders caused by obesity. C57BL/6 mice were fed a high-fat diet (HFD group) for 12 weeks. Concomitantly, some mice were treated with roasted IP (15 mg/ml - HFD + IP) or dimethyl fumarate (DMF) as a positive control (2 mg/ml - HFD + DMF). The control group received standard chow and water ad libitum. Histological analyses of fat tissue and liver, and quantification of mediators related to oxidative stress (Kelch-like ECH-associated protein 1/NF-E2-related factor 2, NADP(H) quinone oxidoreductase-1 [NQO1], heme oxygenase 1 [HO1], and superoxide dismutase) as well as metabolic profile blood biomarkers (glucose, leptin, resistin, high-density lipoproteins [HDLs], and triglycerides) were performed. Metabolic disorders were prevented in mice treated with IP, as evidenced by the observation that glucose, HDL, and resistin levels were similar to those assessed in the control group. Morphological analyses showed that both IP and DMF treatments prevented hepatic steatosis and adipocyte hypertrophy in visceral adipose tissue. Finally, although the antioxidant response stimulated by IP was quite limited, significant effects were found on NQO1 and HO1 expression. In conclusion, IP has promising preventative effects on the development of metabolic disorders caused by obesity.


Subject(s)
Ilex paraguariensis , Metabolic Diseases , Animals , Diet, High-Fat/adverse effects , Liver , Metabolic Diseases/drug therapy , Mice , Mice, Inbred C57BL , Obesity/metabolism , Plant Extracts/metabolism , Plant Extracts/pharmacology
6.
J Nutr Biochem ; 98: 108834, 2021 12.
Article in English | MEDLINE | ID: mdl-34371126

ABSTRACT

Citrate is widely used as a food additive being part of virtually all processed foods. Although considered inert by most of the regulatory agencies in the world, plasma citrate has been proposed to play immunometabolic functions in multiple tissues through altering a plethora of cellular pathways. Here, we used a short-term alimentary intervention (24 hours) with standard chow supplemented with citrate in amount corresponding to that found in processed foods to evaluate its effects on glucose homeostasis and liver physiology in C57BL/6J mice. Animals supplemented with dietary citrate showed glucose intolerance and insulin resistance as revealed by glucose and insulin tolerance tests. Moreover, animals supplemented with citrate in their food displayed fed and fasted hyperinsulinemia and enhanced insulin secretion during an oral glucose tolerance test. Citrate treatment also amplified glucose-induced insulin secretion in vitro in INS1-E cells. Citrate supplemented animals had increased liver PKCα activity and altered phosphorylation at serine or threonine residues of components of insulin signaling including IRS-1, Akt, GSK-3 and FoxO1. Furthermore, citrate supplementation enhanced the hepatic expression of lipogenic genes suggesting increased de novo lipogenesis, a finding that was reproduced after citrate treatment of hepatic FAO cells. Finally, liver inflammation markers were higher in citrate supplemented animals. Overall, the results demonstrate that dietary citrate supplementation in mice causes hyperinsulinemia and insulin resistance both in vivo and in vitro, and therefore call for a note of caution on the use of citrate as a food additive given its potential role in metabolic dysregulation.


Subject(s)
Citric Acid/pharmacology , Inflammation/metabolism , Insulin Resistance , Liver/metabolism , Animals , Citric Acid/adverse effects , Diet , Glucose/metabolism , Glucose Intolerance/metabolism , Glucose Tolerance Test/methods , Glycogen Synthase Kinase 3/metabolism , Hepatocytes/metabolism , Homeostasis , Hyperinsulinism/etiology , Insulin/metabolism , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology
7.
Pulm Pharmacol Ther ; 69: 102053, 2021 08.
Article in English | MEDLINE | ID: mdl-34214692

ABSTRACT

INTRODUCTION: Cigarette smoke (CS) is the main risk factor for the development of chronic obstructive pulmonary disease (COPD) and pulmonary emphysema. The use of antioxidants has emerged as a potential therapeutic strategy to treat airway inflammation and lung diseases. In the current study, we investigated the potential therapeutic impact of diallyl disulfide (Dads) treatment in a murine model of CS-induced emphysema. METHODS: C57BL/6 mice were exposed to CS for 60 consecutive days and treated with vehicle or Dads (30, 60 or 90 mg/kg) by oral gavage for the last 30 days, three times/week. The control group was sham-smoked and received vehicle treatment. All mice were euthanized 24 h after day 60; bronchoalveolar lavage (BAL) was performed and lungs were processed for further experimentation. Histological (HE stained sections, assessment of mean linear intercept (Lm)), biochemical (nitrite, superoxide dismutase (SOD), glutathione transferase (GST), and malondialdehyde (MDA) equivalents), and molecular biology (metalloproteinase (MMP) 12, SOD2, carbonyl reductase 1 (CBR1), nitrotyrosine (PNK), 4-hydroxynonenal (4-HNE), and CYP2E1) analyses were performed. RESULTS: Treatment with Dads dose-dependently reduced CS-induced leukocyte infiltration into the airways (based on BAL fluid counts) and improved lung histology (indicated by a reduction of Lm). Furthermore, CS exposure dramatically reduced the activity of the antioxidant enzymes SOD and GST in lung tissue and increased nitrite and MDA levels in BAL; these effects were all effectively counteracted by Dads treatment. Western blot analysis further confirmed the antioxidant potential of Dads, showing that treatment prevented the CS-induced decrease in SOD2 expression and increase in lung damage markers, such as CBR1, PNK, and 4-HNE. Furthermore, increased MMP12 (an important hallmark of CS-induced emphysema) and CYP2E1 lung protein levels were significantly reduced in mice receiving Dads treatment. CONCLUSION: Our findings demonstrate that treatment with Dads is effective in preventing multiple pathological features of CS-induced emphysema in an in vivo mouse model. In addition, we have identified several proteins/enzymes, including 4-HNE, CBR1, and CYP2E1, that are modifiable by Dads and could represent specific therapeutic targets for the treatment of COPD and emphysema.


Subject(s)
Emphysema , Pulmonary Emphysema , Allyl Compounds , Animals , Bronchoalveolar Lavage Fluid , Disulfides , Lung , Mice , Mice, Inbred C57BL , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/etiology , Pulmonary Emphysema/prevention & control , Smoke/adverse effects , Smoking
8.
Food Res Int ; 123: 697-703, 2019 09.
Article in English | MEDLINE | ID: mdl-31285019

ABSTRACT

The efficacy of probiotic Prato cheese against the inflammatory and oxidative damage in mice organs induced by cigarette smoke exposure was investigated. Forty C57BL/6 male mice were assigned to four groups: (CS) exposed to cigarette smoke and fed regular chow; (CS + C) exposed to cigarette smoke and fed daily conventional cheese ad libitum; (CS + PC) exposed to cigarette smoke and fed daily probiotic (Lactobacillus casei-01) cheese ad libitum; and a control group (C) exposed to ambient smoke-free air and fed regular chow. Bronchoalveolar lavage (BAL), blood, gut and liver homogenates were used for biochemical assays. The (CS + PC) group exhibited fewer BAL leukocytes, reactive oxygen species (ROS), and BAL and gut lipid peroxidation than the (CS) and (CS + C) groups, which had findings similar to the (C) group. Probiotic cheese consumption did not change the red blood cell count, but lower lactate dehydrogenase (LDH) levels in plasma, inducible nitric oxide synthase (iNOS) and peroxynitrite expression were observed compared to the (CS) and (CS + C) groups, with findings similar to the (C) group. These results suggest that probiotic Prato cheese consumption reduced oxidative stress in the lungs, gut, and liver.


Subject(s)
Cheese , Cigarette Smoking , Lung Injury , Probiotics , Animals , Male , Mice , Cheese/microbiology , Cigarette Smoking/adverse effects , Disease Models, Animal , Lacticaseibacillus casei/physiology , Lipid Peroxidation , Lung/pathology , Lung Injury/drug therapy , Mice, Inbred C57BL , Nitric Oxide Synthase/metabolism , Oxidative Stress/drug effects , Probiotics/therapeutic use , Reactive Oxygen Species/metabolism
9.
Inflammation ; 42(2): 526-537, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30411213

ABSTRACT

Air pollution caused by fuel burning contributes to respiratory impairments that may lead to death. We aimed to investigate the effects of biodiesel (DB) burning in mouse lungs. DB particulate matter was collected from the exhaust pipes of a bus engine. Mice were treated with 250 µg or 1000 µg of DB particulate matter by intranasal instillation over 5 consecutive days. We demonstrated that DB particulate matter penetrated the lung in the 250-µg and 1000-µg groups. In addition, the DB particulate matter number in pulmonary parenchyma was 175-fold higher in the 250-µg group and 300-fold higher in the 1000-µg group compared to control mice. The instillation of DB particulate matter increased the macrophage number and protein levels of TNF-alpha in murine lungs. DB particulate matter enhanced ROS production in both exposed groups and the malondialdehyde levels compared to the control group. The protein expression levels of Nrf2, p-NF-kB, and HO-1 were higher in the 250-µg group and lower in the 1000-µg group than in control mice and the 250-µg group. In conclusion, DB particulate matter instillation promotes oxidative stress by activating the Nrf2/HO-1 and inflammation by p-NF-kB/TNF-alpha pathways.


Subject(s)
Environmental Exposure/adverse effects , Lung/metabolism , Oxidative Stress/drug effects , Particulate Matter/adverse effects , Vehicle Emissions/toxicity , Animals , Heme Oxygenase-1/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Membrane Proteins/metabolism , Metabolic Networks and Pathways , Mice , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Particulate Matter/toxicity , Tumor Necrosis Factor-alpha/metabolism
10.
Biomed Pharmacother ; 109: 1547-1555, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30551407

ABSTRACT

High-fat diet (HFD)-induced obesity is a worldwide health problem and can cause lipid accumulation in the liver. We evaluated the hepatoprotective effect of mate tea treatment in mice submitted to an HFD. C57BL/6 mice were fed an HFD for 13 weeks with and without mate tea. A separate group of mice was treated with fenofibrate as a positive control (a regular drug for lipid disorders). Histological analyses, glucose tolerance tests (GTT), and quantification of mediators related to lipid peroxidation, oxidative stress and blood biomarkers for lipid profile were performed. The weight of animals and major organs related to hepatic steatosis was determined, and proinflammatory cytokines and the participation of the Nrf2 pathway and adiponectin were evaluated. Mate tea prevented the accumulation of lipid droplets in hepatocytes as well as weight gain in animals submitted to the HFD. Mate tea treatment also prevented increases in the liver weight, heart weight and amount of visceral and subcutaneous white adipose tissue. Mate tea was able to prevent the deregulation of glucose uptake, as evaluated by GTT, and improved the indicators of oxidative stress, such as nitrite levels, catalase activity, and oxidative damage, as evaluated by protein carbonylation and the MDA levels. Mate tea had an anti-inflammatory effect, preventing the increase of IL-1ß and KC and upregulating the expression of Nrf2. Mate tea prevented insulin increase and HDL cholesterol decrease but did not affect total cholesterol or triglycerides levels. Treatment also prevented adiponectin increase. Mate tea may be a good resource to reduce hepatic steatosis in the future since it has anti-diabetic, anti-inflammatory and antioxidant effects, which prevent the accumulation of fat in the liver.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/drug therapy , Liver/drug effects , Metabolic Diseases/drug therapy , Plant Extracts/pharmacology , Tea/chemistry , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Fatty Liver/metabolism , Glucose Tolerance Test/methods , Hepatocytes/drug effects , Hepatocytes/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Liver/metabolism , Male , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Obesity/drug therapy , Obesity/metabolism , Oxidative Stress/drug effects
11.
Biomed Pharmacother ; 102: 160-168, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29554594

ABSTRACT

Emphysema results in a proteinase - antiproteinase imbalance, inflammation and oxidative stress. Our objective was to investigate whether atorvastatin could repair mouse lungs after elastase-induced emphysema. Vehicle (50 µL) or porcine pancreatic elastase (PPE) was administered on day 1, 3, 5 and 7 at 0.6 U intranasally. Male mice were divided into a control group (sham), PPE 32d (sacrificed 24 h after 32 days), PPE 64d (sacrificed 24 h after 64 days), and atorvastatin 1, 5 and 20 mg treated from day 33 until day 64 and sacrificed 24 h later (A1 mg, A5 mg and A20 mg, respectively). Treatment with atorvastatin was performed via inhalation for 10 min once a day. We observed that emphysema at day 32 was similar to emphysema at day 64. The mean airspace chord length (Lm) indicated a recovery of pulmonary morphology in groups A5 mg and A20 mg, as well as recovery of collagen and elastic fibers in comparison to the PPE group. Bronchoalveolar lavage fluid (BALF) leukocytes were reduced in all atorvastatin-treated groups. However, tissue macrophages were reduced only in the A20 mg group compared with the PPE group, while tissue neutrophils were reduced in the A5 mg and A20 mg groups. The redox balance was restored mainly in the A20 mg group compared with the PPE group. Finally, atorvastatin at doses of 5 and 20 mg reduced nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and matrix metalloproteinase-12 (MMP-12) compared with the PPE group. In conclusion, atorvastatin was able to induce lung tissue repair in emphysematous mice.


Subject(s)
Atorvastatin/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation/drug therapy , Pulmonary Emphysema/drug therapy , Animals , Atorvastatin/administration & dosage , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Dose-Response Relationship, Drug , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Inflammation/pathology , Leukocytes/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Oxidative Stress/drug effects , Pancreatic Elastase/toxicity , Pulmonary Emphysema/physiopathology , Swine , Time Factors
12.
Int Immunopharmacol ; 56: 330-338, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29438939

ABSTRACT

Long-term exposure to cigarette smoke (CS) results in alveolar parenchyma destruction due to chronic inflammatory response and the imbalance between oxidants and antioxidants, and proteases and antiproteases. Emphysema is the main symptom of chronic obstructive pulmonary disease. Current treatment focuses on relieving respiratory symptoms, and inflammation resolution failure is an important pathophysiological element of the disease. Specialized pro-resolving mediators (SPMs) synthesized endogenously during resolution processes demonstrated beneficial effects in murine models of airway inflammation. Here, we aimed to test the SPM AT-RvD1 in a murine model of CS-induced emphysema. AT-RvD1 restored elastic fibers and lung morphology, with reduction in MMP-3, neutrophils, and myeloperoxidase activity and increases in macrophages and IL-10 levels. AT-RvD1 also decreased levels of oxidative stress markers and ROS via upregulation of the Nrf2/Keap1 pathway. Therefore, we suggest that AT-RvD1 causes pro-resolutive action in our murine model of CS-induced emphysema by upregulation of the Nrf2/Keap1 pathway.


Subject(s)
Anti-Inflammatory Agents/metabolism , Docosahexaenoic Acids/metabolism , Emphysema/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Cigarette Smoking/adverse effects , Disease Models, Animal , Docosahexaenoic Acids/chemistry , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Signal Transduction
13.
Bioorg Med Chem ; 25(20): 5557-5568, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28886998

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is an incurable and progressive disease. Emphysema is the principal manifestation of COPD, and the main cause of this condition is cigarette smoke (CS). Natural products have shown antioxidant and anti-inflammatory properties that can prevent acute lung inflammation and emphysema, but there are few reports in the literature regarding therapeutic approaches to emphysema. We hypothesized that supplementation with natural extracts would repair lung damage in emphysema caused by CS exposure. Mice were exposed to 60days of CS and then treated or not with three different natural extracts (mate tea, grape and propolis) orally for additional 60days. Histological analysis revealed significant improvements in lung histoarchitecture, with recovery of alveolar spaces in all groups treated with natural extracts. Propolis was also able to recovery alveolar septa and elastic fibers. Propolis also increased MMP-2 and decreased MMP-12 expression, favoring the process of tissue repair. Additionally, propolis recruited leukocytes, including macrophages, without ROS release. These findings led us to investigate the profile of these macrophages, and we showed that propolis could promote macrophage alternative activation, thus increasing the number of arginase-positive cells and IL-10 levels and favoring an anti-inflammatory microenvironment. We further investigated the participation of Nrf2 in lung repair, but no Nrf2 translocation to the nucleus was observed in lung cells. Proteins and enzymes related to Nrf2 were not altered, other than NQO1, which seemed to be activated by propolis in a Nrf2-independent manner. Finally, propolis downregulated IGF1 expression. In conclusion, propolis promoted lung repair in a mouse emphysema model via macrophage polarization from M1 to M2 in parallel to the downregulation of IGF1 expression in a Nrf2-independent manner.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , NF-E2-Related Factor 2/metabolism , Propolis/pharmacology , Pulmonary Emphysema/drug therapy , Smoking/drug therapy , Animals , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Smoking/metabolism
14.
Free Radic Res ; 51(7-8): 708-722, 2017.
Article in English | MEDLINE | ID: mdl-28776450

ABSTRACT

Asthma is an allergic inflammation driven by the Th2 immune response with release of cytokines such as IL-4 and IL-13, which contribute to the airflow limitations and airway hyperresponsiveness (AHR). The involvement of oxidative stress in this process is well-established, but the specific role of the superoxide anion and nitric oxide in asthma are poorly understood. Thus, the aim of this study was to investigate the mechanisms underlying the superoxide anion/nitric oxide production and detoxification in a murine asthma model. BALB/c male mice were sensitised and challenged with ovalbumin (OVA). Pretreatments with either apocynin (14 mg/kg) or allopurinol (25 mg/kg) (superoxide anion synthesis inhibitors), aminoguanidine (50 mg/kg) (nitric oxide synthesis inhibitor) or diethyldithiocarbamate (100 mg/kg) (superoxide dismutase inhibitor) were performed 1 h before the challenge. Our data showed that apocynin and allopurinol ameliorated AHR and reduced eosinophil peroxidase, as well as IL-4 and IL-13 levels. Apocynin also abrogated leukocyte peribronchiolar infiltrate and increased IL-1ß secretion. Aminoguanidine preserved lung function and shifted the Th2 to the Th1 response with a reduction of IL-4 and IL-13 and increase in IL-1ß production. Diethyldithiocarbamate prevented neither allergen-induced AHR nor eosinophil peroxidase (EPO) generation. All treatments protected against oxidative damage observed by a reduction in TBARS levels. Taken together, these results suggest that AHR in an asthma model can be avoided by the down-regulation of superoxide anion and nitric oxide synthesis in a mechanism that is independent of a redox response. This down-regulation is also associated with a transition in the typical immunological Th2 response toward the Th1 profile.


Subject(s)
Asthma/immunology , Inflammation/immunology , Nitric Oxide/antagonists & inhibitors , Respiratory Hypersensitivity/immunology , Superoxides/antagonists & inhibitors , Acetophenones/administration & dosage , Allopurinol/administration & dosage , Animals , Asthma/metabolism , Asthma/pathology , Disease Models, Animal , Eosinophil Peroxidase/immunology , Eosinophil Peroxidase/metabolism , Guanidines/administration & dosage , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Inflammation/metabolism , Inflammation/pathology , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Mice , Nitric Oxide/immunology , Ovalbumin/immunology , Oxidative Stress/immunology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , Superoxides/immunology , Th1 Cells/immunology , Th2 Cells/immunology
15.
Inflammation ; 40(5): 1487-1496, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28534139

ABSTRACT

Elastase (PPE) is usually used for emphysema models, whereas bleomycin (BLM) is used for fibrosis models. The aim of this study was to investigate the effect of BLM in PPE-induced emphysema, as well as the effect of PPE in BLM-induced fibrosis. C57BL/6 mice were divided into five groups: control, PPE, BLM, PPE + BLM, and BLM + PPE. Mice received saline, PPE (3 U/mouse), or BLM (20 U/kg) by intranasal instillation. Mice from the BLM and BLM + PPE groups received BLM on day 0 and saline or PPE on day 21, respectively. Those in the PPE and PPE + BLM groups received PPE on day 0 and saline or BLM on day 21, respectively. Mice were euthanized on day 42. We performed histology, morphometry in lung sections and ELISA, zymography and western blotting in BAL samples or lung homogenates. In the lungs of PPE + BLM and BLM + PPE groups, we observed inflammation, oxidative stress and expression of MMP-2 and MMP-9. The alveolar enlargement was reduced in the PPE + BLM group, suggesting that the BLM could participate in the alveolar remodeling process. The significance of this result supports future therapeutic approaches targeting extracellular-matrix deposition in patients with emphysema as a way to repair the enlargement of alveoli and airspaces.


Subject(s)
Bleomycin/therapeutic use , Pancreatic Elastase/therapeutic use , Pulmonary Emphysema/drug therapy , Pulmonary Fibrosis/drug therapy , Animals , Bleomycin/adverse effects , Inflammation/chemically induced , Lung/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Oxidative Stress/drug effects , Pancreatic Elastase/adverse effects , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Emphysema/chemically induced , Pulmonary Fibrosis/chemically induced
16.
Inflammation ; 40(4): 1166-1176, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28391514

ABSTRACT

Ovalbumin-induced allergic lung inflammation (ALI) is a condition believed to be mediated by cytokines, extracellular matrix remodeling, and redox imbalance. In this study, we evaluated pulmonary function together with inflammatory markers as interleukin-4 (IL-4), myeloperoxidase (MPO), eosinophil cells, and redox markers in the lungs of BALB/c mice after ovalbumin (OVA) sensitization and challenge. Our results showed an increase in bronchial hyperresponsiveness stimulated by methacholine (Mch), inflammatory cell influx, especially eosinophils together with an increase of high mobility group box 1 (HMGB1) and altered lipid peroxidation (LP) and antioxidant defenses in the OVA group compared to the control group (p ≤ 0.5). Thus, we demonstrated that OVA-induced ALI altered redox status concomitantly with impaired lung function, which was associated with HMGB1 expression and proteolytic remodeling. Taken together all results found here, we may suggest HMGB1 is an important therapeutic target for asthma, once orchestrates the redox signaling, inflammation, and remodeling that contribute to the disease development.


Subject(s)
Asthma/metabolism , Asthma/pathology , HMGB1 Protein/metabolism , Inflammation , Oxidative Stress , Animals , Biomarkers/analysis , Bronchial Hyperreactivity , Eosinophils , Inflammation/diagnosis , Inflammation/immunology , Lipid Peroxidation , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Oxidative Stress/immunology , Proteolysis
17.
Inflammation ; 40(3): 965-979, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28251446

ABSTRACT

Cigarette smoke (CS) induces pulmonary emphysema by inflammation, oxidative stress, and metalloproteinase (MMP) activation. Pharmacological research studies have not focused on tissue repair after the establishment of emphysema but have instead focused on inflammatory stimulation. The aim of our study was to analyze the effects of atorvastatin and simvastatin on mouse lung repair after emphysema caused by CS. Male mice (C57BL/6, n = 45) were divided into the following groups: control (sham-exposed), CSr (mice exposed to 12 cigarettes a day for 60 days and then treated for another 60 days with the vehicle), CSr+A (CSr mice treated with atorvastatin for 60 days), and CSr+S (CSr mice treated with simvastatin for 60 days). The treatment with atorvastatin and simvastatin was administered via inhalation (15 min with 1 mg/mL once a day). Mice were sacrificed 24 h after the completion of the 120-day experimental procedure. We performed biochemical, morphological, and physiological analyses. We observed decreased levels of leukocytes and cytokines in statin-treated mice, accompanied by a reduction in oxidative stress markers. We also observed a morphological improvement confirmed by a mean linear intercept counting in statin-treated mice. Finally, statins also ameliorated lung function. We conclude that inhaled atorvastatin and simvastatin improved lung repair after cigarette smoke-induced emphysema in mice.


Subject(s)
Atorvastatin/pharmacology , Lung/drug effects , Pulmonary Emphysema/drug therapy , Simvastatin/pharmacology , Animals , Atorvastatin/therapeutic use , Male , Mice , Mice, Inbred C57BL , Pulmonary Emphysema/etiology , Simvastatin/therapeutic use , Smoking/adverse effects
18.
Pulm Pharmacol Ther ; 41: 11-18, 2016 12.
Article in English | MEDLINE | ID: mdl-27599597

ABSTRACT

Short-term cigarette smoke (CS) exposure does not cause emphysema; however, some pathogenesis hallmarks are maintained, such as oxidative stress and inflammation. This study aimed to test the efficacy of eucalyptol against short-term CS exposure in mice. C57BL/6 mice were exposed to 12 cigarettes per day for 5 days (CS group). The control group was exposed to sham smoking. Three groups of mice exposed to CS were treated to different concentrations of eucalyptol (1, 3, 10 mg/mL) via inhalation (15 min/daily) for 5 days (CS + 1 mg, CS+3 mg and CS+10 mg groups). CS group and control one were sham treated by using vehicle. The anti-inflammatory and antioxidant effects of eucalyptol were assessed 24 h after the last CS exposure by determining cell counts, measuring cytokine production and performing western blotting, biochemical and histological analyses. Eucalyptol at 3 mg/mL and 10 mg/mL concentrations reduced total leukocyte numbers compared to the CS group (p < 0.001), while macrophage numbers were reduced at all concentrations (p < 0.001). Myeloperoxidase, used as neutrophil marker, was reduced at 3 mg/mL (p < 0.01) and 10 mg/mL (p < 0.05) concentrations. Eucalyptol reduced cytokine levels (IL-1ß, IL-6 and KC) at 3 mg/mL and 10 mg/mL concentrations (p < 0.01) compared to the CS group. The exception was TNF-α, with a reduction only at 10 mg/mL of eucalyptol compared to the CS group (p < 0.001). Additionally, eucalyptol decreased the NF-kappa B p65 subunit at 3 mg/mL and 10 mg/mL compared to the CS group (p < 0.01). Regarding oxidative stress, eucalyptol reduced reactive oxygen species, superoxide dismutase, catalase and malondialdehyde, mainly at 3 mg/mL and 10 mg/mL concentrations compared to the CS group (at least p < 0.05), parallel to reduced glutathione levels at the same concentrations (p < 0.001). Furthermore, treatment with eucalyptol attenuated CS-induced histopathological alterations. Collectively, these results indicate that eucalyptol acts through a mechanism involving decreased oxidative stress, inflammation and the NF-kappa B p65 subunit against CS-induced acute lung inflammation. Thus, eucalyptol may be a potential agent in the treatment of pulmonary inflammation caused by CS in humans.


Subject(s)
Cyclohexanols/pharmacology , Monoterpenes/pharmacology , Oxidative Stress/drug effects , Pneumonia/prevention & control , Smoking/adverse effects , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Antioxidants/administration & dosage , Antioxidants/pharmacology , Cyclohexanols/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Eucalyptol , Inflammation/pathology , Inflammation/prevention & control , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Monoterpenes/administration & dosage , Neutrophils/metabolism , Peroxidase/metabolism , Pneumonia/etiology , Reactive Oxygen Species/metabolism , Smoke/adverse effects , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Acta Histochem ; 118(3): 203-12, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26852294

ABSTRACT

Pulmonary fibrosis (PF) is characterized by excessive accumulation of collagen in the lungs. Emphysema is characterized by loss of the extracellular matrix (ECM) and alveolar enlargement. We studied the co-participation of elastase-induced mild emphysema in bleomycin-induced PF in mice by analyzing oxidative stress, inflammation and lung histology. C57BL/6 mice were divided into four groups: control; bleomycin (0.1U/mouse); elastase (using porcine pancreatic elastase (PPE)+bleomycin (3U/mouse 14 days before 0.1U/mouse of bleomycin; PPE+B); elastase (3U/mouse). Mice were humanely sacrificed 7, 14 and 21 days after treatment with bleomycin or vehicle. PF was observed 14 days and 21 days after bleomycin treatment but was observed after 14 days only in the PPE+B group. In the PPE+B group at 21 days, we observed many alveoli and alveolar septa with few PF areas. We also observed marked and progressive increases of collagens 7, 14 and 21 days after bleomycin treatment whereas, in the PPE+B group, collagen deposition was observed only at 14 days. There was a reduction in activities of the antioxidant enzymes superoxide dismutase (p<0.05), catalase (p<0.01) and glutathione peroxidase (p<0.01) parallel with an increase in nitrite (p<0.01) 21 days after bleomycin treatment compared with the control group. These endpoints were also reduced (p<0.05, p<0.05 and p<0.01, respectively) and increased (p<0.01) in the PPE+B group at 21 days compared with the control group. Interleukin (IL)-1ß expression was upregulated (p<0.01) whereas IL-6 was downregulated (p<0.05) in the PPE+B group at 21 days compared with the control group. PF and emphysema did not coexist in our model of lung disease and despite increased levels of oxidative stress and inflammatory markers after combined stimulus (elastase and bleomycin) overall histology was improved to that of the nearest control group.


Subject(s)
Pancreatic Elastase/pharmacology , Pulmonary Fibrosis/chemically induced , Animals , Bleomycin , Catalase/metabolism , Female , Glutathione Peroxidase/metabolism , Interleukins/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Oxidative Stress , Pulmonary Fibrosis/pathology , Superoxide Dismutase/metabolism
20.
Inflammation ; 37(5): 1355-65, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24609836

ABSTRACT

Our aim was to investigate the effects of four different statins on acute lung inflammation induced by cigarette smoke (CS). C57BL/6 male mice were divided into a control group (sham-smoked) and mice exposed to CS from 12 cigarettes/day for 5 days. Mice exposed to CS were grouped and treated with vehicle (i.p.), atorvastatin (10 mg/kg), pravastatin (10 mg/kg), rosuvastatin (5 mg/kg), or simvastatin (20 mg/kg). Treatment with statins differentially improved the pulmonary response when compared to the CS group. Atorvastatin and pravastatin demonstrated slightly effects on inflammation and oxidative stress. Rosuvastatin demonstrated the best anti-inflammatory effect, whereas simvastatin demonstrated the best antioxidant response.


Subject(s)
Fluorobenzenes/pharmacology , Heptanoic Acids/pharmacology , Lung/metabolism , Oxidative Stress/physiology , Pravastatin/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Simvastatin/pharmacology , Smoking/metabolism , Sulfonamides/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Atorvastatin , Fluorobenzenes/therapeutic use , Heptanoic Acids/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Inhalation Exposure/adverse effects , Lung/drug effects , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Pravastatin/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Rosuvastatin Calcium , Simvastatin/therapeutic use , Smoking/drug therapy , Smoking/pathology , Sulfonamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...