1
|
Song Y, Yang C. Mechanistic advances of hyperoxia-induced immature brain injury. Heliyon 2024; 10:e30005. [PMID: 38694048 PMCID: PMC11058899 DOI: 10.1016/j.heliyon.2024.e30005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/11/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
The impact of hyperoxia-induced brain injury in preterm infants is being increasingly investigated. However, the parameters and protocols used to study this condition in animal models lack consistency. Research is further hampered by the fact that hyperoxia exerts both direct and indirect effects on oligodendrocytes and neurons, with the precise underlying mechanisms remaining unclear. In this article, we aim to provide a comprehensive overview of the conditions used to induce hyperoxia in animal models of immature brain injury. We discuss what is known regarding the mechanisms underlying hyperoxia-induced immature brain injury, focusing on the effects on oligodendrocytes and neurons, and briefly describe therapies that may counteract the effects of hyperoxia. We also identify further studies required to fully elucidate the effects of hyperoxia on the immature brain as well as discuss the leading therapeutic options.
Collapse
Affiliation(s)
- Yue Song
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, China
- Department of Clinical Medicine, The Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Changqiang Yang
- Department of Cardiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, China
- Department of Clinical Medicine, The Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| |
Collapse
|
2
|
Zaghloul N, Cohen NS, Ayasolla KR, Li HL, Kurepa D, Ahmed MN. Galantamine ameliorates hyperoxia-induced brain injury in neonatal mice. Front Neurosci 2023; 17:890015. [PMID: 37424990 PMCID: PMC10323435 DOI: 10.3389/fnins.2023.890015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Prolonged oxygen therapy in preterm infants often leads to cognitive impairment. Hyperoxia leads to excess free radical production with subsequent neuroinflammation, astrogliosis, microgliosis and apoptosis. We hypothesized that Galantamine, an acetyl choline esterase inhibitor and an FDA approved treatment of Alzheimer's disease, will reduce hyperoxic brain injury in neonatal mice and will improve learning and memory. Methods Mouse pups at postnatal day 1 (P1) were placed in a hyperoxia chamber (FiO2 95%) for 7 days. Pups were injected IP daily with Galantamine (5 mg/kg/dose) or saline for 7 days. Results Hyperoxia caused significant neurodegeneration in cholinergic nuclei of the basal forebrain cholinergic system (BFCS), laterodorsal tegmental (LDT) nucleus and nucleus ambiguus (NA). Galantamine ameliorated this neuronal loss. Treated hyperoxic group showed a significant increase of choline acetyl transferase (ChAT) expression and a decrease of acetyl choline esterase activity, thus increasing acetyl choline levels in hyperoxia environment. Hyperoxia increased pro-inflammatory cytokines namely IL -1β, IL-6 and TNF α, HMGB1, NF-κB activation. Galantamine showed its potent anti- inflammatory effect, by blunting cytokines surges among treated group. Treatment with Galantamine increased myelination while reducing apoptosis, microgliosis, astrogliosis and ROS production. Long term neurobehavioral outcomes at P60 showed improved locomotor activity, coordination, learning and memory, along with increased hippocampal volumes on MRI with Galantamine treated versus non treated hyperoxia group. Conclusion Together our findings suggest a potential therapeutic role for Galantamine in attenuating hyperoxia-induced brain injury.
Collapse
Affiliation(s)
- Nahla Zaghloul
- Steele Children's Research Center, Division of Neonatology, Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Naomi S. Cohen
- Neonatology Research Laboratory, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | | | - Hsiu-Ling Li
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, New York, NY, United States
| | - Dalibor Kurepa
- Neonatology Research Laboratory, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Mohamed N. Ahmed
- Steele Children's Research Center, Division of Neonatology, Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
3
|
Guez-Barber D, Eisch AJ, Cristancho AG. Developmental Brain Injury and Social Determinants of Health: Opportunities to Combine Preclinical Models for Mechanistic Insights into Recovery. Dev Neurosci 2023; 45:255-267. [PMID: 37080174 PMCID: PMC10614252 DOI: 10.1159/000530745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/14/2023] [Indexed: 04/22/2023] Open
Abstract
Epidemiological studies show that social determinants of health are among the strongest factors associated with developmental outcomes after prenatal and perinatal brain injuries, even when controlling for the severity of the initial injury. Elevated socioeconomic status and a higher level of parental education correlate with improved neurologic function after premature birth. Conversely, children experiencing early life adversity have worse outcomes after developmental brain injuries. Animal models have provided vital insight into mechanisms perturbed by developmental brain injuries, which have indicated directions for novel therapeutics or interventions. Animal models have also been used to learn how social environments affect brain maturation through enriched environments and early adverse conditions. We recognize animal models cannot fully recapitulate human social circumstances. However, we posit that mechanistic studies combining models of developmental brain injuries and early life social environments will provide insight into pathways important for recovery. Some studies combining enriched environments with neonatal hypoxic injury models have shown improvements in developmental outcomes, but further studies are needed to understand the mechanisms underlying these improvements. By contrast, there have been more limited studies of the effects of adverse conditions on developmental brain injury extent and recovery. Uncovering the biological underpinnings for early life social experiences has translational relevance, enabling the development of novel strategies to improve outcomes through lifelong treatment. With the emergence of new technologies to analyze subtle molecular and behavioral phenotypes, here we discuss the opportunities for combining animal models of developmental brain injury with social construct models to deconvolute the complex interactions between injury, recovery, and social inequity.
Collapse
Affiliation(s)
- Danielle Guez-Barber
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amelia J. Eisch
- Department of Anesthesiology and Critical Care, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana G. Cristancho
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
4
|
Mancardi D, Ottolenghi S, Attanasio U, Tocchetti CG, Paroni R, Pagliaro P, Samaja M. Janus, or the Inevitable Battle Between Too Much and Too Little Oxygen. Antioxid Redox Signal 2022; 37:972-989. [PMID: 35412859 DOI: 10.1089/ars.2021.0232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Significance: Oxygen levels are key regulators of virtually every living mammalian cell, under both physiological and pathological conditions. Starting from embryonic and fetal development, through the growth, onset, and progression of diseases, oxygen is a subtle, although pivotal, mediator of key processes such as differentiation, proliferation, autophagy, necrosis, and apoptosis. Hypoxia-driven modifications of cellular physiology are investigated in depth or for their clinical and translational relevance, especially in the ischemic scenario. Recent Advances: The mild or severe lack of oxygen is, undoubtedly, related to cell death, although abundant evidence points at oscillating oxygen levels, instead of permanent low pO2, as the most detrimental factor. Different cell types can consume oxygen at different rates and, most interestingly, some cells can shift from low to high consumption according to the metabolic demand. Hence, we can assume that, in the intracellular compartment, oxygen tension varies from low to high levels depending on both supply and consumption. Critical Issues: The positive balance between supply and consumption leads to a pro-oxidative environment, with some cell types facing hypoxia/hyperoxia cycles, whereas some others are under fairly constant oxygen tension. Future Directions: Within this frame, the alterations of oxygen levels (dysoxia) are critical in two paradigmatic organs, the heart and brain, under physiological and pathological conditions and the interactions of oxygen with other physiologically relevant gases, such as nitric oxide, can alternatively contribute to the worsening or protection of ischemic organs. Further, the effects of dysoxia are of pivotal importance for iron metabolism. Antioxid. Redox Signal. 37, 972-989.
Collapse
Affiliation(s)
- Daniele Mancardi
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Sara Ottolenghi
- Department of Health Sciences, University of Milano, Milan, Italy
- School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Umberto Attanasio
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Carlo Gabriele Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Interdepartmental Center for Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
| | - Rita Paroni
- Department of Health Sciences, University of Milano, Milan, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Michele Samaja
- Department of Health Sciences, University of Milano, Milan, Italy
- MAGI GROUP, San Felice del Benaco, Italy
| |
Collapse
|
5
|
Siwicka-Gieroba D, Robba C, Gołacki J, Badenes R, Dabrowski W. Cerebral Oxygen Delivery and Consumption in Brain-Injured Patients. J Pers Med 2022; 12:1763. [PMID: 36573716 PMCID: PMC9698645 DOI: 10.3390/jpm12111763] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 12/30/2022] Open
Abstract
Organism survival depends on oxygen delivery and utilization to maintain the balance of energy and toxic oxidants production. This regulation is crucial to the brain, especially after acute injuries. Secondary insults after brain damage may include impaired cerebral metabolism, ischemia, intracranial hypertension and oxygen concentration disturbances such as hypoxia or hyperoxia. Recent data highlight the important role of clinical protocols in improving oxygen delivery and resulting in lower mortality in brain-injured patients. Clinical protocols guide the rules for oxygen supplementation based on physiological processes such as elevation of oxygen supply (by mean arterial pressure (MAP) and intracranial pressure (ICP) modulation, cerebral vasoreactivity, oxygen capacity) and reduction of oxygen demand (by pharmacological sedation and coma or hypothermia). The aim of this review is to discuss oxygen metabolism in the brain under different conditions.
Collapse
Affiliation(s)
- Dorota Siwicka-Gieroba
- Department of Anaesthesiology and Intensive Care, Medical University in Lublin, 20-954 Lublin, Poland
| | - Chiara Robba
- Department of Anesthesiology and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, 16132 Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Jakub Gołacki
- Department of Anaesthesiology and Intensive Care, Medical University in Lublin, 20-954 Lublin, Poland
| | - Rafael Badenes
- Department of Anesthesiology and Surgical-Trauma Intensive Care, Hospital Clinic Universitari, University of Valencia, 46010 Valencia, Spain
| | - Wojciech Dabrowski
- Department of Anaesthesiology and Intensive Care, Medical University in Lublin, 20-954 Lublin, Poland
| |
Collapse
|
6
|
Perinatal Hyperoxia and Developmental Consequences on the Lung-Brain Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5784146. [PMID: 35251477 PMCID: PMC8894035 DOI: 10.1155/2022/5784146] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022]
Abstract
Approximately 11.1% of all newborns worldwide are born preterm. Improved neonatal intensive care significantly increased survival rates over the last decades but failed to reduce the risk for the development of chronic lung disease (i.e., bronchopulmonary dysplasia (BPD)) and impaired neurodevelopment (i.e., encephalopathy of prematurity (EoP)), two major long-term sequelae of prematurity. Premature infants are exposed to relative hyperoxia, when compared to physiological in-utero conditions and, if needed to additional therapeutic oxygen supplementation. Both are associated with an increased risk for impaired organ development. Since the detrimental effects of hyperoxia on the immature retina are known for many years, lung and brain have come into focus in the last decade. Hyperoxia-induced excessive production of reactive oxygen species leading to oxidative stress and inflammation contribute to pulmonary growth restriction and abnormal neurodevelopment, including myelination deficits. Despite a large body of studies, which unraveled important pathophysiological mechanisms for both organs at risk, the majority focused exclusively either on lung or on brain injury. However, considering that preterm infants suffering from BPD are at higher risk for poor neurodevelopmental outcome, an interaction between both organs seems plausible. This review summarizes recent findings regarding mechanisms of hyperoxia-induced neonatal lung and brain injury. We will discuss common pathophysiological pathways, which potentially link both injured organ systems. Furthermore, promises and needs of currently suggested therapies, including pharmacological and regenerative cell-based treatments for BPD and EoP, will be emphasized. Limited therapeutic approaches highlight the urgent need for a better understanding of the mechanisms underlying detrimental effects of hyperoxia on the lung-brain axis in order to pave the way for the development of novel multimodal therapies, ideally targeting both severe preterm birth-associated complications.
Collapse
|
7
|
Dilek M, Orallar H, Cetinkaya A, Bozat G, Pehlivan F, Bekdas M, Kabakus N. Can Excessive Oxygen Cause Hyperactive Behavior Disorder in Preterm Children? Cognitive Effects of Hyperoxia in the Preterm Brain of Rats. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09819-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
8
|
Dapaah-Siakwan F, Zambrano R, Luo S, Duncan MR, Kerr N, Donda K, Vaccari JPDR, Keane RW, Dietrich WD, Benny M, Young K, Wu S. Caspase-1 Inhibition Attenuates Hyperoxia-induced Lung and Brain Injury in Neonatal Mice. Am J Respir Cell Mol Biol 2019; 61:341-354. [DOI: 10.1165/rcmb.2018-0192oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Fredrick Dapaah-Siakwan
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Ronald Zambrano
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Shihua Luo
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Matthew R. Duncan
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Nadine Kerr
- Miami Project to Cure Paralysis
- Department of Physiology and Biophysics, and
| | - Keyur Donda
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Juan Pablo de Rivero Vaccari
- Miami Project to Cure Paralysis
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Robert W. Keane
- Miami Project to Cure Paralysis
- Department of Physiology and Biophysics, and
| | - W. Dalton Dietrich
- Miami Project to Cure Paralysis
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Merline Benny
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Karen Young
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Shu Wu
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| |
Collapse
|
9
|
Buczynski BW, Mai N, Yee M, Allen JL, Prifti L, Cory-Slechta DA, Halterman MW, O'Reilly MA. Lung-Specific Extracellular Superoxide Dismutase Improves Cognition of Adult Mice Exposed to Neonatal Hyperoxia. Front Med (Lausanne) 2018; 5:334. [PMID: 30619855 PMCID: PMC6295554 DOI: 10.3389/fmed.2018.00334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 11/12/2018] [Indexed: 11/13/2022] Open
Abstract
Lung and brain development is often altered in infants born preterm and exposed to excess oxygen, and this can lead to impaired lung function and neurocognitive abilities later in life. Oxygen-derived reactive oxygen species and the ensuing inflammatory response are believed to be an underlying cause of disease because over-expression of some anti-oxidant enzymes is protective in animal models. For example, neurodevelopment is preserved in mice that ubiquitously express human extracellular superoxide dismutase (EC-SOD) under control of an actin promoter. Similarly, oxygen-dependent changes in lung development are attenuated in transgenic SftpcEC−SOD mice that over-express EC-SOD in pulmonary alveolar epithelial type II cells. But whether anti-oxidants targeted to the lung provide protection to other organs, such as the brain is not known. Here, we use transgenic SftpcEC−SOD mice to investigate whether lung-specific expression of EC-SOD also preserves neurodevelopment following exposure to neonatal hyperoxia. Wild type and SftpcEC−SOD transgenic mice were exposed to room air or 100% oxygen between postnatal days 0–4. At 8 weeks of age, we investigated neurocognitive function as defined by novel object recognition, pathologic changes in hippocampal neurons, and microglial cell activation. Neonatal hyperoxia impaired novel object recognition memory in adult female but not male mice. Behavioral deficits were associated with microglial activation, CA1 neuron nuclear contraction, and fiber sprouting within the hilus of the dentate gyrus (DG). Over-expression of EC-SOD in the lung preserved novel object recognition and reduced the observed changes in neuronal nuclear size and myelin basic protein fiber density. It had no effect on the extent of microglial activation in the hippocampus. These findings demonstrate pulmonary expression of EC-SOD preserves short-term memory in adult female mice exposed to neonatal hyperoxia, thus suggesting anti-oxidants designed to alleviate oxygen-induced lung disease such as in preterm infants may also be neuroprotective.
Collapse
Affiliation(s)
- Bradley W Buczynski
- Department of Environmental Medicine, University of Rochester, Rochester, NY, United States
| | - Nguyen Mai
- Department of Neurology, University of Rochester, Rochester, NY, United States
| | - Min Yee
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
| | - Joshua L Allen
- Department of Environmental Medicine, University of Rochester, Rochester, NY, United States
| | - Landa Prifti
- Department of Neurology, University of Rochester, Rochester, NY, United States
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester, Rochester, NY, United States
| | - Marc W Halterman
- Department of Neurology, University of Rochester, Rochester, NY, United States
| | - Michael A O'Reilly
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
| |
Collapse
|
10
|
Terraneo L, Samaja M. Comparative Response of Brain to Chronic Hypoxia and Hyperoxia. Int J Mol Sci 2017; 18:ijms18091914. [PMID: 28880206 PMCID: PMC5618563 DOI: 10.3390/ijms18091914] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/01/2017] [Accepted: 09/03/2017] [Indexed: 12/25/2022] Open
Abstract
Two antithetic terms, hypoxia and hyperoxia, i.e., insufficient and excess oxygen availability with respect to needs, are thought to trigger opposite responses in cells and tissues. This review aims at summarizing the molecular and cellular mechanisms underlying hypoxia and hyperoxia in brain and cerebral tissue, a context that may prove to be useful for characterizing not only several clinically relevant aspects, but also aspects related to the evolution of oxygen transport and use by the tissues. While the response to acute hypoxia/hyperoxia presumably recruits only a minor portion of the potentially involved cell machinery, focusing into chronic conditions, instead, enables to take into consideration a wider range of potential responses to oxygen-linked stress, spanning from metabolic to genic. We will examine how various brain subsystems, including energetic metabolism, oxygen sensing, recruitment of pro-survival pathways as protein kinase B (Akt), mitogen-activated protein kinases (MAPK), neurotrophins (BDNF), erythropoietin (Epo) and its receptors (EpoR), neuroglobin (Ngb), nitric oxide (NO), carbon monoxide (CO), deal with chronic hypoxia and hyperoxia to end-up with the final outcomes, oxidative stress and brain damage. A more complex than expected pattern results, which emphasizes the delicate balance between the severity of the stress imposed by hypoxia and hyperoxia and the recruitment of molecular and cellular defense patterns. While for certain functions the expectation that hypoxia and hyperoxia should cause opposite responses is actually met, for others it is not, and both emerge as dangerous treatments.
Collapse
Affiliation(s)
- Laura Terraneo
- Department of Health Science, University of Milan, I-20142 Milano, Italy.
| | - Michele Samaja
- Department of Health Science, University of Milan, I-20142 Milano, Italy.
| |
Collapse
|
11
|
Wingelaar TT, van Ooij PJAM, van Hulst RA. Oxygen Toxicity and Special Operations Forces Diving: Hidden and Dangerous. Front Psychol 2017; 8:1263. [PMID: 28790955 PMCID: PMC5524741 DOI: 10.3389/fpsyg.2017.01263] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/11/2017] [Indexed: 12/04/2022] Open
Abstract
In Special Operations Forces (SOF) closed-circuit rebreathers with 100% oxygen are commonly utilized for covert diving operations. Exposure to high partial pressures of oxygen (PO2) could cause damage to the central nervous system (CNS) and pulmonary system. Longer exposure time and higher PO2 leads to faster development of more serious pathology. Exposure to a PO2 above 1.4 ATA can cause CNS toxicity, leading to a wide range of neurologic complaints including convulsions. Pulmonary oxygen toxicity develops over time when exposed to a PO2 above 0.5 ATA and can lead to inflammation and fibrosis of lung tissue. Oxygen can also be toxic for the ocular system and may have systemic effects on the inflammatory system. Moreover, some of the effects of oxygen toxicity are irreversible. This paper describes the pathophysiology, epidemiology, signs and symptoms, risk factors and prediction models of oxygen toxicity, and their limitations on SOF diving.
Collapse
Affiliation(s)
- Thijs T Wingelaar
- Diving Medical Center, Royal Netherlands NavyDen Helder, Netherlands.,Department of Anaesthesiology, Academic Medical CenterAmsterdam, Netherlands
| | | | - Rob A van Hulst
- Department of Anaesthesiology, Academic Medical CenterAmsterdam, Netherlands
| |
Collapse
|
12
|
Szyller J, Kozakiewicz M, Siermontowski P. The Influence of Hyperoxia On Heat Shock Proteins Expression and Nitric Oxide Synthase Activity – the Review. POLISH HYPERBARIC RESEARCH 2017. [DOI: 10.1515/phr-2017-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Any stay in an environment with an increased oxygen content (a higher oxygen partial pressure, pO2) and an increased pressure (hyperbaric conditions) leads to an intensification of oxidative stress. Reactive oxygen species (ROS) damage the molecules of proteins, nucleic acids, cause lipid oxidation and are engaged in the development of numerous diseases, including diseases of the circulatory system, neurodegenerative diseases, etc. There are certain mechanisms of protection against unfavourable effects of oxidative stress. Enzymatic and non-enzymatic systems belong to them. The latter include, among others, heat shock proteins (HSP). Their precise role and mechanism of action have been a subject of intensive research conducted in recent years. Hyperoxia and hyperbaria also have an effect on the expression and activity of nitrogen oxide synthase (NOS). Its product - nitrogen oxide (NO) can react with reactive oxygen species and contribute to the development of nitrosative stress. NOS occurs as isoforms in various tissues and exhibit different reactions to the discussed factors. The authors have prepared a brief review of research determining the effect of hyperoxia and hyperbaria on HSP expression and NOS activity.
Collapse
Affiliation(s)
- Jakub Szyller
- DiaLab Medical Laboratories Życzliwa 15-17, 50-001 Wrocław , Poland
| | - Mariusz Kozakiewicz
- Department and Institute of Foodstuff Chemistry, the L. Rydygier Collegium Medicum, Bydgoszcz , Poland
| | - Piotr Siermontowski
- Military Institute of Medicine, Department of Marine and Hyperbaric Medicine, Gdynia , Poland
| |
Collapse
|
13
|
Endesfelder S, Weichelt U, Strauß E, Schlör A, Sifringer M, Scheuer T, Bührer C, Schmitz T. Neuroprotection by Caffeine in Hyperoxia-Induced Neonatal Brain Injury. Int J Mol Sci 2017; 18:E187. [PMID: 28106777 PMCID: PMC5297819 DOI: 10.3390/ijms18010187] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/04/2017] [Accepted: 01/12/2017] [Indexed: 12/11/2022] Open
Abstract
Sequelae of prematurity triggered by oxidative stress and free radical-mediated tissue damage have coined the term "oxygen radical disease of prematurity". Caffeine, a potent free radical scavenger and adenosine receptor antagonist, reduces rates of brain damage in preterm infants. In the present study, we investigated the effects of caffeine on oxidative stress markers, anti-oxidative response, inflammation, redox-sensitive transcription factors, apoptosis, and extracellular matrix following the induction of hyperoxia in neonatal rats. The brain of a rat pups at postnatal Day 6 (P6) corresponds to that of a human fetal brain at 28-32 weeks gestation and the neonatal rat is an ideal model in which to investigate effects of oxidative stress and neuroprotection of caffeine on the developing brain. Six-day-old Wistar rats were pre-treated with caffeine and exposed to 80% oxygen for 24 and 48 h. Caffeine reduced oxidative stress marker (heme oxygenase-1, lipid peroxidation, hydrogen peroxide, and glutamate-cysteine ligase catalytic subunit (GCLC)), promoted anti-oxidative response (superoxide dismutase, peroxiredoxin 1, and sulfiredoxin 1), down-regulated pro-inflammatory cytokines, modulated redox-sensitive transcription factor expression (Nrf2/Keap1, and NFκB), reduced pro-apoptotic effectors (poly (ADP-ribose) polymerase-1 (PARP-1), apoptosis inducing factor (AIF), and caspase-3), and diminished extracellular matrix degeneration (matrix metalloproteinases (MMP) 2, and inhibitor of metalloproteinase (TIMP) 1/2). Our study affirms that caffeine is a pleiotropic neuroprotective drug in the developing brain due to its anti-oxidant, anti-inflammatory, and anti-apoptotic properties.
Collapse
Affiliation(s)
- Stefanie Endesfelder
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Ulrike Weichelt
- Department of Physiology, Charité, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Evelyn Strauß
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Anja Schlör
- Department of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany.
| | - Marco Sifringer
- Department of Anesthesiology and Intensive Care Medicine, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Till Scheuer
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Christoph Bührer
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Thomas Schmitz
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| |
Collapse
|
14
|
Topçu Y, Bayram E, Ozbal S, Yiş U, Tuğyan K, Karaoğlu P, Kumral A, Yılmaz O, Kurul SH. Zonisamide attenuates hyperoxia-induced apoptosis in the developing rat brain. Neurol Sci 2014; 35:1769-75. [PMID: 24916835 DOI: 10.1007/s10072-014-1834-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 05/13/2014] [Indexed: 11/29/2022]
Abstract
Oxygen therapy used in the treatment of perinatal hypoxia induces neurodegeneration in babies with immature antioxidant mechanisms. Zonisamide is a new antiepileptic drug used in childhood intractable seizures. Many studies demonstrated its neuroprotective effects. There is no study evaluating its effect on hyperoxic brain injury. The aim of this study was to investigate the neuroprotective effect of zonisamide on hyperoxia-induced neonatal brain injury. A total of 21 Wistar rat pups were used. The animals were divided into three groups: control group, hyperoxia group, and zonisamide-treated group. The zonisamide-treated group received an intraperitoneal injection of zonisamide. Zonisamide significantly preserved the number of neurons in CA1 and dentate gyrus parts of hippocampus, prefrontal, and parietal cortex. Zonisamide treatment also decreased the number of apoptotic neurons in all examined parts of hippocampus, prefrontal, and parietal cortex. We suggest that zonisamide treatment may be used as a neuroprotective agent in hyperoxic brain injury.
Collapse
Affiliation(s)
- Yasemin Topçu
- Division of Pediatric Neurology, Department of Pediatrics, Dokuz Eylul University Medical Faculty, İzmir, Turkey,
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
As recently as the year 2000, 100% oxygen was recommended to begin resuscitation of depressed newborns in the delivery room. However, the most recent recommendations of the International Liaison Committee on Resuscitation counsel the prudent use of oxygen during resuscitation. In term and preterm infants, oxygen therapy should be guided by pulse oximetry that follows the interquartile range of preductal saturations of healthy term babies after vaginal birth at sea level. This article reviews the literature in this context, which supports the radical but judicious curtailment of the use of oxygen in resuscitation at birth.
Collapse
Affiliation(s)
- Jay P Goldsmith
- Department of Pediatrics, Tulane University, 1430 Tulane Avenue, SL37, New Orleans, LA 70112, USA.
| | | |
Collapse
|
16
|
Brehmer F, Bendix I, Prager S, van de Looij Y, Reinboth BS, Zimmermanns J, Schlager GW, Brait D, Sifringer M, Endesfelder S, Sizonenko S, Mallard C, Bührer C, Felderhoff-Mueser U, Gerstner B. Interaction of inflammation and hyperoxia in a rat model of neonatal white matter damage. PLoS One 2012; 7:e49023. [PMID: 23155446 PMCID: PMC3498343 DOI: 10.1371/journal.pone.0049023] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 10/03/2012] [Indexed: 12/27/2022] Open
Abstract
Intrauterine infection and inflammation are major reasons for preterm birth. The switch from placenta-mediated to lung-mediated oxygen supply during birth is associated with a sudden rise of tissue oxygen tension that amounts to relative hyperoxia in preterm infants. Both infection/inflammation and hyperoxia have been shown to be involved in brain injury of preterm infants. Hypothesizing that they might be additive or synergistic, we investigated the influence of a systemic lipopolysaccharide (LPS) application on hyperoxia-induced white matter damage (WMD) in newborn rats. Three-day-old Wistar rat pups received 0.25 mg/kg LPS i.p. and were subjected to 80% oxygen on P6 for 24 h. The extent of WMD was assessed by immunohistochemistry, western blots, and diffusion tensor (DT) magnetic resonance imaging (MRI). In addition, the effects of LPS and hyperoxia were studied in an in vitro co-culture system of primary rat oligodendrocytes and microglia cells. Both noxious stimuli, hyperoxia, and LPS caused hypomyelination as revealed by western blot, immunohistochemistry, and altered WM microstructure on DT-MRI. Even so, cellular changes resulting in hypomyelination seem to be different. While hyperoxia induces cell death, LPS induces oligodendrocyte maturity arrest without cell death as revealed by TUNEL-staining and immunohistological maturation analysis. In the two-hit scenario cell death is reduced compared with hyperoxia treated animals, nevertheless white matter alterations persist. Concordantly with these in vivo findings we demonstrate that LPS pre-incubation reduced premyelinating-oligodendrocyte susceptibility towards hyperoxia in vitro. This protective effect might be caused by upregulation of interleukin-10 and superoxide dismutase expression after LPS stimulation. Reduced expression of transcription factors controlling oligodendrocyte development and maturation further indicates oligodendrocyte maturity arrest. The knowledge about mechanisms that triggered hypomyelination contributes to a better understanding of WMD in premature born infants.
Collapse
Affiliation(s)
- Felix Brehmer
- Department of Neonatology, Charité University Medical Center, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zaghloul N, Nasim M, Patel H, Codipilly C, Marambaud P, Dewey S, Schiffer WK, Ahmed M. Overexpression of extracellular superoxide dismutase has a protective role against hyperoxia-induced brain injury in neonatal mice. FEBS J 2012; 279:871-81. [PMID: 22240000 DOI: 10.1111/j.1742-4658.2012.08478.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is increasing evidence that hyperoxia, particularly at the time of birth, may result in neurological injury, in particular to the susceptible vasculature of these tissues. This study was aimed at determining whether overexpression of extracellular superoxide dismutase (EC-SOD) is protective against brain injury induced by hyperoxia. Transgenic (TG) mice (with an extra copy of the human extracellular superoxide dismutase gene) and wild-type (WT) neonate mice were exposed to hyperoxia (95% of F(i) o(2) ) for 7 days after birth versus the control group in room air. Brain positron emission tomography (PET) scanning with fludeoxyglucose (FDG) isotope uptake was performed after exposure. To assess apoptosis induced by hyperoxia exposure, caspase 3 ELISA and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining were performed. Quantitative western blot for the following inflammatory markers was performed: glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, macrophage-inhibiting factor, and phospho-AMP-activated protein kinase. PET scanning with FDG isotope uptake showed significantly higher uptake in the WT hyperoxia neonate brain group (0.14 ± 0.03) than in both the TG group (0.09 ± 0.01) and the control group (0.08 ± 0.02) (P< 0.05). Histopathological investigation showed more apoptosis and dead neurons in hippocampus and cerebellum brain sections of WT neonate mice after exposure to hyperoxia than in TG mice; this finding was also confirmed by TUNEL staining. The caspase 3 assay confirmed the finding of more apoptosis in WT hyperoxia neonates (0.814 ± 0.112) than in the TG hyperoxic group (0.579 ± 0.144) (P < 0.05); this finding was also confirmed by TUNEL staining. Quantitative western blotting for the inflammatory and metabolic markers showed significantly higher expression in the WT group than in the TG and control groups. Thus, overexpression of EC-SOD in the neonate brain offers significant protection against hyperoxia-induced brain damage.
Collapse
Affiliation(s)
- Nahla Zaghloul
- Department of Pediatrics, Cohen Children's Medical Center, NS-LIJ, Manhasset, NY 11030, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ferrari DC, Nesic O, Perez-Polo JR. Perspectives on neonatal hypoxia/ischemia-induced edema formation. Neurochem Res 2010; 35:1957-65. [PMID: 21136160 DOI: 10.1007/s11064-010-0308-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2010] [Indexed: 11/30/2022]
Abstract
Neonatal hypoxia/ischemia (HI) is the most common cause of developmental neurological, cognitive and behavioral deficits in children, with hyperoxia (HHI) treatment being a clinical therapy for newborn resuscitation. Although cerebral edema is a common outcome after HI, the mechanisms leading to excessive fluid accumulation in the brain are poorly understood. Given the rigid nature of the bone-encased brain matter, knowledge of edema formation in the brain as a consequence of any injury, as well as the importance of water clearance mechanisms and water and ion homeostasis is important to our understanding of its detrimental effects. Knowledge of the pathological process underlying the appearance of dysfunctional outcomes after development of cerebral edema after neonatal HI in the developing brain and the molecular events triggered will allow a rational assessment of HHI therapy for neonatal HI and determine whether this treatment is beneficial or harmful to the developing infant.
Collapse
|
19
|
Erythropoietin attenuates hyperoxia-induced oxidative stress in the developing rat brain. Brain Behav Immun 2010; 24:792-9. [PMID: 19729061 DOI: 10.1016/j.bbi.2009.08.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 08/27/2009] [Accepted: 08/27/2009] [Indexed: 11/23/2022] Open
Abstract
Oxygen toxicity contributes to the pathogenesis of adverse neurological outcome in survivors of preterm birth in clinical studies. In infant rodent brains, hyperoxia triggers widespread apoptotic neurodegeneration, induces pro-inflammatory cytokines and inhibits growth factor signaling cascades. Since a tissue-protective effect has been observed for recombinant erythropoietin (rEpo), we hypothesized that rEpo would influence hyperoxia-induced oxidative stress in the developing rat brain. The aim of this study was to investigate the level of glutathione (reduced and oxidized), lipid peroxidation and the expression of heme oxygenase-1 (HO-1) and acetylcholinesterase (AChE) after hyperoxia and rEpo treatment. Six-day-old Wistar rats were exposed to 80% oxygen for 2-48 h and received 20,000 IU/kg rEpo intraperitoneally (i.p.). Sex-matched littermates kept under room air and injected with normal saline or rEpo served as controls. Treatment with rEpo significantly reduced hyperoxia-induced upregulation of oxidized glutathione (GSSG) and malondialdehyde, a product of lipid breakdown, whereas reduced glutathione (GSH) was upregulated by rEpo. In parallel, hyperoxia-treated immature rat brains revealed rEpo-suppressible upregulation of synaptic AChE-S as well as of the stress-inducible AChE-R variant, together predicting rEpo-protected cholinergic signaling and restrained inflammatory reactions. Furthermore, treatment with rEpo induced upregulation of HO-1 on mRNA, protein and activity level in the developing rat brain. Our results suggest that rEpo generates its protective effect against oxygen toxicity by a reduction of diverse oxidative stress parameters and by limiting the stressor-inducible changes in both HO-1 and cholinergic functions.
Collapse
|
20
|
D'Agostino DP, Olson JE, Dean JB. Acute hyperoxia increases lipid peroxidation and induces plasma membrane blebbing in human U87 glioblastoma cells. Neuroscience 2009; 159:1011-22. [PMID: 19356685 DOI: 10.1016/j.neuroscience.2009.01.062] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 01/26/2009] [Accepted: 01/27/2009] [Indexed: 11/28/2022]
Abstract
Atomic force microscopy (AFM), malondialdehyde (MDA) assays, and amperometric measurements of extracellular hydrogen peroxide (H(2)O(2)) were used to test the hypothesis that graded hyperoxia induces measurable nanoscopic changes in membrane ultrastructure and membrane lipid peroxidation (MLP) in cultured U87 human glioma cells. U87 cells were exposed to 0.20 atmospheres absolute (ATA) O(2), normobaric hyperoxia (0.95 ATA O(2)) or hyperbaric hyperoxia (HBO(2), 3.25 ATA O(2)) for 60 min. H(2)O(2) (0.2 or 2 mM; 60 min) was used as a positive control for MLP. Cells were fixed with 2% glutaraldehyde immediately after treatment and scanned with AFM in air or fluid. Surface topography revealed ultrastructural changes such as membrane blebbing in cells treated with hyperoxia and H(2)O(2). Average membrane roughness (R(a)) of individual cells from each group (n=35 to 45 cells/group) was quantified to assess ultrastructural changes from oxidative stress. The R(a) of the plasma membrane was 34+/-3, 57+/-3 and 63+/-5 nm in 0.20 ATA O(2), 0.95 ATA O(2) and HBO(2), respectively. R(a) was 56+/-7 and 138+/-14 nm in 0.2 and 2 mM H(2)O(2). Similarly, levels of MDA were significantly elevated in cultures treated with hyperoxia and H(2)O(2) and correlated with O(2)-induced membrane blebbing (r(2)=0.93). Coapplication of antioxidant, Trolox-C (150 microM), significantly reduced membrane R(a) and MDA levels during hyperoxia. Hyperoxia-induced H(2)O(2) production increased 189%+/-5% (0.95 ATA O(2)) and 236%+/-5% (4 ATA O(2)) above control (0.20 ATA O(2)). We conclude that MLP and membrane blebbing increase with increasing O(2) concentration. We hypothesize that membrane blebbing is an ultrastructural correlate of MLP resulting from hyperoxia. Furthermore, AFM is a powerful technique for resolving nanoscopic changes in the plasma membrane that result from oxidative damage.
Collapse
Affiliation(s)
- D P D'Agostino
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, College of Medicine, MDC 8, University of South Florida, 12901 Bruce B. Downs Boulevard, Tampa, FL 33612, USA
| | | | | |
Collapse
|
21
|
Effect of hyperoxia on serine phosphorylation of apoptotic proteins in mitochondrial membranes of the cerebral cortex of newborn piglets. Neurochem Res 2009; 34:1219-25. [PMID: 19169818 DOI: 10.1007/s11064-008-9898-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2008] [Indexed: 10/21/2022]
Abstract
Previous studies have shown that hyperoxia results in cerebral cortical neuronal apoptosis. Studies have also shown that phosphorylation of anti-apoptotic proteins Bcl-2 and Bcl-xl results in loss of their anti-apoptotic potential leading to alteration in mitochondrial membrane permeability and the release of apoptogenic proteins in the neuronal cell of the newborn piglets. The present study tests the hypothesis that cerebral hyperoxia will result in increased serine phosphorylation of apoptotic proteins Bcl-2, Bcl-xl, Bax, and Bad in the mitochondrial membranes of the cerebral cortex of newborn piglets. Twelve newborn piglets were divided into normoxic (Nx, n = 6) exposed to an FiO(2) of 0.21 for 1 h and hyperoxic (Hyx, n = 6) exposed to FiO(2) of 1.0 for 1 h. In the Hyx group, PaO(2) was maintained above 400 mmHg while the Nx group was kept at 80-100 mmHg. Cerebral cortical tissue was harvested and mitochondrial fractions were isolated. Mitochondrial membrane proteins were separated using 12% SDS-PAGE, and probed with anti-serine phosphorylated Bcl-2, Bcl-xl, Bax, and Bad antibodies. Protein bands were detected, analyzed by imaging densitometry and density expressed as absorbance (OD x mm(2)). Phosphorylated Bcl-2 (p-Bcl-2) protein density (OD x mm(2)) was 81.81 +/- 9.24 in Nx and 158.34 +/- 10.66 in Hyx (P < 0.05). Phosphorylated Bcl-xl (p-Bcl-xl) protein density was 52.98 +/- 3.59 in Nx and 99.62 +/- 18.22 in Hyx (P < 0.05). Phosphorylated Bax (p-Bax) protein was 161.13 +/- 6.27 in Nx and 174.21 +/- 15.95 in Hyx (P = NS). Phosphorylated Bad (p-Bad) protein was 166.24 +/- 9.47 in Nx 155.38 +/- 12.32 in Hyx (P = NS). The data show that there is a significant increase in serine phosphorylation of Bcl-2 and Bcl-xl proteins while phosphorylation of Bad and Bax proteins were not altered during hyperoxia in the mitochondrial fraction of the cerebral cortex of newborn piglets. We conclude that hyperoxia results in differential post-translational modification of anti-apoptotic proteins Bcl-2 and Bcl-xl as compared to pro-apoptotic proteins Bax and Bad in mitochondria. We speculate that phosphorylation of Bcl-2 and Bcl-xl will result in loss of their anti-apoptotic potential by preventing their dimerization with Bax leading to activation of the caspase cascade of neuronal death.
Collapse
|
22
|
Dzietko M, Boos V, Sifringer M, Polley O, Gerstner B, Genz K, Endesfelder S, Börner C, Jacotot E, Chauvier D, Obladen M, Bührer C, Felderhoff-Mueser U. A critical role for Fas/CD-95 dependent signaling pathways in the pathogenesis of hyperoxia-induced brain injury. Ann Neurol 2009; 64:664-73. [PMID: 19107989 DOI: 10.1002/ana.21516] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Prematurely born infants are at risk for development of neurocognitive impairment in later life. Oxygen treatment has been recently identified as a trigger of neuronal and oligodendrocyte apoptosis in the developing rodent brain. We investigated the role of the Fas death receptor pathway in oxygen-triggered developmental brain injury. METHODS Six-day-old Wistar rats were exposed to 80% oxygen for various periods (2, 6, 12, 24, 48, and 72 hours), and mice deficient in either Fas (B6.MRL-Tnfrsf6(lpr)) or Fas ligand (B6Smn.C3-Fasl(gld)) and control mice (C57BL/6J) were exposed to 80% oxygen for 24 hours. Polymerase chain reaction, Western blotting, and caspase activity assays of thalamus and cortex tissue were performed. RESULTS Fas and Fas ligand messenger RNA and protein were upregulated. Furthermore, hyperoxia resulted in induction of downstream signaling events of Fas, such as Fas-associated death domain (FADD), the long and short form of FADD-like interleukin-1beta-converting enzyme (FLICE) inhibitory protein (FLIP-L, FLIP-S), and cleavage of caspase-8 and caspase-3. Injection of a selective caspase-8 inhibitor (TRP801, 1mg/kg) at the beginning of hyperoxia blocked subsequent caspase-3 cleavage in this model. B6.MRL-Tnfrsf6(lpr) mice were protected against oxygen-mediated injury, confirming Fas involvement in hyperoxia-induced cell death. Mice deficient in Fas ligand did not differ from control animals in the amount of cell death. INTERPRETATION We conclude that neonatal hyperoxia triggers Fas receptor and its downstream signaling events in a Fas ligand-independent fashion. Lack of functional Fas receptors and selective pharmacological inhibition of caspase-8 prevents activation of caspase-3 and provides significant neuroprotection.
Collapse
Affiliation(s)
- Mark Dzietko
- Department of Neonatology, Charité, Universitätsmedizin-Berlin, Campus Virchow Klinikum, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kaindl AM, Sifringer M, Koppelstaetter A, Genz K, Loeber R, Boerner C, Stuwe J, Klose J, Felderhoff-Mueser U. Erythropoietin protects the developing brain from hyperoxia-induced cell death and proteome changes. Ann Neurol 2009; 64:523-34. [PMID: 19067366 DOI: 10.1002/ana.21471] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Oxygen toxicity has been identified as a risk factor for adverse neurological outcome in survivors of preterm birth. In infant rodent brains, hyperoxia induces disseminated apoptotic neurodegeneration. Because a tissue-protective effect has been observed for recombinant erythropoietin (rEpo), widely used in neonatal medicine for its hematopoietic effect, we examined the effect of rEpo on hyperoxia-induced brain damage. METHODS Six-day-old C57Bl/6 mice or Wistar rats were exposed to hyperoxia (80% O(2)) or normoxia for 24 hours and received rEpo or normal saline injections intraperitoneally. The amount of degenerating cells in their brains was determined by DeOlmos cupric silver staining. Changes of their brain proteome were determined through two-dimensional electrophoresis and mass spectrometry. Western blot, enzyme activity assays and real-time polymerase chain reaction were performed for further analysis of candidate proteins. RESULTS Systemic treatment with 20,000 IE/kg rEpo significantly reduced hyperoxia-induced apoptosis and caspase-2, -3, and -8 activity in the brains of infant rodents. In parallel, rEpo inhibited most brain proteome changes observed in infant mice when hyperoxia was applied exclusively. Furthermore, brain proteome changes after a single systemic rEpo treatment point toward a number of mechanisms through which rEpo may generate its protective effect against oxygen toxicity. These include reduction of oxidative stress and restoration of hyperoxia-induced increased levels of proapoptotic factors, as well as decreased levels of neurotrophins. INTERPRETATION These findings are highly relevant from a clinical perspective because oxygen administration to neonates is often inevitable, and rEpo may serve as an adjunctive neuroprotective therapy.
Collapse
Affiliation(s)
- Angela M Kaindl
- Department of Pediatric Neurology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Refining the role of oxygen administration during delivery room resuscitation: what are the future goals? Semin Fetal Neonatal Med 2008; 13:368-74. [PMID: 18485848 DOI: 10.1016/j.siny.2008.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Oxygen was discovered more than 200 years ago and was thought to be both essential and beneficial for all animal life. Although it is now over 100 years since oxygen was first shown to damage biological tissues exposed to high concentrations, and more than 50 years since it was implicated in the aetiology of retinopathy of prematurity, the use of 100% oxygen was still recommended for the resuscitation of all babies at birth as recently as 2000. However, the 2005 International Liaison Committee on Resuscitation (ILCOR) recommendations allow for the initiation of resuscitation with concentrations of oxygen between 21 and 100%. There are strong arguments in favour of a radical curtailment of the use of oxygen in resuscitation at birth, and for devoting resources to defining the margins of safety for its use in the neonatal period in general.
Collapse
|
25
|
Hyperoxia causes maturation-dependent cell death in the developing white matter. J Neurosci 2008; 28:1236-45. [PMID: 18234901 DOI: 10.1523/jneurosci.3213-07.2008] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Periventricular leukomalacia is the predominant injury in the preterm infant leading to cerebral palsy. Oxygen exposure may be an additional cause of brain injury in these infants. In this study, we investigated pathways of maturation-dependent oligodendrocyte (OL) death induced by hyperoxia in vitro and in vivo. Developing and mature OLs were subjected to 80% oxygen (0-24 h). Lactate dehydrogenase (LDH) assay was used to assess cell viability. Furthermore, 3-, 6-, and 10-d-old rat pups were subjected to 80% oxygen (24 h), and their brains were processed for myelin basic protein staining. Significant cell death was detected after 6-24 h incubation in 80% oxygen in pre-OLs (O4+,O1-), but not in mature OLs (MBP+). Cell death was executed by a caspase-dependent apoptotic pathway and could be blocked by the pan-caspase inhibitor zVAD-fmk. Overexpression of BCL2 (Homo sapiens B-cell chronic lymphocytic leukemia/lymphoma 2) significantly reduced apoptosis. Accumulation of superoxide and generation of reactive oxygen species (ROS) were detected after 2 h of oxygen exposure. Lipoxygenase inhibitors 2,3,5-trimethyl-6-(12-hydroxy-5-10-dodecadiynyl-1,4-benzoquinone and N-benzyl-N-hydroxy-5-phenylpentamide fully protected the cells from oxidative injury. Overexpression of superoxide dismutase (SOD1) dramatically increased injury to pre-OLs but not to mature OLs. We extended these studies by testing the effects of hyperoxia on neonatal white matter. Postnatal day 3 (P3) and P6 rats, but not P10 pups, showed bilateral reduction in MBP (myelin basic protein) expression with 24 h exposure to 80% oxygen. Hyperoxia causes oxidative stress and triggers maturation-dependent apoptosis in pre-OLs, which involves the generation of ROS and caspase activation, and leads to white matter injury in the neonatal rat brain. These observations may be relevant to white matter injury observed in premature infants.
Collapse
|
26
|
Hoehn T, Janssen S, Mani AR, Brauers G, Moore KP, Schadewaldt P, Mayatepek E. Urinary excretion of the nitrotyrosine metabolite 3-nitro-4-hydroxyphenylacetic acid in preterm and term infants. Neonatology 2008; 93:73-6. [PMID: 17684421 DOI: 10.1159/000106783] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Accepted: 05/30/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND Newborn infants are exposed to various sources of oxidative and/or nitrative stress, which refers to either oxidation and/or nitration of endogenous proteins including loss of their original function. Nitrative stress is predominantly caused following synthesis of peroxynitrite. Particularly preterm infants with immature defense mechanisms against free radical injury appear at risk. OBJECTIVE To test the feasibility of quantifying the degradation products of the peroxynitrite marker nitrotyrosine [3-nitro-4-hydroxyphenylacetic acid (NHPA) and para-hydroxyphenylacetic acid (PHPA)] in neonatal urine samples. METHODS NHPA and PHPA were determined by gas chromatography/mass spectroscopy in urinary samples of preterm and term infants (mean gestational age = 28.4 and 39.6 weeks, respectively). RESULTS The urinary NHPA levels were lower in preterm infants in comparison with term infants. When the NHPA levels were adjusted to the urinary PHPA levels, no differences were found between the two groups. CONCLUSIONS Nitrotyrosine can be quantified in urinary samples of even the most immature infants. Nitration of endogenous PHPA in the gastrointestinal tract of term infants may have masked potentially higher levels of NHPA in preterm infants.
Collapse
Affiliation(s)
- Thomas Hoehn
- Neonatology and Paediatric Intensive Care Medicine, Department of General Paediatrics, Heinrich Heine University, Dusseldorf, Germany.
| | | | | | | | | | | | | |
Collapse
|
27
|
Sedowofia K, Giles D, Wade J, Cunningham S, McColm JR, Minns R, McIntosh N. Myelin expression is altered in the brains of neonatal rats reared in a fluctuating oxygen atmosphere. Neonatology 2008; 94:113-22. [PMID: 18332640 DOI: 10.1159/000119721] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 10/25/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND Preterm infants receiving supplemental oxygen therapy experience frequent fluctuations in their blood oxygen levels, the magnitude of which has been associated with the incidence and severity of retinopathy of prematurity in such infants. OBJECTIVE Our objective was to investigate in a relevant animal model whether the immature brain with its poorly vascularised white matter might also be susceptible to injury when exposed to such fluctuations in blood oxygen. METHODS Newborn rats were reared in an atmosphere in which a computer reproduced the oxygen fluctuations derived from the transcutaneous oxygen levels of a 24-week preterm infant who had developed severe retinopathy. Following 14 days of exposure, we measured the expression of active caspase-3, myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP) in the brains comparing with rat pups raised in room air. RESULTS Compared to room air controls, at day 14, the expression of active caspase-3 was increased by up to 162% (significant increase in 7 of 9 regions), MBP decreased by up to 70% (significant in the hypothalamus only) and GFAP increased by up to 103% (significant in 6 of 7 regions. On day 21, following 7 days of reparative recovery, GFAP levels in most areas of oxygen-exposed brains had returned to near control levels. There were no longer significant differences in caspase-3 levels apart from the cerebral cortex, cerebellum and striatum. In contrast, MBP expression was now much higher in most regions of the treated brains compared to controls. CONCLUSION We conclude that fluctuations in blood oxygen, observed in preterm survivors, may constitute a source of injury to the white matter and corpus striatum of the developing brain and contribute to the neurological sequelae in extremely premature infants.
Collapse
Affiliation(s)
- Kofi Sedowofia
- Child Life and Health Section, Division of Reproductive and Developmental Sciences, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | | |
Collapse
|
28
|
Groenendaal F, Vles J, Lammers H, De Vente J, Smit D, Nikkels PGJ. Nitrotyrosine in human neonatal spinal cord after perinatal asphyxia. Neonatology 2008; 93:1-6. [PMID: 17652956 DOI: 10.1159/000106432] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Accepted: 05/09/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND Spinal cord injury has been reported after perinatal asphyxia in full-term neonates. OBJECTIVES To examine the role of excessive nitric oxide production in perinatal spinal cord injury. SUBJECTS AND METHODS Tissue samples of 18 full-term neonates who died of hypoxic-ischemic encephalopathy were analyzed for the presence of nitrotyrosine (NT). RESULTS NT was demonstrated in 5 of these 18 neonates. In addition, activated caspase 3, a marker of apoptosis, and CD68, as a marker of inflammation, could be demonstrated in some infants. CONCLUSIONS excessive nitric oxide production and subsequent NT formation is seen in spinal cord tissue after severe perinatal asphyxia. This finding may be relevant for the development of neuroprotective strategies.
Collapse
Affiliation(s)
- Floris Groenendaal
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
29
|
Hoehn T, Gratopp A, Raehse K, Koehne P. Effects of hyperoxia and nitric oxide on endogenous nitric oxide production in polymorphonuclear leukocytes. Neonatology 2008; 94:132-7. [PMID: 18332642 DOI: 10.1159/000119723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Accepted: 11/21/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND Exposure to hyperoxia and nitric oxide (NO) occur frequently during the treatment of neonatal hypoxic pulmonary failure. OBJECTIVE The aim of the study was to quantify the endogenous synthesis of NO in neonatal polymorphonuclear neutrophils following exposure to hyperoxia and NO in vitro. METHODS Neonatal cord blood was exposed to room air, 25, 30 and 100% oxygen and 10 or 20 ppm NO added to the different oxygen concentrations for up to 30 min. 4,5-Diaminofluorescein diacetate (DAF-2 DA) is an intracellular dye used to measure real-time changes in NO levels in vivo. The molecular structure of DAF-2 DA changes upon contact with NO to its oxidized and fluorescent form diaminofluorescein-triazol (DAF-2T) and after being hydrolyzed by intracellular esterases cannot leave the cell. DAF-2 DA signals following equilibration with room air were used as controls. RESULTS Exposure to 100% oxygen increased NO production significantly when compared to 20 ppm NO plus 100% oxygen (p = 0.031) and to 20 ppm NO alone (p = 0.006). 10 ppm NO produced a similar effect. Significant increases in NO production were also noticed following exposure to 25% oxygen. This increase was already present after 10 min of oxygen exposure. CONCLUSION These findings support the propagated avoidance of hyperoxia not only in preterm infants, but also in term neonates.
Collapse
Affiliation(s)
- Thomas Hoehn
- Neonatology and Pediatric Intensive Care Medicine, Department of General Pediatrics, Heinrich Heine University, Dusseldorf, Germany.
| | | | | | | |
Collapse
|
30
|
Abstract
UNLABELLED Education in oxygenation and in how oxygen is given to newborns needs to increase. Treatment with oxygen should no longer be considered proverbial and customary, regardless of our 'past experience' or consensus recommendations in clinical guidelines, since oxygen may lead to acute or chronic health effects. CONCLUSION Inappropriate oxygen use is a neonatal health hazard associated with aging, DNA damage and cancer, retinopathy of prematurity, injury to the developing brain, infection and others. Neonatal exposure to pure O2, even if brief, or to pulse oximetry >95% when breathing supplemental O2 must be avoided as much as possible.
Collapse
Affiliation(s)
- Augusto Sola
- Mid Atlantic Neonatology Associates and Morristown Memorial Hospital, Morristown, NJ, USA.
| | | | | |
Collapse
|
31
|
Gerstner B, Bührer C, Rheinländer C, Polley O, Schüller A, Berns M, Obladen M, Felderhoff-Mueser U. Maturation-dependent oligodendrocyte apoptosis caused by hyperoxia. J Neurosci Res 2006; 84:306-15. [PMID: 16673399 DOI: 10.1002/jnr.20880] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In the immature human brain, periventricular leukomalacia (PVL) is the predominant white matter injury underlying the development of cerebral palsy. PVL has its peak incidence during a well-defined period in human brain development (23-32 weeks postconceptional age) characterized by extensive oligodendrocyte migration and maturation. We hypothesized that the dramatic rise of oxygen tissue tension associated with mammalian birth and additional oxygen exposure of the preterm infant during intensive care may be harmful to immature oligodendrocytes (OLs). We therefore investigated the effects of hyperoxia on rat oligodendroglia cells in vitro and in vivo. Immature OLs (OLN-93), their progenitors [preoligodendrocytes (pre-OL)], and mature OLs were subjected to 80% hyperoxia (24-96 hr). Flow cytometry was used to assess cell death. Cell viability was measured by metabolism of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium (MTT). In addition, 6-day-old rat pups were subjected to 80% oxygen (24 hr) and then sacrificed, and their brains were processed for immunfluorescence staining. Apoptosis was detected at various stages (annexin-V, activated caspase-3) after 24-48 hr of incubation in 80% oxygen in pre- and immature OLs. Mature OLs were resistant to oxygen exposure. These results were confirmed by MTT assay. This cell death was blocked by administration of the pan-caspase inhibitor zVAD-fmk. Degeneration of OLs was confirmed in 7-day-old rat brains by positive staining for activated caspase-3. Hyperoxia triggers maturation-dependent apoptosis in immature and pre-OLs and involves caspase activation. This mechanism may be relevant to the white matter injury observed in infants born preterm.
Collapse
Affiliation(s)
- Bettina Gerstner
- Department of Neonatology, Charité Campus Virchow-Klinikum, Berlin, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Richards EM, Rosenthal RE, Kristian T, Fiskum G. Postischemic hyperoxia reduces hippocampal pyruvate dehydrogenase activity. Free Radic Biol Med 2006; 40:1960-70. [PMID: 16716897 PMCID: PMC2570699 DOI: 10.1016/j.freeradbiomed.2006.01.022] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 01/20/2006] [Accepted: 01/23/2006] [Indexed: 01/04/2023]
Abstract
The pyruvate dehydrogenase complex (PDHC) is a mitochondrial matrix enzyme that catalyzes the oxidative decarboxylation of pyruvate and represents the sole bridge between anaerobic and aerobic cerebral energy metabolism. Previous studies demonstrating loss of PDHC enzyme activity and immunoreactivity during reperfusion after cerebral ischemia suggest that oxidative modifications are involved. This study tested the hypothesis that hyperoxic reperfusion exacerbates loss of PDHC enzyme activity, possibly due to tyrosine nitration or S-nitrosation. We used a clinically relevant canine ventricular fibrillation cardiac arrest model in which, after resuscitation and ventilation on either 100% O2 (hyperoxic) or 21-30% O2 (normoxic), animals were sacrificed at 2 h reperfusion and the brains removed for enzyme activity and immunoreactivity measurements. Animals resuscitated under hyperoxic conditions exhibited decreased PDHC activity and elevated 3-nitrotyrosine immunoreactivity in the hippocampus but not the cortex, compared to nonischemic controls. These measures were unchanged in normoxic animals. In vitro exposure of purified PDHC to peroxynitrite resulted in a dose-dependent loss of activity and increased nitrotyrosine immunoreactivity. These results support the hypothesis that oxidative stress contributes to loss of hippocampal PDHC activity during cerebral ischemia and reperfusion and suggest that PDHC is a target of peroxynitrite.
Collapse
Affiliation(s)
- Erica M. Richards
- Department of Anesthesiology, University of Maryland, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
| | - Robert E. Rosenthal
- Program in Trauma, Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland, Baltimore, MD 21201, USA
| | - Gary Fiskum
- Department of Anesthesiology, University of Maryland, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
- Corresponding author. Department of Anesthesiology, University of Maryland, Baltimore, MD 21201, USA. Fax: +1 410 706 2550. E-mail address: (G. Fiskum)
| |
Collapse
|
33
|
Zemlyak I, Nimon V, Brooke S, Moore T, McLaughlin J, Sapolsky R. Gene therapy in the nervous system with superoxide dismutase. Brain Res 2006; 1088:12-8. [PMID: 16630587 DOI: 10.1016/j.brainres.2006.02.109] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2005] [Revised: 10/20/2005] [Accepted: 02/26/2006] [Indexed: 01/23/2023]
Abstract
Neuronal death following necrotic insults involves the generation of reactive oxygen species (ROS). We investigated the effects of antioxidant gene therapy on ROS accumulation after exposure to either sodium cyanide, kainic acid or oxygen glucose deprivation (OGD). Specifically, we generated herpes simplex virus-1 amplicon vector expressing the gene for the antioxidant enzyme CuZnSOD. Overexpression of this gene in primary hippocampal cultures resulted in increased enzymatic activity of the corresponding protein. CuZnSOD significantly protected hippocampal neurons against sodium cyanide insult and the subsequent lipid peroxidation. However, it did not protect against OGD- or kainic-acid-induced toxicity. Moreover, CuZnSOD significantly worsened the toxicity, hydrogen peroxide accumulation and lipid peroxidation induced by kainic acid. As a possible explanation for this surprising worsening, CuZnSOD overexpression increased glutathione peroxidase activity in the presence of sodium cyanide but had no effect on catalase or glutathione peroxidase activity in the presence of kainic acid. Thus, cells were unlikely to be able to detoxify the excess hydrogen peroxide produced as a result of the CuZnSOD overexpression. These studies can be viewed as a cautionary note concerning gene therapy intervention against necrotic insults.
Collapse
Affiliation(s)
- Ilona Zemlyak
- Dept. of Biological Sciences, Stanford University, Gilbert Lab, MC 5020, Stanford, CA 94305-5020, USA
| | | | | | | | | | | |
Collapse
|
34
|
Seifert EL, Sant Anna GM, Rohlicek CV. Effect of body warming on regional blood flow distribution in conscious hypoxic one-month-old rabbits. Neonatology 2006; 90:104-12. [PMID: 16549907 DOI: 10.1159/000092069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Accepted: 10/27/2005] [Indexed: 11/19/2022]
Abstract
BACKGROUND Previous experiments have shown that warming hypoxic infants reduces total peripheral vascular resistance. This suggests that the usual vasoconstriction of less essential vascular beds during hypoxia may be reduced and that the normal redistribution of blood flow to more vital organs may be compromised. OBJECTIVE Evaluate the effect of body warming during hypoxia on the distribution of blood flow. METHODS The fluorescent microsphere technique was used to compare regional blood flow in 1-month-old rabbits during systemic hypoxia (10% inspired O2) with (n = 9) and without (n = 10) body warming. Blood flow was measured in brain, stomach, small intestine, hindlimb muscle, skin, and kidneys. Arterial blood pressure, whole-body O2 consumption, arterial blood O2 saturation and blood gases were also measured. MEASUREMENTS AND MAIN RESULTS In hypoxia all animals decreased body temperature (-2 degrees C). With hypoxia blood flow increased to brain and hindlimb muscle; decreased to stomach, small intestine, and kidneys, and was unchanged in skin. The increase in brain-blood flow maintained O2 delivery at normoxic levels. Rewarming to the normoxic body temperature significantly changed blood flow in hypoxia. Brain blood flow increased by 102 +/- 30% (mean +/- SEM) thereby increasing O2 delivery by 50 +/- 23% above normoxic values. Blood flow also increased to skin, stomach, and small intestine. However, O2 delivery to these tissues remained below normoxic levels. CONCLUSIONS Warming during hypoxia may impose an additional cardiovascular demand. The changes in the pattern of blood flow distribution with mild warming during hypoxia support the hypothesis that warming represents a significant heat stress.
Collapse
Affiliation(s)
- Erin L Seifert
- Montreal Children's Hospital, McGill UniversityResearch Institute, McGill University Health Centre, Montreal, Canada
| | | | | |
Collapse
|
35
|
Kaindl AM, Sifringer M, Zabel C, Nebrich G, Wacker MA, Felderhoff-Mueser U, Endesfelder S, von der Hagen M, Stefovska V, Klose J, Ikonomidou C. Acute and long-term proteome changes induced by oxidative stress in the developing brain. Cell Death Differ 2005; 13:1097-109. [PMID: 16254572 DOI: 10.1038/sj.cdd.4401796] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The developing mammalian brain experiences a period of rapid growth during which various otherwise innocuous environmental factors cause widespread apoptotic neuronal death. To gain insight into developmental events influenced by a premature exposure to high oxygen levels and identify proteins engaged in neurodegenerative and reparative processes, we analyzed mouse brain proteome changes at P7, P14 and P35 caused by an exposure to hyperoxia at P6. Changes detected in the brain proteome suggested that hyperoxia leads to oxidative stress and apoptotic neuronal death. These changes were consistent with results of histological and biochemical evaluation of the brains, which revealed widespread apoptotic neuronal death and increased levels of protein carbonyls. Furthermore, we detected changes in proteins involved in synaptic function, cell proliferation and formation of neuronal connections, suggesting interference of oxidative stress with these developmental events. These effects are age-dependent, as they did not occur in mice subjected to hyperoxia in adolescence.
Collapse
Affiliation(s)
- A M Kaindl
- Department of Pediatric Neurology, Charité, University Medical School, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Felderhoff-Mueser U, Sifringer M, Polley O, Dzietko M, Leineweber B, Mahler L, Baier M, Bittigau P, Obladen M, Ikonomidou C, Bührer C. Caspase-1-processed interleukins in hyperoxia-induced cell death in the developing brain. Ann Neurol 2004; 57:50-9. [PMID: 15622543 DOI: 10.1002/ana.20322] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Infants born prematurely may develop neurocognitive deficits without an obvious cause. Oxygen, which is widely used in neonatal medicine, constitutes one possible contributing neurotoxic factor, because it can trigger neuronal apoptosis in the developing brain of rodents. We hypothesized that two caspase-1-processed cytokines, interleukin (IL)-1beta and IL-18, are involved in oxygen-induced neuronal cell death. Six-day-old Wistar rats or C57/BL6 mice were exposed to 80% oxygen for various time periods (2, 6, 12, 24, and 48 hours). Neuronal cell death in the brain, as assessed by Fluoro-Jade B and silver staining, peaked at 12 to 24 hours and was preceded by a marked increase in mRNA and protein levels of caspase 1, IL-1beta, IL-18, and IL-18 receptor alpha (IL-18Ralpha). Intraperitoneal injection of recombinant human IL-18-binding protein, a specific inhibitor of IL-18, attenuated hyperoxic brain injury. Mice deficient in IL-1 receptor-associated kinase 4 (IRAK-4), which is pivotal for both IL-1beta and IL-18 signal transduction, were protected against oxygen-mediated neurotoxicity. These findings causally link IL-1beta and IL-18 to hyperoxia-induced cell death in the immature brain. These cytokines might serve as useful targets for therapeutic approaches aimed at preserving neuronal function in the immature brain, which is exquisitely sensitive to a variety of iatrogenic measures including oxygen.
Collapse
Affiliation(s)
- Ursula Felderhoff-Mueser
- Department of Neonatology, Charité, Campus Virchow Klinikum, Humboldt University Medical Center, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|