1
|
Li C, Jiang M, Chen Z, Hu Q, Liu Z, Wang J, Yin X, Wang J, Wu M. The neuroprotective effects of normobaric oxygen therapy after stroke. CNS Neurosci Ther 2024; 30:e14858. [PMID: 39009510 PMCID: PMC11250159 DOI: 10.1111/cns.14858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Stroke, including ischemic and hemorrhagic stroke, is a severe and prevalent acute cerebrovascular disease. The development of hypoxia following stroke can trigger a cascade of pathological events, including mitochondrial dysfunction, energy deficiency, oxidative stress, neuroinflammation, and excitotoxicity, all of which are often associated with unfavorable prognosis. Nonetheless, a noninvasive intervention, referred to as normobaric hyperoxia (NBO), is known to have neuroprotective effects against stroke. RESULTS NBO can exert neuroprotective effects through various mechanisms, such as the rescue of hypoxic tissues, preservation of the blood-brain barrier, reduction of brain edema, alleviation of neuroinflammation, improvement of mitochondrial function, mitigation of oxidative stress, reduction of excitotoxicity, and inhibition of apoptosis. These mechanisms may help improve the prognosis of stroke patients. CONCLUSIONS This review summarizes the mechanism by which hypoxia causes brain injury and how NBO can act as a neuroprotective therapy to treat stroke. We conclude that NBO has significant potential for treating stroke and may represent a novel therapeutic strategy.
Collapse
Affiliation(s)
- Chuan Li
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Min Jiang
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Zhiying Chen
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Qiongqiong Hu
- Department of NeurologyZhengzhou Central Hospital, Zhengzhou UniversityZhengzhouHenanChina
| | - Ziying Liu
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Junmin Wang
- Department of Human AnatomySchool of Basic Medical Sciences, Zhengzhou UniversityZhengzhouHenanChina
| | - Xiaoping Yin
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Jian Wang
- Department of Human AnatomySchool of Basic Medical Sciences, Zhengzhou UniversityZhengzhouHenanChina
| | - Moxin Wu
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| |
Collapse
|
2
|
Inguscio CR, Dalla Pozza E, Dando I, Boschi F, Tabaracci G, Angelini O, Picotti PM, Malatesta M, Cisterna B. Mitochondrial Features of Mouse Myoblasts Are Finely Tuned by Low Doses of Ozone: The Evidence In Vitro. Int J Mol Sci 2023; 24:ijms24108900. [PMID: 37240245 DOI: 10.3390/ijms24108900] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The mild oxidative stress induced by low doses of gaseous ozone (O3) activates the antioxidant cell response through the nuclear factor erythroid 2-related factor 2 (Nrf2), thus inducing beneficial effects without cell damage. Mitochondria are sensitive to mild oxidative stress and represent a susceptible O3 target. In this in vitro study, we investigated the mitochondrial response to low O3 doses in the immortalized, non-tumoral muscle C2C12 cells; a multimodal approach including fluorescence microscopy, transmission electron microscopy and biochemistry was used. Results demonstrated that mitochondrial features are finely tuned by low O3 doses. The O3 concentration of 10 μg maintained normal levels of mitochondria-associated Nrf2, promoted the mitochondrial increase of size and cristae extension, reduced cellular reactive oxygen species (ROS) and prevented cell death. Conversely, in 20 μg O3-treated cells, where the association of Nrf2 with the mitochondria drastically dropped, mitochondria underwent more significant swelling, and ROS and cell death increased. This study, therefore, adds original evidence for the involvement of Nrf2 in the dose-dependent response to low O3 concentrations not only as an Antioxidant Response Elements (ARE) gene activator but also as a regulatory/protective factor of mitochondrial function.
Collapse
Affiliation(s)
- Chiara Rita Inguscio
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, I-37134 Verona, Italy
| | - Elisa Dalla Pozza
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, I-37134 Verona, Italy
| | - Ilaria Dando
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, I-37134 Verona, Italy
| | - Federico Boschi
- Department of Engineering for Innovation Medicine, University of Verona, I-37134 Verona, Italy
| | | | | | | | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, I-37134 Verona, Italy
| | - Barbara Cisterna
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, I-37134 Verona, Italy
| |
Collapse
|
3
|
Kandasamy J, Li R, Vamesu BM, Olave N, Halloran B, Jilling T, Ballinger SW, Ambalavanan N. Mitochondrial DNA Variations Modulate Alveolar Epithelial Mitochondrial Function and Oxidative Stress in Newborn Mice Exposed to Hyperoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541177. [PMID: 37292719 PMCID: PMC10245974 DOI: 10.1101/2023.05.17.541177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Oxidative stress is an important contributor to bronchopulmonary dysplasia (BPD), a form of chronic lung disease that is the most common morbidity in very preterm infants. Mitochondrial functional differences due to inherited and acquired mutations influence the pathogenesis of disorders in which oxidative stress plays a critical role. We previously showed using mitochondrial-nuclear exchange (MNX) mice that mitochondrial DNA (mtDNA) variations modulate hyperoxia-induced lung injury severity in a model of BPD. In this study, we studied the effects of mtDNA variations on mitochondrial function including mitophagy in alveolar epithelial cells (AT2) from MNX mice. We also investigated oxidant and inflammatory stress as well as transcriptomic profiles in lung tissue in mice and expression of proteins such as PINK1, Parkin and SIRT3 in infants with BPD. Our results indicate that AT2 from mice with C57 mtDNA had decreased mitochondrial bioenergetic function and inner membrane potential, increased mitochondrial membrane permeability and were exposed to higher levels of oxidant stress during hyperoxia compared to AT2 from mice with C3H mtDNA. Lungs from hyperoxia-exposed mice with C57 mtDNA also had higher levels of pro-inflammatory cytokines compared to lungs from mice with C3H mtDNA. We also noted changes in KEGG pathways related to inflammation, PPAR and glutamatergic signaling, and mitophagy in mice with certain mito-nuclear combinations but not others. Mitophagy was decreased by hyperoxia in all mice strains, but to a greater degree in AT2 and neonatal mice lung fibroblasts from hyperoxia-exposed mice with C57 mtDNA compared to C3H mtDNA. Finally, mtDNA haplogroups vary with ethnicity, and Black infants with BPD had lower levels of PINK1, Parkin and SIRT3 expression in HUVEC at birth and tracheal aspirates at 28 days of life when compared to White infants with BPD. These results indicate that predisposition to neonatal lung injury may be modulated by variations in mtDNA and mito-nuclear interactions need to be investigated to discover novel pathogenic mechanisms for BPD.
Collapse
|
4
|
Alva R, Mirza M, Baiton A, Lazuran L, Samokysh L, Bobinski A, Cowan C, Jaimon A, Obioru D, Al Makhoul T, Stuart JA. Oxygen toxicity: cellular mechanisms in normobaric hyperoxia. Cell Biol Toxicol 2022; 39:111-143. [PMID: 36112262 PMCID: PMC9483325 DOI: 10.1007/s10565-022-09773-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/07/2022] [Indexed: 12/15/2022]
Abstract
In clinical settings, oxygen therapy is administered to preterm neonates and to adults with acute and chronic conditions such as COVID-19, pulmonary fibrosis, sepsis, cardiac arrest, carbon monoxide poisoning, and acute heart failure. In non-clinical settings, divers and astronauts may also receive supplemental oxygen. In addition, under current standard cell culture practices, cells are maintained in atmospheric oxygen, which is several times higher than what most cells experience in vivo. In all the above scenarios, the elevated oxygen levels (hyperoxia) can lead to increased production of reactive oxygen species from mitochondria, NADPH oxidases, and other sources. This can cause cell dysfunction or death. Acute hyperoxia injury impairs various cellular functions, manifesting ultimately as physiological deficits. Chronic hyperoxia, particularly in the neonate, can disrupt development, leading to permanent deficiencies. In this review, we discuss the cellular activities and pathways affected by hyperoxia, as well as strategies that have been developed to ameliorate injury.
Collapse
Affiliation(s)
- Ricardo Alva
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Maha Mirza
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Adam Baiton
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Lucas Lazuran
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Lyuda Samokysh
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Ava Bobinski
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Cale Cowan
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Alvin Jaimon
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Dede Obioru
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Tala Al Makhoul
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Jeffrey A Stuart
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
5
|
Hong JY, Kim MN, Kim EG, Lee JW, Kim HR, Kim SY, Lee SM, Kim YH, Kim KW, Sohn MH. Clusterin Deficiency Exacerbates Hyperoxia-Induced Acute Lung Injury. Cells 2021; 10:944. [PMID: 33921872 PMCID: PMC8073575 DOI: 10.3390/cells10040944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/07/2021] [Accepted: 04/16/2021] [Indexed: 02/05/2023] Open
Abstract
Exposure to high oxygen concentrations leads to generation of excessive reactive oxygen species, causing cellular injury and multiple organ dysfunctions and is associated with a high mortality rate. Clusterin (CLU) is a heterodimeric glycoprotein that mediates several intracellular signaling pathways, including cell death and inflammation. However, the role of CLU in the pathogenesis of hyperoxic acute lung injury (HALI) is unknown. Wild-type (WT) and CLU-deficient mice and cultured human airway epithelial cells were used. Changes in cell death- and inflammation-related molecules with or without hyperoxia exposure in cells and animals were determined. Hyperoxia induced an increase in CLU expression in mouse lungs and human airway epithelial cells. Mice lacking CLU had increased HALI and mortality rate compared with WT mice. In vitro, CLU-disrupted cells showed enhanced release of cytochrome c, Bax translocation, cell death and inflammatory cytokine expression. However, treatment with recombinant CLU attenuated hyperoxia-induced apoptosis. Moreover, the Kyoto Encyclopedia of Genes and Genomes and Gene Ontology analyses revealed metabolic pathways, hematopoietic cell lineage, response to stress and localization and regulation of immune system that were differentially regulated between WT and CLU-/- mice. These results demonstrate that prolonged hyperoxia-induced lung injury is associated with CLU expression and that CLU replenishment may alleviate hyperoxia-induced cell death.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Myung Hyun Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine,134 Sinchon-Dong, Seoul 03722, Korea; (J.Y.H.); (M.N.K.); (E.G.K.); (J.W.L.); (H.R.K.); (S.Y.K.); (S.M.L.); (Y.H.K.); (K.W.K.)
| |
Collapse
|
6
|
Mitochondrial Dysfunction and Permeability Transition in Neonatal Brain and Lung Injuries. Cells 2021; 10:cells10030569. [PMID: 33807810 PMCID: PMC7999701 DOI: 10.3390/cells10030569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 01/16/2023] Open
Abstract
This review discusses the potential mechanistic role of abnormally elevated mitochondrial proton leak and mitochondrial bioenergetic dysfunction in the pathogenesis of neonatal brain and lung injuries associated with premature birth. Providing supporting evidence, we hypothesized that mitochondrial dysfunction contributes to postnatal alveolar developmental arrest in bronchopulmonary dysplasia (BPD) and cerebral myelination failure in diffuse white matter injury (WMI). This review also analyzes data on mitochondrial dysfunction triggered by activation of mitochondrial permeability transition pore(s) (mPTP) during the evolution of perinatal hypoxic-ischemic encephalopathy. While the still cryptic molecular identity of mPTP continues to be a subject for extensive basic science research efforts, the translational significance of mitochondrial proton leak received less scientific attention, especially in diseases of the developing organs. This review is focused on the potential mechanistic relevance of mPTP and mitochondrial dysfunction to neonatal diseases driven by developmental failure of organ maturation or by acute ischemia-reperfusion insult during development.
Collapse
|
7
|
Sidramagowda Patil S, Hernández-Cuervo H, Fukumoto J, Krishnamurthy S, Lin M, Alleyn M, Breitzig M, Narala VR, Soundararajan R, Lockey RF, Kolliputi N, Galam L. Alda-1 Attenuates Hyperoxia-Induced Acute Lung Injury in Mice. Front Pharmacol 2021; 11:597942. [PMID: 33597876 PMCID: PMC7883597 DOI: 10.3389/fphar.2020.597942] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022] Open
Abstract
Acute lung injury (ALI), a milder form of acute respiratory distress syndrome (ARDS), is a leading cause of mortality in older adults with an increasing prevalence. Oxygen therapy, is a common treatment for ALI, involving exposure to a high concentration of oxygen. Unfortunately, hyperoxia induces the formation of reactive oxygen species which can cause an increase in 4-HNE (4-hydroxy 2 nonenal), a toxic byproduct of lipid peroxidation. Mitochondrial aldehyde dehydrogenase 2 (ALDH2) serves as an endogenous shield against oxidative stress-mediated damage by clearing 4-HNE. Alda-1 [(N-(1, 3 benzodioxol-5-ylmethyl)-2, 6- dichloro-benzamide)], a small molecular activator of ALDH2, protects against reactive oxygen species-mediated oxidative stress by promoting ALDH2 activity. As a result, Alda-1 shields against ischemic reperfusion injury, heart failure, stroke, and myocardial infarction. However, the mechanisms of Alda-1 in hyperoxia-induced ALI remains unclear. C57BL/6 mice implanted with Alzet pumps received Alda-1 in a sustained fashion while being exposed to hyperoxia for 48 h. The mice displayed suppressed immune cell infiltration, decreased protein leakage and alveolar permeability compared to controls. Mechanistic analysis shows that mice pretreated with Alda-1 also experience decreased oxidative stress and enhanced levels of p-Akt and mTOR pathway associated proteins. These results show that continuous delivery of Alda-1 protects against hyperoxia-induced lung injury in mice.
Collapse
Affiliation(s)
- Sahebgowda Sidramagowda Patil
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Helena Hernández-Cuervo
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jutaro Fukumoto
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sudarshan Krishnamurthy
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Muling Lin
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Matthew Alleyn
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mason Breitzig
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Brown School, Washington University, St. Louis, MO, United States
| | | | - Ramani Soundararajan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Richard F Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Lakshmi Galam
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
8
|
Zou L, Linck V, Zhai YJ, Galarza-Paez L, Li L, Yue Q, Al-Khalili O, Bao HF, Ma HP, Thai TL, Jiao J, Eaton DC. Knockout of mitochondrial voltage-dependent anion channel type 3 increases reactive oxygen species (ROS) levels and alters renal sodium transport. J Biol Chem 2017; 293:1666-1675. [PMID: 29180450 DOI: 10.1074/jbc.m117.798645] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/27/2017] [Indexed: 11/06/2022] Open
Abstract
It has been suggested that voltage-dependent anion channels (VDACs) control the release of superoxide from mitochondria. We have previously shown that reactive oxygen species (ROS) such as superoxide (O2̇̄) and hydrogen peroxide (H2O2) stimulate epithelial sodium channels (ENaCs) in sodium-transporting epithelial tissue, including cortical collecting duct (CCD) principal cells. Therefore, we hypothesized that VDACs could regulate ENaC by modulating cytosolic ROS levels. Herein, we find that VDAC3-knockout(KO) mice can maintain normal salt and water balance on low-salt and high-salt diets. However, on a high-salt diet for 2 weeks, VDAC3-KO mice had significantly higher systolic blood pressure than wildtype mice. Consistent with this observation, after a high-salt diet for 2 weeks, ENaC activity in VDAC3-KO mice was significantly higher than wildtype mice. EM analysis disclosed a significant morphological change of mitochondria in the CCD cells of VDAC3-KO mice compared with wildtype mice, which may have been caused by mitochondrial superoxide overload. Of note, compared with wildtype animals, ROS levels in VDAC3-KO animals fed a normal or high-salt diet were consistently and significantly increased in renal tubules. Both the ROS scavenger 1-oxyl-2,2,6,6-tetramethyl-4-hydroxypiperidine (TEMPOL) and the mitochondrial ROS scavenger (2-(2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mito-TEMPO) could reverse the effect of high-salt on ENaC activity and systolic blood pressure in the VDAC3-KO mice. Mito-TEMPO partially correct the morphological changes in VDAC3-KO mice. Our results suggest that knocking out mitochondrial VDAC3 increases ROS, alters renal sodium transport, and leads to hypertension.
Collapse
Affiliation(s)
- Li Zou
- From the Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China.,the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Valerie Linck
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Yu-Jia Zhai
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Laura Galarza-Paez
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Linda Li
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Qiang Yue
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Otor Al-Khalili
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Hui-Fang Bao
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - He-Ping Ma
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Tiffany L Thai
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Jundong Jiao
- From the Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China, .,the Institute of Nephrology, Harbin Medical University, Harbin, China
| | - Douglas C Eaton
- the Department of Physiology and the Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia 30322, and
| |
Collapse
|
9
|
Wang Z, Zhou L, Zheng X, Chen G, Pan R, Li J, Liu W. Autophagy protects against PI3K/Akt/mTOR-mediated apoptosis of spinal cord neurons after mechanical injury. Neurosci Lett 2017; 656:158-164. [DOI: 10.1016/j.neulet.2017.07.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/25/2017] [Accepted: 07/20/2017] [Indexed: 12/18/2022]
|
10
|
Kuhn H, Nieuwenhuijsen H, Karthe B, Wirtz H. Stretch-induced apoptosis in rat alveolar epithelial cells is mediated by the intrinsic mitochondrial pathway. Exp Lung Res 2017; 43:49-56. [DOI: 10.1080/01902148.2017.1287228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Hartmut Kuhn
- Department of Respiratory Medicine, University of Leipzig, Leipzig, Germany
| | | | - Bianca Karthe
- Department of Respiratory Medicine, University of Leipzig, Leipzig, Germany
| | - Hubert Wirtz
- Department of Respiratory Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
11
|
Huang R, Zhao L, Chen H, Yin RH, Li CY, Zhan YQ, Zhang JH, Ge CH, Yu M, Yang XM. Megakaryocytic differentiation of K562 cells induced by PMA reduced the activity of respiratory chain complex IV. PLoS One 2014; 9:e96246. [PMID: 24817082 PMCID: PMC4015910 DOI: 10.1371/journal.pone.0096246] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 04/06/2014] [Indexed: 01/14/2023] Open
Abstract
Mitochondria are involved in the regulation of cell differentiation processes, but its function changes and molecular mechanisms are not yet clear. In this study, we found that mitochondrial functions changed obviously when K562 cells were induced to megakaryocytic differentiation by phorbol 12-myristate 13-acetate (PMA). During the cell differentiation, the reactive oxygen species (ROS) level was increased, mitochondrial membrane potential declined and respiratory chain complex IV activity was decreased. Treatment with specific inhibitor of mitochondrial respiratory chain complex IV led to a significant inhibition in mitochondrial membrane potential and reduction of PMA-induced cell differentiation. However, treatment with cyclosporine A, a stabilization reagent of mitochondrial membrane potential, did not improve the down-regulation of mitochondrial respiratory chain complex IV induced by PMA. Furthermore, we found that the level of the complex IV core subunit COX3 and mitochondrial transport-related proteins Tim9 and Tim10 were decreased during the differentiation of K562 cells induced by PMA, suggesting an important role of these factors in mitochondrial functional changes. Our results suggest that changes in mitochondrial functions are involved in the PMA-induced K562 cell differentiation process, and the maintenance of the steady-state of mitochondrial functions plays a critical role in the regulation of cell differentiation.
Collapse
Affiliation(s)
- Rui Huang
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Long Zhao
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Chen
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Rong-Hua Yin
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chang-Yan Li
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yi-Qun Zhan
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jian-Hong Zhang
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chang-hui Ge
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Miao Yu
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiao-Ming Yang
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
12
|
Sturrock A, Seedahmed E, Mir-Kasimov M, Boltax J, McManus ML, Paine R. GM-CSF provides autocrine protection for murine alveolar epithelial cells from oxidant-induced mitochondrial injury. Am J Physiol Lung Cell Mol Physiol 2011; 302:L343-51. [PMID: 22140071 DOI: 10.1152/ajplung.00276.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Exposure of mice to hyperoxia induces alveolar epithelial cell (AEC) injury, acute lung injury and death. Overexpression of granulocyte-macrophage colony-stimulating factor (GM-CSF) in the lung protects against these effects, although the mechanisms are not yet clear. Hyperoxia induces cellular injury via effects on mitochondrial integrity, associated with induction of proapoptotic members of the Bcl-2 family. We hypothesized that GM-CSF protects AEC through effects on mitochondrial integrity. MLE-12 cells (a murine type II cell line) and primary murine type II AEC were subjected to oxidative stress by exposure to 80% oxygen and by exposure to H(2)O(2). Exposure to H(2)O(2) induced cytochrome c release and decreased mitochondrial reductase activity in MLE-12 cells. Incubation with GM-CSF significantly attenuated these effects. Protection induced by GM-CSF was associated with Akt activation. GM-CSF treatment also resulted in increased expression of the antiapoptotic Bcl-2 family member, Mcl-1. Primary murine AEC were significantly more tolerant of oxidative stress than MLE-12 cells. In contrast to MLE-12 cells, primary AEC expressed significant GM-CSF at baseline and demonstrated constitutive activation of Akt and increased baseline expression of Mcl-1. Treatment with exogenous GM-CSF further increased Akt activation and Mcl-1 expression in primary AEC. Conversely, suppression of AEC GM-CSF expression by use of GM-CSF-specific small interfering RNA resulted in decreased tolerance of oxidative stress, Furthermore, silencing of Mcl-1 prevented GM-CSF-induced protection. We conclude that GM-CSF protects alveolar epithelial cells against oxidative stress-induced mitochondrial injury via the Akt pathway and its downstream components, including Mcl-1. Epithelial cell-derived GM-CSF may contribute to intrinsic defense mechanisms limiting lung injury.
Collapse
Affiliation(s)
- Anne Sturrock
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Univ. of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | | | | | | | | | |
Collapse
|
13
|
Tuder RM, Hunt JM, Schmidt EP. Hyperoxia and apoptosis. Too much of a good thing? Am J Respir Crit Care Med 2011; 183:964-5. [PMID: 21498818 DOI: 10.1164/rccm.201010-1756ed] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
14
|
Métrailler-Ruchonnet I, Pagano A, Carnesecchi S, Khatib K, Herrera P, Donati Y, Bron C, Barazzone C. Bcl-2 overexpression in type II epithelial cells does not prevent hyperoxia-induced acute lung injury in mice. Am J Physiol Lung Cell Mol Physiol 2010; 299:L312-22. [DOI: 10.1152/ajplung.00212.2009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Bcl-2 is an anti-apoptotic molecule preventing oxidative stress damage and cell death. We have previously shown that Bcl-2 is able to prevent hyperoxia-induced cell death when overexpressed in a murine fibrosarcoma cell line L929. We hypothesized that its specific overexpression in pulmonary epithelial type II cells could prevent hyperoxia-induced lung injury by protecting the epithelial side of the alveolo-capillary barrier. In the present work, we first showed that in vitro Bcl-2 can rescue murine pulmonary epithelial cells (MLE12) from oxygen-induced cell apoptosis, as shown by analysis of LDH release, annexin V/propidium staining, and caspase-3 activity. We then generated transgenic mice overexpressing specifically Bcl-2 in lung epithelial type II cells under surfactant protein C (SP-C) promoter (Tg-Bcl-2) and exposed them to hyperoxia. Bcl-2 did not hinder hyperoxia-induced mitochondria and DNA oxidative damage of type II cell in vivo. Accordingly, lung damage was identical in both Tg-Bcl-2 and littermate mice strains, as measured by lung weight, bronchoalveolar lavage, and protein content. Nevertheless, we observed a significant lower number of TUNEL-positive cells in type II cells isolated from Tg-Bcl-2 mice exposed to hyperoxia compared with cells isolated from littermate mice. In summary, these results show that although Bcl-2 overexpression is able to prevent hyperoxia-induced cell death at single cell level in vitro and ex vivo, it is not sufficient to prevent cell death of parenchymal cells and to protect the lung from acute damage in mice.
Collapse
Affiliation(s)
| | - Alessandra Pagano
- Institut National de la Santé et de la Recherche Médicale UMR 911, Centre de Recherche en Oncologie biologique et en Oncopharmacologie, Université Aix-Marseille, France; and
| | - Stéphanie Carnesecchi
- Departments of 1Pediatrics and
- Pathology-Immunology, Medical School, University of Geneva, Switzerland
| | - Karim Khatib
- Pathology-Immunology, Medical School, University of Geneva, Switzerland
| | - Pedro Herrera
- Department of Genetic Medicine and Development, Medical School, University of Geneva, Switzerland
| | - Yves Donati
- Departments of 1Pediatrics and
- Pathology-Immunology, Medical School, University of Geneva, Switzerland
| | - Camille Bron
- Departments of 1Pediatrics and
- Pathology-Immunology, Medical School, University of Geneva, Switzerland
| | - Constance Barazzone
- Departments of 1Pediatrics and
- Pathology-Immunology, Medical School, University of Geneva, Switzerland
| |
Collapse
|
15
|
Tyurina YY, Tyurin VA, Kaynar AM, Kapralova VI, Wasserloos K, Li J, Mosher M, Wright L, Wipf P, Watkins S, Pitt BR, Kagan VE. Oxidative lipidomics of hyperoxic acute lung injury: mass spectrometric characterization of cardiolipin and phosphatidylserine peroxidation. Am J Physiol Lung Cell Mol Physiol 2010; 299:L73-85. [PMID: 20418384 DOI: 10.1152/ajplung.00035.2010] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Reactive oxygen species have been shown to play a significant role in hyperoxia-induced acute lung injury, in part, by inducing apoptosis of pulmonary endothelium. However, the signaling roles of phospholipid oxidation products in pulmonary endothelial apoptosis have not been studied. Using an oxidative lipidomics approach, we identified individual molecular species of phospholipids involved in the apoptosis-associated peroxidation process in a hyperoxic lung. C57BL/6 mice were killed 72 h after exposure to hyperoxia (100% oxygen). We found that hyperoxia-induced apoptosis (documented by activation of caspase-3 and -7 and histochemical terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling staining of pulmonary endothelium) was accompanied by nonrandom oxidation of pulmonary lipids. Two anionic phospholipids, mitochondria-specific cardiolipin (CL) and extramitochondrial phosphatidylserine (PS), were the two major oxidized phospholipids in hyperoxic lung. Using electrospray ionization mass spectrometry, we identified several oxygenation products in CL and PS. Quantitative assessments revealed a significant decrease of CL and PS molecular species containing C(18:2), C(20:4), C(22:5), and C(22:6) fatty acids. Similarly, exposure of mouse pulmonary endothelial cells (MLEC) to hyperoxia (95% oxygen; 72 h) resulted in activation of caspase-3 and -7 and significantly decreased the content of CL molecular species containing C(18:2) and C(20:4) as well as PS molecular species containing C(22:5) and C(22:6). Oxygenated molecular species were found in the same two anionic phospholipids, CL and PS, in MLEC exposed to hyperoxia. Treatment of MLEC with a mitochondria-targeted radical scavenger, a conjugate of hemi-gramicidin S with nitroxide, XJB-5-131, resulted in significantly lower oxidation of both CL and PS and a decrease in hyperoxia-induced changes in caspase-3 and -7 activation. We speculate that cytochrome c driven oxidation of CL and PS is associated with the signaling role of these oxygenated species participating in the execution of apoptosis and clearance of pulmonary endothelial cells, thus contributing to hyperoxic lung injury.
Collapse
Affiliation(s)
- Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Dept. of Environmental and Occupational Health, Univ. of Pittsburgh, Bridgeside Point, 100 Technology Drive, Suite 350, Pittsburgh, PA 15219, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
There are many organelles within a cell, each with individual responsibilities required for life. Of these organelles, the mitochondria are the hemi of the cell, producing the energy necessary for cell function. Reactive oxygen species can cause mitochondrial dysfunction and contribute to many diseases often seen in emergency departments. When reactive oxygen species are produced, the mitochondria undergo functional and structural changes causing the release of cytochrome c. Cytochrome c is responsible for activating apoptotic pathways leading to cell death. Apoptosis, or programmed cell death, is needed to maintain homeostasis in the body; however, when this occurs prematurely by an increase in reactive oxygen species production, many pathological conditions can occur. Clinicians in emergency departments caring for patients with different diseases should consider that the mitochondria may play an important role in patients' recovery. For instance, myocardial infarctions and burns are two examples of altered physiologic states that play a role in mitochondrial dysfunction. Awareness of the different treatments that target the mitochondria will prepare emergency department clinicians to better care for their patients.
Collapse
|
17
|
Carnesecchi S, Deffert C, Pagano A, Garrido-Urbani S, Métrailler-Ruchonnet I, Schäppi M, Donati Y, Matthay MA, Krause KH, Barazzone Argiroffo C. NADPH oxidase-1 plays a crucial role in hyperoxia-induced acute lung injury in mice. Am J Respir Crit Care Med 2009; 180:972-81. [PMID: 19661248 PMCID: PMC2778156 DOI: 10.1164/rccm.200902-0296oc] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 08/05/2009] [Indexed: 02/07/2023] Open
Abstract
RATIONALE Hyperoxia-induced acute lung injury has been used for many years as a model of oxidative stress mimicking clinical acute lung injury and the acute respiratory distress syndrome. Excess quantities of reactive oxygen species (ROS) are responsible for oxidative stress-induced lung injury. ROS are produced by mitochondrial chain transport, but also by NADPH oxidase (NOX) family members. Although NOX1 and NOX2 are expressed in the lungs, their precise function has not been determined until now. OBJECTIVES To determine whether NOX1 and NOX2 contribute in vivo to hyperoxia-induced acute lung injury. METHODS Wild-type and NOX1- and NOX2-deficient mice, as well as primary lung epithelial and endothelial cells, were exposed to room air or 100% O(2) for 72 hours. MEASUREMENTS AND MAIN RESULTS Lung injury was significantly prevented in NOX1-deficient mice, but not in NOX2-deficient mice. Hyperoxia-dependent ROS production was strongly reduced in lung sections, in isolated epithelial type II cells, and lung endothelial cells from NOX1-deficient mice. Concomitantly, lung cell death in situ and in primary cells was markedly decreased in NOX1-deficient mice. In wild-type mice, hyperoxia led to phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK), two mitogen-activated protein kinases involved in cell death signaling, and to caspase-3 activation. In NOX1-deficient mice, JNK phosphorylation was blunted, and ERK phosphorylation and caspase-3 activation were decreased. CONCLUSIONS NOX1 is an important contributor to ROS production and cell death of the alveolocapillary barrier during hyperoxia and is an upstream actor in oxidative stress-induced acute lung injury involving JNK and ERK pathways in mice.
Collapse
Affiliation(s)
- Stéphanie Carnesecchi
- Department of Pediatrics and Department of Pathology and Immunology, Centre Médical Universitaire, 1, rue Michel Servet, 1211 Geneva 4, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Tyurin VA, Tyurina YY, Jung MY, Tungekar MA, Wasserloos KJ, Bayir H, Greenberger JS, Kochanek PM, Shvedova AA, Pitt B, Kagan VE. Mass-spectrometric analysis of hydroperoxy- and hydroxy-derivatives of cardiolipin and phosphatidylserine in cells and tissues induced by pro-apoptotic and pro-inflammatory stimuli. J Chromatogr B Analyt Technol Biomed Life Sci 2009; 877:2863-72. [PMID: 19328050 PMCID: PMC2723191 DOI: 10.1016/j.jchromb.2009.03.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 03/04/2009] [Accepted: 03/06/2009] [Indexed: 10/21/2022]
Abstract
Oxidation of two anionic phospholipids--cardiolipin (CL) in mitochondria and phosphatidylserine (PS) in extramitochondrial compartments--is important signaling event, particularly during the execution of programmed cell death and clearance of apoptotic cells. Quantitative analysis of CL and PS oxidation products is central to understanding their molecular mechanisms of action. We combined the identification of diverse phospholipid molecular species by ESI-MS with quantitative assessments of lipid hydroperoxides using a fluorescence HPLC-based protocol. We characterized CL and PS oxidation products formed in a model system (cyt c/H(2)O(2)), in apoptotic cells (neurons, pulmonary artery endothelial cells) and mouse lung under inflammatory/oxidative stress conditions (hyperoxia, inhalation of single walled carbon nanotubes). Our results demonstrate the usefulness of this approach for quantitative assessments, identification of individual molecular species and structural characterization of anionic phospholipids that are involved in oxidative modification in cells and tissues.
Collapse
Affiliation(s)
- Vladimir A. Tyurin
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh PA
- Departments of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh PA
| | - Yulia Y. Tyurina
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh PA
- Departments of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh PA
| | - Mi-Yeon Jung
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh PA
- Departments of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh PA
| | - Muhammad A. Tungekar
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh PA
- Departments of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh PA
| | - Karla J. Wasserloos
- Departments of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh PA
| | - Hülya Bayir
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh PA
- Departments of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh PA
- Critical Care Medicine, University of Pittsburgh, Pittsburgh PA
| | | | | | | | - Bruce Pitt
- Departments of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh PA
| | - Valerian E. Kagan
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh PA
- Departments of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh PA
| |
Collapse
|
19
|
Prows DR, Winterberg AV, Gibbons WJ, Burzynski BB, Liu C, Nick TG. Reciprocal backcross mice confirm major loci linked to hyperoxic acute lung injury survival time. Physiol Genomics 2009; 38:158-68. [PMID: 19417010 PMCID: PMC2712219 DOI: 10.1152/physiolgenomics.90392.2008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 04/30/2009] [Indexed: 11/22/2022] Open
Abstract
Morbidity and mortality associated with acute lung injury (ALI) and acute respiratory distress syndrome remain substantial. Although many candidate genes have been tested, a clear understanding of the pathogenesis is lacking, as is our ability to predict individual outcome. Because ALI is a complex disease, single gene approaches cannot easily identify effectors that must be treated concurrently. We employed a strategy to help identify critical genes and gene combinations involved in ALI mortality. Using hyperoxia to induce ALI, a mouse model for genetic analyses of ALI survival time was identified: C57BL/6J (B) mice are sensitive (i.e., die early), whereas 129X1/SvJ (S) mice are significantly more resistant, but with low penetrance. Segregation analysis of reciprocal F(2) mice generated from B and S strains revealed significant sex, cross, and parent of origin effects. Quantitative trait locus (QTL) analysis identified five chromosomal regions significantly linked to hyperoxic ALI survival time (named Shali1-Shali5). Further analyses demonstrated that both parental strains contribute resistance alleles to their offspring and that the phenotype demonstrated parent of origin effects. To validate earlier findings, we generated and tested mice from all eight possible B-S-derived backcrosses. Results from segregation and QTL analyses of 935 backcrosses, alone and combined with the previous 840 B-S-derived F(2) population, further supported the highly significant QTLs on chromosomes 1 (Shali1) and 4 (Shali2) and confirmed that the sex, cross, and parent of origin all contribute to survival time with hyperoxic ALI.
Collapse
Affiliation(s)
- Daniel R Prows
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
AIMS Mitochondrial fusion and fission are essential processes for preservation of normal mitochondrial function. We hypothesized that fusion proteins would be decreased in heart failure (HF), as the mitochondria in HF have been reported to be small and dysfunctional. METHODS AND RESULTS Expression of optic atrophy 1 (OPA1), a mitochondrial fusion protein, was decreased in both human and rat HF, as observed by western blotting. OPA1 is important for maintaining normal cristae structure and function, for preserving the inner membrane structure and for protecting cells from apoptosis. Confocal and electron microscopy studies demonstrated that the mitochondria in the failing hearts were small and fragmented, consistent with decreased fusion. OPA1 mRNA levels did not differ between failing and normal hearts, suggesting post-transcriptional control. Simulated ischaemia in the cardiac myogenic cell line H9c2 cells reduced OPA protein levels. Reduction of OPA1 expression with shRNA resulted in increased apoptosis and fragmentation of the mitochondria. Overexpression of OPA1 increased mitochondrial tubularity, but did not protect against simulated ischaemia-induced apoptosis. Cytochrome c release from the mitochondria was increased both with reduction in OPA1 and with overexpression of OPA1. CONCLUSION This is the first report, to our knowledge, of changes in mitochondrial fusion/fission proteins in cardiovascular disease. These changes have implications for mitochondrial function and apoptosis, contributing to the cell loss which is part of the downward progression of the failing heart.
Collapse
Affiliation(s)
- Le Chen
- Molecular and Cellular Cardiology, Department of Medicine, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
21
|
|
22
|
Carraway MS, Suliman HB, Kliment C, Welty-Wolf KE, Oury TD, Piantadosi CA. Mitochondrial biogenesis in the pulmonary vasculature during inhalational lung injury and fibrosis. Antioxid Redox Signal 2008; 10:269-75. [PMID: 17999632 PMCID: PMC2268979 DOI: 10.1089/ars.2007.1910] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cell survival and injury repair is facilitated by mitochondrial biogenesis; however, the role of this process in lung repair is unknown. We evaluated mitochondrial biogenesis in the mouse lung in two injuries that cause acute inflammation and in two that cause chronic inflammation and pulmonary fibrosis. By using reporter mice that express green fluorescent protein (GFP) exclusively in mitochondria, we tracked mitochondrial biogenesis and correlated it with histologic lung injury, proliferation, and fibrosis. At 72 hours after acute LPS or continuous exposure to hyperoxia (Fio2, 1.0), the lungs showed diffuse infiltration by inflammatory cells in the alveolar region. In reporter mice, patchy new mitochondrial fluorescence was found in the alveolar region but was most prominent and unexpected in perivascular regions. At 14 days after instillation of asbestos or bleomycin, diffuse chronic inflammation had developed, and green fluorescence appeared in inflammatory cells in the expanded interstitium and was most intense in smooth muscle cells of pulmonary vessels. In all four lung injuries, mitochondrial fluorescence colocalized with mitochondrial superoxide dismutase, but not with proliferating cell nuclear antigen. These data indicate that vascular mitochondrial biogenesis is activated in diverse inhalational lung injuries along with oxidative stress. This finding indicates a unique and unexpected mechanism of metabolic adaptation to pulmonary fibrotic injuries.
Collapse
Affiliation(s)
- Martha S Carraway
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Yamada T, Iwasaki Y, Nagata K, Fushiki S, Nakamura H, Marunaka Y, Yodoi J. Thioredoxin-1 protects against hyperoxia-induced apoptosis in cells of the alveolar walls. Pulm Pharmacol Ther 2007; 20:650-9. [PMID: 17045827 DOI: 10.1016/j.pupt.2006.07.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Revised: 07/21/2006] [Accepted: 07/31/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND The mechanisms of hyperoxia-induced lung injury remain poorly defined. Thioredoxin-1 (TRX-1) is a small ubiquitous protein that acts as an important radical scavenger. We investigated the effect of TRX-1 on apoptosis in hyperoxia-induced lung injury. METHODS Mice were exposed to 98% O(2) to produce a model of hyperoxia-induced lung injury. Using transgenic mice overexpressing human TRX-1 (hTRX-1), we assessed lung structure (n=4 per group), immunohistochemical staining for 8-hydroxy-deoxyguanosine (n=4 per group), TUNEL staining (n=5 per group), cytokine (n=5 per group) of IL-1beta and IL-6, and protein (n=6 per group) and m-RNA levels (n=4 per group) (or both) of cytochrome c, Bcl-2, Bax, p21, and p53 in the lungs. RESULTS After exposure to hyperoxia, hTRX-1 transgenic mice had significantly decreased alveolar damage. The apoptotic index was significantly lower in hTRX-1 transgenic mice than in wild-type (WT) mice after exposure to hyperoxia. Protein expression of cytochrome c in the lung was significantly lower in hTRX-1 transgenic mice than in WT mice after exposure to hyperoxia. Protein expression and m-RNA levels of Bcl-2 in the lung were significantly higher in hTRX-1 transgenic mice than in WT mice after exposure to hyperoxia. TRX-1 had no effect on the protein and m-RNA levels of Bax and p21. The protein and m-RNA levels of p53 was unaffected by hyperoxia in hTRX-1 transgenic mice. The cytokine level of IL-6 was significantly higher in hTRX-1 transgenic mice than in WT mice after exposure to hyperoxia. TRX-1 had no effect on cytokine level of IL-1beta. CONCLUSIONS These findings suggest that overexpression of hTRX-1 protects against hyperoxia-induced apoptosis in cells of the alveolar walls. The up-regulating Bcl-2 protein is considered to be one of antiapoptotic effects of TRX-1 in hyperoxia-induced lung injury.
Collapse
Affiliation(s)
- Tadaaki Yamada
- Department of Respiratory Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan.
| | | | | | | | | | | | | |
Collapse
|
24
|
Johnston A, Whitehouse T. Are Mitochondria Responsible for Improved Outcomes in Recent Studies? Intensive Care Med 2007. [DOI: 10.1007/978-0-387-49518-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
25
|
Abstract
PURPOSE OF REVIEW Hyperoxic inspired gas is essential for patients with hypoxic respiratory failure; it is also suspected, however, as a contributor to the pathogenesis of acute lung injury. Several recent studies in humans, animals, and cell culture have identified mechanisms by which hyperoxia may exert deleterious effects on critically ill patients. This review identifies relevant new findings regarding hyperoxic lung injury in the context of providing guidance for future clinical studies. RECENT FINDINGS Recent studies have clarified the roles of both receptor-mediated and mitochondrial cell death pathways in experimental hyperoxic lung injury. Studies in animals demonstrate that hyperoxia interacts with mechanical stretch to augment ventilator-induced lung injury. Finally, studies in humans implicate hyperoxia in impairment of host defense responses to infections. SUMMARY Although hyperoxia has not been conclusively identified as a clinically important cause of lung injury in humans, animal data strongly implicate it. Reports of interaction effects between hyperoxia and both mechanical ventilation and host defense suggest that clinical studies of hyperoxia must take these variables into account. Accumulating data about how hyperoxia initiates cell death provide guidance for development of both biomarkers to identify hyperoxia-induced injury and pharmacological interventions to limit hyperoxia's adverse effects.
Collapse
Affiliation(s)
- William A Altemeier
- Department of Medicine, University of Washington, Seattle, Washington 98195-6522, USA.
| | | |
Collapse
|
26
|
Wang X, Wang Y, Kim HP, Choi AMK, Ryter SW. FLIP inhibits endothelial cell apoptosis during hyperoxia by suppressing Bax. Free Radic Biol Med 2007; 42:1599-609. [PMID: 17448907 DOI: 10.1016/j.freeradbiomed.2007.02.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 02/16/2007] [Accepted: 02/20/2007] [Indexed: 02/08/2023]
Abstract
High oxygen tension (hyperoxia) causes pulmonary cell death, involving apoptosis, necrosis, or mixed death phenotypes, though the underlying mechanisms remain unclear. In mouse lung endothelial cells (MLEC) hyperoxia activates both extrinsic (Fas-dependent) and intrinsic (mitochondria-dependent) apoptotic pathways. We examined the hypothesis that FLIP, an inhibitor of caspase-8, can protect endothelial cells against the lethal effects of hyperoxia. Hyperoxia caused the time-dependent downregulation of FLIP in MLEC. Overexpression of FLIP attenuated intracellular reactive oxygen species generation during hyperoxia exposure, by downregulating extracellular-regulated kinase-1/2 activation and p47(phox) expression. FLIP prevented hyperoxia-induced trafficking of the death-inducing signal complex from the Golgi apparatus to the plasma membrane. Furthermore, FLIP blocked the activations of caspase-8/Bid, caspases -3/-9, and inhibited the mitochondrial translocation and activation of Bax, resulting in protection against hyperoxia-induced cell death. Under normoxic conditions, FLIP expression increased the phosphorylation of p38 mitogen-activated protein kinase leading to increased phosphorylation of Bax during hyperoxic stress. Furthermore, FLIP expression markedly inhibited protein kinase C activation and expression of distinct protein kinase C isoforms (alpha, eta, and zeta), and stabilized an interaction of PKC with Bax. In conclusion, FLIP exerted novel inhibitory effects on extrinsic and intrinsic apoptotic pathways, which significantly protected endothelial cells from the lethal effects of hyperoxia.
Collapse
Affiliation(s)
- Xue Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, 3459 Fifth Avenue, MUH 628NW, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
27
|
Bem RA, Bos AP, Matute-Bello G, van Tuyl M, van Woensel JBM. Lung epithelial cell apoptosis during acute lung injury in infancy. Pediatr Crit Care Med 2007; 8:132-7. [PMID: 17273113 DOI: 10.1097/01.pcc.0000257207.02408.67] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
CONTEXT Apoptosis of lung epithelial cells is implicated in the pathogenesis of acute lung injury. Most research on this subject has focused on adults. Very little is known about a potential interaction of this process with lung development in children. OBJECTIVE To summarize the current literature on lung epithelial cell apoptosis and common causes of acute lung injury in infants and young children and to identify new areas of research. DESIGN A Medline-based literature search. RESULTS AND CONCLUSIONS Few studies have focused on lung epithelial cell apoptosis during common causes of acute lung injury in children. Nevertheless, the limited literature suggests that this may be an important mechanism during respiratory distress syndrome of infants and viral respiratory tract infection. Apoptosis is an essential process during lung development and maturation. Insufficient attention has been paid to potential consequences of this for the short- and long-term outcomes of acute lung injury.
Collapse
Affiliation(s)
- Reinout A Bem
- Pediatric Intensive Care Unit, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
28
|
Xu D, Guthrie JR, Mabry S, Sack TM, Truog WE. Mitochondrial aldehyde dehydrogenase attenuates hyperoxia-induced cell death through activation of ERK/MAPK and PI3K-Akt pathways in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 2006; 291:L966-75. [PMID: 16782756 DOI: 10.1152/ajplung.00045.2006] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Oxygen toxicity is one of the major risk factors in the development of the chronic lung disease or bronchopulmonary dysplasia in premature infants. Using proteomic analysis, we discovered that mitochondrial aldehyde dehydrogenase (mtALDH or ALDH2) was downregulated in neonatal rat lung after hyperoxic exposure. To study the role of mtALDH in hyperoxic lung injury, we overexpressed mtALDH in human lung epithelial cells (A549) and found that mtALDH significantly reduced hyperoxia-induced cell death. Compared with control cells (Neo-A549), the necrotic cell death in mtALDH-overexpressing cells (mtALDH-A549) decreased from 25.3 to 6.5%, 50.5 to 9.1%, and 52.4 to 15.1% after 24-, 48-, and 72-h hyperoxic exposure, respectively. The levels of intracellular and mitochondria-derived reactive oxygen species (ROS) in mtALDH-A549 cells after hyperoxic exposure were significantly lowered compared with Neo-A549 cells. mtALDH overexpression significantly stimulated extracellular signal-regulated kinase (ERK) phosphorylation under normoxic and hyperoxic conditions. Inhibition of ERK phosphorylation partially eliminated the protective effect of mtALDH in hyperoxia-induced cell death, suggesting ERK activation by mtALDH conferred cellular resistance to hyperoxia. mtALDH overexpression augmented Akt phosphorylation and maintained the total Akt level in mtALDH-A549 cells under normoxic and hyperoxic conditions. Inhibition of phosphatidylinositol 3-kinase (PI3K) activation by LY294002 in mtALDH-A549 cells significantly increased necrotic cell death after hyperoxic exposure, indicating that PI3K-Akt activation by mtALDH played an important role in cell survival after hyperoxia. Taken together, these data demonstrate that mtALDH overexpression attenuates hyperoxia-induced cell death in lung epithelial cells through reduction of ROS, activation of ERK/MAPK, and PI3K-Akt cell survival signaling pathways.
Collapse
Affiliation(s)
- Dong Xu
- Neonatology Research Laboratory, Children's Mercy Hospital, Pediatric Research Center, 4th Floor, 2401 Gillham Rd., Kansas City, MO 64108, USA.
| | | | | | | | | |
Collapse
|
29
|
Husari AW, Dbaibo GS, Bitar H, Khayat A, Panjarian S, Nasser M, Bitar FF, El-Sabban M, Zaatari G, Mroueh SM. Apoptosis and the activity of ceramide, Bax and Bcl-2 in the lungs of neonatal rats exposed to limited and prolonged hyperoxia. Respir Res 2006; 7:100. [PMID: 16869980 PMCID: PMC1559609 DOI: 10.1186/1465-9921-7-100] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Accepted: 07/26/2006] [Indexed: 01/02/2023] Open
Abstract
Background The aim of the study is to examine the effect of limited and prolonged hyperoxia on neonatal rat lung. This is done by examining the morphologic changes of apoptosis, the expression of ceramide, an important mediator of apoptosis, the expression of inflammatory mediators represented by IL-1β and the expression of 2 proto-oncogenes that appear to modulate apoptosis (Bax and Bcl-2). Methods Newborn rats were placed in chambers containing room air or oxygen above 90% for 7 days. The rats were sacrificed at 3, 7 or 14 days and their lungs removed. Sections were fixed, subjected to TUNEL, Hoechst, and E-Cadherin Staining. Sections were also incubated with anti-Bcl-2 and anti-Bax antisera. Bcl-2 and Bax were quantitated by immunohistochemistry. Lipids were extracted, and ceramide measured through a modified diacylglycerol kinase assay. RT-PCR was utilized to assess IL-1β expression. Results TUNEL staining showed significant apoptosis in the hyperoxia-exposed lungs at 3 days only. Co-staining of the apoptotic cells with Hoechst, and E-Cadherin indicated that apoptotic cells were mainly epithelial cells. The expression of Bax and ceramide was significantly higher in the hyperoxia-exposed lungs at 3 and 14 days of age, but not at 7 days. Bcl-2 was significantly elevated in the hyperoxia-exposed lungs at 3 and 14 days. IL-1β expression was significantly increased at 14 days. Conclusion Exposure of neonatal rat lung to hyperoxia results in early apoptosis documented by TUNEL assay. The early rise in Bax and ceramide appears to overcome the anti-apoptotic activity of Bcl-2. Further exposure did not result in late apoptotic changes. This suggests that apoptotic response to hyperoxia is time sensitive. Prolonged hyperoxia results in acute lung injury and the shifting balance of ceramide, Bax and Bcl-2 may be related to the evolution of the inflammatory process.
Collapse
Affiliation(s)
- Ahmad W Husari
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| | - Ghassan S Dbaibo
- Department of Pediatrics, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
- Department of Biochemistry, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| | - Hala Bitar
- Department of Pediatrics, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| | - Aline Khayat
- Department of Pediatrics, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| | - Shoghag Panjarian
- Department of Pediatrics, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| | - Michel Nasser
- Department of Physiology, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| | - Fadi F Bitar
- Department of Pediatrics, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| | - Marwan El-Sabban
- Department of Human Morphology, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| | - Ghazi Zaatari
- Pathology Department, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| | - Salman M Mroueh
- Department of Pediatrics, American University of Beirut-Medical Center, P.O. Box: 113-6044, Beirut 1107 2802, Lebanon
| |
Collapse
|
30
|
Carlson DE, Pumplin DW, Ghavam S, Fiedler SM, Chiu WC, Scalea TM. ATP accelerates respiration of mitochondria from rat lung and suppresses their release of hydrogen peroxide. J Bioenerg Biomembr 2006; 37:327-38. [PMID: 16341777 DOI: 10.1007/s10863-005-8644-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2005] [Accepted: 10/04/2005] [Indexed: 11/26/2022]
Abstract
Lung mitochondria were isolated by differential centrifugation from pentobarbital-anesthetized male rats. One to three millimolar Mg2+-ATP increased the consumption of oxygen of lung mitochondria oxidizing 10 mM succinate > fourfold (P < 0.01) whereas ATP increased the respiration of liver mitochondria by < 35%. ATP also hyperpolarized partially uncoupled lung mitochondria in the presence of the mitochondria-specific antagonist, oligomycin. However, only 20% of the ATPase activity in the lung mitochondria was blocked by oligomycin compared to a blockade of 91% for liver mitochondria. We investigated the effect of reducing the non-mitochondrial ATPase activity in the lung preparation. A purer suspension of lung mitochondria from a Percoll gradient was inhibited 95% by oligomycin. The volume fraction identified as mitochondria by electron microscopy in this suspension (73.6+/- 3.5%) did not differ from that for liver mitochondria (69.1+/- 4.9%). ATP reduced the mean area of the mitochondrial profiles in this Percoll fraction by 15% (P <0.01) and increased its state 3 respiration with succinate as substrate by 1.5-fold (P < 0.01) with no change in the state 4 respiration measured after carboxyatractyloside. Hence, ATP increased the respiratory control ratio (state 3/state 4, P <0.01). In contrast, state 3 respiration with the complex 1-selective substrates, glutamate and malate, did not change with addition of ATP. The acceleration of respiration by ATP was accompanied by decreased production of H2O2. Thus ATP-dependent processes that increase respiration appear to improve lung mitochondrial function while minimizing the release of reactive oxygen species.
Collapse
Affiliation(s)
- Drew E Carlson
- Program in Trauma and Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Pagano A, Pitteloud C, Reverdin C, Métrailler-Ruchonnet I, Donati Y, Barazzone Argiroffo C. Poly(ADP-ribose)polymerase Activation Mediates Lung Epithelial Cell DeathIn Vitrobut Is Not Essential in Hyperoxia-Induced Lung Injury. Am J Respir Cell Mol Biol 2005; 33:555-64. [PMID: 16151053 DOI: 10.1165/rcmb.2004-0361oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Hyperoxia induces extensive DNA damage and lung cell death by apoptotic and nonapoptotic pathways. We analyzed the regulation of Poly(ADP-ribose)polymerase-1 (PARP-1), a nuclear enzyme activated by DNA damage, and its relation to cell death during hyperoxia in vitro and in vivo. In lung epithelial-derived A549 cells, which are known to die by necrosis when exposed to oxygen, a minimal amount of PARP-1 was cleaved, correlating with the absence of active caspase-3. Conversely, in primary lung fibroblasts, which die mainly by apoptosis, the complete cleavage of PARP-1 was concomitant to the induction of active caspase-3, as assessed by Western blot and caspase activity. Blockade of caspase activity by Z-VAD reduced the amount of cleaved PARP-1 in fibroblasts. Hyperoxia induced PARP activity in both cell types, as revealed by poly-ADP-ribose accumulation. In A549 cells, the final outcome of necrosis was dependent on PARP activity because it was prevented by the PARP inhibitor 3-aminobenzamide. In contrast, apoptosis of lung fibroblasts was not sensitive to 3-aminobenzamide and was not affected by PARP-1 deletion. In vivo, despite evidence of PARP activation in hyperoxia-exposed mouse lungs, absence of PARP-1 did not change the extent of lung damage, arguing for redundant oxidative stress-induced cell death pathways.
Collapse
Affiliation(s)
- Alessandra Pagano
- Departments of Pediatrics and Pathology, Centre Médical Universitaire, 1 rue Michel Servet, 1211 Geneva 4, Switzerland
| | | | | | | | | | | |
Collapse
|
32
|
Koshika T, Hirayama Y, Ohkubo Y, Mutoh S, Ishizaka A. Tacrolimus (FK506) has protective actions against murine bleomycin-induced acute lung injuries. Eur J Pharmacol 2005; 515:169-78. [PMID: 15894307 DOI: 10.1016/j.ejphar.2005.03.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2004] [Revised: 03/22/2005] [Accepted: 03/30/2005] [Indexed: 02/06/2023]
Abstract
The effects of tacrolimus on murine acute lung injury were tested, especially in comparison to dexamethasone. Acute lung injury was induced by intratracheal instillation of bleomycin. Oral tacrolimus significantly improved survival rates of bleomycin-exposed mice, while cyclosporin A or dexamethasone did not. After instillation of bleomycin (day 0), a migration of neutrophils into alveolar spaces peaked on day 3, with concomitant increases of chemokines. On day 6, marked morphological changes in the lungs were observed. All these changes were significantly inhibited by tacrolimus. Furthermore, DNA ladder and immunohistochemical analyses of lungs showed that apoptosis of lung cells appeared on day 6 and was abolished only by the treatment of tacrolimus. These results suggest that both anti-inflammatory and anti-apoptotic action of tacrolimus contribute to improvement of bleomycin-induced acute lung injury.
Collapse
Affiliation(s)
- Tadatsura Koshika
- Department of Immunology and Inflammation, Medicinal Biology Research Laboratories, Fujisawa Pharmaceutical Co., Ltd., 2-1-6 Kashima, Yodogawa-ku, Osaka 532-8514, Japan.
| | | | | | | | | |
Collapse
|
33
|
Roper JM, Gehen SC, Staversky RJ, Hollander MC, Fornace AJ, O'Reilly MA. Loss of Gadd45a does not modify the pulmonary response to oxidative stress. Am J Physiol Lung Cell Mol Physiol 2005; 288:L663-71. [PMID: 15653712 DOI: 10.1152/ajplung.00355.2004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [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 exposure to high levels of oxygen (hyperoxia) injures and kills microvascular endothelial and alveolar type I epithelial cells. In contrast, significant death of airway and type II epithelial cells is not observed at mortality, suggesting that these cell types may express genes that protect against oxidative stress and damage. During a search for genes induced by hyperoxia, we previously reported that airway and alveolar type II epithelial cells uniquely express the growth arrest and DNA damage ( Gadd) 45a gene. Because Gadd45a has been implicated in protection against genotoxic stress, adult Gadd45a (+/+) and Gadd45a (−/−) mice were exposed to hyperoxia to investigate whether it protected epithelial cells against oxidative stress. During hyperoxia, Gadd45a deficiency did not affect loss of airway epithelial expression of Clara cell secretory protein or type II epithelial cell expression of pro-surfactant protein C. Likewise, Gadd45a deficiency did not alter recruitment of inflammatory cells, edema, or overall mortality. Consistent with Gadd45a not affecting the oxidative stress response, p21Cip1/WAF1and heme oxygenase-1 were comparably induced in Gadd45a (+/+) and Gadd45a (−/−) mice. Additionally, Gadd45a deficiency did not affect oxidative DNA damage or apoptosis as assessed by oxidized guanine and terminal deoxyneucleotidyl transferase-mediated dUTP nick-end labeling staining. Overexpression of Gadd45a in human lung adenocarcinoma cells did not affect viability or survival during exposure, whereas it was protective against UV-radiation. We conclude that increased tolerance of airway and type II epithelial cells to hyperoxia is not attributed solely to expression of Gadd45a.
Collapse
Affiliation(s)
- Jason M Roper
- Departments of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
34
|
Guthmann F, Wissel H, Schachtrup C, Tölle A, Rüdiger M, Spener F, Rüstow B. Inhibition of TNFalpha in vivo prevents hyperoxia-mediated activation of caspase 3 in type II cells. Respir Res 2005; 6:10. [PMID: 15663790 PMCID: PMC548140 DOI: 10.1186/1465-9921-6-10] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2004] [Accepted: 01/21/2005] [Indexed: 01/22/2023] Open
Abstract
Background The mechanisms during the initial phase of oxygen toxicity leading to pulmonary tissue damage are incompletely known. Increase of tumour necrosis factor alpha (TNFalpha) represents one of the first pulmonary responses to hyperoxia. We hypothesised that, in the initial phase of hyperoxia, TNFalpha activates the caspase cascade in type II pneumocytes (TIIcells). Methods Lung sections or freshly isolated TIIcells of control and hyperoxic treated rats (48 hrs) were used for the determination of TNFalpha (ELISA), TNF-receptor 1 (Western blot) and activity of caspases 8, 3, and 9 (colorimetrically). NF-kappaB activation was determined by EMSA, by increase of the p65 subunit in the nuclear fraction, and by immunocytochemistry using a monoclonal anti-NF-kappaB-antibody which selectively stained the activated, nuclear form of NF-kappa B. Apoptotic markers in lung tissue sections (TUNEL) and in TIIcells (cell death detection ELISA, Bax, Bcl-2, mitochondrial membrane potential, and late and early apoptotic cells) were measured using commercially available kits. Results In vivo, hyperoxia activated NF-kappaB and increased the expression of TNFalpha, TNF-receptor 1 and the activity of caspase 8 and 3 in freshly isolated TIIcells. Intratracheal application of anti-TNFalpha antibodies prevented the increase of TNFRI and of caspase 3 activity. Under hyperoxia, there was neither a significant change of cytosolic cytochrome C or of caspase 9 activity, nor an increase in apoptosis of TIIcells. Hyperoxia-induced activation of caspase 3 gradually decreased over two days of normoxia without increasing apoptosis. Therefore, activation of caspase 3 is a temporary effect in sublethal hyperoxia and did not mark the "point of no return" in TIIcells. Conclusion In the initiation phase of pulmonary oxygen toxicity, an increase of TNFalpha and its receptor TNFR1 leads to the activation of caspase 8 and 3 in TIIcells. Together with the hyperoxic induced increase of Bax and the decrease of the mitochondrial membrane potential, activation of caspase 3 can be seen as sensitisation for apoptosis. Eliminating the TNFalpha effect in vivo by anti-TNFalpha antibodies prevents the pro-apoptotic sensitisation of TIIcells.
Collapse
Affiliation(s)
- Florian Guthmann
- Humboldt-Universität zu Berlin, Klinik für Neonatologie, Charité Campus Mitte, D-10098 Berlin, Germany
| | - Heide Wissel
- Humboldt-Universität zu Berlin, Klinik für Neonatologie, Charité Campus Mitte, D-10098 Berlin, Germany
| | - Christian Schachtrup
- Westfälische Wilhelms-Universität Münster, Institut für Biochemie, Wilhelm-Klemm-Str. 2, D-48149 Münster, Germany
| | - Angelika Tölle
- Humboldt-Universität zu Berlin, Klinik für Neonatologie, Charité Campus Mitte, D-10098 Berlin, Germany
| | - Mario Rüdiger
- Humboldt-Universität zu Berlin, Klinik für Neonatologie, Charité Campus Mitte, D-10098 Berlin, Germany
| | - Friedrich Spener
- Westfälische Wilhelms-Universität Münster, Institut für Biochemie, Wilhelm-Klemm-Str. 2, D-48149 Münster, Germany
| | - Bernd Rüstow
- Humboldt-Universität zu Berlin, Klinik für Neonatologie, Charité Campus Mitte, D-10098 Berlin, Germany
| |
Collapse
|
35
|
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: 3.0] [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
|