1
|
Oxidative Stress, Environmental Pollution, and Lifestyle as Determinants of Asthma in Children. BIOLOGY 2023; 12:biology12010133. [PMID: 36671825 PMCID: PMC9856068 DOI: 10.3390/biology12010133] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
Exposure to cigarette smoke, allergens, viruses, and other environmental contaminants, as well as a detrimental lifestyle, are the main factors supporting elevated levels of airway oxidative stress. Elevated oxidative stress results from an imbalance in reactive oxygen species (ROS) production and efficiency in antioxidant defense systems. Uncontrolled increased oxidative stress amplifies inflammatory processes and tissue damage and alters innate and adaptive immunity, thus compromising airway homeostasis. Oxidative stress events reduce responsiveness to corticosteroids. These events can increase risk of asthma into adolescence and prompt evolution of asthma toward its most severe forms. Development of new therapies aimed to restore oxidant/antioxidant balance and active interventions aimed to improve physical activity and quality/quantity of food are all necessary strategies to prevent asthma onset and avoid in asthmatics evolution toward severe forms of the disease.
Collapse
|
2
|
Mineral Micronutrients in Asthma. Nutrients 2021; 13:nu13114001. [PMID: 34836256 PMCID: PMC8625329 DOI: 10.3390/nu13114001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 01/01/2023] Open
Abstract
Asthma represents one of the most common medical issues in the modern world. It is a chronic inflammatory disease characterized by persistent inflammation of the airways and disturbances in redox status, leading to hyperresponsiveness of bronchi and airway obstruction. Apart from classical risk factors such as air pollution, family history, allergies, or obesity, disturbances of the levels of micronutrients lead to impairments in the defense mechanisms of the affected organism against oxidative stress and proinflammatory stimuli. In the present review, the impact of micronutrients on the prevalence, severity, and possible risk factors of asthma is discussed. Although the influence of classical micronutrients such as selenium, copper, or zinc are well known, the effects of those such as iodine or manganese are only rarely mentioned. As a consequence, the aim of this paper is to demonstrate how disturbances in the levels of micronutrients and their supplementation might affect the course of asthma.
Collapse
|
3
|
Kiskurno S, Ryan RM, Paturi B, Wang H, Kumar VH. Antioxidant MnTBAP does not protect adult mice from neonatal hyperoxic lung injury. Respir Physiol Neurobiol 2020; 282:103545. [PMID: 32927098 DOI: 10.1016/j.resp.2020.103545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 11/21/2022]
Abstract
BACKGROUND Oxygen therapy and mechanical ventilation are important predisposing factors for the development of bronchopulmonary dysplasia (BPD), leading to increased morbidity and mortality in premature infants. Oxygen toxicity mediated by reactive oxygen species (ROS) may play an important part in the development of BPD. We studied the effects of MnTBAP, a catalytic antioxidant on airway responsiveness and alveolar simplification in adult mice following neonatal hyperoxia. METHODS Mice litters were randomized to 85 %O2 or room air (RA) on D3 for 12 days to receive either MnTBAP (10 mg/kg/d) or saline intraperitoneally. Methacholine challenge (MCC) performed at 8 and 12 weeks of age by whole-body plethysmography to assess airway reactivity. Alveolarization quantified on lung sections by radial alveolar count (RAC) and mean linear intercept (MLI). Cell counts assessed from bronchoalveolar lavage (BAL) performed at 15 weeks. RESULTS Mice exposed to hyperoxia and MnTBAP (OXMN) had significantly higher airway reactivity post-MCC at 8 weeks compared to RA and O2 groups. At 12 weeks, airway reactivity was higher post-MCC in both hyperoxia and OXMN groups. MnTBAP did not attenuate hyperoxia-induced airway reactivity in adult mice. Hyperoxia exposed mice demonstrated large and distended alveoli on histopathology at 2 and 15 weeks. MnTBAP did not ameliorate hyperoxia-induced lung injury as assessed by RAC/MLI. Absolute lymphocyte count was significantly higher in BAL in the hyperoxia and OXMN groups. CONCLUSIONS MnTBAP, a catalytic antioxidant, did not afford protection from hyperoxia-induced lung injury in adult mice.
Collapse
Affiliation(s)
- Sergei Kiskurno
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - Rita M Ryan
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - Babu Paturi
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - Huamei Wang
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | | |
Collapse
|
4
|
Sin KR, Ko SG, Kim CJ, Maeng TW, Choe SJ, Ri KR. Reduction of peroxynitrite by some manganoporphyrins of AEOL series: DFT approach with dispersion correction and NBO analysis. J Inorg Biochem 2020; 214:111299. [PMID: 33152662 DOI: 10.1016/j.jinorgbio.2020.111299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/21/2020] [Accepted: 10/24/2020] [Indexed: 11/18/2022]
Abstract
The AEOL series of manganoporphyrins (MnP; AEOL compounds were named by US Aeolus pharmaceuticals) designed as superoxide dismutase mimic are well-known for their powerful catalytic activity to neutralize reactive oxygen and nitrogen species. Reductive oxygen atom cleavage from peroxynitrite (ONOO-) to form NO2 in aqueous solution by some AEOL compounds (AEOL-10113, AEOL-10150, AEOL-11114 and AEOL-11203) was studied by DFT/M06-2X computations with D3 dispersion correction and gCP (geometrical counterpoise correction) for basis set superposition error. DFT computation showed that AEOL-10150 can form the most stable association complex {MnP…OONO} among four AEOL models. AEOL-10150 complex with ONOO- has the lowest deformation energy. In AEOL compounds and their association complexes with ONOO-, Mn atom prefered the high spin state (S = 2) to the intermediate spin state (S = 1). Natural bond orbital analysis showed that electron transfer from the most negative oxygen atom in ONOO- to Mn atom in MnP has the biggest interaction energy among all kinds of donor-acceptor interactions between ONOO- and MnP.
Collapse
Affiliation(s)
- Kye-Ryong Sin
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea.
| | - Sun-Gyong Ko
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Chol-Jin Kim
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Tae-Won Maeng
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Sung-Jub Choe
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Kum-Ryong Ri
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| |
Collapse
|
5
|
Li Z, Wang J, Zheng B, Guo Z. Impact of combined ultrasound-microwave treatment on structural and functional properties of golden threadfin bream (Nemipterus virgatus) myofibrillar proteins and hydrolysates. ULTRASONICS SONOCHEMISTRY 2020; 65:105063. [PMID: 32199256 DOI: 10.1016/j.ultsonch.2020.105063] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 03/07/2020] [Accepted: 03/08/2020] [Indexed: 05/13/2023]
Abstract
The effects of microwave, ultrasound and combined ultrasound-microwave (UM) treatment with different intensities on structural and hydrolysis properties of myofibrillar protein (MP) were investigated. Freeradical scavenging ability, angiotensin-I-converting enzyme (ACE) inhibitory activity, and cellular antioxidant and anti-inflammatory abilities of the related bioactive peptides were also evaluated. Raman spectroscopic analysis indicated that MP molecule tended to unfold and stretch with increasing in β-turn and random coil content under mild microwave (100 W), ultrasound (100-200 W) and combined UM treatments. Meanwhile, differential scanning calorimetry (DSC) and thermogravimetric analysis (TGA) revealed these treatments could also improve the thermal stability against heat-induced denaturation and degeneration. The 200 W ultrasound treatment clearly increased MP solubility by disrupting the highly-ordered aggregates into smaller filament and fragment structures. The 300 W ultrasound coupled with 100 W microwave treatment further enhanced these effects. The resulting partially denatured structure induced by suitable ultrasound and combined UM treatments increased the susceptibility of MP to exogenous enzymes, thereby accelerating hydrolytic process and yielding a high peptide concentration in MP hydrolysates. MP peptides could effectively inhibit free radical and ACE activity, which also improved the ability of antioxidant defence system, and suppressed the production of proinflammatory cytokines in RAW 264.7 cells stimulated by H2O2. The combination of 100 W microwave and 300 W ultrasound treatment was optimal method for generating bioactive MP peptides with the strongest multi-activity effects against H2O2-induced cell damage.
Collapse
Affiliation(s)
- Zhiyu Li
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, Fujian 350002, China; China-Ireland International Cooperation Centre for Food Material Science and Structure Design, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Jianyi Wang
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, Fujian 350002, China; China-Ireland International Cooperation Centre for Food Material Science and Structure Design, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Baodong Zheng
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, Fujian 350002, China; China-Ireland International Cooperation Centre for Food Material Science and Structure Design, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China; State Key Laboratory of Food Safety and Technology for Meat Products, Xiamen, Fujian 361100, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Zebin Guo
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, Fujian 350002, China; China-Ireland International Cooperation Centre for Food Material Science and Structure Design, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China; State Key Laboratory of Food Safety and Technology for Meat Products, Xiamen, Fujian 361100, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China.
| |
Collapse
|
6
|
Passi M, Shahid S, Chockalingam S, Sundar IK, Packirisamy G. Conventional and Nanotechnology Based Approaches to Combat Chronic Obstructive Pulmonary Disease: Implications for Chronic Airway Diseases. Int J Nanomedicine 2020; 15:3803-3826. [PMID: 32547029 PMCID: PMC7266405 DOI: 10.2147/ijn.s242516] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the most prevalent obstructive lung disease worldwide characterized by decline in lung function. It is associated with airway obstruction, oxidative stress, chronic inflammation, mucus hypersecretion, and enhanced autophagy and cellular senescence. Cigarette smoke being the major risk factor, other secondary risk factors such as the exposure to air pollutants, occupational exposure to gases and fumes in developing countries, also contribute to the pathogenesis of COPD. Conventional therapeutic strategies of COPD are based on anti-oxidant and anti-inflammatory drugs. However, traditional anti-oxidant pharmacological therapies are commonly used to alleviate the impact of COPD as they have many associated repercussions such as low diffusion rate and inappropriate drug pharmacokinetics. Recent advances in nanotechnology and stem cell research have shed new light on the current treatment of chronic airway disease. This review is focused on some of the anti-oxidant therapies currently used in the treatment and management of COPD with more emphasis on the recent advances in nanotechnology-based therapeutics including stem cell and gene therapy approaches for the treatment of chronic airway disease such as COPD and asthma.
Collapse
Affiliation(s)
- Mehak Passi
- Nanobiotechnology Laboratory, Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Sadia Shahid
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | | | - Isaac Kirubakaran Sundar
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14623, USA
| | - Gopinath Packirisamy
- Nanobiotechnology Laboratory, Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.,Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| |
Collapse
|
7
|
Chikuma K, Arima K, Asaba Y, Kubota R, Asayama S, Sato K, Kawakami H. The potential of lipid-polymer nanoparticles as epigenetic and ROS control approaches for COPD. Free Radic Res 2019; 54:829-840. [PMID: 31809610 DOI: 10.1080/10715762.2019.1696965] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a lung disease caused by an inflammatory response to various inhaled toxins, especially cigarette smoke. Reactive oxygen species (ROS) and epigenetic abnormality are intimately related to the pathology of COPD, and the overproduction of ROS results in a decrease of histone deacetylase 2 (HDAC2), leading to glucocorticoid resistance. Therefore, a novel treatment that simultaneously reduces ROS level and glucocorticoid resistance is urgently needed. In this study, we developed a codelivery system using core-shell type lipid-polymer nanoparticles (LPNs) composed of a poly(lactic acid) (PLA) core encapsulating a potent antioxidant Mn-porphyrin dimer (MnPD) and a cationic lipid (DOTAP) shell that binds HDAC2-encoding plasmid DNA (pHDAC2), as a new therapeutic approach toward COPD. The transfection of pHDAC2 combined with the elimination of ROS by MnPD exhibited a significant enhancement of intracellular HDAC2 expression levels, suggesting that the multi-antioxidative activity of MnPD plays a crucial role in the expression of HDAC2. Moreover, treatment with LPNs efficiently ameliorated the steroid resistance in COPD models in vitro as evidenced by the lowered expression levels of IL-8. Recovery from mitochondrial dysfunction may be the mechanism underlying the action of LPNs. The PLA-MnPD/DOTAP/pHDAC2 system proposed offers a new therapeutic approach for COPD based on the synergism of ROS elimination and HDAC2 expression.
Collapse
Affiliation(s)
- Kosuke Chikuma
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Kohe Arima
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Yutaro Asaba
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Riku Kubota
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Shoichiro Asayama
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Kiyoshi Sato
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Hiroyoshi Kawakami
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
8
|
Vézina FA, Cantin AM. Antioxidants and Chronic Obstructive Pulmonary Disease. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2018; 5:277-288. [PMID: 30723785 DOI: 10.15326/jcopdf.5.4.2018.0133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Antioxidants represent an attractive therapeutic avenue for individuals with chronic obstructive pulmonary disease (COPD). Cigarette smoke, the major cause of COPD, contains very high concentrations of gaseous and soluble oxidants that can directly induce cell injury and death. Furthermore, particulate matter in cigarette smoke activates lung macrophages that subsequently attract neutrophils. Both neutrophils and macrophages from the lungs of cigarette smokers continuously release large amounts of superoxide and hydrogen peroxide through the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex. Once individuals with COPD stop smoking, the neutrophilic inflammation in the airways and lung parenchyma persists, as do the markers of oxidative stress. Several animal models of cigarette smoke-induced injury have provided evidence that various antioxidants may prevent inflammation and morphological changes associated with COPD however, evidence of benefit in patients is less abundant. Although oxidants can inactivate alpha-1 antitrypsin and other protective proteins, damage lung tissue, and increase mucus production, they also are essential for killing pathogens and resolving inflammation. This review will examine the pre-clinical and clinical evidence of a role for antioxidants in the therapy of patients with COPD.
Collapse
Affiliation(s)
- Félix-Antoine Vézina
- Respiratory Division, Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, Quebec Canada
| | - André M Cantin
- Respiratory Division, Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, Quebec Canada.,Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Quebec, Canada
| |
Collapse
|
9
|
De Grove KC, Provoost S, Brusselle GG, Joos GF, Maes T. Insights in particulate matter-induced allergic airway inflammation: Focus on the epithelium. Clin Exp Allergy 2018; 48:773-786. [PMID: 29772098 DOI: 10.1111/cea.13178] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 05/02/2018] [Accepted: 05/02/2018] [Indexed: 01/01/2023]
Abstract
Outdoor air pollution is a major environmental health problem throughout the world. In particular, exposure to particulate matter (PM) has been associated with the development and exacerbation of several respiratory diseases, including asthma. Although the adverse health effects of PM have been demonstrated for many years, the underlying mechanisms have not been fully identified. In this review, we focus on the role of the lung epithelium and specifically highlight multiple cytokines in PM-induced respiratory responses. We describe the available literature on the topic including in vitro studies, findings in humans (ie observations in human cohorts, human controlled exposure and ex vivo studies) and in vivo animal studies. In brief, it has been shown that exposure to PM modulates the airway epithelium and promotes the production of several cytokines, including IL-1, IL-6, IL-8, IL-25, IL-33, TNF-α, TSLP and GM-CSF. Further, we propose that PM-induced type 2-promoting cytokines are important mediators in the acute and aggravating effects of PM on airway inflammation. Targeting these cytokines could therefore be a new approach in the treatment of asthma.
Collapse
Affiliation(s)
- K C De Grove
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - S Provoost
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - G G Brusselle
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - G F Joos
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - T Maes
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
10
|
Kim DE, Lee Y, Kim M, Lee S, Jon S, Lee SH. Bilirubin nanoparticles ameliorate allergic lung inflammation in a mouse model of asthma. Biomaterials 2017. [PMID: 28624706 DOI: 10.1016/j.biomaterials.2017.06.014] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although asthma, a chronic inflammatory airway disease, is relatively well-managed by inhaled corticosteroids, the side effects associated with the long-term use of these agents precipitate the need for alternative therapeutic options based on differing modes of action. Bilirubin, a potent endogenous antioxidant, and anti-inflammatory molecule have been shown to ameliorate asthmatic symptoms; however, its clinical translation has been limited owing to its water insolubility and associated potential toxicity. Here we report the first application of bilirubin-based nanoparticles (BRNPs) as a nanomedicine for the treatment of allergic lung inflammatory disease. BRNPs were prepared directly from self-assembly of PEGylated bilirubin in aqueous solution and had a hydrodynamic diameter of ∼100 nm. Because allergen-specific type 2 T-helper (Th2) cells play a key role in the pathogenesis and progression of allergic asthma, the effects of BRNPs on Th2 immune responses were investigated both in vivo and in vitro. BRNPs after intravenous injection (i.v.) showed much higher serum concentration and a longer circulation time of bilirubin than the intraperitoneal injection (i.p.) of BRNPs or unconjugated bilirubin (UCB). The anti-asthmatic effects of BRNPs were assessed in a mouse model of allergen-induced asthma. Compared with UCB, treatment with BRNPs suppressed the symptoms of experimental allergic asthma and dramatically ameliorated Th2-related allergic lung inflammation. Consistent with these results, BRNPs caused a reduction of Th2 cell populations and the expression of related cytokines by antibody-stimulated CD4+ T cells in vitro. Therefore, our results establish BRNPs as an important immunomodulatory agent that may be useful as a therapeutic for allergic lung inflammatory disease and other immune-mediated disorders.
Collapse
Affiliation(s)
- Dong Eon Kim
- Biomedical Science and Engineering Interdisciplinary Program, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea; Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Yonghyun Lee
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - MinGyo Kim
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Soyoung Lee
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Sangyong Jon
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea; Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea.
| | - Seung-Hyo Lee
- Biomedical Science and Engineering Interdisciplinary Program, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea; Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea.
| |
Collapse
|
11
|
Abstract
Pulmonary oxidant stress plays an important pathogenetic role in disease conditions including acute lung injury/adult respiratory distress syndrome (ALI/ARDS), hyperoxia, ischemia-reperfusion, sepsis, radiation injury, lung transplantation, COPD, and inflammation. Reactive oxygen species (ROS), released from activated macrophages and leukocytes or formed in the pulmonary epithelial and endothelial cells, damage the lungs and initiate cascades of pro-inflammatory reactions propagating pulmonary and systemic stress. Diverse molecules including small organic compounds (e.g. gluthatione, tocopherol (vitamin E), flavonoids) serve as natural antioxidants that reduce oxidized cellular components, decompose ROS and detoxify toxic oxidation products. Antioxidant enzymes can either facilitate these antioxidant reactions (e.g. peroxidases using glutathione as a reducing agent) or directly decompose ROS (e.g. superoxide dismutases [SOD] and catalase). Many antioxidant agents are being tested for treatment of pulmonary oxidant stress. The administration of small antioxidants via the oral, intratracheal and vascular routes for the treatment of short- and long-term oxidant stress showed rather modest protective effects in animal and human studies. Intratracheal and intravascular administration of antioxidant enzymes are being currently tested for the treatment of acute oxidant stress. For example, intratracheal administration of recombinant human SOD is protective in premature infants exposed to hyperoxia. However, animal and human studies show that more effective delivery of drugs to cells experiencing oxidant stress is needed to improve protection. Diverse delivery systems for antioxidants including liposomes, chemical modifications (e.g. attachment of masking pegylated [PEG]-groups) and coupling to affinity carriers (e.g. antibodies against cellular adhesion molecules) are being employed and currently tested, mostly in animal and, to a limited extent, in humans, for the treatment of oxidant stress. Further studies are needed, however, in order to develop and establish effective applications of pulmonary antioxidant interventions useful in clinical practice. Although beyond the scope of this review, antioxidant gene therapies may eventually provide a strategy for the management of subacute and chronic pulmonary oxidant stress.
Collapse
Affiliation(s)
- Melpo Christofidou-Solomidou
- Institute of Environmental Medicine and Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
12
|
Lawrence LA, Mulligan JK, Roach C, Pasquini WN, Soler ZM, Banglawala SM, Karnezis TT, Gudis DA, Schlosser RJ. Superoxide dismutase reduces the inflammatory response to Aspergillus and Alternaria in human sinonasal epithelial cells derived from patients with chronic rhinosinusitis. Am J Rhinol Allergy 2016; 29:89-93. [PMID: 25785747 DOI: 10.2500/ajra.2015.29.4155] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Aspergillus fumigatus and Alternaria alternata are ubiquitous environmental fungal allergens that can exacerbate airway inflammation and contribute to the disease process in patients with chronic rhinosinusitis (CRS). These antigens have been shown to induce human sinonasal epithelial cells (HSNECs) to promote a proinflammatory response, but what is unclear is a means by which to reduce these effects. Inhaled pathogens can induce HSNECs to produce reactive oxygen species (ROS) that trigger cytokine production. OBJECTIVE This study aimed to determine whether the free radical scavenger superoxide dismutase (SOD) could reduce HSNEC-derived inflammation, as measured by interleukin (IL)-6 and IL-8 production, in response to Aspergillus or Alternaria exposure. METHODS Sinus tissue explants were collected at the time of surgery from control patients (n = 7) and patients with CRS with nasal polyps (CRSwNP) (n = 9). HSNECs were cultured from the explants and treated with Aspergillus, Alternaria, and SOD for 24 hours. Cell supernatants and lysates were collected, and IL-6 and IL-8 concentrations were measured using enzyme-linked immunosorbent assay. RESULTS In control and CRSwNP HSNECs, Aspergillus and Alternaria both increased cytokine production (p < 0.05), as measured by IL-6 and IL-8 concentration. SOD treatment reduced the inflammatory response to fungal antigen exposure from CRSwNP HSNECs but not control HSNECs. In CRSwNP patients, SOD significantly decreased IL-6 and IL-8 production after Alternaria exposure and IL-8 after Aspergillus exposure (p < 0.05). CONCLUSIONS When HSNECs from CRSwNP patients are treated with SOD concurrently with Aspergillus or Alternaria, SOD treatment decreases the fungal antigen-induced inflammatory response. The ability to attenuate inflammation induced by common fungal allergens with SOD treatment could provide a novel therapeutic or preventative approach for patients with CRS or other allergic inflammatory airway diseases.
Collapse
Affiliation(s)
- Lauren A Lawrence
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Carlsten C, MacNutt MJ, Zhang Z, Sava F, Pui MM. Anti-oxidant N-acetylcysteine diminishes diesel exhaust-induced increased airway responsiveness in person with airway hyper-reactivity. Toxicol Sci 2014; 139:479-87. [PMID: 24814479 DOI: 10.1093/toxsci/kfu040] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Inhalation of diesel exhaust (DE) at moderate concentrations causes increased airway responsiveness in asthmatics and increased airway resistance in both healthy and asthmatic subjects, but the effect of baseline airway responsiveness and anti-oxidant supplementation on this dynamic is unknown. OBJECTIVES We aimed to determine if changes in airway responsiveness due to DE are attenuated by thiol anti-oxidant supplementation, particularly in those with underlying airway hyper-responsiveness. METHODS Participants took N-acetylcysteine (600 mg) or placebo capsules three times daily for 6 days. On the last of these 6 days, participants were exposed for 2 h to either filtered air (FA) or DE (300 μg/m(3) of particulate matter smaller than 2.5 microns). Twenty-six non-smokers were studied under each of three experimental conditions (filtered air with placebo, diesel exhaust with placebo, and diesel exhaust with N-acetylcysteine) using a randomized, double-blind, crossover design, with a 2-week washout between conditions. Methacholine challenge was performed pre-exposure (baseline airway responsiveness) and post-exposure (effect of exposure). RESULTS Anti-oxidant supplementation reduced baseline airway responsiveness in hyper-responsive individuals by 20% (p = 0.001). In hyper-responsive individuals, airway responsiveness increased 42% following DE compared with FA (p = 0.03) and this increase was abrogated with anti-oxidant supplementation (diesel exhaust with N-acetylcysteine vs. filtered air with placebo, p = 0.85). CONCLUSIONS Anti-oxidant (N-acetylcysteine) supplementation protects against increased airway responsiveness associated with DE inhalation and reduces need for supplement bronchodilators in those with baseline airway hyper-responsiveness. Individuals with variants in genes of oxidative stress metabolism when exposed to DE are protected from increases in airway responsiveness if taking anti-oxidant supplementation.
Collapse
|
14
|
Bunyavanich S, Schadt EE, Himes BE, Lasky-Su J, Qiu W, Lazarus R, Ziniti JP, Cohain A, Linderman M, Torgerson DG, Eng CS, Pino-Yanes M, Padhukasahasram B, Yang JJ, Mathias RA, Beaty TH, Li X, Graves P, Romieu I, Navarro BDR, Salam MT, Vora H, Nicolae DL, Ober C, Martinez FD, Bleecker ER, Meyers DA, Gauderman WJ, Gilliland F, Burchard EG, Barnes KC, Williams LK, London SJ, Zhang B, Raby BA, Weiss ST. Integrated genome-wide association, coexpression network, and expression single nucleotide polymorphism analysis identifies novel pathway in allergic rhinitis. BMC Med Genomics 2014; 7:48. [PMID: 25085501 PMCID: PMC4127082 DOI: 10.1186/1755-8794-7-48] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 06/04/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Allergic rhinitis is a common disease whose genetic basis is incompletely explained. We report an integrated genomic analysis of allergic rhinitis. METHODS We performed genome wide association studies (GWAS) of allergic rhinitis in 5633 ethnically diverse North American subjects. Next, we profiled gene expression in disease-relevant tissue (peripheral blood CD4+ lymphocytes) collected from subjects who had been genotyped. We then integrated the GWAS and gene expression data using expression single nucleotide (eSNP), coexpression network, and pathway approaches to identify the biologic relevance of our GWAS. RESULTS GWAS revealed ethnicity-specific findings, with 4 genome-wide significant loci among Latinos and 1 genome-wide significant locus in the GWAS meta-analysis across ethnic groups. To identify biologic context for these results, we constructed a coexpression network to define modules of genes with similar patterns of CD4+ gene expression (coexpression modules) that could serve as constructs of broader gene expression. 6 of the 22 GWAS loci with P-value ≤ 1x10-6 tagged one particular coexpression module (4.0-fold enrichment, P-value 0.0029), and this module also had the greatest enrichment (3.4-fold enrichment, P-value 2.6 × 10-24) for allergic rhinitis-associated eSNPs (genetic variants associated with both gene expression and allergic rhinitis). The integrated GWAS, coexpression network, and eSNP results therefore supported this coexpression module as an allergic rhinitis module. Pathway analysis revealed that the module was enriched for mitochondrial pathways (8.6-fold enrichment, P-value 4.5 × 10-72). CONCLUSIONS Our results highlight mitochondrial pathways as a target for further investigation of allergic rhinitis mechanism and treatment. Our integrated approach can be applied to provide biologic context for GWAS of other diseases.
Collapse
Affiliation(s)
- Supinda Bunyavanich
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 10029 New York, NY, USA
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 10029 New York, NY, USA
| | - Blanca E Himes
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Weiliang Qiu
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Ross Lazarus
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Medical Bioinformatics, Baker IDI, Melbourne, Australia
| | - John P Ziniti
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Ariella Cohain
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 10029 New York, NY, USA
| | - Michael Linderman
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 10029 New York, NY, USA
| | - Dara G Torgerson
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Celeste S Eng
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Maria Pino-Yanes
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- IBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Badri Padhukasahasram
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, USA
| | - James J Yang
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - Rasika A Mathias
- Departments of Medicine and Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Terri H Beaty
- Departments of Medicine and Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Xingnan Li
- Center for Genomics, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Penelope Graves
- Arizona Respiratory Center and BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | | | | | - M Towhid Salam
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hita Vora
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dan L Nicolae
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Fernando D Martinez
- Arizona Respiratory Center and BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Eugene R Bleecker
- Center for Genomics, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Deborah A Meyers
- Center for Genomics, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - W James Gauderman
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Frank Gilliland
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Esteban G Burchard
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Kathleen C Barnes
- Departments of Medicine and Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - L Keoki Williams
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, USA
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, USA
| | - Stephanie J London
- Division of Intramural Research, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle, Park, NC, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 10029 New York, NY, USA
| | - Benjamin A Raby
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Aravamudan B, Kiel A, Freeman M, Delmotte P, Thompson M, Vassallo R, Sieck GC, Pabelick CM, Prakash YS. Cigarette smoke-induced mitochondrial fragmentation and dysfunction in human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2014; 306:L840-54. [PMID: 24610934 DOI: 10.1152/ajplung.00155.2013] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The balance between mitochondrial fission and fusion is crucial for mitochondria to perform its normal cellular functions. We hypothesized that cigarette smoke (CS) disrupts this balance and enhances mitochondrial dysfunction in the airway. In nonasthmatic human airway smooth muscle (ASM) cells, CS extract (CSE) induced mitochondrial fragmentation and damages their networked morphology in a concentration-dependent fashion, via increased expression of mitochondrial fission protein dynamin-related protein 1 (Drp1) and decreased fusion protein mitofusin (Mfn) 2. CSE effects on Drp1 vs. Mfn2 and mitochondrial network morphology involved reactive oxygen species (ROS), activation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt), protein kinase C (PKC) and proteasome pathways, as well as transcriptional regulation via factors such as NF-κB and nuclear erythroid 2-related factor 2. Inhibiting Drp1 prevented CSE effects on mitochondrial networks and ROS generation, whereas blocking Mfn2 had the opposite, detrimental effect. In ASM from asmatic patients, mitochondria exhibited substantial morphological defects at baseline and showed increased Drp1 but decreased Mfn2 expression, with exacerbating effects of CSE. Overall, these results highlight the importance of mitochondrial networks and their regulation in the context of cellular changes induced by insults such as inflammation (as in asthma) or CS. Altered mitochondrial fission/fusion proteins have a further potential to influence parameters such as ROS and cell proliferation and apoptosis relevant to airway diseases.
Collapse
Affiliation(s)
- Bharathi Aravamudan
- Division of Anesthesia Research, Dept. of Anesthesiology, 4-184 W. Joseph SMH, Mayo Clinic, Rochester, MN 55905.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Hung CY, Huang FL, Shi LS, Ka SM, Wang JY, Tsai YC, Hung TJ, Ye YL. The Ethanol Extract of Osmanthus fragrans Flowers Reduces Oxidative Stress and Allergic Airway Inflammation in an Animal Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:304290. [PMID: 24386002 PMCID: PMC3872418 DOI: 10.1155/2013/304290] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 11/24/2022]
Abstract
The Osmanthus fragrans flower, a popular herb in Eastern countries, contains several antioxidant compounds. Ben Cao Gang Mu, traditional Chinese medical literature, describes the usefulness of these flowers for phlegm and stasis reduction, arrest of dysentery with blood in the bowel, and stomachache and diarrhea treatment. However, modern evidence regarding the therapeutic efficacy of these flowers is limited. This study was aimed at assessing the antioxidative effects of the ethanol extract of O. fragrans flowers (OFE) in vivo and evaluating its antioxidant maintenance and therapeutic effect on an allergic airway inflammation in mice. After OFE's oral administration to mice, the values obtained in the oxygen radical absorbance capacity assay as well as the glutathione concentration in the lungs and spleens of mice increased while thiobarbituric acid reactive substances decreased significantly, indicating OFE's significant in vivo antioxidant activity. OFE was also therapeutically efficacious in a mouse model of ovalbumin-induced allergic airway inflammation. Orally administered OFE suppressed ovalbumin-specific IgE production and inflammatory cell infiltration in the lung. Moreover, the antioxidative state of the mice improved. Thus, our findings confirm the ability of the O. fragrans flowers to reduce phlegm and suggest that OFE may be useful as an antiallergic agent.
Collapse
Affiliation(s)
- Chien-Ya Hung
- Department of Food Nutrition, Chung Hwa University of Medical Technology, Tainan 71703, Taiwan
| | - Fu-Long Huang
- Graduate Institute of Food Science, National Chiayi University, Chiayi 60004, Taiwan
| | - Li-Shian Shi
- Department of Biotechnology, National Formosa University, Yunlin 63201, Taiwan
| | - Shuk-Man Ka
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei 11490, Taiwan
| | - Jing-Yao Wang
- Department of Biotechnology, National Formosa University, Yunlin 63201, Taiwan
| | - Yu-Cheng Tsai
- Department of Food Nutrition, Chung Hwa University of Medical Technology, Tainan 71703, Taiwan
| | - Tsung-Jen Hung
- Department of Graduate Institute of Biomedical Science, Chung Hwa University of Medical Technology, Tainan 71703, Taiwan
| | - Yi-Ling Ye
- Department of Biotechnology, National Formosa University, Yunlin 63201, Taiwan
| |
Collapse
|
17
|
Villegas LR, Kluck D, Field C, Oberley-Deegan RE, Woods C, Yeager ME, El Kasmi KC, Savani RC, Bowler RP, Nozik-Grayck E. Superoxide dismutase mimetic, MnTE-2-PyP, attenuates chronic hypoxia-induced pulmonary hypertension, pulmonary vascular remodeling, and activation of the NALP3 inflammasome. Antioxid Redox Signal 2013; 18:1753-64. [PMID: 23240585 PMCID: PMC3619229 DOI: 10.1089/ars.2012.4799] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS Pulmonary hypertension (PH) is characterized by an oxidant/antioxidant imbalance that promotes abnormal vascular responses. Reactive oxygen species, such as superoxide (O(2)(•-)), contribute to the pathogenesis of PH and vascular responses, including vascular remodeling and inflammation. This study sought to investigate the protective role of a pharmacological catalytic antioxidant, a superoxide dismutase (SOD) mimetic (MnTE-2-PyP), in hypoxia-induced PH, vascular remodeling, and NALP3 (NACHT, LRR, and PYD domain-containing protein 3)-mediated inflammation. RESULTS Mice (C57/BL6) were exposed to hypobaric hypoxic conditions, while subcutaneous injections of MnTE-2-PyP (5 mg/kg) or phosphate-buffered saline (PBS) were given 3× weekly for up to 35 days. SOD mimetic-treated groups demonstrated protection against increased right ventricular systolic pressure, indirect measurements of pulmonary artery pressure, and RV hypertrophy. Vascular remodeling was assessed by Ki67 staining to detect vascular cell proliferation, α-smooth muscle actin staining to analyze small vessel muscularization, and hyaluronan (HA) measurements to assess extracellular matrix modulation. Activation of the NALP3 inflammasome pathway was measured by NALP3 expression, caspase-1 activation, and interleukin 1-beta (IL-1β) and IL-18 production. Hypoxic exposure increased PH, vascular remodeling, and NALP3 inflammasome activation in PBS-treated mice, while mice treated with MnTE-2-PyP showed an attenuation in each of these endpoints. INNOVATION This study is the first to demonstrate activation of the NALP3 inflammasome with cleavage of caspase-1 and release of active IL-1 β and IL-18 in chronic hypoxic PH, as well as its attenuation by the SOD mimetic, MnTE-2-PyP. CONCLUSION The ability of the SOD mimetic to scavenge extracellular O(2)(•-) supports our previous observations in EC-SOD-overexpressing mice that implicate extracellular oxidant/antioxidant imbalance in hypoxic PH and implicates its role in hypoxia-induced inflammation.
Collapse
Affiliation(s)
- Leah R Villegas
- Department of Pediatrics, University of Colorado, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Molecular mechanisms of reactive oxygen species-related pulmonary inflammation and asthma. Mol Immunol 2013; 56:57-63. [PMID: 23665383 DOI: 10.1016/j.molimm.2013.04.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 04/07/2013] [Indexed: 02/07/2023]
Abstract
Asthma is a highly relevant disorder that can be induced by many environmental factors such as allergens and pollutants. One of the most critical pathological symptoms of asthma is airway inflammation. In order to identify a cause of respiratory inflammation, we thoroughly examine the unique role of reactive oxygen species (ROS). Evidence supports that the inhalation of aggravating compounds such as allergens can promote the increased generation of ROS. Accordingly, ROS have a proven role in the cellular signaling cascades of many respiratory diseases that cause respiratory inflammation, including asthma. Although there is no known cure for asthma, current treatments effectively lessen the inflammation symptom. Based on the investigations of asthma pathogenesis and the mechanism of ROS formation, we have identified several novel anti-inflammatory therapeutic treatments, shedding light on a fundamental understanding for the cure of this disorder. In this review, we will outline the pathogenesis of asthma and its relationship to ROS, oxidative stress, and pulmonary inflammation.
Collapse
|
19
|
Sohn SW, Jung JW, Lee SY, Kang HR, Park HW, Min KU, Cho SH. Expression pattern of GSTP1 and GSTA1 in the pathogenesis of asthma. Exp Lung Res 2013; 39:173-81. [PMID: 23647087 DOI: 10.3109/01902148.2013.789572] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Reactive oxygen species (ROS) are known aggravating factors for airway inflammation in asthma. Glutathione S-transferases (GSTs) detoxify ROS and toxic compounds in environmental exposures. However, little is known about the regulation of GST and expression of GST subtypes in asthma. The aim of this study was to evaluate how GSTs are regulated in asthma. We observed total GST activity and expression of GST subtypes in murine asthma models and GST expressions in induced sputum cells of asthmatics. Total GST activity was increased in BAL fluids of OVA-treated murine asthma model. GSTP and GSTA are highly expressed in peribronchiolar mononuclear inflammatory cells and epithelial cells in OVA-treated mice. GSTM are expressed in epithelial cells in both OVA and PBS-treated groups. GSTP1 mRNA expression was increased in the lung of OVA-treated mice compared with PBS-treated mice. GSTA1, GSTM1, and GSTT1 mRNA expressions were not different between both groups. GSTA1 mRNA expression was increased in induced sputum cells of asthmatics compared with healthy controls. GSTP1, GSTM1, and GSTT1 mRNA expressions were not different between asthmatics and healthy controls. In asthmatics, GSTP1 and GSTA1 mRNA expressions were higher in induced sputum cells of asthmatics with PC20 ≤ 4 mg/ml than those with PC20 > 4 mg/ml. GSTM1 and GSTT1 mRNA expressions were not different between two groups. These findings suggest that GSTs are upregulated in the airways of asthmatics in response to increased oxidative stress. GSTP and GSTA are thought to play an important role in protecting the airways of asthmatics compared with GSTM and GSTT.
Collapse
Affiliation(s)
- Seong-Wook Sohn
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
20
|
Fabian E, Pölöskey P, Kósa L, Elmadfa I, Réthy LA. Nutritional supplements and plasma antioxidants in childhood asthma. Wien Klin Wochenschr 2013; 125:309-15. [PMID: 23636616 DOI: 10.1007/s00508-013-0359-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 03/20/2013] [Indexed: 12/30/2022]
Abstract
OBJECTIVE This study investigated the relationship of plasma antioxidants to airway inflammation and systemic oxidative stress in children suffering from atopic asthma with consideration of the intake of nutritional supplements. SUBJECTS AND RESEARCH METHODS A total of 35 asthmatic children (AG) and 21 healthy controls (CG) participated in this study. Plasma levels of vitamins A and E, β-carotene, coenzyme Q10 and malondialdehyde (MDA) were analyzed with high-performance liquid chromatography (HPLC); the total antioxidant capacity (TAC) was measured photometrically, and selenium was determined by atomic absorption spectroscopy (AAS). The volume of fractionated exhaled nitric oxide (FeNO) was measured with the NIOX nitric oxide monitoring system. RESULTS The plasma antioxidants vitamins A and E, selenium, and coenzyme Q10 but not β-carotene were significantly (p < 0.05) lower in asthmatics than in controls. Further, asthmatic children had significantly reduced plasma concentrations of TAC (p < 0.01), significantly enhanced levels of MDA (p < 0.001), and exhaled a significantly (p < 0.001) higher mean volume of FENO than healthy children. Regular intake of supplements had a significant positive influence on plasma vitamin E (p < 0.01), selenium (p < 0.01), TAC (p < 0.05), MDA (p < 0.01), and FENO (p < 0.01) in asthmatics but not in controls. Additionally, significant negative associations of vitamin E and MDA (AG: p < 0.01; CG: p < 0.05), and vitamin E and FENO (AG: p < 0.05; CG: p > 0.05) were identified. CONCLUSION These results indicate that nutritional supplements beneficially modulate plasma antioxidants and thus might have a positive influence on systemic redox balance and subsequently, pulmonary inflammation in asthmatic children.
Collapse
Affiliation(s)
- Elisabeth Fabian
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
| | | | | | | | | |
Collapse
|
21
|
Rahman I, Kinnula VL. Strategies to decrease ongoing oxidant burden in chronic obstructive pulmonary disease. Expert Rev Clin Pharmacol 2012; 5:293-309. [PMID: 22697592 DOI: 10.1586/ecp.12.16] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading cause of mortality and morbidity globally, and its development is mainly associated with tobacco/biomass smoke-induced oxidative stress. Hence, targeting systemic and local oxidative stress with agents that can balance the antioxidant/redox system can be expected to be useful in the treatment of COPD. Preclinical and clinical trials have revealed that antioxidants/redox modulators can detoxify free radicals and oxidants, control expression of redox and glutathione biosynthesis genes, chromatin remodeling and inflammatory gene expression; and are especially useful in preventing COPD exacerbations. In this review, various novel approaches and problems associated with these approaches in COPD are reviewed.
Collapse
Affiliation(s)
- Irfan Rahman
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | |
Collapse
|
22
|
Mao XW, Crapo JD, Gridley DS. Mitochondrial Oxidative Stress-Induced Apoptosis and Radioprotection in Proton-Irradiated Rat Retina. Radiat Res 2012; 178:118-25. [DOI: 10.1667/rr2821.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
23
|
Jungsuwadee P, Weaver MR, Gally F, Oberley-Deegan RE. The metalloporphyrin antioxidant, MnTE-2-PyP, inhibits Th2 cell immune responses in an asthma model. Int J Mol Sci 2012; 13:9785-9797. [PMID: 22949830 PMCID: PMC3431828 DOI: 10.3390/ijms13089785] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/13/2012] [Accepted: 07/26/2012] [Indexed: 01/24/2023] Open
Abstract
MnTE-2-PyP, a superoxide dismutase mimetic, inhibited OVA-induced airway inflammation in mice suggesting an effect on Th2 responsiveness. Thus, we hypothesized that MnTE-2-PyP may alter dendritic cell-Th2 interactions. Bone marrow derived dendritic cells (DC) and OVA(323-339)-specific Th2 cells were cultured separately in the presence or absence of MnTE-2-PyP for 3 days prior to the co-culturing of the two cell types in the presence of an OVA(323-339) peptide and in some cases stimulated with CD3/CD28. MnTE-2-PyP-pretreated DC inhibited IL-4, IL-5 and IFNγ production and inhibited Th2 cell proliferation in the DC-Th2 co-culturing system in the presence of the OVA(323-339) peptide. Similar results were obtained using the CD3/CD28 cell-activation system; the addition of MnTE-2-PyP inhibited Th2 cell proliferation. MnTE-2-PyP suppressed CD25 expression on OVA-specific Th2 cells, which implied that MnTE-2-PyP can inhibit the activation of Th2 cells. MnTE-2-PyP also down-regulated co-stimulatory molecules: CD40, CD80 and CD86 on immature DC. Our studies suggest that the major mechanism by which MnTE-2-PyP inhibits airway inflammation is by acting on the DC and suppressing Th2 cell proliferation and activation.
Collapse
Affiliation(s)
- Paiboon Jungsuwadee
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; E-Mails: (M.R.W.); (F.G.); (R.E.O.-D.)
- Department of Biopharmaceutical Sciences, College of Pharmacy, Roosevelt University, Schaumburg Campus, Schaumburg, IL 60173, USA
- Department of Medicine, University of Colorado Health Science Center, Aurora, CO 80045, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-847-330-4506; Fax: +1-847-330-4257
| | - Michael R. Weaver
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; E-Mails: (M.R.W.); (F.G.); (R.E.O.-D.)
| | - Fabienne Gally
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; E-Mails: (M.R.W.); (F.G.); (R.E.O.-D.)
| | - Rebecca E. Oberley-Deegan
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; E-Mails: (M.R.W.); (F.G.); (R.E.O.-D.)
| |
Collapse
|
24
|
Terziev L, Dancheva V, Shopova V, Stavreva G. Antioxidant effect of MnTE-2-PyP on lung in asthma mice model. ScientificWorldJournal 2012; 2012:379360. [PMID: 22654599 PMCID: PMC3361234 DOI: 10.1100/2012/379360] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 12/28/2011] [Indexed: 12/02/2022] Open
Abstract
AIM To investigate the effects of MnTE-2-PyP on some markers of antioxidant defence system in asthma mice model. MATERIAL AND METHODS The animals were divided into four groups: group 1, controls; group 2, injected with ovalbumin, group 3, treated with MnTE-2-PyP, and group 4, treated with ovalbumin and MnTE-2-PyP. The activities of superoxide dismutase, catalase, glutathione peroxidase and nonprotein sulfhydryl groups content (NPSH) were determined in lung homogenate. RESULTS The activities of superoxide dismutase and catalase in group 2 decreased significantly as compared to control group. The decrease of the same enzymes in group 4 was lower and significant as compared to group 2. Changes in the glutathione peroxidase activity showed a similar dynamics. The NPSH groups content decreased in group 2. In group 4 this decrease was relatively lower as compared to group 2. CONCLUSIONS The application of MnTE-2-PyP mitigated the effects of oxidative stress in asthma mice model.
Collapse
Affiliation(s)
- Lyudmil Terziev
- Sector of Clinical Immunology and Allergology, Pelven Medical University, 5800 Pleven, Bulgaria.
| | | | | | | |
Collapse
|
25
|
Rahman I, MacNee W. Antioxidant pharmacological therapies for COPD. Curr Opin Pharmacol 2012; 12:256-65. [PMID: 22349417 DOI: 10.1016/j.coph.2012.01.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 01/26/2012] [Indexed: 12/28/2022]
Abstract
Increased oxidative stress occurs in the lungs and systemically in COPD, which plays a role in many of the pathogenic mechanisms in COPD. Hence, targeting local lung and systemic oxidative stress with agents that modulate the antioxidants/redox system or boost endogenous antioxidants would be a useful therapeutic approach in COPD. Thiol antioxidants (N-acetyl-l-cysteine [NAC] and N-acystelyn, carbocysteine, erdosteine, and fudosteine) have been used to increase lung thiol content. Modulation of cigarette smoke (CS) induced oxidative stress and its consequent cellular changes have also been reported to be effected by synthetic molecules, such as spin traps (α-phenyl-N-tert-butyl nitrone), catalytic antioxidants (superoxide dismutase [ECSOD] mimetics), porphyrins, and lipid peroxidation and protein carbonylation blockers/inhibitors (edaravone and lazaroids/tirilazad). Preclinical and clinical trials have shown that these antioxidants can reduce oxidative stress, affect redox and glutathione biosynthesis genes, and proinflammatory gene expression. In this review the approaches to enhance lung antioxidants in COPD and the potential beneficial effects of antioxidant therapy on the course of the disease are discussed.
Collapse
Affiliation(s)
- Irfan Rahman
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, NY, USA.
| | | |
Collapse
|
26
|
Rahman I. Pharmacological antioxidant strategies as therapeutic interventions for COPD. Biochim Biophys Acta Mol Basis Dis 2011; 1822:714-28. [PMID: 22101076 DOI: 10.1016/j.bbadis.2011.11.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/01/2011] [Accepted: 11/02/2011] [Indexed: 10/15/2022]
Abstract
Cigarette/tobacco smoke/biomass fuel-induced oxidative and aldehyde/carbonyl stress are intimately associated with the progression and exacerbation of chronic obstructive pulmonary disease (COPD). Therefore, targeting systemic and local oxidative stress with antioxidants/redox modulating agents, or boosting the endogenous levels of antioxidants are likely to have beneficial effects in the treatment/management of COPD. Various antioxidant agents, such as thiol molecules (glutathione and mucolytic drugs, such as N-acetyl-L-cysteine and N-acystelyn, erdosteine, fudosteine, ergothioneine, and carbocysteine), have been reported to modulate various cellular and biochemical aspects of COPD. These antioxidants have been found to scavenge and detoxify free radicals and oxidants, regulate of glutathione biosynthesis, control nuclear factor-kappaB (NF-kappaB) activation, and hence inhibiting inflammatory gene expression. Synthetic molecules, such as specific spin traps like α-phenyl-N-tert-butyl nitrone, a catalytic antioxidant (ECSOD mimetic), porphyrins (AEOL 10150 and AEOL 10113), and a superoxide dismutase mimetic M40419, iNOS and myeloperoxidase inhibitors, lipid peroxidation inhibitors/blockers edaravone, and lazaroids/tirilazad have also been shown to have beneficial effects by inhibiting cigarette smoke-induced inflammatory responses and other carbonyl/oxidative stress-induced cellular alterations. A variety of oxidants, free radicals, and carbonyls/aldehydes are implicated in the pathogenesis of COPD, it is therefore, possible that therapeutic administration or supplementation of multiple antioxidants and/or boosting the endogenous levels of antioxidants will be beneficial in the treatment of COPD. This review discusses various novel pharmacological approaches adopted to enhance lung antioxidant levels, and various emerging beneficial and/or prophylactic effects of antioxidant therapeutics in halting or intervening the progression of COPD. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.
Collapse
Affiliation(s)
- Irfan Rahman
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, NY 14642, USA.
| |
Collapse
|
27
|
Batinic-Haberle I, Rajic Z, Tovmasyan A, Ye X, Leong KW, Dewhirst MW, Vujaskovic Z, Benov L, Spasojevic I. Diverse functions of cationic Mn(III) N-substituted pyridylporphyrins, recognized as SOD mimics. Free Radic Biol Med 2011; 51:1035-53. [PMID: 21616142 PMCID: PMC3178885 DOI: 10.1016/j.freeradbiomed.2011.04.046] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 03/30/2011] [Accepted: 04/29/2011] [Indexed: 10/18/2022]
Abstract
Oxidative stress, a redox imbalance between the endogenous reactive species and antioxidant systems, is common to numerous pathological conditions such as cancer, central nervous system injuries, radiation injury, diabetes etc. Therefore, compounds able to reduce oxidative stress have been actively sought for over 3 decades. Superoxide is the major species involved in oxidative stress either in its own right or through its progeny, such as ONOO⁻, H₂O₂, •OH, CO₃•⁻, and •NO₂. Hence, the very first compounds developed in the late 1970-ies were the superoxide dismutase (SOD) mimics. Thus far the most potent mimics have been the cationic meso Mn(III) N-substituted pyridylporphyrins and N,N'-disubstituted imidazolylporphyrins (MnPs), some of them with k(cat)(O₂·⁻) similar to the k(cat) of SOD enzymes. Most frequently studied are ortho isomers MnTE-2-PyP⁵⁺, MnTnHex-2-PyP⁵⁺, and MnTDE-2-ImP⁵⁺. The ability to disproportionate O₂·⁻ parallels their ability to remove the other major oxidizing species, peroxynitrite, ONOO⁻. The same structural feature that gives rise to the high k(cat)(O₂·⁻) and k(red)(ONOO⁻), allows MnPs to strongly impact the activation of the redox-sensitive transcription factors, HIF-1α, NF-κB, AP-1, and SP-1, and therefore modify the excessive inflammatory and immune responses. Coupling with cellular reductants and other redox-active endogenous proteins seems to be involved in the actions of Mn porphyrins. While hydrophilic analogues, such as MnTE-2-PyP⁵⁺ and MnTDE-2-ImP⁵⁺ are potent in numerous animal models of diseases, the lipophilic analogues, such as MnTnHex-2-PyP⁵⁺, were developed to cross blood brain barrier and target central nervous system and critical cellular compartments, mitochondria. The modification of its structure, aimed to preserve the SOD-like potency and lipophilicity, and diminish the toxicity, has presently been pursued. The pulmonary radioprotection by MnTnHex-2-PyP⁵⁺ was the first efficacy study performed successfully with non-human primates. The Phase I toxicity clinical trials were done on amyotrophic lateral sclerosis patients with N,N'-diethylimidazolium analogue, MnTDE-2-ImP⁵⁺ (AEOL10150). Its aggressive development as a wide spectrum radioprotector by Aeolus Pharmaceuticals has been supported by USA Federal government. The latest generation of compounds, bearing oxygens in pyridyl substituents is presently under aggressive development for cancer and CNS injuries at Duke University and is supported by Duke Translational Research Institute, The Wallace H. Coulter Translational Partners Grant Program, Preston Robert Tisch Brain Tumor Center at Duke, and National Institute of Allergy and Infectious Diseases. Metal center of cationic MnPs easily accepts and donates electrons as exemplified in the catalysis of O₂·⁻ dismutation. Thus such compounds may be equally good anti- and pro-oxidants; in either case the beneficial therapeutic effects may be observed. Moreover, while the in vivo effects may appear antioxidative, the mechanism of action of MnPs that produced such effects may be pro-oxidative; the most obvious example being the inhibition of NF-κB. The experimental data therefore teach us that we need to distinguish between the mechanism/s of action/s of MnPs and the effects we observe. A number of factors impact the type of action of MnPs leading to favorable therapeutic effects: levels of reactive species and oxygen, levels of endogenous antioxidants (enzymes and low-molecular compounds), levels of MnPs, their site of accumulation, and the mutual encounters of all of those species. The complexity of in vivo redox systems and the complex redox chemistry of MnPs challenge and motivate us to further our understanding of the physiology of the normal and diseased cell with ultimate goal to successfully treat human diseases.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
- Corresponding authors: Ines Batinic-Haberle, Ph. D. Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8718, . Ivan Spasojevic, Ph. D. Department of Medicine, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-8311, Fax: 919-684-8380,
| | - Zrinka Rajic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiaodong Ye
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Mark W. Dewhirst
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait School of Medicine, Kuwait
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Corresponding authors: Ines Batinic-Haberle, Ph. D. Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8718, . Ivan Spasojevic, Ph. D. Department of Medicine, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-8311, Fax: 919-684-8380,
| |
Collapse
|
28
|
Deshane J, Zmijewski JW, Luther R, Gaggar A, Deshane R, Lai JF, Xu X, Spell M, Estell K, Weaver CT, Abraham E, Schwiebert LM, Chaplin DD. Free radical-producing myeloid-derived regulatory cells: potent activators and suppressors of lung inflammation and airway hyperresponsiveness. Mucosal Immunol 2011; 4:503-18. [PMID: 21471960 PMCID: PMC3694614 DOI: 10.1038/mi.2011.16] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Levels of reactive free radicals are elevated in the airway during asthmatic exacerbations, but their roles in the pathophysiology of asthma remain unclear. We have identified subsets of myeloid-derived suppressor-like cells as key sources of nitric oxide and superoxide in the lungs of mice with evolving experimental allergic airway inflammation and established these cells as master regulators of the airway inflammatory response. The profiles of free radicals they produced depended on expression of inducible nitric oxide synthase (iNOS), arginase, and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. These radicals controlled the pro- and anti-inflammatory potential of these cells, and also regulated the reciprocal pattern of their infiltration into the lung. The nitric oxide-producing cells were Ly-6C(+)Ly-6G(-) and they downmodulated T-cell activation, recruited T(reg) cells, and dramatically downregulated antigen-induced airway hyperresponsiveness. The superoxide-producing cells were Ly-6C(-)Ly-6G(+) and they expressed proinflammatory activities, exacerbating airway hyperresponsiveness in a superoxide-dependent fashion. A smaller population of Ly-6C(+)Ly-6G(+) cells also suppressed T-cell responses, but in an iNOS- and arginase-independent fashion. These regulatory myeloid cells represent important targets for asthma therapy.
Collapse
Affiliation(s)
- Jessy Deshane
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294
,Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294
,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Jaroslaw W. Zmijewski
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294
,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Rita Luther
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Amit Gaggar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294
,Veteran Affairs Medical Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Rohit Deshane
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Jen-Feng Lai
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Xin Xu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Marion Spell
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294
,Center for AIDS Research, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Kim Estell
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Casey T Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Edward Abraham
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294
,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Lisa M. Schwiebert
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, Alabama 35294
,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - David D. Chaplin
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294
,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
,Arthritis and Musculoskeletal Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
29
|
Fabian E, Pölöskey P, Kósa L, Elmadfa I, Réthy LA. Activities of antioxidant enzymes in relation to oxidative and nitrosative challenges in childhood asthma. J Asthma 2011; 48:351-7. [PMID: 21401389 DOI: 10.3109/02770903.2011.560319] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND This study aimed to investigate the relationship between antioxidant enzyme activities, extent of airway inflammation, and systemic oxidative stress in children suffering from atopic asthma. METHODS A total of 35 asthmatic (AG) and 21 healthy children (CG) participated in this study. The volume of fractionated exhaled NO (Fe(no)) was measured with the NIOX test system. The activities of the erythrocyte antioxidant enzymes superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px), and the total antioxidant capacity (TAC) were determined photometrically. Plasma interleukin (IL)-6 was measured using ELISA; malondialdehyde (MDA) levels were analyzed using HPLC. RESULTS Compared to healthy controls, asthmatic children exhaled a significantly (p < .001) higher mean volume of Fe(no), had significantly reduced plasma concentrations of TAC (p = .006), and significantly enhanced levels of MDA (p < .001) and IL-6 (p = .012). SOD (p = .027), CAT (p < .001), and GSH-Px (p = .005) were significantly less active in the asthma group and significantly negatively associated with Fe(no) (SOD/Fe(no): p = .017; CAT/Fe(no): p = .008; GSH-Px/Fe(no): p = .001); the oxidative stress marker MDA showed such correlations in both investigated groups (SOD/MDA: AG: p = .001, CG: p = .381; CAT/MDA: AG: p = .003, CG: p = .020; GSH-Px/MDA: AG: p = .006, CG: p = .011). Furthermore, there was a significant (p< .01) positive correlation between MDA/Fe(no) and MDA/IL-6 observed in both groups. CONCLUSIONS These results indicate that inflammation of the bronchial tree, reflected by increased NO formation in the airways and enhanced systemic oxidative stress, is related to an alteration of antioxidant enzyme activities in childhood asthma. Modulating the activity of antioxidant enzymes may therefore have beneficial effects on pulmonary and systemic antioxidant defense mechanisms and could reduce airway inflammation and oxidative stress in asthmatics.
Collapse
Affiliation(s)
- Elisabeth Fabian
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria.
| | | | | | | | | |
Collapse
|
30
|
Choi MJ, Kim BK, Park KY, Yokozawa T, Song YO, Cho EJ. Anti-aging effects of cyanidin under a stress-induced premature senescence cellular system. Biol Pharm Bull 2010; 33:421-6. [PMID: 20190403 DOI: 10.1248/bpb.33.421] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The anti-aging effects of cyanidin were investigated under stress-induced premature senescence (SIPS) using WI-38 human diploid fibroblasts. WI-38 cells that were treated with 300 microM H(2)O(2) showed losses of cell viability, increased lipid peroxidation, and shortened cell lifespans. However, treatment with cyanidin attenuated cellular oxidative stress through increase of cell viability and the inhibition of lipid peroxidation. In addition, the life spans of young-, middle-, and old-aged WI-38 cells were prolonged by cyanidin treatment. Furthermore, H(2)O(2)-treated WI-38 cells significantly increased mRNA and protein expressions of nuclear factor-kappaB, cyclooxygenase-2, and inducible nitric oxide synthase, while those treated with cyanidin had significantly decreased expressions. These results suggest that cyanidin may delay the aging process by attenuating oxidative stress under the SIPS cellular model.
Collapse
Affiliation(s)
- Mi Jin Choi
- Department of Food Science and Nutrition, Research Institute of Ecology for the Elderly, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 609-735, Korea
| | | | | | | | | | | |
Collapse
|
31
|
Batinić-Haberle I, Rebouças JS, Spasojević I. Superoxide dismutase mimics: chemistry, pharmacology, and therapeutic potential. Antioxid Redox Signal 2010; 13:877-918. [PMID: 20095865 PMCID: PMC2935339 DOI: 10.1089/ars.2009.2876] [Citation(s) in RCA: 390] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Oxidative stress has become widely viewed as an underlying condition in a number of diseases, such as ischemia-reperfusion disorders, central nervous system disorders, cardiovascular conditions, cancer, and diabetes. Thus, natural and synthetic antioxidants have been actively sought. Superoxide dismutase is a first line of defense against oxidative stress under physiological and pathological conditions. Therefore, the development of therapeutics aimed at mimicking superoxide dismutase was a natural maneuver. Metalloporphyrins, as well as Mn cyclic polyamines, Mn salen derivatives and nitroxides were all originally developed as SOD mimics. The same thermodynamic and electrostatic properties that make them potent SOD mimics may allow them to reduce other reactive species such as peroxynitrite, peroxynitrite-derived CO(3)(*-), peroxyl radical, and less efficiently H(2)O(2). By doing so SOD mimics can decrease both primary and secondary oxidative events, the latter arising from the inhibition of cellular transcriptional activity. To better judge the therapeutic potential and the advantage of one over the other type of compound, comparative studies of different classes of drugs in the same cellular and/or animal models are needed. We here provide a comprehensive overview of the chemical properties and some in vivo effects observed with various classes of compounds with a special emphasis on porphyrin-based compounds.
Collapse
Affiliation(s)
- Ines Batinić-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
32
|
O’Neill HC, White CW, Veress LA, Hendry-Hofer TB, Loader JE, Min E, Huang J, Rancourt RC, Day BJ. Treatment with the catalytic metalloporphyrin AEOL 10150 reduces inflammation and oxidative stress due to inhalation of the sulfur mustard analog 2-chloroethyl ethyl sulfide. Free Radic Biol Med 2010; 48:1188-96. [PMID: 20138141 PMCID: PMC2847650 DOI: 10.1016/j.freeradbiomed.2010.01.039] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 12/23/2009] [Accepted: 01/28/2010] [Indexed: 11/20/2022]
Abstract
Sulfur mustard (bis-2-(chloroethyl) sulfide; SM) is a highly reactive vesicating and alkylating chemical warfare agent. A SM analog, 2-chloroethyl ethyl sulfide (CEES), has been utilized to elucidate mechanisms of toxicity and as a screen for therapeutics. Previous studies with SM and CEES have demonstrated a role for oxidative stress as well as decreased injury with antioxidant treatment. We tested whether posttreatment with the metalloporphyrin catalytic antioxidant AEOL 10150 would improve outcome in CEES-induced lung injury. Anesthetized rats inhaled 5% CEES for 15 min via a nose-only inhalation system. At 1 and 9 h after CEES exposure, rats were given AEOL 10150 (5 mg/kg, sc). At 18 h post-CEES exposure BALF lactate dehydrogenase activity, protein, IgM, red blood cells, and neutrophils were elevated but were decreased by AEOL 10150 treatment. Lung myeloperoxidase activity was increased after CEES inhalation and was ameliorated by AEOL 10150. The lung oxidative stress markers 8-OHdG and 4-HNE were elevated after CEES exposure and significantly decreased by AEOL 10150 treatment. These findings demonstrate that CEES inhalation increased lung injury, inflammation, and oxidative stress, and AEOL 10150 was an effective rescue agent. Further investigation utilizing catalytic antioxidants as treatment for SM inhalation injury is warranted.
Collapse
Affiliation(s)
- Heidi C. O’Neill
- Department of Pharmaceutical Sciences, University of Colorado at Denver
| | - Carl W. White
- Department of Pediatrics National Jewish Health, University of Colorado at Denver
- Department of Pharmaceutical Sciences, University of Colorado at Denver
| | - Livia A. Veress
- Health Sciences Center and The Children’s Hospital Denver Aurora, CO USA
| | - Tara B. Hendry-Hofer
- Department of Pediatrics National Jewish Health, University of Colorado at Denver
| | - Joan E. Loader
- Department of Pediatrics National Jewish Health, University of Colorado at Denver
| | - Elysia Min
- Department of Medicine, University of Colorado at Denver
| | - Jie Huang
- Department of Medicine, University of Colorado at Denver
| | - Raymond C. Rancourt
- Department of Pediatrics National Jewish Health, University of Colorado at Denver
| | - Brian J. Day
- Department of Medicine, University of Colorado at Denver
- Department of Pharmaceutical Sciences, University of Colorado at Denver
| |
Collapse
|
33
|
Ma X, He P, Sun P, Han P. Lipoic acid: an immunomodulator that attenuates glycinin-induced anaphylactic reactions in a rat model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2010; 58:5086-92. [PMID: 20302377 DOI: 10.1021/jf904403u] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The purpose of this study was to evaluate the attenuation effects of consuming a small dose of lipoic acid on soybean glycinin-induced hypersensitivity using a rat model. Sensitized by gavage with glycinin, weaned rats were orally supplemented with the diet containing 12.5 mug of lipoic acid/g (per diet). Results showed that untreated, sensitized rats demonstrated an increase in the level of serum IgE, the level of histamine release, and incidence of diarrhea and reduced growth performance compared with the controls (P < 0.05). Lipoic acid significantly (P < 0.05) improved growth performance while reducing mast cell numbers, the level of serum IgE, and the level of histamine release. Lipoic acid supplementation altered the balance of cytokines, attenuated the Th2-type immune response, and amended the ratio of CD4(+) to CD8(+) T cells (P < 0.05). Our results suggest that a small dose of lipoic acid has the potential to be an immunomodulator to prevent soybean-induced allergies.
Collapse
Affiliation(s)
- Xi Ma
- National Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China.
| | | | | | | |
Collapse
|
34
|
Makinde AY, Rizvi A, Crapo JD, Pearlstein RD, Slater JM, Gridley DS. A Metalloporphyrin Antioxidant Alters Cytokine Responses after Irradiation in a Prostate Tumor Model. Radiat Res 2010; 173:441-52. [DOI: 10.1667/rr1765.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
35
|
Comhair SAA, Erzurum SC. Redox control of asthma: molecular mechanisms and therapeutic opportunities. Antioxid Redox Signal 2010; 12:93-124. [PMID: 19634987 PMCID: PMC2824520 DOI: 10.1089/ars.2008.2425] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An imbalance in reducing and oxidizing (redox) systems favoring a more oxidative environment is present in asthma and linked to the pathophysiology of the defining symptoms and signs including airflow limitation, hyper-reactivity, and airway remodeling. High levels of hydrogen peroxide, nitric oxide ((*)NO), and 15-F(2t)-isoprostane in exhaled breath, and excessive oxidative protein products in lung epithelial lining fluid, peripheral blood, and urine provide abundant evidence for pathologic oxidizing processes in asthma. Parallel studies document loss of reducing potential by nonenzymatic and enzymatic antioxidants. The essential first line antioxidant enzymes superoxide dismutases (SOD) and catalase are reduced in asthma as compared to healthy individuals, with lowest levels in those patients with the most severe asthma. Loss of SOD and catalase activity is related to oxidative modifications of the enzymes, while other antioxidant gene polymorphisms are linked to susceptibility to develop asthma. Monitoring of exhaled (*)NO has entered clinical practice because it is useful to optimize asthma care, and a wide array of other biochemical oxidative and nitrative biomarkers are currently being evaluated for asthma monitoring and phenotyping. Novel therapeutic strategies that target correction of redox abnormalities show promise for the treatment of asthma.
Collapse
Affiliation(s)
- Suzy A A Comhair
- Pathobiology, Lerner Research Institute, and the Respiratory Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA.
| | | |
Collapse
|
36
|
Aguilera-Aguirre L, Bacsi A, Saavedra-Molina A, Kurosky A, Sur S, Boldogh I. Mitochondrial dysfunction increases allergic airway inflammation. THE JOURNAL OF IMMUNOLOGY 2009; 183:5379-87. [PMID: 19786549 DOI: 10.4049/jimmunol.0900228] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The prevalence of allergies and asthma among the world's population has been steadily increasing due to environmental factors. It has been described that exposure to ozone, diesel exhaust particles, or tobacco smoke exacerbates allergic inflammation in the lungs. These environmental oxidants increase the levels of cellular reactive oxygen species (ROS) and induce mitochondrial dysfunction in the airway epithelium. In this study, we investigated the involvement of preexisting mitochondrial dysfunction in the exacerbation of allergic airway inflammation. After cellular oxidative insult induced by ragweed pollen extract (RWE) exposure, we have identified nine oxidatively damaged mitochondrial respiratory chain-complex and associated proteins. Out of these, the ubiquinol-cytochrome c reductase core II protein (UQCRC2) was found to be implicated in mitochondrial ROS generation from respiratory complex III. Mitochondrial dysfunction induced by deficiency of UQCRC2 in airway epithelium of sensitized BALB/c mice prior the RWE challenge increased the Ag-induced accumulation of eosinophils, mucin levels in the airways, and bronchial hyperresponsiveness. Deficiency of UQCRC1, another oxidative damage-sensitive complex III protein, did not significantly alter cellular ROS levels or the intensity of RWE-induced airway inflammation. These observations suggest that preexisting mitochondrial dysfunction induced by oxidant environmental pollutants is responsible for the severe symptoms in allergic airway inflammation. These data also imply that mitochondrial defects could be risk factors and may be responsible for severe allergic disorders in atopic individuals.
Collapse
Affiliation(s)
- Leopoldo Aguilera-Aguirre
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | | | | | | | | | | |
Collapse
|
37
|
Sood A, Qualls C, Arynchyn A, Beckett WS, Gross MD, Steffes MW, Smith LJ, Holvoet P, Thyagarajan B, Jacobs DR. Obesity-asthma association: is it explained by systemic oxidant stress? Chest 2009; 136:1055-1062. [PMID: 19592473 DOI: 10.1378/chest.09-0493] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND The mechanism for the obesity-asthma association is unknown. This study evaluated the hypothesis that systemic oxidant stress explains this association. METHODS This cross-sectional study used year-20 follow-up evaluation data of 2,865 eligible participants in the Coronary Artery Risk Development in Young Adults (CARDIA) cohort. Current asthma was self-reported. Oxidant stress primarily was assessed by plasma F2-isoprostane concentrations. Obesity measures included categories of BMI and dual-energy x-ray absorptiometry-assessed fat mass index (FMI) and lean mass index (LMI). Logistic and linear regressions were used for analyses. RESULTS Asthma was associated with higher plasma F2-isoprostane concentrations (p = 0.049); however, this association was not significant when adjusted for either gender or BMI. The BMI-asthma association was seen only among women (p = 0.03; gender-specific interaction, p = 0.01), and this association was not explained by plasma F2-isoprostane levels. Similarly, both FMI and LMI were positively associated with asthma in women (p = 0.20 and 0.01, respectively). These associations also were not explained by plasma F2-isoprostane levels. Similar results were obtained when plasma levels of oxidized low-density lipoprotein were used instead of F2-isoprostane levels to study the BMI-asthma association at the year-15 evaluation. CONCLUSIONS Systemic oxidant stress, primarily assessed by plasma F2-isoprostane concentrations, was not independently associated with asthma and, therefore, may not explain the obesity-asthma association in women. The asthma-oxidant stress association is confounded by gender and obesity. This study is limited by the inability to measure airway oxidant stress. It is possible that another (as yet undetermined) measure of systemic oxidant stress may be more relevant in asthma.
Collapse
Affiliation(s)
- Akshay Sood
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM.
| | - Clifford Qualls
- Clinical Translational Sciences Center, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Alexander Arynchyn
- Department of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | | - Myron D Gross
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Michael W Steffes
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Lewis J Smith
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Paul Holvoet
- Department of Experimental Surgery and Anesthesiology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - David R Jacobs
- Division of Epidemiology, University of Minnesota, Minneapolis, MN; Institute for Nutrition Research, University of Oslo, Oslo, Norway
| |
Collapse
|
38
|
Nuclear factor kappaB, airway epithelium, and asthma: avenues for redox control. Ann Am Thorac Soc 2009; 6:249-55. [PMID: 19387025 DOI: 10.1513/pats.200806-054rm] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A wealth of recent studies points to the importance of airway epithelial cells in the orchestration of inflammatory responses in the allergic inflamed lung. Studies also point to a role of oxidative stress in the pathophysiology of chronic inflammatory diseases. This article provides a perspective on the significance of airway epithelial cells in allergic inflammation, and reviews the relevance of the transcription factor, nuclear factor kappaB, herein. We also provide the reader with a perspective on the role that oxidants can play in lung homeostasis, and address the concept of "redox biology." In addition, we review recent evidence that highlights potential inhibitory roles of oxidants on nuclear factor kappaB activation and inflammation, and discuss recent assays that have become available to probe the functional roles of oxidants in lung biology.
Collapse
|
39
|
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with a high incidence of morbidity and mortality. Cigarette smoke-induced oxidative stress is intimately associated with the progression and exacerbation of COPD and therefore targeting oxidative stress with antioxidants or boosting the endogenous levels of antioxidants is likely to have beneficial outcome in the treatment of COPD. Among the various antioxidants tried so far, thiol antioxidants and mucolytic agents, such as glutathione, N-acetyl-L-cysteine, N-acystelyn, erdosteine, fudosteine and carbocysteine; Nrf2 activators; and dietary polyphenols (curcumin, resveratrol, and green tea catechins/quercetin) have been reported to increase intracellular thiol status along with induction of GSH biosynthesis. Such an elevation in the thiol status in turn leads to detoxification of free radicals and oxidants as well as inhibition of ongoing inflammatory responses. In addition, specific spin traps, such as alpha-phenyl-N-tert-butyl nitrone, a catalytic antioxidant (ECSOD mimetic), porphyrins (AEOL 10150 and AEOL 10113), and a SOD mimetic M40419 have also been reported to inhibit cigarette smoke-induced inflammatory responses in vivo in the lung. Since a variety of oxidants, free radicals and aldehydes are implicated in the pathogenesis of COPD, it is possible that therapeutic administration of multiple antioxidants and mucolytics will be effective in management of COPD. However, a successful outcome will critically depend upon the choice of antioxidant therapy for a particular clinical phenotype of COPD, whose pathophysiology should be first properly understood. This article will review the various approaches adopted to enhance lung antioxidant levels, antioxidant therapeutic advances and recent past clinical trials of antioxidant compounds in COPD.
Collapse
Affiliation(s)
- Irfan Rahman
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
40
|
Nadeem A, Masood A, Siddiqui N. Oxidant--antioxidant imbalance in asthma: scientific evidence, epidemiological data and possible therapeutic options. Ther Adv Respir Dis 2009; 2:215-35. [PMID: 19124374 DOI: 10.1177/1753465808094971] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Prevalence of asthma has increased considerably in recent decades throughout the world especially in developed countries. Airway inflammation is thought to be prime cause for repeated episodes of airway obstruction in asthmatics. Several studies have shown that reactive oxygen species (ROS) play a key role in initiation as well as amplification of inflammation in asthmatic airways. Excessive ROS production in asthma leads to alteration in key enzymatic as well as nonenzymatic antioxidants such as glutathione, vitamins C and E, beta-carotene, uric acid, thioredoxin, superoxide dismutases, catalase, and glutathione peroxidases leading to oxidant-antioxidant imbalance in airways. Oxidant-antioxidant imbalance leads to pathophysiological effects associated with asthma such as vascular permeability, mucus hypersecretion, smooth muscle contraction, and epithelial shedding. Epidemiological data also support the scientific evidence of oxidant-antioxidant imbalance in asthmatics. Therefore, the supplementation of antioxidants to boost the endogenous antioxidants or scavenge excessive ROS production could be utilized to dampen/prevent the inflammatory response in asthma by restoring oxidant-antioxidant balance. This review summarizes the scientific and epidemiological evidence linking asthma with oxidant-antioxidant imbalance and possible antioxidant strategies that can be used therapeutically for better management of asthma.
Collapse
Affiliation(s)
- Ahmed Nadeem
- Department of Physiology and Pharmacology, Health Sciences Center North West Virginia University Morgantown WV 26506, USA.
| | | | | |
Collapse
|
41
|
Mao XW, Crapo JD, Mekonnen T, Lindsey N, Martinez P, Gridley DS, Slater JM. Radioprotective effect of a metalloporphyrin compound in rat eye model. Curr Eye Res 2009; 34:62-72. [PMID: 19172472 DOI: 10.1080/02713680802546948] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE The purpose of this study was to evaluate the efficacy of the antioxidant Mn (III) tetrakis (N-ethylpyridinium-2-yl) porphyrin (MnTE-2-PyP) in protecting ocular tissue and retinal microvasculature from radiation damage. MATERIALS AND METHODS 75 rats were treated with Mn TE-2-PyP at 2.5 micro g/injection into one eye an hour before proton irradiation. The radiation was delivered in a single fraction to total doses of 8 Gray (Gy) or 28 Gy; Rats were sacrificed 3 days and 3, 6, 9, and 12 months thereafter for histology and quantification of photoreceptor cell populations and retinal capillary changes. RESULTS By 6 months following radiation, there was significant loss of retinal outer and inner nuclear layers in eyes receiving radiation only (8 and 28 Gy) (p < 0.05) compared to their controls and to the eyes of rats treated with radiation plus metalloporphyrin. Retinal microvessel length density decreased significantly 6 months following 28 Gy (p < 0.05) compared to their controls and to MnTE-2-PyP treated rats. By 12 months following irradiation, irradiated eyes showed extensive damage to the photoreceptor layer, whereas the eyes of animals receiving radiation plus MnTE-2-PyP showed almost no morphological damage. MnTE-2-PyP treatment also suppressed radiation-induced apoptosis in our study. CONCLUSIONS These results demonstrated that MnTE-2-PyP protected both photoreceptors and retinal capillaries from radiation damage, suggesting that this metalloporphyrin antioxidant is effective in regulating the damage induced by proton radiation.
Collapse
Affiliation(s)
- X W Mao
- Department of Radiation Medicine, Molecular Radiation Biology Laboratories, Loma Linda University and Medical Center, Loma Linda, California, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Yu DH, Kim MO, Kim SH, Shin MJ, Kim BS, Kim HJ, Lee SR, Lee SG, Yoo SA, Kim WU, Hyun BH, Park YS, Kim TY, Ryoo ZY. The Therapeutic Effect of Extracellular Superoxide Dismutase (EC-SOD) Mouse Embryonic Fibroblast (MEF) on Collagen-Induced Arthritis (CIA) Mice. Cell Transplant 2008; 17:1371-80. [DOI: 10.3727/096368908787648029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Rheumatoid arthritis is a chronic inflammatory disease. The generation of reactive oxygen species (ROS) within an inflamed joint has been suggested as playing a significant pathogenic role. Extracellular superoxide dismutase (EC-SOD) is a major scavenger enzyme of ROS, which has received growing attention for its therapeutic potential. To investigate the therapeutic effect of EC-SOD in mice with collagen-induced arthritis (CIA), we used mouse embryonic fibroblast (MEF) of transgenic mice that overexpresses EC-SOD on the skin by using hK14 promoter. DBA/1 mice that had been treated with bovine type II collagen were administrated subcutaneous injections of EC-SOD transgenic MEF (each at 1.4 × 106 cells) on days 28, 35, and 42 after primary immunization. To test EC-SOD activity, blood samples were collected in each group on day 49. The EC-SOD activity was nearly 1.5-fold higher in the transgenic MEF-treated group than in the non-transgenic MEF-treated group (p < 0.05). The severity of arthritis in mice was scored in a double-blind manner, with each paw being assigned a separate clinical score. The severity of arthritis in EC-SOD transgenic MEF-treated mice was significantly suppressed in the arthritic clinical score (p < 0.05). To investigate the alteration of cytokine levels, ELISA was used to measure blood samples. Levels of IL-1β and TNF-α were reduced in the transgenic MEF-treated group (p < 0.05). Abnormalities of the joints were examined by H&E staining. There were no signs of inflammation except for mild hyperplasia of the synovium in the transgenic MEF-treated group. The proliferation of CII-specific T cells was lower in the transgenic MEF-treated mice than in those in the other groups. The transfer of EC-SOD transgenic MEF has shown a therapeutic effect in CIA mice and this approach may be a safer and more effective form of therapy for rheumatoid arthritis.
Collapse
Affiliation(s)
- Dong Hoon Yu
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, 702-701, Korea
| | - Myoung Ok Kim
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, 702-701, Korea
| | - Sung Hyun Kim
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, 702-701, Korea
| | - Mi Jung Shin
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, 702-701, Korea
| | - Bong Soo Kim
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, 702-701, Korea
| | - Hei Jung Kim
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, 702-701, Korea
| | - Sang Ryeul Lee
- Department of Immunology and Dermatology, College of Medicine, Catholic University, Seoul, 137-040, Korea
| | - Sang Gyu Lee
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, 702-701, Korea
| | - Seung-Ah Yoo
- Division of Rheumatology, Department of Internal Medicine, Catholic University of Korea, Seoul, Korea
| | - Wan Uk Kim
- Division of Rheumatology, Department of Internal Medicine, Catholic University of Korea, Seoul, Korea
| | - Byung Hwa Hyun
- Disease Model Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea
| | - Young Sik Park
- School of Life and Food Sciences, Kyungpook National University, Daegu, 702-701, Korea
| | - Tae Yoon Kim
- Department of Immunology and Dermatology, College of Medicine, Catholic University, Seoul, 137-040, Korea
| | - Zae Young Ryoo
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, 702-701, Korea
| |
Collapse
|
43
|
Tripathi P, Singh BP, Arora N. Mutated glutathione-S-transferase reduced airway inflammation by limiting oxidative stress and Th2 response. Free Radic Biol Med 2008; 45:1413-9. [PMID: 18786635 DOI: 10.1016/j.freeradbiomed.2008.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 07/24/2008] [Accepted: 08/05/2008] [Indexed: 11/20/2022]
Abstract
Oxidative stress is an important factor in the pathogenesis of asthma. Furthermore, antioxidants like GST are reduced in asthma patients. In the present study, the therapeutic effects of exogenous GST and mGST were evaluated in a mice model. GST mutated at residues 21/27 has reduced IgE binding with similar enzyme activity as that of GST. To evaluate the therapeutic effects of GST, BALB/c mice were immunized and challenged with ovalbumin. Mice were given GST, mGST, and alpha-lipoic acid by inhalation and sacrificed on Day 31 to evaluate inflammation and oxidative stress. Mice treated with mGST showed significantly reduced total cell count (P<0.01) and eosinophils (P<0.01) in BALF compared to GST- or PBS-treated groups. The lung inflammation score was lowest for the mGST-treated group along with reduced IL-4 (P<0.01) and OVA-specific IgE than other groups. Oxidative stress as per the lipid peroxidation level in BALF of mGST-treated mice was reduced significantly in comparison to PBS- or GST-treated mice. In conclusion, inhalation of mGST reduced airway inflammation in mice. Mutated GST with reduced allergenicity has better therapeutic potential and can be explored as an adjunct therapy in asthma.
Collapse
Affiliation(s)
- Prabhanshu Tripathi
- Institute of Genomics and Integrative Biology, Mall Road, Delhi 110 007, India
| | | | | |
Collapse
|
44
|
Day BJ. Catalase and glutathione peroxidase mimics. Biochem Pharmacol 2008; 77:285-96. [PMID: 18948086 DOI: 10.1016/j.bcp.2008.09.029] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 09/18/2008] [Accepted: 09/19/2008] [Indexed: 12/14/2022]
Abstract
Overproduction of the reactive oxygen species (ROS) superoxide (O(2)(-)) and hydrogen peroxide (H(2)O(2)) are increasingly implicated in human disease and aging. ROS are also being explored as important modulating agents in a number of cell signaling pathways. Earlier work has focused on development of small catalytic scavengers of O(2)(-), commonly referred to as superoxide dismutase (SOD) mimetics. Many of these compounds also have substantial abilities to catalytically scavenge H(2)O(2) and peroxynitrite (ONOO(-)). Peroxides have been increasingly shown to disrupt cell signaling cascades associated with excessive inflammation associated with a wide variety of human diseases. Early studies with enzymatic scavengers like SOD frequently reported little or no beneficial effect in biologic models unless SOD was combined with catalase or a peroxidase. Increasing attention has been devoted to developing catalase or peroxidase mimetics as a way to treat overt inflammation associated with the pathophysiology of many human disorders. This review will focus on recent development of catalytic scavengers of peroxides and their potential use as therapeutic agents for pulmonary, cardiovascular, neurodegenerative and inflammatory disorders.
Collapse
Affiliation(s)
- Brian J Day
- Department of Medicine, National Jewish Health, Departments of Medicine, Immunology & Pharmaceutical Sciences, University of Colorado Health Sciences Center, Denver, CO 80206, USA.
| |
Collapse
|
45
|
Nambiar S, Viswanathan S, Zachariah B, Hanumanthappa N, Magadi SG. Oxidative stress in prehypertension: rationale for antioxidant clinical trials. Angiology 2008; 60:221-34. [PMID: 18796443 DOI: 10.1177/0003319708319781] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Prehypertension has been recently described as an independent category of blood pressure. Mounting evidence suggests that blood pressure in the prehypertensive range is associated with an increased risk of developing hypertension and cardiovascular disease. Several reports have assigned a critical role for oxidative stress in these disease processes. This review focuses on the clinical and experimental studies done in prehypertension and hypertension within the context of oxidative stress. This article also provides insights into why diverse therapeutic interventions, which have in common the ability to reduce oxidative stress, can impede or delay the onset of hypertension in prehypertension subjects.
Collapse
Affiliation(s)
- Selvaraj Nambiar
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605 006, India
| | | | | | | | | |
Collapse
|
46
|
Abstract
There has been a marked increase in the global prevalence, morbidity, and mortality of asthma, and its associated economic burden has also grown over the last 40 years. Approximately 300 million people worldwide currently have asthma, and its prevalence increases by 50% every decade. Airway inflammation is the most proximate cause of the recurrent episodes of airflow limitation in asthma. Recent research has revealed that numerous biologically active proinflammatory mediators are responsible for the pathogenesis of asthma. Among these mediators, there is increasing evidence that endogenous or exogenous reactive oxygen species (ROS) and reactive nitrogen species (RNS) are responsible for the airway inflammation of asthma. Many reports have shown that there is an excessive production of ROS and RNS in the airways of asthmatic individuals compared with healthy subjects. Excessively produced ROS and RNS have been reported to lead to airway inflammation, airway hyper-responsiveness, airway microvascular hyperpermeability, tissue injury, and remodeling in animal models and human studies. Although human lungs have a potent antioxidant system, excessive oxidative and nitrative stress leads to an imbalance of oxidants/antioxidants. This review describes the rapidly accruing data linking oxidative and nitrative events to the pathogenesis of bronchial asthma.
Collapse
Affiliation(s)
- Hisatoshi Sugiura
- Third Department of Internal Medicine, Wakayama Medical University School of Medicine, Wakayama City, Wakayama, Japan
| | | |
Collapse
|
47
|
Thompson PJ, Chung F. Respirology year-in-review 2006: Basic science. Respirology 2007; 12:184-90. [PMID: 17298449 DOI: 10.1111/j.1440-1843.2007.01045.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The year 2006 was a good year for basic science publications in Respirology with a lot of the studies being relevant to clinical practice. In this respect many of the publications focused on biomarkers of disease and so much so that these have been discussed at the end of this review. The majority of manuscripts are related to airway diseases, respiratory infections, interstitial lung diseases and lung cancers, and are discussed under these headings.
Collapse
Affiliation(s)
- Philip J Thompson
- Lung Institute of Western Australia, Centre for Asthma, Allergy & Respiratory Research, University of Western Australia, Perth, Western Australia, Australia.
| | | |
Collapse
|
48
|
Cantin AM, White TB, Cross CE, Forman HJ, Sokol RJ, Borowitz D. Antioxidants in cystic fibrosis. Conclusions from the CF antioxidant workshop, Bethesda, Maryland, November 11-12, 2003. Free Radic Biol Med 2007; 42:15-31. [PMID: 17157190 PMCID: PMC2696206 DOI: 10.1016/j.freeradbiomed.2006.09.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Revised: 09/11/2006] [Accepted: 09/25/2006] [Indexed: 02/07/2023]
Abstract
Although great strides are being made in the care of individuals with cystic fibrosis (CF), this condition remains the most common fatal hereditary disease in North America. Numerous links exist between progression of CF lung disease and oxidative stress. The defect in CF is the loss of function of the transmembrane conductance regulator (CFTR) protein; recent evidence that CFTR expression and function are modulated by oxidative stress suggests that the loss may result in a poor adaptive response to oxidants. Pancreatic insufficiency in CF also increases susceptibility to deficiencies in lipophilic antioxidants. Finally the airway infection and inflammatory processes in the CF lung are potential sources of oxidants that can affect normal airway physiology and contribute to the mechanisms causing characteristic changes associated with bronchiectasis and loss of lung function. These multiple abnormalities in the oxidant/antioxidant balance raise several possibilities for therapeutic interventions that must be carefully assessed.
Collapse
Affiliation(s)
- André M. Cantin
- Pulmonary Division, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec, Canada J1H 5N4
| | | | - Carroll E. Cross
- Pulmonary-Critical Care Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Henry Jay Forman
- Division of Natural Sciences, University of California, Merced, CA, USA
| | - Ronald J. Sokol
- Pediatric Gastroenterology, Hepatology and Nutrition, University of Colorado School of Medicine and The Children's Hospital, Denver, CO, USA
| | - Drucy Borowitz
- Pediatric Pulmonology, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
49
|
Suchankova J, Voprsalova M, Kottova M, Semecky V, Visnovsky P. Effects of oral alpha-tocopherol on lung response in rat model of allergic asthma. Respirology 2006; 11:414-21. [PMID: 16771910 DOI: 10.1111/j.1440-1843.2006.00864.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE AND BACKGROUND Asthma is a chronic inflammatory disease in which an oxidant/antioxidant imbalance plays an important role. d-alpha-tocopherol (biologically the most active form of vitamin E) has redox properties and by scavenging the free radicals can act as an antioxidant. The aim of this study was to examine the effects of orally administered alpha-tocopherol in a rat model of allergic asthma. METHODOLOGY Actively sensitized rats (OA) were treated with alpha-tocopherol (400 mg/kg/day for 10 days) or vehicle; 1 h after the last dose, they were challenged with antigen aerosol. The antigen-induced airway hyperresponsiveness to direct bronchoconstrictor (serotonin), the inflammatory cell infiltrate and histological changes were determined 1 or 24 h after the antigen challenge. RESULTS Alpha-tocopherol pretreatment was not significantly effective at reducing the studied parameters when compared with controls, even though there was a tendency to a reduction in bronchial responsiveness and in eosinophil and neutrophil infiltration. CONCLUSION Alpha-tocopherol when administered in the chosen study design in an animal model of asthma had no major effect on airway inflammation. The effect of antioxidants deserves further evaluation.
Collapse
Affiliation(s)
- Jana Suchankova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Prague, Czech Republic.
| | | | | | | | | |
Collapse
|
50
|
Snelgrove RJ, Edwards L, Williams AE, Rae AJ, Hussell T. In the Absence of Reactive Oxygen Species, T Cells Default to a Th1 Phenotype and Mediate Protection against PulmonaryCryptococcus neoformansInfection. THE JOURNAL OF IMMUNOLOGY 2006; 177:5509-16. [PMID: 17015737 DOI: 10.4049/jimmunol.177.8.5509] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In recent years, the prevalence of invasive fungal infections has increased, attributed mostly to the rising population of immunocompromised individuals. Cryptococcus neoformans has been one of the most devastating, with an estimated 6-8% of AIDS-infected patients succumbing to Cryptococcus-associated meningitis. Reactive oxygen species (ROS) are potent antimicrobial agents but also play a significant role in regulating immune cell phenotype, but cause immunopathology when produced in excess. We now show that mice lacking phagocyte NADPH oxidase have heightened macrophage and Th1 responses and improved pathogen containment within pulmonary granulomatous lesions. Consequently, dissemination of this fungus to the brain is diminished, an effect that is independent of IL-12. Similar results are described using the metalloporphyrin antioxidant manganese(III) tetrakis(N-ethyl pyridinium-2-yl)porphyrin, which also promoted a protective Th1 response and reduced dissemination to the brain. These findings are in sharp contrast to the protective potential of ROS against other fungal pathogens, and highlight the pivotal role that ROS can fulfill in shaping the profile of the host's immune response.
Collapse
Affiliation(s)
- Robert J Snelgrove
- Kennedy Institute of Rheumatology, Imperial College London, Hammersmith, London, United Kingdom
| | | | | | | | | |
Collapse
|