1
|
Safety aspects of natural food additives frequently used at their maximum levels in South Korea. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-021-00191-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
2
|
Oxygen Toxicity to the Immature Lung-Part I: Pathomechanistic Understanding and Preclinical Perspectives. Int J Mol Sci 2021; 22:ijms222011006. [PMID: 34681665 PMCID: PMC8540649 DOI: 10.3390/ijms222011006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 01/13/2023] Open
Abstract
In utero, the fetus and its lungs develop in a hypoxic environment, where HIF-1α and VEGFA signaling constitute major determinants of further development. Disruption of this homeostasis after preterm delivery and extrauterine exposure to high fractions of oxygen are among the key events leading to bronchopulmonary dysplasia (BPD). Reactive oxygen species (ROS) production constitutes the initial driver of pulmonary inflammation and cell death, altered gene expression, and vasoconstriction, leading to the distortion of further lung development. From preclinical studies mainly performed on rodents over the past two decades, the deleterious effects of oxygen toxicity and the injurious insults and downstream cascades arising from ROS production are well recognized. This article provides a concise overview of disease drivers and different therapeutic approaches that have been successfully tested within experimental models. Despite current studies, clinical researchers are still faced with an unmet clinical need, and many of these strategies have not proven to be equally effective in clinical trials. In light of this challenge, adapting experimental models to the complexity of the clinical situation and pursuing new directions constitute appropriate actions to overcome this dilemma. Our review intends to stimulate research activities towards the understanding of an important issue of immature lung injury.
Collapse
|
3
|
Liu Z, Xu S, Ji Z, Xu H, Zhao W, Xia Z, Xu R. Mechanistic study of mtROS-JNK-SOD2 signaling in bupivacaine-induced neuron oxidative stress. Aging (Albany NY) 2021; 12:13463-13476. [PMID: 32658869 PMCID: PMC7377901 DOI: 10.18632/aging.103447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/23/2020] [Indexed: 12/27/2022]
Abstract
Manganese superoxide dismutase (SOD2) is a key enzyme to scavenge free radical superoxide in the mitochondrion. SOD2 deficiency leads to oxidative injury in cells. Bupivacaine, a local anesthetic commonly used in clinic, could induce neurotoxic injury via oxidative stress. The role and the mechanism of SOD2 regulation in bupivacaine-induced oxidative stress remains unclear. Here, bupivacaine was used to treat Sprague-Dawley rats with intrathecal injection and culture human neuroblastoma cells for developing vivo injury model and vitro injury model. The results showed that bupivacaine caused the over-production of mitochondrial reactive oxygen species (mtROS), the activation of C-Jun N-terminal kinase (JNK), and the elevation of SOD2 transcription. Decrease of mtROS with N-acetyl-L-cysteine attenuated the activation of JNK and the increase of SOD2 transcription. Inhibition of JNK signaling with a small interfering RNA (siRNA) or with sp600125 down-regulated the increase of SOD2 transcription. SOD2 gene knock-down exacerbated bupivacaine-induced mtROS generation and neurotoxic injury but had no effect on JNK phosphorylation. Mito-TEMPO (a mitochondria-targeted antioxidant) could protect neuron against bupivacaine-induced toxic injury. Collectively, our results confirm that mtROS stimulates the transcription of SOD2 via activating JNK signaling in bupivacaine-induced oxidative stress. Enhancing antioxidant ability of SOD2 might be crucial in combating bupivacaine-induced neurotoxic injury.
Collapse
Affiliation(s)
- Zhongjie Liu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shiyuan Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhonghua Ji
- Department of Anesthesiology, Affiliated Zhuhai Hospital of Jinan University, Zhuhai, Guangdong Province, China
| | - Huali Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wei Zhao
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Rui Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
4
|
Metabolic Remodelling: An Accomplice for New Therapeutic Strategies to Fight Lung Cancer. Antioxidants (Basel) 2019; 8:antiox8120603. [PMID: 31795465 PMCID: PMC6943435 DOI: 10.3390/antiox8120603] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022] Open
Abstract
Metabolic remodelling is a hallmark of cancer, however little has been unravelled in its role in chemoresistance, which is a major hurdle to cancer control. Lung cancer is a leading cause of death by cancer, mainly due to the diagnosis at an advanced stage and to the development of resistance to therapy. Targeted therapeutic agents combined with comprehensive drugs are commonly used to treat lung cancer. However, resistance mechanisms are difficult to avoid. In this review, we will address some of those therapeutic regimens, resistance mechanisms that are eventually developed by lung cancer cells, metabolic alterations that have already been described in lung cancer and putative new therapeutic strategies, and the integration of conventional drugs and genetic and metabolic-targeted therapies. The oxidative stress is pivotal in this whole network. A better understanding of cancer cell metabolism and molecular adaptations underlying resistance mechanisms will provide clues to design new therapeutic strategies, including the combination of chemotherapeutic and targeted agents, considering metabolic intervenients. As cancer cells undergo a constant metabolic adaptive drift, therapeutic regimens must constantly adapt.
Collapse
|
5
|
Perez M, Robbins ME, Revhaug C, Saugstad OD. Oxygen radical disease in the newborn, revisited: Oxidative stress and disease in the newborn period. Free Radic Biol Med 2019; 142:61-72. [PMID: 30954546 PMCID: PMC6791125 DOI: 10.1016/j.freeradbiomed.2019.03.035] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/06/2019] [Accepted: 03/29/2019] [Indexed: 12/28/2022]
Abstract
Thirty years ago, there was an emerging appreciation for the significance of oxidative stress in newborn disease. This prompted a renewed interest in the impact of oxygen therapy for the newborn in the delivery room and beyond, especially in premature infants. Today, the complexity of oxidative stress both in normal regulation and pathology is better understood, especially as it relates to neonatal mitochondrial oxidative stress responses to hyperoxia. Mitochondria are recipients of oxidative damage and have a propensity for oxidative self-injury that has been implicated in the pathogenesis of neonatal lung diseases. Similarly, both intrauterine growth restriction (IUGR) and macrosomia are associated with mitochondrial dysfunction and oxidative stress. Additionally, reoxygenation with 100% O2 in a hypoxic-ischemic newborn lamb model increased the production of pro-inflammatory cytokines in the brain. Moreover, the interplay between inflammation and oxidative stress in the newborn is better understood because of animal studies. Transcriptomic analyses have found a number of genes to be differentially expressed in murine models of bronchopulmonary dysplasia (BPD). Epigenetic changes have also been detected both in animal models of BPD and premature infants exposed to oxygen. Antioxidant therapy to prevent newborn disease has not been very successful; however, new therapeutic principles, like melatonin, are under investigation.
Collapse
Affiliation(s)
- Marta Perez
- Division of Neonatology, Stanley Manne Children's Research Institute, Ann and Robert H Lurie Children's Hospital, Chicago, IL, United States; Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Mary E Robbins
- Division of Neonatology, Stanley Manne Children's Research Institute, Ann and Robert H Lurie Children's Hospital, Chicago, IL, United States; Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Cecilie Revhaug
- Department of Pediatric Research, University of Oslo, Oslo University Hospital, Norway
| | - Ola D Saugstad
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States; Department of Pediatric Research, University of Oslo, Oslo University Hospital, Norway.
| |
Collapse
|
6
|
Valencia AM, Abrantes MA, Hasan J, Aranda JV, Beharry KD. Reactive Oxygen Species, Biomarkers of Microvascular Maturation and Alveolarization, and Antioxidants in Oxidative Lung Injury. REACTIVE OXYGEN SPECIES (APEX, N.C.) 2018; 6:373-388. [PMID: 30533532 DOI: 10.20455/ros.2018.867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The lungs of extremely low gestational age neonates (ELGANs) are deficient in pulmonary surfactant and are incapable of efficient gas exchange necessary for successful transition from a hypoxic intrauterine environment to ambient air. To improve gas exchange and survival, ELGANs often receive supplemental oxygen with mechanical ventilation which disrupts normal lung developmental processes, including microvascular maturation and alveolarization. Factors that regulate these developmental processes include vascular endothelial growth factor and matrix metalloproteinases, both of which are influenced by generation of oxygen byproducts, or reactive oxygen species (ROS). ELGANs are also deficient in antioxidants necessary to scavenge excessive ROS. Thus, the accumulation of ROS in the preterm lungs exposed to prolonged hyperoxia, results in inflammation and development of bronchopulmonary dysplasia (BPD), a form of chronic lung disease (CLD). Despite advances in neonatal care, BPD/CLD remains a major cause of neonatal morbidity and mortality. The underlying mechanisms are not completely understood, and the benefits of current therapeutic interventions are limited. The association between ROS and biomarkers of microvascular maturation and alveolarization, as well as antioxidant therapies in the setting of hyperoxia-induced neonatal lung injury are reviewed in this article.
Collapse
Affiliation(s)
- Arwin M Valencia
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Saddleback Memorial Hospital, Laguna Hills, CA 92653, USA
| | - Maria A Abrantes
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Kaiser Permanente, Anaheim, CA 92806, USA
| | - Jamal Hasan
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Miller's Children's and Women's Hospital, Long Beach, CA 90806, USA
| | - Jacob V Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA.,Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Kay D Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA.,Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA
| |
Collapse
|
7
|
Resseguie EA, Brookes PS, O’Reilly MA. SMG-1 kinase attenuates mitochondrial ROS production but not cell respiration deficits during hyperoxia. Exp Lung Res 2017; 43:229-239. [PMID: 28749708 PMCID: PMC5956894 DOI: 10.1080/01902148.2017.1339143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/02/2017] [Indexed: 12/29/2022]
Abstract
PURPOSE Supplemental oxygen (hyperoxia) used to treat individuals in respiratory distress causes cell injury by enhancing the production of toxic reactive oxygen species (ROS) and inhibiting mitochondrial respiration. The suppressor of morphogenesis of genitalia (SMG-1) kinase is activated during hyperoxia and promotes cell survival by phosphorylating the tumor suppressor p53 on serine 15. Here, we investigate whether SMG-1 and p53 blunt this vicious cycle of progressive ROS production and decline in mitochondrial respiration seen during hyperoxia. MATERIALS AND METHODS Human lung adenocarcinoma A549 and H1299 or colon carcinoma HCT116 cells were depleted of SMG-1, UPF-1, or p53 using RNA interference, and then exposed to room air (21% oxygen) or hyperoxia (95% oxygen). Immunoblotting was used to evaluate protein expression; a Seahorse Bioanalyzer was used to assess cellular respiration; and flow cytometry was used to evaluate fluorescence intensity of cells stained with mitochondrial or redox sensitive dyes. RESULTS Hyperoxia increased mitochondrial and cytoplasmic ROS and suppressed mitochondrial respiration without changing mitochondrial mass or membrane potential. Depletion of SMG-1 or its cofactor, UPF1, significantly enhanced hyperoxia-induced mitochondrial but not cytosolic ROS abundance. They did not affect mitochondrial mass, membrane potential, or hyperoxia-induced deficits in mitochondrial respiration. Genetic depletion of p53 in A549 cells and ablation of the p53 gene in H1299 or HCT116 cells revealed that SMG-1 influences mitochondrial ROS through activation of p53. CONCLUSIONS Our findings show that hyperoxia does not promote a vicious cycle of progressive mitochondrial ROS and dysfunction because SMG-1-p53 signaling attenuates production of mitochondrial ROS without preserving respiration. This suggests antioxidant therapies that blunt ROS production during hyperoxia may not suffice to restore cellular respiration.
Collapse
Affiliation(s)
- Emily A. Resseguie
- Department of Environmental Medicine, The University of Rochester, Rochester, New York, USA
| | - Paul S. Brookes
- Department of Anesthesiology, The University of Rochester, Rochester, New York, USA
| | - Michael A. O’Reilly
- Department of Environmental Medicine, The University of Rochester, Rochester, New York, USA
- Department of Pediatrics, The University of Rochester, Rochester, New York, USA
| |
Collapse
|
8
|
Potteti HR, Rajasekaran S, Rajamohan SB, Tamatam CR, Reddy NM, Reddy SP. Sirtuin 1 Promotes Hyperoxia-Induced Lung Epithelial Cell Death Independent of NF-E2-Related Factor 2 Activation. Am J Respir Cell Mol Biol 2017; 54:697-706. [PMID: 26465873 DOI: 10.1165/rcmb.2014-0056oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Lung epithelial cell damage accompanied by death is a cardinal feature of toxicant- and prooxidant-induced acute lung injury. The transcription factor nuclear factor (erythroid-derived 2)-like 2 (NEF2L2 or NRF2) activates several antioxidant enzymes (AOEs) and prosurvival genes in response to oxidant stress, and its deficiency enhances susceptibility to hyperoxic lung injury and other oxidant-induced lung pathologies. Sirtuin 1 (SIRT1) regulates cell growth and survival in response to both physiological and pathological stresses by selectively deacetylating multiple proteins required for chromatin remodeling and transcription; therefore, we sought to examine potential SIRT1-NRF2 cross-talk in the regulation of AOE expression during hyperoxia-induced lung epithelial cell death. Unexpectedly, pharmacological inhibition or small interfering RNA-mediated depletion of SIRT1 caused a reduction in cell death, accompanied by reduced levels of NRF2-dependent AOE expression in chronic hyperoxia. NRF2 acetylation was markedly and transiently higher in cells exposed to acute (6 h) hyperoxia. Sirtinol blocked this acute effect, but NRF2 acetylation was low or undetectable in cells exposed to chronic hyperoxia (24-36 h) both with and without sirtinol. SIRT1 activation by resveratrol augmented hyperoxia-induced death in cells with NRF2 deficiency. SIRT1 inhibition or depletion led to a reduced activation of the cell-death executioner caspase 3, whereas caspase inhibition prevented death. Consistent with these results, sirtinol attenuated hyperoxia-induced lung alveolar permeability and toxicity in vivo. Collectively, these results reveal that, in chronic hyperoxia, SIRT1 promotes hyperoxia-induced lung epithelial cell damage and death by altering pro- and antiapoptotic balance, not by dampening optimal NRF2-dependent AOE expression.
Collapse
Affiliation(s)
- Haranatha R Potteti
- Division of Developmental Biology and Basic Research, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Subbiah Rajasekaran
- Division of Developmental Biology and Basic Research, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Senthilkumar B Rajamohan
- Division of Developmental Biology and Basic Research, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Chandramohan R Tamatam
- Division of Developmental Biology and Basic Research, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Narsa M Reddy
- Division of Developmental Biology and Basic Research, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Sekhar P Reddy
- Division of Developmental Biology and Basic Research, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
9
|
Patel VS, Sampat V, Espey MG, Sitapara R, Wang H, Yang X, Ashby CR, Thomas DD, Mantell LL. Ascorbic Acid Attenuates Hyperoxia-Compromised Host Defense against Pulmonary Bacterial Infection. Am J Respir Cell Mol Biol 2016; 55:511-520. [PMID: 27120084 DOI: 10.1165/rcmb.2015-0310oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Supraphysiological concentrations of oxygen (hyperoxia) can compromise host defense and increase susceptibility to bacterial infections, causing ventilator-associated pneumonia. The phagocytic activity of macrophages is impaired by hyperoxia-induced increases in the levels of reactive oxygen species (ROS) and extracellular high-mobility group box protein B1 (HMGB1). Ascorbic acid (AA), an essential nutrient and antioxidant, has been shown to be beneficial in various animal models of ROS-mediated diseases. The aim of this study was to determine whether AA could attenuate hyperoxia-compromised host defense and improve macrophage functions against bacterial infections. C57BL/6 male mice were exposed to hyperoxia (≥98% O2, 48 h), followed by intratracheal inoculation with Pseudomonas aeruginosa, and simultaneous intraperitoneal administration of AA. AA (50 mg/kg) significantly improved bacterial clearance in the lungs and airways, and significantly reduced HMGB1 accumulation in the airways. The incubation of RAW 264.7 cells (a macrophage-like cell line) with AA (0-1,000 μM) before hyperoxic exposure (95% O2) stabilized the phagocytic activity of macrophages in a concentration-dependent manner. The AA-enhanced macrophage function was associated with significantly decreased production of intracellular ROS and accumulation of extracellular HMGB1. These data suggest that AA supplementation can prevent or attenuate the development of ventilator-associated pneumonia in patients receiving oxygen support.
Collapse
Affiliation(s)
- Vivek S Patel
- 1 Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, Queens, New York
| | - Vaishali Sampat
- 1 Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, Queens, New York
| | | | - Ravikumar Sitapara
- 1 Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, Queens, New York
| | - Haichao Wang
- 3 The Feinstein Institute for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, New York; and
| | - Xiaojing Yang
- 1 Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, Queens, New York
| | - Charles R Ashby
- 1 Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, Queens, New York
| | - Douglas D Thomas
- 4 Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois
| | - Lin L Mantell
- 1 Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, Queens, New York.,3 The Feinstein Institute for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, New York; and
| |
Collapse
|
10
|
Manganese superoxide dismutase deficiency triggers mitochondrial uncoupling and the Warburg effect. Oncogene 2014; 34:4229-37. [PMID: 25362851 PMCID: PMC4859767 DOI: 10.1038/onc.2014.355] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 09/07/2014] [Accepted: 09/19/2014] [Indexed: 12/22/2022]
Abstract
Manganese superoxide dismutase (MnSOD) is a mitochondrially localized primary antioxidant enzyme, known to be essential for the survival of aerobic life and to have important roles in tumorigenesis. Here, we show that MnSOD deficiency in skin tissues of MnSOD-heterozygous knockout (Sod2(+/-)) mice leads to increased expresson of uncoupling proteins (UCPs). When MnSOD is deficient, superoxide radical and its resulting reactive oxygen species (ROS) activate ligand binding to peroxisome proliferator-activated receptor alpha (PPARα), suggesting that the activation of PPARα signaling is a major mechanism underlying MnSOD-dependent UCPs expression that consequently triggers the PI3K/Akt/mTOR pathway, leading to increased aerobic glycolysis. Knockdown of UCPs and mTOR suppresses lactate production and increases ATP levels, suggesting that UCPs contribute to increased glycolysis. These results highlight the existence of a free radical-mediated mechanism that activates mitochondria uncoupling to reduce ROS production, which precedes the glycolytic adaptation described as the Warburg Effect.
Collapse
|
11
|
Poonyagariyagorn HK, Metzger S, Dikeman D, Mercado AL, Malinina A, Calvi C, McGrath-Morrow S, Neptune ER. Superoxide dismutase 3 dysregulation in a murine model of neonatal lung injury. Am J Respir Cell Mol Biol 2014; 51:380-90. [PMID: 24673633 DOI: 10.1165/rcmb.2013-0043oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD), a common chronic respiratory disease that occurs after premature birth, is believed to be secondary to oxidative damage from hyperoxia and inflammation, which leads to impaired alveolar formation and chronic lung dysfunction. We hypothesized that extracellular superoxide dismutase (SOD)3, an antioxidant uniquely targeted to the extracellular matrix (ECM) and alveolar fluid, might have a different response (down-regulation) to hyperoxic injury and recovery in room air (RA), thereby contributing to the persistent airspace injury and inflammation. We used a murine BPD model using postnatal hyperoxia (O2) (4 or 5 d) followed by short-term recovery (14 d) in RA, which mimics the durable effects after injury during alveolar development. This was associated with significantly increased mRNA expression for antioxidant genes mediated by nuclear factor erythroid 2-related factor (Nrf2) in the O2 (n = 4) versus RA group (n = 5). SOD3, an Nrf2-independent antioxidant, was significantly reduced in the O2-exposed mice compared with RA. Immunohistochemistry revealed decreased and disrupted SOD3 deposition in the alveolar ECM of O2-exposed mice. Furthermore, this distinct hyperoxic antioxidant and injury profile was reproducible in murine lung epithelial 12 cells exposed to O2. Overexpression of SOD3 rescued the injury measures in the O2-exposed cells. We establish that reduced SOD3 expression correlates with alveolar injury measures in the recovered neonatal hyperoxic lung, and SOD3 overexpression attenuates hyperoxic injury in an alveolar epithelial cell line. Such findings suggest a candidate mechanism for the pathogenesis of BPD that may lead to targeted interventions.
Collapse
|
12
|
Abstract
Acute lung injury (ALI) and its more severe form of clinical manifestation, the acute respiratory distress syndrome is associated with significant dysfunction in air exchange due to inflammation of the lung parenchyma. Several factors contribute to the inflammatory process, including hypoxia (inadequate oxygen), hyperoxia (higher than normal partial pressure of oxygen), inflammatory mediators (such as cytokines), infections (viral and bacterial), and environmental conditions (such as cigarette smoke or noxious gases). However, studies over the past several decades suggest that oxidants formed in the various cells of the lung including endothelial, alveolar, and epithelial cells as well as lung macrophages and neutrophils in response to the factors mentioned above mediate the pathogenesis of ALI. Oxidants modify cellular proteins, lipids, carbohydrates, and DNA to cause their aberrant function. For example, oxidation of lipids changes membrane permeability. Interestingly, recent studies also suggest that spatially and temporally regulated production of oxidants plays an important role antimicrobial defense and immunomodulatory function (such as transcription factor activation). To counteract the oxidants an arsenal of antioxidants exists in the lung to maintain the redox status, but when overwhelmed tissue injury and exacerbation of inflammation occurs. We present below the current understanding of the pathogenesis of oxidant-mediated ALI.
Collapse
Affiliation(s)
- J Vidya Sarma
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | |
Collapse
|
13
|
Genetic evidence of an evolutionarily conserved role for Nrf2 in the protection against oxidative stress. Mol Cell Biol 2012; 32:4455-61. [PMID: 22949501 DOI: 10.1128/mcb.00481-12] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transcription factor Nrf2 is considered a master regulator of antioxidant defense in mammals. However, it is unclear whether this concept is applicable to nonmammalian vertebrates, because no animal model other than Nrf2 knockout mice has been generated to examine the effects of Nrf2 deficiency. Here, we characterized a recessive loss-of-function mutant of Nrf2 (nrf2(fh318)) in a lower vertebrate, the zebrafish (Danio rerio). In keeping with the findings in the mouse model, nrf2(fh318) mutants exhibited reduced induction of the Nrf2 target genes in response to oxidative stress and electrophiles but were viable and fertile, and their embryos developed normally. The nrf2(fh318) larvae displayed enhanced sensitivity to oxidative stress and electrophiles, especially peroxides, and pretreatment with an Nrf2-activating compound, sulforaphane, decreased peroxide-induced lethality in the wild type but not nrf2(fh318) mutants, indicating that resistance to oxidative stress is highly dependent on Nrf2 functions. These results reveal an evolutionarily conserved role of vertebrate Nrf2 in protection against oxidative stress. Interestingly, there were no significant differences between wild-type and nrf2(fh318) larvae with regard to their sensitivity to superoxide and singlet oxygen generators, suggesting that the importance of Nrf2 in oxidative stress protection varies based on the type of reactive oxygen species (ROS).
Collapse
|
14
|
Abstract
INTRODUCTION Oxygen exposure plays an important role in the pathogenesis of bronchopulmonary dysplasia (BPD). The phosphodiesterase inhibitor pentoxifylline (PTX) has anti-inflammatory and antifibrotic effects in multiple organs. It was hypothesized that PTX would have a protective effect on hyperoxia-induced lung injury (HILI). METHODS Newborn Sprague-Dawley rats were exposed to >95% oxygen (O(2)) and injected subcutaneously with normal saline (NS) or PTX (75 mg/kg) twice a day for 9 d. NS-injected, room air-exposed pups were controls. At days 4 and 9, lung tissue was collected to assess edema, antioxidant enzyme (AOE) activities, and vascular endothelial growth factor (VEGF) expression. At day 9, pulmonary macrophage infiltration, vascularization, and alveolarization were also examined. RESULTS At day 9, treatment with PTX significantly increased survival from 54% to 88% during hyperoxia. Treatment with PTX significantly decreased lung edema and macrophage infiltration. PTX treatment increased lung AOE activities including those of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX). Furthermore, PTX treatment also increased the gene expression of VEGF189 and VEGF165, increased VEGF protein expression, and improved pulmonary vascularization. DISCUSSION These data indicate that the reduced lung edema and inflammation, increased AOE activities, and improved vascularization may be responsible for the improved survival with PTX during hyperoxia. PTX may be a potential therapy in reducing some of the features of BPD in preterm newborns.
Collapse
|
15
|
Budinger GRS, Mutlu GM, Urich D, Soberanes S, Buccellato LJ, Hawkins K, Chiarella SE, Radigan KA, Eisenbart J, Agrawal H, Berkelhamer S, Hekimi S, Zhang J, Perlman H, Schumacker PT, Jain M, Chandel NS. Epithelial cell death is an important contributor to oxidant-mediated acute lung injury. Am J Respir Crit Care Med 2011; 183:1043-54. [PMID: 20959557 PMCID: PMC3086743 DOI: 10.1164/rccm.201002-0181oc] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 10/15/2010] [Indexed: 01/11/2023] Open
Abstract
RATIONALE Acute lung injury and the acute respiratory distress syndrome are characterized by increased lung oxidant stress and apoptotic cell death. The contribution of epithelial cell apoptosis to the development of lung injury is unknown. OBJECTIVES To determine whether oxidant-mediated activation of the intrinsic or extrinsic apoptotic pathway contributes to the development of acute lung injury. METHODS Exposure of tissue-specific or global knockout mice or cells lacking critical components of the apoptotic pathway to hyperoxia, a well-established mouse model of oxidant-induced lung injury, for measurement of cell death, lung injury, and survival. MEASUREMENTS AND MAIN RESULTS We found that the overexpression of SOD2 prevents hyperoxia-induced BAX activation and cell death in primary alveolar epithelial cells and prolongs the survival of mice exposed to hyperoxia. The conditional loss of BAX and BAK in the lung epithelium prevented hyperoxia-induced cell death in alveolar epithelial cells, ameliorated hyperoxia-induced lung injury, and prolonged survival in mice. By contrast, Cyclophilin D-deficient mice were not protected from hyperoxia, indicating that opening of the mitochondrial permeability transition pore is dispensable for hyperoxia-induced lung injury. Mice globally deficient in the BH3-only proteins BIM, BID, PUMA, or NOXA, which are proximal upstream regulators of BAX and BAK, were not protected against hyperoxia-induced lung injury suggesting redundancy of these proteins in the activation of BAX or BAK. CONCLUSIONS Mitochondrial oxidant generation initiates BAX- or BAK-dependent alveolar epithelial cell death, which contributes to hyperoxia-induced lung injury.
Collapse
Affiliation(s)
- G. R. Scott Budinger
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Gökhan M. Mutlu
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Daniela Urich
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Saul Soberanes
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Leonard J. Buccellato
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Keenan Hawkins
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sergio E. Chiarella
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Kathryn A. Radigan
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - James Eisenbart
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Hemant Agrawal
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sara Berkelhamer
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Siegfried Hekimi
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jianke Zhang
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Harris Perlman
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Paul T. Schumacker
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Manu Jain
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Navdeep S. Chandel
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
16
|
Zhao HW, Zhou D, Haddad GG. Antimicrobial peptides increase tolerance to oxidant stress in Drosophila melanogaster. J Biol Chem 2010; 286:6211-8. [PMID: 21148307 DOI: 10.1074/jbc.m110.181206] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It is well appreciated that reactive oxygen species (ROS) are deleterious to mammals, including humans, especially when generated in abnormally large quantities from cellular metabolism. Whereas the mechanisms leading to the production of ROS are rather well delineated, the mechanisms underlying tissue susceptibility or tolerance to oxidant stress remain elusive. Through an experimental selection over many generations, we have previously generated Drosophila melanogaster flies that tolerate tremendous oxidant stress and have shown that the family of antimicrobial peptides (AMPs) is over-represented in these tolerant flies. Furthermore, we have also demonstrated that overexpression of even one AMP at a time (e.g. Diptericin) allows wild-type flies to survive much better in hyperoxia. In this study, we used a number of experimental approaches to investigate the potential mechanisms underlying hyperoxia tolerance in flies with AMP overexpression. We demonstrate that flies with Diptericin overexpression resist oxidative stress by increasing antioxidant enzyme activities and preventing an increase in ROS levels after hyperoxia. Depleting the GSH pool using buthionine sulfoximine limits fly survival, thus confirming that enhanced survival observed in these flies is related to improved redox homeostasis. We conclude that 1) AMPs play an important role in tolerance to oxidant stress, 2) overexpression of Diptericin changes the cellular redox balance between oxidant and antioxidant, and 3) this change in redox balance plays an important role in survival in hyperoxia.
Collapse
Affiliation(s)
- Huiwen W Zhao
- Division of Respiratory Medicine, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
17
|
Zanchetta S, Resende LADL, Bentlin MR, Rugulo LM, Trindade CEP. Conductive hearing loss in children with bronchopulmonary dysplasia: a longitudinal follow-up study in children aged between 6 and 24 months. Early Hum Dev 2010; 86:385-9. [PMID: 20554131 DOI: 10.1016/j.earlhumdev.2010.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Revised: 05/04/2010] [Accepted: 05/05/2010] [Indexed: 10/19/2022]
Abstract
AIMS To determine the occurrence of isolated and recurrent episodes of conductive hearing loss (CHL) during the first two years of life in very low birth weight (VLBW) infants with and without bronchopulmonary dysplasia (BPD). STUDY DESIGN, SUBJECTS AND OUTCOME MEASURES: In a longitudinal clinical study, 187 children were evaluated at 6, 9, 12, 15 18 and 24 months of age by visual reinforcement audiometry, tympanometry and auditory brain response system. RESULTS Of the children with BPD, 54.5% presented with episodes of CHL, as opposed to 34.7% of the children without BPD. This difference was found to be statistically significant. The recurrent or persistent episodes were more frequent among children with BPD (25.7%) than among those without BPD (8.3%). The independent variables that contributed to this finding were small for gestational age and a 5 min Apgar score. CONCLUSIONS Recurrent CHL episodes are more frequent among VLBW infants with BPD than among VLBW infants without BPD.
Collapse
Affiliation(s)
- Sthella Zanchetta
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck Surgery, School of Medicine of Ribeirão Preto-University of São Paulo, Ribeirão Preto, SP, Brazil.
| | | | | | | | | |
Collapse
|
18
|
Abstract
Reactive oxygen species (ROS) serve as cell signaling molecules for normal biologic processes. However, the generation of ROS can also provoke damage to multiple cellular organelles and processes, which can ultimately disrupt normal physiology. An imbalance between the production of ROS and the antioxidant defenses that protect cells has been implicated in the pathogenesis of a variety of diseases, such as cancer, asthma, pulmonary hypertension, and retinopathy. The nature of the injury will ultimately depend on specific molecular interactions, cellular locations, and timing of the insult. This review will outline the origins of endogenous and exogenously generated ROS. The molecular, cellular, pathologic, and physiologic targets will then be discussed with a particular emphasis on aspects relevant to child development. Finally, antioxidant defenses that scavenge ROS and mitigate associated toxicities will be presented, with a discussion of potential therapeutic approaches for the prevention and/or treatment of human diseases using enzymatic and nonenzymatic antioxidants.
Collapse
Affiliation(s)
- Richard L Auten
- Department of Pediatrics, Duke Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
19
|
Joseph A, Li Y, Koo HC, Davis JM, Pollack S, Kazzaz JA. Superoxide dismutase attenuates hyperoxia-induced interleukin-8 induction via AP-1. Free Radic Biol Med 2008; 45:1143-9. [PMID: 18692129 DOI: 10.1016/j.freeradbiomed.2008.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Revised: 05/30/2008] [Accepted: 07/10/2008] [Indexed: 10/21/2022]
Abstract
Exposure of lung epithelial cells to hyperoxia results in the generation of excess reactive oxygen species (ROS), cell damage, and production of proinflammatory cytokines (interleukin-8; IL-8). Although activation of the NF-kappaB and c-Jun N-terminal kinase (JNK)/activator protein (AP)-1 transcription pathways occurs in hyperoxia, it is unclear whether activation of the AP-1 pathway has a direct impact on IL-8 production and whether overexpression of superoxide dismutase (SOD) can mitigate these proinflammatory processes. A549 cells were exposed to 95% O(2), and ROS production, AP-1 activation, and IL-8 levels were determined. Experimental groups included cells transduced with a recombinant adenovirus encoding CuZnSOD or MnSOD (two- to threefold increased activity) or transfected with a JNK1 small interfering RNA (RNAi). Hyperoxia resulted in significant increases in ROS generation, AP-1 activation, and IL-8 production, which were significantly attenuated by overexpression of either MnSOD or CuZnSOD. JNK1 RNAi also moderated IL-8 induction. The data indicate that activation of JNK1/AP-1 and subsequent IL-8 induction in hyperoxia are mediated by intracellular ROS, with SOD having significant protective effects.
Collapse
Affiliation(s)
- Ansamma Joseph
- CardioPulmonary Research Institute, Department of Medicine, Winthrop University Hospital, State University of New York Stony Brook School of Medicine, Mineola, NY 11501, USA
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
Interstitial lung disease encompasses a large group of chronic lung disorders associated with excessive tissue remodeling, scarring, and fibrosis. The evidence of a redox imbalance in lung fibrosis is substantial, and the rationale for testing antioxidants as potential new therapeutics for lung fibrosis is appealing. Current animal models of lung fibrosis have clear involvement of ROS in their pathogenesis. New classes of antioxidant agents divided into catalytic antioxidant mimetics and antioxidant scavengers are being developed. The catalytic antioxidant class is based on endogenous antioxidant enzymes and includes the manganese-containing macrocyclics, porphyrins, salens, and the non-metal-containing nitroxides. The antioxidant scavenging class is based on endogenous antioxidant molecules and includes the vitamin E analogues, thiols, lazaroids, and polyphenolic agents. Numerous studies have shown oxidative stress to be associated with many interstitial lung diseases and that these agents are effective in attenuating fibroproliferative responses in the lung of animals and humans.
Collapse
Affiliation(s)
- Brian J Day
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Medical and Research Center, Denver, Colorado 80206, USA.
| |
Collapse
|
21
|
15-Deoxy-Delta12,14-prostaglandin J2 induces chemokine expression, oxidative stress and microfilament reorganization in bovine mammary epithelial cells. J DAIRY RES 2008; 75:55-63. [PMID: 18226310 DOI: 10.1017/s0022029907003056] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The roles of the pro-adipogenic ligands of the transcription factor Peroxisome Proliferator Activated Receptor gamma (PPARG) in regulating innate immune responses in bovine mammary epithelial cells (bMEC) were investigated using quantitative real-time PCR. The analyses revealed that 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2) enhanced the expression of Interleukin 8 (IL-8) and Chemokine (C-X-C motif) ligand 6 (CXCL6) in these cells in a dose-dependent manner. 15d-PGJ2 also induced the expression of transcripts encoding proteins involved in oxidative stress, including Ferritin heavy chain and Superoxide dismutase 1, as well as substantial microfilament reorganization. In contrast, synthetic PPARG agonists displayed a different and much smaller range of effects on the cells, causing down-regulation of Interleukin 1-beta, Interleukin 6 and IL-8 and increased expression of Chemokine (C-C motif) ligand 2 (CCL2) and Tumour necrosis factor alpha (TNFalpha). In an independent analysis, the cells were pre-incubated with PPARG agonists followed by lipopolysaccharide stimulation. This study revealed that troglitazone increased the responsiveness of the cells to lipopolysaccharide resulting in up-regulation of Interleukin 1-beta, TNFalpha, IL-8, CCL2 and CXCL6 while 15d-PGJ2 caused down-regulation of TNFalpha, CCL2 and CXCL6. These findings are relevant to understanding the anti-inflammatory potential of the PPARG ligands and underline different mechanisms of action of 15d-PGJ2 and troglitazone in bMEC. Furthermore, the present results demonstrate that the generation of pro-inflammatory mediators can be modulated by currently available therapeutic agents and may therefore be of value in the treatment of mastitis in ruminants.
Collapse
|
22
|
Arita Y, Kazzaz JA, Joseph A, Koo HC, Li Y, Davis JM. Antioxidants improve antibacterial function in hyperoxia-exposed macrophages. Free Radic Biol Med 2007; 42:1517-23. [PMID: 17448898 PMCID: PMC1963462 DOI: 10.1016/j.freeradbiomed.2007.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Revised: 01/02/2007] [Accepted: 02/09/2007] [Indexed: 01/24/2023]
Abstract
Hyperoxia and pulmonary infections are well known to increase the risk of acute and chronic lung injury in newborn infants, but it is not clear whether hyperoxia directly increases the risk of pneumonia. The purpose of this study was to examine: (1) the effects of hyperoxia and antioxidant enzymes on inflammation and bacterial clearance in mononuclear cells and (2) developmental differences between adult and neonatal mononuclear cells in response to hyperoxia. Mouse macrophages were exposed to either room air or 95% O2 for 24 h and then incubated with Pseudomonas aeruginosa. After 1 h, bacterial adherence, phagocytosis, and macrophage inflammatory protein (MIP)-1alpha production were analyzed. Bacterial adherence increased 5.8-fold (p < 0.0001), phagocytosis decreased 60% (p < 0.05), and MIP-1alpha production increased 49% (p < 0.05) in response to hyperoxia. Overexpression of MnSOD or catalase significantly decreased bacterial adherence by 30.5%, but only MnSOD significantly improved bacterial phagocytosis and attenuated MIP-1alpha production. When monocytes from newborns and adults were exposed to hyperoxia, phagocytosis was impaired in both groups. However, adult monocytes were significantly more impaired than neonatal monocytes. Data indicate that hyperoxia significantly increases bacterial adherence while impairing function of mononuclear cells, with adult cells being more impaired than neonatal cells. MnSOD reduces bacterial adherence and inflammation and improves bacterial phagocytosis in mononuclear cells in response to hyperoxia, which should minimize the development of oxidant-induced lung injury as well as reducing nosocomial infections.
Collapse
Affiliation(s)
- Yuko Arita
- CardioPulmonary Research Institute, and the Departments of Pediatrics, Medicine, and Thoracic-Cardiovascular Surgery, Winthrop University Hospital, SUNY Stony Brook School of Medicine, Mineola NY
| | - Jeffrey A. Kazzaz
- CardioPulmonary Research Institute, and the Departments of Pediatrics, Medicine, and Thoracic-Cardiovascular Surgery, Winthrop University Hospital, SUNY Stony Brook School of Medicine, Mineola NY
| | - Ansamma Joseph
- CardioPulmonary Research Institute, and the Departments of Pediatrics, Medicine, and Thoracic-Cardiovascular Surgery, Winthrop University Hospital, SUNY Stony Brook School of Medicine, Mineola NY
| | - Hshi-chi Koo
- Department of Pediatrics, Tufts University School of Medicine, Boston, MA
| | - Yuchi Li
- CardioPulmonary Research Institute, and the Departments of Pediatrics, Medicine, and Thoracic-Cardiovascular Surgery, Winthrop University Hospital, SUNY Stony Brook School of Medicine, Mineola NY
| | - Jonathan M. Davis
- Department of Pediatrics, Tufts University School of Medicine, Boston, MA
| |
Collapse
|
23
|
Chandel NS, Budinger GRS. The cellular basis for diverse responses to oxygen. Free Radic Biol Med 2007; 42:165-74. [PMID: 17189822 DOI: 10.1016/j.freeradbiomed.2006.10.048] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Revised: 09/27/2006] [Accepted: 10/24/2006] [Indexed: 11/22/2022]
Abstract
Mammalian cells have divergent responses to varying oxygen levels. Cells exposed to low oxygen levels (hypoxia) activate the transcription factor hypoxia-inducible factor-1 (HIF-1) as an adaptive response. Cells exposed to hypoxia do not undergo senescence or cell death and do not diminish ATP levels. By contrast, cells exposed to high oxygen levels (hyperoxia) undergo senescence and cell death and decrease their ATP levels, yet do not activate HIF-1. Despite these divergent responses with respect to senescence, cell death, metabolism, and gene expression, the signaling events in both systems are mediated by the generation of mitochondrial-derived reactive oxygen species (ROS). This perspective reviews the role of signaling through mitochondrial ROS in hypoxic and hyperoxic environments.
Collapse
Affiliation(s)
- Navdeep S Chandel
- Department of Medicine and Department of Cell & Molecular Biology, Northwestern University, McGaw Pavilion M-334, 240 East Huron Avenue, Chicago, IL 60611, USA
| | | |
Collapse
|
24
|
RANDHIR REENA, VATTEM DHIRAJA, SHETTY KALIDAS. ANTIOXIDANT ENZYME RESPONSE STUDIES IN H2O2-STRESSED PORCINE MUSCLE TISSUE FOLLOWING TREATMENT WITH FAVA BEAN SPROUT EXTRACT AND L-DOPA. J Food Biochem 2006. [DOI: 10.1111/j.1745-4514.2006.00090.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
25
|
Arita Y, Harkness SH, Kazzaz JA, Koo HC, Joseph A, Melendez JA, Davis JM, Chander A, Li Y. Mitochondrial localization of catalase provides optimal protection from H2O2-induced cell death in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 2006; 290:L978-86. [PMID: 16387755 DOI: 10.1152/ajplung.00296.2005] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Reactive oxygen species (ROS) can cause cell injury and death via mitochondrial-dependent pathways, and supplementation with antioxidants has been shown to ameliorate these processes. The c-Jun NH2-terminal kinase (JNK) pathway has been shown to play a critical role in ROS-induced cell death. To determine if targeting catalase (CAT) to the mitochondria provides better protection than cytosolic expression against H2O2-induced injury, the following two approaches were taken: 1) adenoviral-mediated transduction was performed using cytosolic (CCAT) or mitochondrial (MCAT) CAT cDNAs and 2) stable cell lines were generated overexpressing CAT in mitochondria ( n = 3). Cells were exposed to 250 μM H2O2, and cell survival, mitochondrial function, cytochrome c release, and JNK activity were analyzed. Although all viral transduced cells had a transient twofold increase in CAT activity, MCAT cells had significantly higher survival rates, the best mitochondrial function, and lowest JNK activity compared with CCAT and LacZ controls. The improved protection with MCAT was observed in primary type II lung epithelial cells and in transformed lung epithelial cells. In the three stable cell lines, cell survival directly correlated with extent of mitochondrial localization ( r = 0.60572, P < 0.05) and not overall CAT activity ( r = −0.45501, P < 0.05). Data indicate that targeting of antioxidants directly to the mitochondria is more effective in protecting lung epithelial cells against ROS-induced injury. This has important implications in antioxidant supplementation trials to prevent ROS-induced lung injury in critically ill patients.
Collapse
Affiliation(s)
- Yuko Arita
- Department of Pediatrics, CardioPulmonary Research Institute, Winthrop University Hospital, SUNY Stony Brook School of Medicine, Suite 505, 222 Station Plaza North, Mineola, NY 11501, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
McAdams RM, Mustafa SB, Shenberger JS, Dixon PS, Henson BM, DiGeronimo RJ. Cyclic stretch attenuates effects of hyperoxia on cell proliferation and viability in human alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 2006; 291:L166-74. [PMID: 16461433 PMCID: PMC2683386 DOI: 10.1152/ajplung.00160.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The treatment of severe lung disease often requires the use of high concentrations of oxygen coupled with the need for assisted ventilation, potentially exposing the pulmonary epithelium to both reactive oxygen species and nonphysiological cyclic stretch. Whereas prolonged hyperoxia is known to cause increased cell injury, cyclic stretch may result in either cell proliferation or injury depending on the pattern and degree of exposure to mechanical deformation. How hyperoxia and cyclic stretch interact to affect the pulmonary epithelium in vitro has not been previously investigated. This study was performed using human alveolar epithelial A549 cells to explore the combined effects of cyclic stretch and hyperoxia on cell proliferation and viability. Under room air conditions, cyclic stretch did not alter cell viability at any time point and increased cell number after 48 h compared with unstretched controls. After exposure to prolonged hyperoxia, cell number and [(3)H]thymidine incorporation markedly decreased, whereas evidence of oxidative stress and nonapoptotic cell death increased. The combination of cyclic stretch with hyperoxia significantly mitigated the negative effects of prolonged hyperoxia alone on measures of cell proliferation and viability. In addition, cyclic stretch resulted in decreased levels of oxidative stress over time in hyperoxia-exposed cells. Our results suggest that cyclic stretch, as applied in this study, can minimize the detrimental effects of hyperoxia on alveolar epithelial A549 cells.
Collapse
Affiliation(s)
- Ryan M McAdams
- Department of Pediatrics, Wilford Hall United States Air Force Medical Center, Lackland Air Force Base, San Antonio, TX 78236, USA
| | | | | | | | | | | |
Collapse
|
27
|
Kinsella JP, Parker TA, Davis JM, Abman SH. Superoxide dismutase improves gas exchange and pulmonary hemodynamics in premature lambs. Am J Respir Crit Care Med 2005; 172:745-9. [PMID: 15947289 PMCID: PMC2718553 DOI: 10.1164/rccm.200501-146oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Oxidant stress may increase the severity of respiratory distress syndrome (RDS) after premature birth by altering vasoreactivity and increasing lung edema, but the acute effects of superoxide dismutase (SOD) treatment on gas exchange, lung compliance (CL), and pulmonary vascular resistance in premature animals with RDS are unknown. OBJECTIVE We studied the effects of intratracheal recombinant human SOD treatment (rhSOD) on gas exchange, CL, and pulmonary hemodynamics in 46 premature lambs with RDS. METHODS After C-section delivery, lambs were randomly assigned to treatment with SOD (2.5-10 mg/kg) with or without inhaled nitric oxide (iNO, 5 ppm), and mechanically ventilated for 4 hours. At the end of the study, pressure-volume curves and wet-dry lung weights were measured to assess CL and edema, respectively. MAIN RESULTS Despite an initial rise in Pa(O(2)), Pa(O(2)) in control animals progressively declined over the 4-hour treatment period (Pa(O(2)) = 25.0 +/- 7.5 mm Hg at 4 hours). In comparison with control animals, early treatment with SOD at 5 and 10 mg/kg improved Pa(O(2)) at 4 hours (167 +/- 44 and 269 +/- 33 mm Hg, respectively; p < 0.05 vs. control), but did not decrease lung edema or improve CL. In contrast, late treatment with SOD did not improve Pa(O(2)). Treatment with iNO increased Pa(O(2)) (196 +/- 22 vs. 25 +/- 8 mm Hg, control animals; p < 0.01), but the response to iNO was not augmented by combined therapy (SOD + iNO). After 4 hours of ventilation with FI(O(2)) = 1.00, rhSOD treatment lowered pulmonary vascular resistance compared with control animals. CONCLUSIONS Early intratracheal rhSOD treatment improves oxygenation in premature lambs with RDS and prevents the development of pulmonary hypertension.
Collapse
Affiliation(s)
- John P Kinsella
- Department of Pediatrics, Pediatric Heart-Lung Center, Section of Neonatology, University of Colorado School of Medicine, Denver, Colorado, USA.
| | | | | | | |
Collapse
|
28
|
Koo HC, Davis JM, Li Y, Hatzis D, Opsimos H, Pollack S, Strayer MS, Ballard PL, Kazzaz JA. Effects of transgene expression of superoxide dismutase and glutathione peroxidase on pulmonary epithelial cell growth in hyperoxia. Am J Physiol Lung Cell Mol Physiol 2005; 288:L718-26. [PMID: 15579623 DOI: 10.1152/ajplung.00456.2003] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Prolonged exposure to supraphysiological oxygen concentrations results in the generation of reactive oxygen species, which can cause significant lung injury in critically ill patients. Supplementation with human recombinant antioxidant enzymes (AOE) may mitigate hyperoxic lung injury, but it is unclear which combination and concentration will optimally protect pulmonary epithelial cells. First, stable cell lines were generated in alveolar epithelial cells (MLE12) overexpressing one or more of the following AOE: Mn superoxide dismutase (MnSOD), CuZnSOD, or glutathione peroxidase 1. Next, A549 cells were transduced with 50–300 particles/cell of recombinant adenovirus containing either LacZ or each of the three AOE (alone or in combination). Cells were then exposed to 95% O2 for up to 3 days, with cell number and viability determined daily. Overexpression of either MnSOD (primarily mitochondrial) or CuZnSOD (primarily cytosolic) reversed the growth inhibitory effects of hyperoxia within the first 48 h of exposure, resulting in a significant increase in viable cells ( P < 0.05), with 1.5- to 3-fold increases in activity providing optimal protection. Protection from mitochondrial oxidation was confirmed by assessing aconitase activity, which was significantly improved in cells overexpressing MnSOD ( P < 0.05). Data indicate that optimal protection from hyperoxic injury occurs in cells coexpressing MnSOD and glutathione peroxidase 1, with prevention of mitochondrial oxidation being a critical factor. This has important implications for clinical trials in preterm infants receiving SOD supplementation to prevent acute and chronic lung injury.
Collapse
Affiliation(s)
- Hshi-Chi Koo
- CardioPulmonary Research Institute, Winthrop University Hospital, 222 Station Plaza North, Suite 604, Mineola, NY 11501, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Fessel JP, Jackson Roberts L. Isofurans: novel products of lipid peroxidation that define the occurrence of oxidant injury in settings of elevated oxygen tension. Antioxid Redox Signal 2005; 7:202-9. [PMID: 15650408 DOI: 10.1089/ars.2005.7.202] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We recently reported the discovery of isofurans, novel products of free radical-induced peroxidation of arachidonic acid that exhibit favored formation with increasing oxygen concentrations. In this review, the biochemistry of isofuran formation is compared with that of isoprostanes, with an emphasis on the mechanistic basis for the favored formation of isofurans at elevated oxygen tensions. In addition, the formation of isofurans in various disease states in vivo is also discussed. Parkinson's disease is presented as a disease model involving mitochondrial dysfunction, a situation in which quantification of isofurans can provide a uniquely sensitive indicator of oxidant injury. Measurement of isofurans has also provided unexpected insights into the earliest events in hyperoxic lung injury, an important clinical problem in which measurement of isofurans might prove to be uniquely valuable in the evaluation of approaches to limit this injury. These two settings are then used as models to suggest a variety of other pathological settings in which measurement of isofurans together with isoprostanes could provide a complete and robust picture of oxidative stress status in ongoing and future investigations.
Collapse
Affiliation(s)
- Joshua P Fessel
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232-6602, USA
| | | |
Collapse
|
30
|
Viemann D, Strey A, Janning A, Jurk K, Klimmek K, Vogl T, Hirono K, Ichida F, Foell D, Kehrel B, Gerke V, Sorg C, Roth J. Myeloid-related proteins 8 and 14 induce a specific inflammatory response in human microvascular endothelial cells. Blood 2004; 105:2955-62. [PMID: 15598812 DOI: 10.1182/blood-2004-07-2520] [Citation(s) in RCA: 241] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myeloid-related protein 8 (MRP8) and MRP14, S100 proteins secreted by activated phagocytes, bind specifically to endothelial cells. The endothelial response to MRP8/MRP14, however, is unknown. Using oligonucleotide microarray analysis, we show for the first time that MRP8/MRP14 induce a thrombogenic, inflammatory response in human microvascular endothelial cells by increasing the transcription of proinflammatory chemokines and adhesion molecules and by decreasing the expression of cell junction proteins and molecules involved in monolayer integrity. All changes on the gene expression level could be confirmed using biochemical and functional assays. We demonstrated that the expression of MRP8/MRP14 closely correlated with the inflammatory activity in systemic vasculitis, confirming the important role of these proteins for distinct inflammatory reactions in endothelia. MRP8/MRP14 may represent novel targets for anti-inflammatory strategies.
Collapse
Affiliation(s)
- Dorothee Viemann
- Institute of Experimental Dermatology, Integrated Functional Genomics, and the Interdisciplinary Clinical Research Center, University Hospital Muenster, University of Muenster, Röntgenstrasse 21, 48149 Muenster, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Arita Y, Joseph A, Koo HC, Li Y, Palaia TA, Davis JM, Kazzaz JA. Superoxide dismutase moderates basal and induced bacterial adherence and interleukin-8 expression in airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2004; 287:L1199-206. [PMID: 15286004 DOI: 10.1152/ajplung.00457.2003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Bacterial infection of the tracheobronchial tree is a frequent, serious complication in patients receiving treatment with oxygen and mechanical ventilation, resulting in increased morbidity and mortality. Using human airway epithelial cell culture models, we examined the effect of hyperoxia on bacterial adherence and the expression of interleukin-8 (IL-8), an important mediator involved in the inflammatory process. A 24-h exposure to 95% O2increased Pseudomonas aeruginosa (PA) adherence 57% in A549 cells ( P < 0.01) and 115% in 16HBE cells ( P < 0.01) but had little effect on Staphylococcus aureus (SA) adherence. Exposure to hyperoxia, followed by a 1-h incubation with SA, further enhanced PA adherence ( P < 0.01), suggesting that hyperoxia and SA colonization may enhance the susceptibility of lung epithelial cells to gram-negative infections. IL-8 expression was also increased in cells exposed to both hyperoxia and PA. Stable or transient overexpression of manganese superoxide dismutase reduced both basal and stimulated levels of PA adherence and IL-8 levels in response to exposure to either hyperoxia or PA. These data indicate that hyperoxia increases susceptibility to infection and that the pathways are mediated by reactive oxygen species. Therapeutic intervention strategies designed to prevent accumulation of intracellular reactive oxygen species may reduce opportunistic pulmonary infections.
Collapse
Affiliation(s)
- Yuko Arita
- CardioPulmonary Research Institute, Department of Medicine, Division of Pulmonary and Critical Care Medicine, Winthrop-University Hospital, SUNY Stony Brook School of Medicine, Mineola, NY 11501, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Franek WR, Morrow DMP, Zhu H, Vancurova I, Miskolci V, Darley-Usmar K, Simms HH, Mantell LL. NF-kappaB protects lung epithelium against hyperoxia-induced nonapoptotic cell death-oncosis. Free Radic Biol Med 2004; 37:1670-9. [PMID: 15477018 DOI: 10.1016/j.freeradbiomed.2004.08.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2004] [Accepted: 08/12/2004] [Indexed: 01/26/2023]
Abstract
Prolonged exposure to hyperoxia induces pulmonary epithelial cell death and acute lung injury. Although both apoptotic and nonapoptotic morphologies are observed in hyperoxic animal lungs, nonapoptotic cell death had only been recorded in transformed lung epithelium cultured in hyperoxia. To test whether the nonapoptotic characteristics in hyperoxic animal lungs are direct effects of hyperoxia, the mode of cell death was determined both morphologically and biochemically in human primary lung epithelium exposed to 95% O(2). In contrast to characteristics observed in apoptotic cells, hyperoxia induced swelling of nuclei and an increase in cell size, with no evidence for any augmentation in the levels of either caspase-3 activity or annexin V incorporation. These data suggest that hyperoxia can directly induce nonapoptotic cell death in primary lung epithelium. Although hyperoxia-induced nonapoptotic cell death was associated with NF-kappaB activation, it is unknown whether NF-kappaB activation plays any causal role in nonapoptotic cell death. This study shows that inhibition of NF-kappaB activation can accelerate hyperoxia-induced epithelial cell death in both primary and transformed lung epithelium. Corresponding to the reduced cell survival in hyperoxia, the levels of MnSOD were also low in NF-kappaB-deficient cells. These results demonstrate that NF-kappaB protects lung epithelial cells from hyperoxia-induced nonapoptotic cell death.
Collapse
Affiliation(s)
- William R Franek
- Department of Surgery, North Shore University Hospital, New York University School of Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Oxidative Stress/Antioxidant Status in HeaIth and Disease. OXYGEN/NITROGEN RADICALS 2004. [DOI: 10.1201/b14147-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
34
|
Haddad JJ. Redox and oxidant-mediated regulation of apoptosis signaling pathways: immuno-pharmaco-redox conception of oxidative siege versus cell death commitment. Int Immunopharmacol 2004; 4:475-93. [PMID: 15099526 DOI: 10.1016/j.intimp.2004.02.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Revised: 11/10/2003] [Accepted: 02/04/2004] [Indexed: 11/21/2022]
Abstract
The mechanisms controlling apoptosis remain largely obscure. Because apoptosis is an integral part of the developmental program and is frequently the end-result of a temporal course of cellular events, it is referred to as programmed cell death. While there is considerable variation in the signals and requisite cellular metabolic events necessary to induce apoptosis in diverse cell types, the morphological features associated with apoptosis are highly conserved. Free radicals, particularly reactive oxygen species (ROS), have been proposed as common mediators for apoptosis. Many agents that induce apoptosis are either oxidants or stimulators of cellular oxidative metabolism. Conversely, many inhibitors of apoptosis have antioxidant activities or enhance cellular antioxidant defenses. Mammalian cells, therefore, exist in a state of oxidative siege in which survival requires an optimum balance of oxidants and antioxidants. The respiratory tract is subjected to a variety of environmental stresses, including oxidizing agents, particulates and airborne microorganisms that, together, may injure structural and functional lung components and thereby jeopardize the primary lung function of gas exchange. To cope with this challenge, the lung has developed elaborate defense mechanisms that include inflammatory-immune pathways as well as efficient antioxidant defense systems. In the absence of adequate antioxidant defenses, the damage produced is detected by the cell leading to the activation of genes responsible for the regulation of apoptosis, conceivably through stress-responsive transcription factors. Oxidative stress, in addition, may cause a shift in cellular redox state, which thereby modifies the nature of the stimulatory signal and which results in cell death as opposed to proliferation. ROS/redox modifications, therefore, may disrupt signal transduction pathways, can be perceived as abnormal and, under some conditions, may trigger apoptosis.
Collapse
Affiliation(s)
- John J Haddad
- Severinghaus-Radiometer Research Laboratories, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA.
| |
Collapse
|
35
|
Kinnula VL, Crapo JD. Superoxide dismutases in malignant cells and human tumors. Free Radic Biol Med 2004; 36:718-44. [PMID: 14990352 DOI: 10.1016/j.freeradbiomed.2003.12.010] [Citation(s) in RCA: 239] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2003] [Revised: 12/01/2003] [Accepted: 12/12/2003] [Indexed: 02/06/2023]
Abstract
Reactive oxygen metabolites have multifactorial effects on the regulation of cell growth and the capacity of malignant cells to invade. Overexpression of the superoxide dismutases (SODs) in vitro increases cell differentiation, decreases cell growth and proliferation, and can reverse a malignant phenotype to a nonmalignant one. The situation in vivo is more complex due to multiple interactions of tumor cells with their environment. Numerous in vivo studies show that the superoxide dismutases can be highly expressed in aggressive human solid tumors. Furthermore, high SOD has occasionally been associated with a poor prognosis and with resistance to cytotoxic drugs and radiation. Most of the apparent conflicts between the above in vitro and in vivo observations can be reconciled by considering the net redox status of tumor cells in different environments. Administering high concentrations of SOD to cells in vitro is usually associated with a non- or less malignant phenotype, whereas secondary induction of SOD in tumors in vivo can be associated with an aggressive malignant transformation probably due to the altered (oxidative) redox state in the malignant cells. This concept suggests that for many types of tumors antioxidants could be used to diminish the invasive capability of malignant cells.
Collapse
Affiliation(s)
- Vuokko L Kinnula
- Division of Pulmonary Medicine, Department of Medicine, University of Helsinki and Helsinki University Hospital, FIN-0029 Helsinki, Finland.
| | | |
Collapse
|
36
|
Roper JM, Mazzatti DJ, Watkins RH, Maniscalco WM, Keng PC, O'Reilly MA. In vivo exposure to hyperoxia induces DNA damage in a population of alveolar type II epithelial cells. Am J Physiol Lung Cell Mol Physiol 2004; 286:L1045-54. [PMID: 14729512 DOI: 10.1152/ajplung.00376.2003] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It is well established that hyperoxia injures and kills alveolar endothelial and type I epithelial cells of the lung. Although type II epithelial cells remain morphologically intact, it remains unclear whether they are also damaged. DNA integrity was investigated in adult mice whose type II cells were identified by their endogenous expression of pro-surfactant protein C or transgenic expression of enhanced green fluorescent protein. In mice exposed to room air, punctate perinuclear 8-oxoguanine staining was detected in approximately 4% of all alveolar cells and in 30% of type II cells. After 48 or 72 h of hyperoxia, 8-oxoguanine was detected in 11% of all alveolar cells and in >60% of type II cells. 8-Oxoguanine colocalized by confocal microscopy with the mitochondrial transmembrane protein cytochrome oxidase subunit 1. Type II cells isolated from hyperoxic lungs exhibited nuclear DNA strand breaks by comet assay even though they were viable and morphologically indistinguishable from cells isolated from lungs exposed to room air. These data reveal that type II cells exposed to in vivo hyperoxia have oxidized and fragmented DNA. Because type II cells are essential for lung remodeling, our findings raise the possibility that they are proficient in DNA repair.
Collapse
Affiliation(s)
- Jason M Roper
- Dept. of Pediatrics, Box 850, School of Medicine and Dentistry, Univ. of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
37
|
Paine R, Wilcoxen SE, Morris SB, Sartori C, Baleeiro CEO, Matthay MA, Christensen PJ. Transgenic overexpression of granulocyte macrophage-colony stimulating factor in the lung prevents hyperoxic lung injury. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 163:2397-406. [PMID: 14633611 PMCID: PMC1892382 DOI: 10.1016/s0002-9440(10)63594-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Granulocyte macrophage-colony stimulating factor (GM-CSF) plays an important role in pulmonary homeostasis, with effects on both alveolar macrophages and alveolar epithelial cells. We hypothesized that overexpression of GM-CSF in the lung would protect mice from hyperoxic lung injury by limiting alveolar epithelial cell injury. Wild-type C57BL/6 mice and mutant mice in which GM-CSF was overexpressed in the lung under control of the SP-C promoter (SP-C-GM mice) were placed in >95% oxygen. Within 6 days, 100% of the wild-type mice had died, while 70% of the SP-C-GM mice remained alive after 10 days in hyperoxia. Histological assessment of the lungs at day 4 revealed less disruption of the alveolar wall in SP-C-GM mice compared to wild-type mice. The concentration of albumin in bronchoalveolar lavage fluid after 4 days in hyperoxia was significantly lower in SP-C-GM mice than in wild-type mice, indicating preservation of alveolar epithelial barrier properties in the SP-C-GM mice. Alveolar fluid clearance was preserved in SP-C-GM mice in hyperoxia, but decreased significantly in hyperoxia-exposed wild-type mice. Staining of lung tissue for caspase 3 demonstrated increased apoptosis in alveolar wall cells in wild-type mice in hyperoxia compared to mice in room air. In contrast, SP-C-GM mice exposed to hyperoxia demonstrated only modest increase in alveolar wall apoptosis compared to room air. Systemic treatment with GM-CSF (9 micro g/kg/day) during 4 days of hyperoxic exposure resulted in decreased apoptosis in the lungs compared to placebo. In studies using isolated murine type II alveolar epithelial cells, treatment with GM-CSF greatly reduced apoptosis in response to suspension culture. In conclusion, overexpression of GM-CSF enhances survival of mice in hyperoxia; this effect may be explained by preservation of alveolar epithelial barrier function and fluid clearance, at least in part because of reduction in hyperoxia-induced apoptosis of cells in the alveolar wall.
Collapse
Affiliation(s)
- Robert Paine
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan 48105, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Buccellato LJ, Tso M, Akinci OI, Chandel NS, Budinger GRS. Reactive oxygen species are required for hyperoxia-induced Bax activation and cell death in alveolar epithelial cells. J Biol Chem 2003; 279:6753-60. [PMID: 14625274 DOI: 10.1074/jbc.m310145200] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Exposure of animals to hyperoxia results in respiratory failure and death within 72 h. Histologic evaluation of the lungs of these animals demonstrates epithelial apoptosis and necrosis. Although the generation of reactive oxygen species (ROS) is widely thought to be responsible for the cell death observed following exposure to hyperoxia, it is not clear whether they act upstream of activation of the cell death pathway or whether they are generated as a result of mitochondrial membrane permeabilization and caspase activation. We hypothesized that the generation of ROS was required for hyperoxia-induced cell death upstream of Bax activation. In primary rat alveolar epithelial cells, we found that exposure to hyperoxia resulted in the generation of ROS that was completely prevented by the administration of the combined superoxide dismutase/catalase mimetic EUK-134 (Eukarion, Inc., Bedford, MA). Exposure to hyperoxia resulted in the activation of Bax at the mitochondrial membrane, cytochrome c release, and cell death. The administration of EUK-134 prevented Bax activation, cytochrome c release, and cell death. In a mouse lung epithelial cell line (MLE-12), the overexpression of Bcl-XL protected cells against hyperoxia by preventing the activation of Bax at the mitochondrial membrane. We conclude that exposure to hyperoxia results in Bax activation at the mitochondrial membrane and subsequent cytochrome c release. Bax activation at the mitochondrial membrane requires the generation of ROS and can be prevented by the overexpression of Bcl-XL.
Collapse
Affiliation(s)
- Leonard J Buccellato
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | |
Collapse
|
39
|
Romashko J, Horowitz S, Franek WR, Palaia T, Miller EJ, Lin A, Birrer MJ, Scott W, Mantell LL. MAPK pathways mediate hyperoxia-induced oncotic cell death in lung epithelial cells. Free Radic Biol Med 2003; 35:978-93. [PMID: 14556862 DOI: 10.1016/s0891-5849(03)00494-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cell injury and cell death of pulmonary epithelium plays an important role in the pathogenesis of acute lung injury in animals exposed to prolonged hyperoxia. The aim of this study was to decipher the molecular mechanisms modulating cell death induced by hyperoxia in lung epithelium. Cell death is thought to be either apoptotic, with shrinking phenotypes and activated caspases, or oncotic, with swelling organelles. Exposure to 95% O2 (hyperoxia) induced cell death of MLE-12 cells with cellular as well as nuclear swelling, cytosolic vacuolation, and loss of mitochondrial structure and enzyme function. Neither elevated caspase-3 activity nor phosphatidylserine translocation were detected, suggesting that in hyperoxia, MLE-12 cells die via oncosis rather than apoptosis. In addition, hyperoxia triggered a sustained activation of the transcription factor AP-1, as well as mitogen-activated protein kinase (MAPK) family members p38 and JNK. Importantly, survival of MLE-12 cells in hyperoxia was significantly enhanced when either AP-1, p38, or JNK activation was inhibited by either specific inhibitors or dominant negative DNA constructs, indicating that in lung epithelial cells hyperoxia induces a program-driven oncosis, involving AP-1, JNK, and p38 MAPK. Interestingly, hydrogen peroxide-induced oxidative apoptosis of MLE-12 cells, with a shrinking nuclear morphology and activated caspase-3 activity, is also mediated by AP-1, JNK, and p38. Therefore, our data indicate that although they have divergent downstream events, oxidative oncosis and apoptosis share upstream JNK/p38 and AP-1 pathways, which could be used as potential targets for reducing hyperoxic inflammatory lung injury.
Collapse
Affiliation(s)
- John Romashko
- Department of Surgery, North Shore University Hospital, New York University School of Medicine, Manhasset, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Abstract
The lungs are directly exposed to higher oxygen concentrations than most other tissues. Increased oxidative stress is a significant part of the pathogenesis of obstructive lung diseases such as asthma and chronic obstructive pulmonary disease, parenchymal lung diseases (e.g., idiopathic pulmonary fibrosis and lung granulomatous diseases), and lung malignancies. Lung tissue is protected against these oxidants by a variety of antioxidant mechanisms among which the superoxide dismutases (SODs) are the only ones converting superoxide radicals to hydrogen peroxide. There are three SODs: cytosolic copper-zinc, mitochondrial manganese, and extracellular SODs. These enzymes have specific distributions and functions. Their importance in protecting lung tissue has been confirmed in transgenic and knockout animal studies. Relatively few studies have been conducted on these enzymes in the normal human lung or in human lung diseases. Most human studies suggest that there is induction of manganese SOD and, possibly, extracellular SOD during inflammatory, but not fibrotic, phases of parenchymal lung diseases and that both copper-zinc SOD and manganese SOD may be downregulated in asthmatic airways. Many previous antioxidant therapies have been disappointing, but newly characterized SOD mimetics are being shown to protect against oxidant-related lung disorders in animal models.
Collapse
|
42
|
Saibara T, Toda K, Wakatsuki A, Ogawa Y, Ono M, Onishi S. Protective effect of 3-methyl-1-phenyl-2-pyrazolin-5-one, a free radical scavenger, on acute toxicity of paraquat in mice. Toxicol Lett 2003; 143:51-4. [PMID: 12697380 DOI: 10.1016/s0378-4274(03)00113-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Paraquat poisoning results in damages of multiple organs including liver, kidney and lung, and antioxidants have been proven to have protective effects. As a novel free radical scavenger, 3-methyl-1-phenyl-2-pyrazolin-5-one (MCI-186), was introduced to clinical use recently, its protective effect was studied on acute toxicity of paraquat in male ddY mice. When paraquat (175 mg/kg) were given orally, the survival rate was only 8% on the 6th day of paraquat ingestion in Control Group mice. Protective effect of MCI-186 was most evident and the survival rate was 42% on the 6th day and 38% on the 14th day of paraquat ingestion, respectively, when mice were treated immediately. A delay of 30 min in treatment resulted in an abrupt reduction of the survival rate. These results suggested that MCI-186 used in acute phase of paraquat intoxication might serve as a clinically available antidote for attenuating paraquat toxicity.
Collapse
Affiliation(s)
- Toshiji Saibara
- First Department of Internal Medicine, Kochi Medical School, Nankoku 783-8505, Japan.
| | | | | | | | | | | |
Collapse
|
43
|
O'Reilly PJ, Hickman-Davis JM, Davis IC, Matalon S. Hyperoxia impairs antibacterial function of macrophages through effects on actin. Am J Respir Cell Mol Biol 2003; 28:443-50. [PMID: 12654633 DOI: 10.1165/rcmb.2002-0153oc] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Oxidative stress may impair alveolar macrophage function in patients with inflammatory lung diseases or those exposed to high concentrations of oxygen. We investigated putative mechanisms of injury to macrophages by oxidative stress, using RAW 264.7 cells exposed to 95% oxygen for 48 h. Hyperoxia-exposed macrophages were less able to phagocytose and kill Klebsiella pneumoniae than normoxic controls, despite increased production of nitric oxide, a free radical important in pathogen killing. Exposure of macrophages to hyperoxia had marked effects on the actin cytoskeleton, including increased actin polymerization, loss of cortical actin, formation of stress fibers, de novo synthesis of actin, and actin oxidation. Hyperoxia induced changes in cell morphology, with increased cell size and pseudopod formation. Exposure of macrophages to jasplakinolide, an agent that increases actin polymerization, also impaired their ability to phagocytose Klebsiella. Alveolar macrophages isolated from mice exposed to 100% oxygen for 84 h also demonstrated impaired phagocytic function, as well as similar effects on the actin cytoskeleton and cell morphology to macrophages exposed to hyperoxia in vitro. We conclude that oxidative stress in vitro and in vivo impairs macrophage antibacterial function through effects on actin.
Collapse
Affiliation(s)
- Philip J O'Reilly
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Alabama at Birmingham, 35205-3703, USA
| | | | | | | |
Collapse
|
44
|
Davis JM, Parad RB, Michele T, Allred E, Price A, Rosenfeld W. Pulmonary outcome at 1 year corrected age in premature infants treated at birth with recombinant human CuZn superoxide dismutase. Pediatrics 2003; 111:469-76. [PMID: 12612223 DOI: 10.1542/peds.111.3.469] [Citation(s) in RCA: 203] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To examine whether treatment of premature infants with intratracheal recombinant human CuZn superoxide dismutase (r-h CuZnSOD) reduces bronchopulmonary dysplasia and improves pulmonary outcome at 1 year corrected age. DESIGN Three hundred two premature infants (600-1200 g birth weight) treated with exogenous surfactant at birth for respiratory distress syndrome were randomized to receive either intratracheal r-h CuZnSOD (5 mg/kg in 2 mL/kg saline) or placebo every 48 hours (as long as intubation was required) for up to 1 month of age. Short-term, as well as longer-term pulmonary outcome was assessed. RESULTS There were no differences between groups in the incidence of death or the development of bronchopulmonary dysplasia (oxygen requirement with an Edwards chest radiograph score of >or=3) at 28 days of life or 36 weeks' postmenstrual age. r-h CuZnSOD was well-tolerated and not associated with significant increases in any adverse event. At a median of 1 year corrected age, health assessments and physical examinations were performed on 209 (80%) surviving infants, with complete data available on 189 infants. Thirty-seven percent of placebo-treated infants had repeated episodes of wheezing or other respiratory illness severe enough to require treatment with asthma medications such as bronchodilators and/or corticosteroids compared with 24% of r-h CuZnSOD-treated infants, a 36% reduction. In infants <27 weeks' gestation, 42% treated with placebo received asthma medications compared with 19% of r-h CuZnSOD-treated infants, a 55% decrease. Infants <27 weeks' gestation who received r-h CuZnSOD also had a 55% decrease in emergency department visits and a 44% decrease in subsequent hospitalizations. Growth measurements and the results of physical examinations were comparable between groups. CONCLUSIONS These data indicate that treatment at birth with r-h CuZnSOD may reduce early pulmonary injury, resulting in improved clinical status when measured at 1 year corrected age. r-h CuZnSOD appears to be a safe and effective therapy that improves pulmonary outcome in high-risk premature infants.
Collapse
Affiliation(s)
- Jonathan M Davis
- Department of Pediatrics (Neonatology), CardioPulmonary Research Institute, Winthrop University Hospital, SUNY Stony Brook School of Medicine, Mineola, New York 11501, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Davis JM. Role of oxidant injury in the pathogenesis of neonatal lung disease. ACTA PAEDIATRICA (OSLO, NORWAY : 1992). SUPPLEMENT 2003; 91:23-5. [PMID: 12200893 DOI: 10.1111/j.1651-2227.2002.tb00156.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
UNLABELLED Bronchopulmonary dysplasia (BPD) is a chronic lung disease that develops in newborn infants treated with oxygen and mechanical ventilation for a primary lung disorder. With significant improvements in survival of many critically ill infants, BPD has become an extremely important complication of newborn intensive care. The pathogenesis of BPD is complex and involves a variety of causative factors. However, increasing evidence has suggested that an oxidative insult could be an extremely important component of the injury process. Premature infants are especially sensitive to oxidant injury since they are exposed to supraphysiological concentrations of oxygen at birth while endogenous antioxidant enzyme activity may be relatively deficient. CONCLUSION Superoxide dismutase is an antioxidant enzyme that has been shown in numerous basic and clinical studies to protect cells against oxidant injury.
Collapse
Affiliation(s)
- Jonathan M Davis
- Division of Neonatology, Winthrop University Hospital, SUNY at Stony Brook School of Medicine, 259 First St, Mineola, NY 11501, USA
| |
Collapse
|
46
|
Fessel JP, Porter NA, Moore KP, Sheller JR, Roberts LJ. Discovery of lipid peroxidation products formed in vivo with a substituted tetrahydrofuran ring (isofurans) that are favored by increased oxygen tension. Proc Natl Acad Sci U S A 2002; 99:16713-8. [PMID: 12482927 PMCID: PMC139209 DOI: 10.1073/pnas.252649099] [Citation(s) in RCA: 185] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Free radicals have been implicated in the pathogenesis of an increasing number of diseases. Lipids, which undergo peroxidation, are major targets of free radical attack. We report the discovery of a pathway of lipid peroxidation that forms a series of isomers in vivo that are characterized by a substituted tetrahydrofuran ring structure, termed isofurans (IsoFs). We have proposed two distinct pathways by which IsoFs can be formed based on 18O2 and H2 18O labeling studies. Measurement of F2-isoprostanes (IsoPs), prostaglandin F2-like compounds formed nonenzymatically as products of lipid peroxidation, is considered one of the most reliable approaches for assessing oxidative stress status in vivo. However, one limitation with this approach is that the formation of IsoPs becomes limited at high oxygen tension. In contrast, the formation of IsoFs becomes increasingly favored as oxygen tension increases. IsoFs are present at readily detectable levels in normal fluids and tissues, and levels increase dramatically in CCl4-treated rats, an animal model of oxidant injury. The ratio of IsoFs to IsoPs in major organs varies according to normal steady-state tissue oxygenation. In addition, IsoFs show a marked increase early in the course of hyperoxia-induced lung injury, whereas IsoPs do not significantly increase. We propose that combined measurement of IsoFs and IsoPs should provide a more reliable index of oxidant stress severity than quantification of either alone because of the opposing modulation of the two pathways by oxygen tension, which can vary widely in different organs and disease states.
Collapse
Affiliation(s)
- Joshua P Fessel
- Departments of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | |
Collapse
|
47
|
Abstract
This review describes production and effects of reactive oxygen species (ROS) on airway function. ROS are important in many physiological processes but can also have detrimental effects on airway cells and tissues when produced in high quantities or during the absence of sufficient amounts of anti-oxidants. Therefore, these mediators play a prominent role in the pathogenesis of various inflammatory airway disorders, including asthma. Effects of ROS on airway function in asthma have been studied with isolated airway cells and tissues and with animal models and patients. With the use of inhibitors, transgenic animals and measurements of the release of ROS within the airways, it became clear that oxidative stress contributes to the initiation and worsening of inflammatory respiratory disorders.
Collapse
Affiliation(s)
- P A Henricks
- Department of Pharmacology and Pathophysiology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands.
| | | |
Collapse
|
48
|
Gomez-Sanchez EP, Gomez-Sanchez CE. Central hypertensinogenic effects of glycyrrhizic acid and carbenoxolone. Exp Biol Med (Maywood) 1993; 234:263-77. [PMID: 1476186 DOI: 10.3181/0805-rm-178] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The apparent mineralocorticoid excess syndrome of patients ingesting large amounts of licorice or its derivatives is thought to be caused by the antagonism by these compounds of the enzyme 11 beta-hydroxysteroid dehydrogenase (11 beta-HSD). 11 beta-HSD inactivates cortisol and corticosterone, allowing the more abundantly produced glucocorticoids access to the mineralocorticoid receptor (MR) in the kidney, where they act as mineralocorticoids. We have found that the infusion of both glycyrrhizic acid, an active principle of licorice, and carbenoxolone, a synthetic analogue, into a lateral ventricle of the brain [intracerebroventricular (icv)] of a rat, at a dose less than that which has an effect when infused subcutaneously, produces hypertension. Furthermore, the hypertension produced by the oral administration of carbenoxolone or glycyrrhizic acid is blocked by the icv administration of RU 28318, an MR antagonist, at a dose below that which has an effect on blood pressure when infused subcutaneously. While the oral administration caused saline polydipsia and polyuria typical of chronic systemic mineralocorticoid excess, the icv licorice derivatives produced hypertension without affecting saline appetite. Sensitizing the rats to mineralocorticoid hypertension by renal mass reduction and increasing salt consumption was not necessary for the production of hypertension. These findings provide additional evidence for a central role in blood pressure control by mineralocorticoids that is distinct from their renal effects. They also suggest that more is involved in licorice-induced hypertension than only inhibition of 11 beta-HSD.
Collapse
|