351
|
Wagener KC, Kolbrink B, Dietrich K, Kizina KM, Terwitte LS, Kempkes B, Bao G, Müller M. Redox Indicator Mice Stably Expressing Genetically Encoded Neuronal roGFP: Versatile Tools to Decipher Subcellular Redox Dynamics in Neuropathophysiology. Antioxid Redox Signal 2016; 25:41-58. [PMID: 27059697 PMCID: PMC4931743 DOI: 10.1089/ars.2015.6587] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Reactive oxygen species (ROS) and downstream redox alterations not only mediate physiological signaling but also neuropathology. For long, ROS/redox imaging was hampered by a lack of reliable probes. Genetically encoded redox sensors overcame this gap and revolutionized (sub)cellular redox imaging. Yet, the successful delivery of sensor-coding DNA, which demands transfection/transduction of cultured preparations or stereotaxic microinjections of each subject, remains challenging. By generating transgenic mice, we aimed to overcome limiting cultured preparations, circumvent surgical interventions, and to extend effectively redox imaging to complex and adult preparations. RESULTS Our redox indicator mice widely express Thy1-driven roGFP1 (reduction-oxidation-sensitive green fluorescent protein 1) in neuronal cytosol or mitochondria. Negative phenotypic effects of roGFP1 were excluded and its proper targeting and functionality confirmed. Redox mapping by ratiometric wide-field imaging reveals most oxidizing conditions in CA3 neurons. Furthermore, mitochondria are more oxidized than cytosol. Cytosolic and mitochondrial roGFP1s reliably report cell endogenous redox dynamics upon metabolic challenge or stimulation. Fluorescence lifetime imaging yields stable, but marginal, response ranges. We therefore developed automated excitation ratiometric 2-photon imaging. It offers superior sensitivity, spatial resolution, and response dynamics. INNOVATION AND CONCLUSION Redox indicator mice enable quantitative analyses of subcellular redox dynamics in a multitude of preparations and at all postnatal stages. This will uncover cell- and compartment-specific cerebral redox signals and their defined alterations during development, maturation, and aging. Cross-breeding with other disease models will reveal molecular details on compartmental redox homeostasis in neuropathology. Combined with ratiometric 2-photon imaging, this will foster our mechanistic understanding of cellular redox signals in their full complexity. Antioxid. Redox Signal. 25, 41-58.
Collapse
Affiliation(s)
- Kerstin C Wagener
- 1 Institut für Neuro- und Sinnesphysiologie, Georg-August-Universität Göttingen , Universitätsmedizin, Göttingen, Germany
| | - Benedikt Kolbrink
- 1 Institut für Neuro- und Sinnesphysiologie, Georg-August-Universität Göttingen , Universitätsmedizin, Göttingen, Germany
| | - Katharina Dietrich
- 1 Institut für Neuro- und Sinnesphysiologie, Georg-August-Universität Göttingen , Universitätsmedizin, Göttingen, Germany
| | - Kathrin M Kizina
- 1 Institut für Neuro- und Sinnesphysiologie, Georg-August-Universität Göttingen , Universitätsmedizin, Göttingen, Germany
| | - Lukas S Terwitte
- 1 Institut für Neuro- und Sinnesphysiologie, Georg-August-Universität Göttingen , Universitätsmedizin, Göttingen, Germany
| | - Belinda Kempkes
- 1 Institut für Neuro- und Sinnesphysiologie, Georg-August-Universität Göttingen , Universitätsmedizin, Göttingen, Germany
| | - Guobin Bao
- 2 Institut für Neurophysiologie und Zelluläre Biophysik, Georg-August-Universität Göttingen , Universitätsmedizin, Göttingen, Germany
| | - Michael Müller
- 1 Institut für Neuro- und Sinnesphysiologie, Georg-August-Universität Göttingen , Universitätsmedizin, Göttingen, Germany
| |
Collapse
|
352
|
Pietrobono S, Morandi A, Gagliardi S, Gerlini G, Borgognoni L, Chiarugi P, Arbiser JL, Stecca B. Down-Regulation of SOX2 Underlies the Inhibitory Effects of the Triphenylmethane Gentian Violet on Melanoma Cell Self-Renewal and Survival. J Invest Dermatol 2016; 136:2059-2069. [PMID: 27373978 DOI: 10.1016/j.jid.2016.06.610] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/09/2016] [Accepted: 06/21/2016] [Indexed: 12/14/2022]
Abstract
Human melanomas contain a population of tumor-initiating cells that are able to maintain the growth of the tumor. We previously showed that the embryonic transcription factor SOX2 is essential for self-renewal and tumorigenicity of human melanoma-initiating cells. However, targeting a transcription factor is still challenging. Gentian violet (GV) is a cationic triphenylmethane dye with potent antifungal and antibacterial activity. Recently, a combination therapy of imiquimod and GV has shown an inhibitory effect against melanoma metastases. Whether and how GV affects melanoma cells remains unknown. Here we show that GV represses melanoma stem cell self-renewal through inhibition of SOX2. Mechanistically, GV hinders EGFR activation and inhibits the signal transducer and activator of transcription-3 [(STAT3)/SOX2] axis. Importantly, we show that GV treatment decreases STAT3 phosphorylation at residue tyrosine 705, thus preventing the translocation of STAT3 into the nucleus and its binding to SOX2 promoter. In addition, GV affects melanoma cell growth by promoting mitochondrial apoptosis and G2 cell cycle arrest. This study shows that in melanoma, GV affects both the stem cell and the tumor bulk compartments, suggesting the potential use of GV in treating human melanoma alone or in combination with targeted therapy and/or immunotherapy.
Collapse
Affiliation(s)
- Silvia Pietrobono
- Core Research Laboratory-Istituto Toscano Tumori, Viale Pieraccini 6, Florence, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Sinforosa Gagliardi
- Core Research Laboratory-Istituto Toscano Tumori, Viale Pieraccini 6, Florence, Italy
| | - Gianni Gerlini
- Plastic Surgery Unit, S.M. Annunziata Hospital-Regional Melanoma Referral Center, Istituto Toscano Tumori, Florence, Italy
| | - Lorenzo Borgognoni
- Plastic Surgery Unit, S.M. Annunziata Hospital-Regional Melanoma Referral Center, Istituto Toscano Tumori, Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Jack L Arbiser
- Department of Dermatology, Atlanta Veterans Administration Medical Center, Atlanta, Georgia, USA; Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Barbara Stecca
- Core Research Laboratory-Istituto Toscano Tumori, Viale Pieraccini 6, Florence, Italy; Department of Oncology, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
353
|
Busija DW, Rutkai I, Dutta S, Katakam PV. Role of Mitochondria in Cerebral Vascular Function: Energy Production, Cellular Protection, and Regulation of Vascular Tone. Compr Physiol 2016; 6:1529-48. [PMID: 27347901 DOI: 10.1002/cphy.c150051] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitochondria not only produce energy in the form of ATP to support the activities of cells comprising the neurovascular unit, but mitochondrial events, such as depolarization and/or ROS release, also initiate signaling events which protect the endothelium and neurons against lethal stresses via pre-/postconditioning as well as promote changes in cerebral vascular tone. Mitochondrial depolarization in vascular smooth muscle (VSM), via pharmacological activation of the ATP-dependent potassium channels on the inner mitochondrial membrane (mitoKATP channels), leads to vasorelaxation through generation of calcium sparks by the sarcoplasmic reticulum and subsequent downstream signaling mechanisms. Increased release of ROS by mitochondria has similar effects. Relaxation of VSM can also be indirectly achieved via actions of nitric oxide (NO) and other vasoactive agents produced by endothelium, perivascular and parenchymal nerves, and astroglia following mitochondrial activation. Additionally, NO production following mitochondrial activation is involved in neuronal preconditioning. Cerebral arteries from female rats have greater mitochondrial mass and respiration and enhanced cerebral arterial dilation to mitochondrial activators. Preexisting chronic conditions such as insulin resistance and/or diabetes impair mitoKATP channel relaxation of cerebral arteries and preconditioning. Surprisingly, mitoKATP channel function after transient ischemia appears to be retained in the endothelium of large cerebral arteries despite generalized cerebral vascular dysfunction. Thus, mitochondrial mechanisms may represent the elusive signaling link between metabolic rate and blood flow as well as mediators of vascular change according to physiological status. Mitochondrial mechanisms are an important, but underutilized target for improving vascular function and decreasing brain injury in stroke patients. © 2016 American Physiological Society. Compr Physiol 6:1529-1548, 2016.
Collapse
Affiliation(s)
- David W Busija
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Ibolya Rutkai
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Somhrita Dutta
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Prasad V Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
354
|
Itani HA, Dikalova AE, McMaster WG, Nazarewicz RR, Bikineyeva AT, Harrison DG, Dikalov SI. Mitochondrial Cyclophilin D in Vascular Oxidative Stress and Hypertension. Hypertension 2016; 67:1218-27. [PMID: 27067720 PMCID: PMC4865418 DOI: 10.1161/hypertensionaha.115.07085] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/08/2016] [Indexed: 12/20/2022]
Abstract
Vascular superoxide (O˙2 (-)) and inflammation contribute to hypertension. The mitochondria are an important source of O˙2 (-); however, the regulation of mitochondrial O˙2 (-) and the antihypertensive potential of targeting the mitochondria remain poorly defined. Angiotensin II and inflammatory cytokines, such as interleukin 17A and tumor necrosis factor-α (TNFα) significantly contribute to hypertension. We hypothesized that angiotensin II and cytokines co-operatively induce cyclophilin D (CypD)-dependent mitochondrial O˙2 (-) production in hypertension. We tested whether CypD inhibition attenuates endothelial oxidative stress and reduces hypertension. CypD depletion in CypD(-/-) mice prevents overproduction of mitochondrial O˙2 (-) in angiotensin II-infused mice, attenuates hypertension by 20 mm Hg, and improves vascular relaxation compared with wild-type C57Bl/6J mice. Treatment of hypertensive mice with the specific CypD inhibitor Sanglifehrin A reduces blood pressure by 28 mm Hg, inhibits production of mitochondrial O˙2 (-) by 40%, and improves vascular relaxation. Angiotensin II-induced hypertension was associated with CypD redox activation by S-glutathionylation, and expression of the mitochondria-targeted H2O2 scavenger, catalase, abolished CypD S-glutathionylation, prevented stimulation mitochondrial O˙2 (-), and attenuated hypertension. The functional role of cytokine-angiotensin II interplay was confirmed by co-operative stimulation of mitochondrial O˙2 (-) by 3-fold in cultured endothelial cells and impairment of aortic relaxation incubated with combination of angiotensin II, interleukin 17A, and tumor necrosis factor-α which was prevented by CypD depletion or expression of mitochondria-targeted SOD2 and catalase. These data support a novel role of CypD in hypertension and demonstrate that targeting CypD decreases mitochondrial O˙2 (-), improves vascular relaxation, and reduces hypertension.
Collapse
Affiliation(s)
- Hana A Itani
- From the Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Anna E Dikalova
- From the Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - William G McMaster
- From the Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Rafal R Nazarewicz
- From the Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Alfiya T Bikineyeva
- From the Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - David G Harrison
- From the Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Sergey I Dikalov
- From the Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
355
|
Song MJ, Davidovich N, Lawrence GG, Margulies SS. Superoxide mediates tight junction complex dissociation in cyclically stretched lung slices. J Biomech 2016; 49:1330-1335. [PMID: 26592435 PMCID: PMC4864146 DOI: 10.1016/j.jbiomech.2015.10.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/20/2015] [Accepted: 10/21/2015] [Indexed: 12/25/2022]
Abstract
We found that stretching Type I rat alveolar epithelial cell (RAEC) monolayers at magnitudes that correspond to high tidal-volume mechanical ventilation results in the production of reactive oxygen species, including nitric oxide and superoxide. Scavenging superoxide with Tiron eliminated the stretch-induced increase in cell monolayer permeability, and similar results were reported for rats ventilated at large tidal volumes, suggesting that oxidative stress plays an important role in barrier impairment in ventilator-induced lung injury associated with large stretch and tidal volumes. In this communication we show that mechanisms that involve oxidative injury are also present in a novel precision cut lung slices (PCLS) model under identical mechanical loads. PCLSs from healthy rats were stretched cyclically to 37% change in surface area for 1 hour. Superoxide was visualized using MitoSOX. To evaluate functional relationships, in separate stretch studies superoxide was scavenged using Tiron or mito-Tempo. PCLS and RAEC permeability was assessed as tight junction (TJ) protein (occludin, claudin-4 and claudin-7) dissociation from zona occludins-1 (ZO-1) via co-immunoprecipitation and Western blot, after 1h (PCLS) or 10min (RAEC) of stretch. Superoxide was increased significantly in PCLS, and Tiron and mito-Tempo dramatically attenuated the response, preventing claudin-4 and claudin-7 dissociation from ZO-1. Using a novel PCLS model for ventilator-induced lung injury studies, we have shown that uniform, biaxial, cyclic stretch generates ROS in the slices, and that superoxide scavenging that can protect the lung tissue under stretch conditions. We conclude that PCLS offer a valuable platform for investigating antioxidant treatments to prevent ventilation-induced lung injury.
Collapse
Affiliation(s)
- Min Jae Song
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Nurit Davidovich
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Gladys G Lawrence
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan S Margulies
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
356
|
Mitochondrial H2O2 in Lung Antigen-Presenting Cells Blocks NF-κB Activation to Prevent Unwarranted Immune Activation. Cell Rep 2016; 15:1700-14. [PMID: 27184852 DOI: 10.1016/j.celrep.2016.04.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/10/2016] [Accepted: 04/15/2016] [Indexed: 12/21/2022] Open
Abstract
Inhalation of environmental antigens such as allergens does not always induce inflammation in the respiratory tract. While antigen-presenting cells (APCs), including dendritic cells and macrophages, take up inhaled antigens, the cell-intrinsic molecular mechanisms that prevent an inflammatory response during this process, such as activation of the transcription factor NF-κB, are not well understood. Here, we show that the nuclear receptor PPARγ plays a critical role in blocking NF-κB activation in response to inhaled antigens to preserve immune tolerance. Tolerance induction promoted mitochondrial respiration, generation of H2O2, and suppression of NF-κB activation in WT, but not PPARγ-deficient, APCs. Forced restoration of H2O2 in PPARγ-deficient cells suppressed IκBα degradation and NF-κB activation. Conversely, scavenging reactive oxygen species from mitochondria promoted IκBα degradation with loss of regulatory and promotion of inflammatory T cell responses in vivo. Thus, communication between PPARγ and the mitochondria maintains immune quiescence in the airways.
Collapse
|
357
|
Park JB, Nagar H, Choi S, Jung SB, Kim HW, Kang SK, Lee JW, Lee JH, Park JW, Irani K, Jeon BH, Song HJ, Kim CS. IDH2 deficiency impairs mitochondrial function in endothelial cells and endothelium-dependent vasomotor function. Free Radic Biol Med 2016; 94:36-46. [PMID: 26898144 DOI: 10.1016/j.freeradbiomed.2016.02.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/09/2016] [Accepted: 02/15/2016] [Indexed: 02/05/2023]
Abstract
Mitochondrial NADP(+)-dependent isocitrate dehydrogenase (IDH2) plays an essential role protecting cells against oxidative stress-induced damage. A deficiency in IDH2 leads to mitochondrial dysfunction and the production of reactive oxygen species (ROS) in cardiomyocytes and cancer cells. However, the function of IDH2 in vascular endothelial cells is mostly unknown. In this study the effects of IDH2 deficiency on mitochondrial and vascular function were investigated in endothelial cells. IDH2 knockdown decreased the expression of mitochondrial oxidative phosphorylation (OXPHOS) complexes I, II and III, which lead to increased mitochondrial superoxide. In addition, the levels of fission and fusion proteins (Mfn-1, OPA-1, and Drp-1) were significantly altered and MnSOD expression also was decreased by IDH2 knockdown. Furthermore, knockdown of IDH2 decreased eNOS phosphorylation and nitric oxide (NO) concentration in endothelial cells. Interestingly, treatment with Mito-TEMPO, a mitochondrial-specific superoxide scavenger, recovered mitochondrial fission-fusion imbalance and blunted mitochondrial superoxide production, and reduced the IDH2 knockdown-induced decrease in MnSOD expression, eNOS phosphorylation and NO production in endothelial cells. Endothelium-dependent vasorelaxation was impaired, and the concentration of bioavailable NO decreased in the aortic ring in IDH2 knockout mice. These findings suggest that IDH2 deficiency induces endothelial dysfunction through the induction of dynamic mitochondrial changes and impairment in vascular function.
Collapse
Affiliation(s)
- Jung-Bum Park
- Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-131, Republic of Korea
| | - Harsha Nagar
- Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-131, Republic of Korea
| | - Sujeong Choi
- Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-131, Republic of Korea
| | - Saet-Byel Jung
- Department of Endocrinology, Chungnam National University, Daejeon 301-131, Republic of Korea
| | - Hyun-Woo Kim
- Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-131, Republic of Korea
| | - Shin Kwang Kang
- Department of Thoracic and Cardiovascular Surgery, Chungnam National University, Daejeon 301-131, Republic of Korea
| | - Jun Wan Lee
- Emergency ICU, Regional Emergency Center, Chungnam National University, Daejeon 301-131, Republic of Korea
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong 339-700, Republic of Korea
| | - Jeen-Woo Park
- School of Life Sciences, College of Natural Science, Kyungpook National University, Taegu 702-701, Republic of Korea
| | - Kaikobad Irani
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Byeong Hwa Jeon
- Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-131, Republic of Korea
| | - Hee-Jung Song
- Department of Neurology, School of Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea.
| | - Cuk-Seong Kim
- Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-131, Republic of Korea.
| |
Collapse
|
358
|
Giam B, Kaye DM, Rajapakse NW. Role of Renal Oxidative Stress in the Pathogenesis of the Cardiorenal Syndrome. Heart Lung Circ 2016; 25:874-80. [PMID: 27132623 DOI: 10.1016/j.hlc.2016.02.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/19/2016] [Accepted: 02/24/2016] [Indexed: 10/24/2022]
Abstract
Renal dysfunction and heart failure commonly co-exist; it is termed the cardiorenal syndrome (CRS). This combination of renal and cardiac impairment presents a substantial clinical challenge and is associated with adverse prognosis. The pathogenesis of the CRS is complex, including chronic activation of the renin-angiotensin-aldosterone system (RAAS) and the sympathetic nervous system, together with reduced renal perfusion. Chronic activation of the RAAS can impair mitochondrial function, and increase mitochondrial derived oxidative stress which in turn can lead to renal injury and sodium and water retention. For example, it has been shown that exogenous Ang II augments renal mitochondrial oxidative stress, reduces GFR and induces albuminuria in rats with heart failure. Administration of Ang II also augmented renal mitochondrial dysfunction in aged mice. Current treatments for CRS, including angiotensin-converting enzyme inhibitors, exert limited renal protection if any at all. Therefore, novel treatments particularly those that can target renal mechanisms downstream to chronic activation of the renal renin-angiotensin system are likely to exert renoprotection in the setting of CRS.
Collapse
Affiliation(s)
- Beverly Giam
- Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, Vic., Australia; Central Clinical School, Monash University, Melbourne, Vic., Australia.
| | - David M Kaye
- Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, Vic., Australia; Department of Medicine, Monash University, Melbourne, Vic., Australia
| | - Niwanthi W Rajapakse
- Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, Vic., Australia; Department of Physiology, Monash University, Melbourne, Vic., Australia
| |
Collapse
|
359
|
Amino acids trigger down-regulation of superoxide via TORC pathway in the midgut of Rhodnius prolixus. Biosci Rep 2016; 36:BSR20160061. [PMID: 26945025 PMCID: PMC4832317 DOI: 10.1042/bsr20160061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 03/03/2016] [Indexed: 01/20/2023] Open
Abstract
Sensing incoming nutrients is an important and critical event for intestinal cells to sustain life of the whole organism. The TORC is a major protein complex involved in monitoring the nutritional status and is activated by elevated amino acid concentrations. An important feature of haematophagy is that huge amounts of blood are ingested in a single meal, which results in the release of large quantities of amino acids, together with the haemoglobin prosthetic group, haem, which decomposes hydroperoxides and propagates oxygen-derived free radicals. Our previous studies demonstrated that reactive oxygen species (ROS) levels were diminished in the mitochondria and midgut of the Dengue fever mosquito, Aedes aegypti, immediately after a blood meal. We proposed that this mechanism serves to avoid oxidative damage that would otherwise be induced by haem following a blood meal. Studies also performed in mosquitoes have shown that blood or amino acids controls protein synthesis through TORC activation. It was already proposed, in different models, a link between ROS and TOR, however, little is known about TOR signalling in insect midgut nor about the involvement of ROS in this pathway. Here, we studied the effect of a blood meal on ROS production in the midgut of Rhodnius prolixus We observed that blood meal amino acids decreased ROS levels in the R. prolixus midgut immediately after feeding, via lowering mitochondrial superoxide production and involving the amino acid-sensing TORC pathway.
Collapse
|
360
|
Haidasz EA, Meng D, Amorati R, Baschieri A, Ingold KU, Valgimigli L, Pratt DA. Acid Is Key to the Radical-Trapping Antioxidant Activity of Nitroxides. J Am Chem Soc 2016; 138:5290-8. [DOI: 10.1021/jacs.6b00677] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Evan A. Haidasz
- Department
of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Derek Meng
- Department
of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Riccardo Amorati
- Department
of Chemistry “G. Ciamician”, University of Bologna, Bologna I-40126, Italy
| | - Andrea Baschieri
- Department
of Chemistry “G. Ciamician”, University of Bologna, Bologna I-40126, Italy
| | - Keith U. Ingold
- National Research Council of Canada, Ottawa, Ontario K1A 0R6, Canada
| | - Luca Valgimigli
- Department
of Chemistry “G. Ciamician”, University of Bologna, Bologna I-40126, Italy
| | - Derek A. Pratt
- Department
of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
361
|
Vergeade A, Bertram CC, Bikineyeva AT, Zackert WE, Zinkel SS, May JM, Dikalov SI, Roberts LJ, Boutaud O. Cardiolipin fatty acid remodeling regulates mitochondrial function by modifying the electron entry point in the respiratory chain. Mitochondrion 2016; 28:88-95. [PMID: 27085476 DOI: 10.1016/j.mito.2016.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 03/24/2016] [Accepted: 04/07/2016] [Indexed: 01/18/2023]
Abstract
Modifications of cardiolipin (CL) levels or compositions are associated with changes in mitochondrial function in a wide range of pathologies. We have made the discovery that acetaminophen remodels CL fatty acids composition from tetralinoleoyl to linoleoyltrioleoyl-CL, a remodeling that is associated with decreased mitochondrial respiration. Our data show that CL remodeling causes a shift in electron entry from complex II to the β-oxidation electron transfer flavoprotein quinone oxidoreductase (ETF/QOR) pathway. These data demonstrate that electron entry in the respiratory chain is regulated by CL fatty acid composition and provide proof-of-concept that pharmacological intervention can be used to modify CL composition.
Collapse
Affiliation(s)
- Aurelia Vergeade
- Department of Pharmacology, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States
| | - Clinton C Bertram
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States
| | - Alfiya T Bikineyeva
- Department of Medicine, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States
| | - William E Zackert
- Department of Pharmacology, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States
| | - Sandra S Zinkel
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States; Department of Medicine, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States
| | - James M May
- Department of Medicine, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States
| | - Sergey I Dikalov
- Department of Medicine, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States
| | - L Jackson Roberts
- Department of Pharmacology, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States; Department of Medicine, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States
| | - Olivier Boutaud
- Department of Pharmacology, Vanderbilt University Medical Center, 536 Robinson Research Building, Nashville, TN 37232-6602, United States.
| |
Collapse
|
362
|
Patel D, Alhawaj R, Kelly MR, Accarino JJO, Lakhkar A, Gupte SA, Sun D, Wolin MS. Potential role of mitochondrial superoxide decreasing ferrochelatase and heme in coronary artery soluble guanylate cyclase depletion by angiotensin II. Am J Physiol Heart Circ Physiol 2016; 310:H1439-47. [PMID: 27037373 DOI: 10.1152/ajpheart.00859.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/28/2016] [Indexed: 02/07/2023]
Abstract
Oxidation of the soluble guanylate cyclase (sGC) heme promotes loss of regulation by nitric oxide (NO) and depletion of sGC. We hypothesized that angiotensin II (ANG II) stimulation of mitochondrial superoxide by its type 1 receptor could function as a potential inhibitor of heme biosynthesis by ferrochelatase, and this could decrease vascular responsiveness to NO by depleting sGC. These processes were investigated in a 24-h organoid culture model of bovine coronary arteries (BCA) with 0.1 μM ANG II. Treatment of BCA with ANG II increased mitochondrial superoxide, depleted mitochondrial superoxide dismutase (SOD2), ferrochelatase, and cytochrome oxidase subunit 4, and sGC, associated with impairment of relaxation to NO. These processes were attenuated by organoid culture with 8-bromo-cGMP and/or δ-aminolevulinic acid (a stimulator of sGC by protoporphyrin IX generation and heme biosynthesis). Organoid culture with Mito-TEMPOL, a scavenger of mitochondrial matrix superoxide, also attenuated ANG II-elicited ferrochelatase depletion and loss of relaxation to NO, whereas organoid culture with Tempol, an extramitochondrial scavenger of superoxide, attenuated the loss of relaxation to NO by ANG II, but not ferrochelatase depletion, suggesting cytosolic superoxide could be an initiating factor in the loss of sGC regulation by NO. The depletion of cytochrome oxidase subunit 4 and sGC (but not catalase) suggests that sGC expression may be very sensitive to depletion of heme caused by ANG II disrupting ferrochelatase activity by increasing mitochondrial superoxide. In addition, cGMP-dependent activation of protein kinase G appears to attenuate these ANG II-stimulated processes through both preventing SOD2 depletion and increases in mitochondrial and extramitochondrial superoxide.
Collapse
Affiliation(s)
- Dhara Patel
- Department of Physiology, Translational Center for Pulmonary Hypertension, New York Medical College, Valhalla, New York and
| | - Raed Alhawaj
- Department of Physiology, Translational Center for Pulmonary Hypertension, New York Medical College, Valhalla, New York and
| | - Melissa R Kelly
- Department of Physiology, Translational Center for Pulmonary Hypertension, New York Medical College, Valhalla, New York and
| | - John J O Accarino
- Department of Physiology, Translational Center for Pulmonary Hypertension, New York Medical College, Valhalla, New York and
| | - Anand Lakhkar
- Department of Pharmacology, Translational Center for Pulmonary Hypertension, New York Medical College, Valhalla, New York
| | - Sachin A Gupte
- Department of Pharmacology, Translational Center for Pulmonary Hypertension, New York Medical College, Valhalla, New York
| | - Dong Sun
- Department of Physiology, Translational Center for Pulmonary Hypertension, New York Medical College, Valhalla, New York and
| | - Michael S Wolin
- Department of Physiology, Translational Center for Pulmonary Hypertension, New York Medical College, Valhalla, New York and
| |
Collapse
|
363
|
Du K, Farhood A, Jaeschke H. Mitochondria-targeted antioxidant Mito-Tempo protects against acetaminophen hepatotoxicity. Arch Toxicol 2016; 91:761-773. [PMID: 27002509 DOI: 10.1007/s00204-016-1692-0] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/10/2016] [Indexed: 02/06/2023]
Abstract
Acetaminophen (APAP) hepatotoxicity is characterized by an extensive mitochondrial oxidant stress. However, its importance as a drug target has not been clarified. To investigate this, fasted C57BL/6J mice were treated with 300 mg/kg APAP and the mitochondria-targeted antioxidant Mito-Tempo (MT) was given 1.5 h later. APAP caused severe liver injury in mice, as indicated by the increase in plasma ALT activities and centrilobular necrosis. MT dose-dependently reduced the injury. Importantly, MT did not affect APAP-protein adducts formation, glutathione depletion or c-jun N-terminal kinase activation and its mitochondrial translocation. In contrast, hepatic glutathione disulfide and peroxynitrite formation were dose-dependently reduced by MT, indicating its effective mitochondrial oxidant stress scavenging capacity. Consequently, mitochondrial translocation of Bax and release of mitochondrial intermembrane proteins such as apoptosis-inducing factor were prevented, and nuclear DNA fragmentation was eliminated. To demonstrate the importance of mitochondria-specific antioxidant property of MT, we compared its efficacy with Tempo, which has the same pharmacological mode of action as MT but lacks the mitochondria targeting moiety. In contrast to the dramatic protection by MT, the same molar dose of Tempo did not significantly reduce APAP hepatotoxicity. In contrast, even a 3 h post-treatment with MT reduced 70 % of the injury, and the combination of MT with N-acetylcysteine (NAC) provided superior protection than NAC alone. We conclude that MT protects against APAP overdose in mice by attenuating the mitochondrial oxidant stress and preventing peroxynitrite formation and the subsequent mitochondrial dysfunction. MT is a promising therapeutic agent for APAP overdose patients.
Collapse
Affiliation(s)
- Kuo Du
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, KS, 66160, USA
| | - Anwar Farhood
- Department of Pathology, St. David's North Austin Medical Center, Austin, TX, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, KS, 66160, USA.
| |
Collapse
|
364
|
c-Src, ERK1/2 and Rho kinase mediate hydrogen peroxide-induced vascular contraction in hypertension: role of TXA2, NAD(P)H oxidase and mitochondria. J Hypertens 2016; 33:77-87. [PMID: 25380156 DOI: 10.1097/hjh.0000000000000383] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIM : The aim of this study was to analyse the signalling pathways involved in H2O2 vascular responses in hypertension. METHODS Vascular function, thromboxane A2 (TXA2) production, oxidative stress and protein expression were determined in mesenteric resistance arteries (MRAs) from hypertensive (spontaneously hypertensive rats, SHR) and normotensive Wistar Kyoto (WKY) rats. RESULTS H2O2 and the TP agonist U46619 induced greater contractile responses in MRA from SHR than WKY. Moreover, H2O2 increased TXA2 production more in SHR than in WKY. The c-Src inhibitor PP1 reduced H2O2 and U46619-induced contraction and TXA2 release in both strains. The ERK1/2 inhibitor PD98059 reduced H2O2 but not U46619-induced contraction only in SHR arteries. The Rho kinase inhibitor Y26372 reduced H2O2 and U46619-induced contractions only in SHR arteries. Basal c-Src, ERK1/2 and Rho kinase expression were greater in MRA from SHR than WKY. In SHR, the combination of PD98059 with the TP antagonist SQ29548 but not with Y27632 inhibited the H2O2 contraction more than each inhibitor alone. H2O2 and U46619 increased NAD(P)H oxidase activity and O2 production and decreased mitochondrial membrane potential in vessels from SHR. The effects induced by H2O2 were abolished by inhibitors of TXA2 synthase, ERK1/2 and c-Src. The mitochondrial antioxidant mitoTEMPO reduced H2O2-induced contraction and NAD(P)H oxidase activation. CONCLUSION In arteries from WKY, c-Src mediates H2O2 contractile responses by modulating TXA2 release and TXA2 effect. In SHR, H2O2 induces c-Src dependent TXA2 release that provokes vascular contractile responses through Rho kinase, c-Src and O2 from NAD(P)H Oxidase and mitochondria. Moreover, ERK1/2 activation contributes to H2O2 contraction in SHR through effects on mitochondria/NAD(P)H Oxidase.
Collapse
|
365
|
Shirai T, Nazarewicz RR, Wallis BB, Yanes RE, Watanabe R, Hilhorst M, Tian L, Harrison DG, Giacomini JC, Assimes TL, Goronzy JJ, Weyand CM. The glycolytic enzyme PKM2 bridges metabolic and inflammatory dysfunction in coronary artery disease. J Exp Med 2016; 213:337-54. [PMID: 26926996 PMCID: PMC4813677 DOI: 10.1084/jem.20150900] [Citation(s) in RCA: 391] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 02/01/2016] [Indexed: 12/25/2022] Open
Abstract
Abnormal glucose metabolism and enhanced oxidative stress accelerate cardiovascular disease, a chronic inflammatory condition causing high morbidity and mortality. Here, we report that in monocytes and macrophages of patients with atherosclerotic coronary artery disease (CAD), overutilization of glucose promotes excessive and prolonged production of the cytokines IL-6 and IL-1β, driving systemic and tissue inflammation. In patient-derived monocytes and macrophages, increased glucose uptake and glycolytic flux fuel the generation of mitochondrial reactive oxygen species, which in turn promote dimerization of the glycolytic enzyme pyruvate kinase M2 (PKM2) and enable its nuclear translocation. Nuclear PKM2 functions as a protein kinase that phosphorylates the transcription factor STAT3, thus boosting IL-6 and IL-1β production. Reducing glycolysis, scavenging superoxide and enforcing PKM2 tetramerization correct the proinflammatory phenotype of CAD macrophages. In essence, PKM2 serves a previously unidentified role as a molecular integrator of metabolic dysfunction, oxidative stress and tissue inflammation and represents a novel therapeutic target in cardiovascular disease.
Collapse
Affiliation(s)
- Tsuyoshi Shirai
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Rafal R Nazarewicz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305 Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Barbara B Wallis
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Rolando E Yanes
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Ryu Watanabe
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Marc Hilhorst
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Lu Tian
- Division of Biostatistics, Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA 94305
| | - David G Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - John C Giacomini
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Themistocles L Assimes
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
366
|
Asensio-López MC, Soler F, Sánchez-Más J, Pascual-Figal D, Fernández-Belda F, Lax A. Early oxidative damage induced by doxorubicin: Source of production, protection by GKT137831 and effect on Ca(2+) transporters in HL-1 cardiomyocytes. Arch Biochem Biophys 2016; 594:26-36. [PMID: 26906075 DOI: 10.1016/j.abb.2016.02.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/04/2016] [Accepted: 02/18/2016] [Indexed: 01/26/2023]
Abstract
In atrial-derived HL-1 cells, ryanodine receptor and Na(+)/Ca(2+)-exchanger were altered early by 5 μM doxorubicin. The observed effects were an increase of cytosolic Ca(2+) at rest, ensuing ryanodine receptor phosphorylation, and the slowing of Ca(2+) transient decay after caffeine addition. Doxorubicin triggered a linear rise of reactive oxygen species (ROS) with no early effect on mitochondrial inner membrane potential. Doxorubicin and ROS were both detected in mitochondria by colocalization with fluorescence probes and doxorubicin-induced ROS was totally blocked by mitoTEMPO. The NADPH oxidase activity in the mitochondrial fraction was sensitive to inhibition by GKT137831, and doxorubicin-induced ROS decreased gradually as the GKT137831 concentration added in preincubation was increased. When doxorubicin-induced ROS was prevented by GKT137831, the kinetic response revealed a permanent degree of protection that was consistent with mitochondrial NADPH oxidase inhibition. In contrast, the ROS induction by doxorubicin after melatonin preincubation was totally eliminated at first but the effect was completely reversed with time. Limiting the source of ROS production is a better alternative for dealing with oxidative damage than using ROS scavengers. The short-term effect of doxorubicin on Ca(2+) transporters involved in myocardiac contractility was dependent on oxidative damage, and so the impairment was subsequent to ROS production.
Collapse
Affiliation(s)
- Mari C Asensio-López
- Cardiología Clínica y Experimental, Departamento de Medicina Interna, Facultad de Medicina, Universidad de Murcia, Campus de El Palmar, 30120, Murcia, Spain
| | - Fernando Soler
- Departamento de Bioquímica y Biología Molecular A, Universidad de Murcia, Campus de Espinardo, 30071, Murcia, Spain
| | - Jesús Sánchez-Más
- Cardiología Clínica y Experimental, Departamento de Medicina Interna, Facultad de Medicina, Universidad de Murcia, Campus de El Palmar, 30120, Murcia, Spain
| | - Domingo Pascual-Figal
- Cardiología Clínica y Experimental, Departamento de Medicina Interna, Facultad de Medicina, Universidad de Murcia, Campus de El Palmar, 30120, Murcia, Spain; Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, El Palmar, Murcia, Spain
| | - Francisco Fernández-Belda
- Departamento de Bioquímica y Biología Molecular A, Universidad de Murcia, Campus de Espinardo, 30071, Murcia, Spain.
| | - Antonio Lax
- Cardiología Clínica y Experimental, Departamento de Medicina Interna, Facultad de Medicina, Universidad de Murcia, Campus de El Palmar, 30120, Murcia, Spain
| |
Collapse
|
367
|
Abstract
OBJECTIVES Reactive oxygen species-mediated cell death contributes to the pathophysiology of cardiovascular disease and myocardial dysfunction. We recently showed that mitochondrial NADP+-dependent isocitrate dehydrogenase (IDH2) functions as an antioxidant and anti-apoptotic protein by supplying NADPH to antioxidant systems. METHODS In the present study, we demonstrated that H2O2-induced apoptosis and hypertrophy of H9c2 cardiomyoblasts was markedly exacerbated by small interfering RNA (siRNA) specific for IDH2. RESULTS Attenuated IDH2 expression resulted in the modulation of cellular and mitochondrial redox status, mitochondrial function, and cellular oxidative damage. MitoTEMPO, a mitochondria-targeted antioxidant, efficiently suppressed increased caspase-3 activity, increased cell size, and depletion of cellular GSH levels in IDH2 siRNA-transfected cells that were treated with H2O2. DISCUSSION These results indicated that the disruption of cellular redox balance might be responsible for the enhanced H2O2-induced apoptosis and hypertrophy of cultured cardiomyocytes by the attenuated IDH2 expression.
Collapse
Affiliation(s)
- Hyeong Jun Ku
- a School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group , Kyungpook National University , Taegu , Korea
| | - Jeen-Woo Park
- a School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group , Kyungpook National University , Taegu , Korea
| |
Collapse
|
368
|
Laitano O, Ahn B, Patel N, Coblentz PD, Smuder AJ, Yoo JK, Christou DD, Adhihetty PJ, Ferreira LF. Pharmacological targeting of mitochondrial reactive oxygen species counteracts diaphragm weakness in chronic heart failure. J Appl Physiol (1985) 2016; 120:733-42. [PMID: 26846552 DOI: 10.1152/japplphysiol.00822.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/28/2016] [Indexed: 12/15/2022] Open
Abstract
Diaphragm muscle weakness in chronic heart failure (CHF) is caused by elevated oxidants and exacerbates breathing abnormalities, exercise intolerance, and dyspnea. However, the specific source of oxidants that cause diaphragm weakness is unknown. We examined whether mitochondrial reactive oxygen species (ROS) cause diaphragm weakness in CHF by testing the hypothesis that CHF animals treated with a mitochondria-targeted antioxidant have normal diaphragm function. Rats underwent CHF or sham surgery. Eight weeks after surgeries, we administered a mitochondrial-targeted antioxidant (MitoTEMPO; 1 mg·kg(-1)·day(-1)) or sterile saline (Vehicle). Left ventricular dysfunction (echocardiography) pre- and posttreatment and morphological abnormalities were consistent with the presence of CHF. CHF elicited a threefold (P < 0.05) increase in diaphragm mitochondrial H2O2 emission, decreased diaphragm glutathione content by 23%, and also depressed twitch and maximal tetanic force by ∼20% in Vehicle-treated animals compared with Sham (P < 0.05 for all comparisons). Diaphragm mitochondrial H2O2 emission, glutathione content, and twitch and maximal tetanic force were normal in CHF animals receiving MitoTEMPO. Neither CHF nor MitoTEMPO altered the diaphragm protein levels of antioxidant enzymes: superoxide dismutases (CuZn-SOD or MnSOD), glutathione peroxidase, and catalase. In both Vehicle and MitoTEMPO groups, CHF elicited a ∼30% increase in cytochrome c oxidase activity, whereas there were no changes in citrate synthase activity. Our data suggest that elevated mitochondrial H2O2 emission causes diaphragm weakness in CHF. Moreover, changes in protein levels of antioxidant enzymes or mitochondrial content do not seem to mediate the increase in mitochondria H2O2 emission in CHF and protective effects of MitoTEMPO.
Collapse
Affiliation(s)
- Orlando Laitano
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Bumsoo Ahn
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Nikhil Patel
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Philip D Coblentz
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Ashley J Smuder
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Jeung-Ki Yoo
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Demetra D Christou
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Peter J Adhihetty
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| |
Collapse
|
369
|
Billings EA, Lee CS, Owen KA, D'Souza RS, Ravichandran KS, Casanova JE. The adhesion GPCR BAI1 mediates macrophage ROS production and microbicidal activity against Gram-negative bacteria. Sci Signal 2016; 9:ra14. [PMID: 26838550 PMCID: PMC4894535 DOI: 10.1126/scisignal.aac6250] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The detection of microbes and initiation of an innate immune response occur through pattern recognition receptors (PRRs), which are critical for the production of inflammatory cytokines and activation of the cellular microbicidal machinery. In particular, the production of reactive oxygen species (ROS) by the NADPH oxidase complex is a critical component of the macrophage bactericidal machinery. We previously characterized brain-specific angiogenesis inhibitor 1 (BAI1), a member of the adhesion family of G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors (GPCRs), as a PRR that mediates the selective phagocytic uptake of Gram-negative bacteria by macrophages. We showed that BAI1 promoted phagosomal ROS production through activation of the Rho family guanosine triphosphatase (GTPase) Rac1, thereby stimulating NADPH oxidase activity. Primary BAI1-deficient macrophages exhibited attenuated Rac GTPase activity and reduced ROS production in response to several Gram-negative bacteria, resulting in impaired microbicidal activity. Furthermore, in a peritoneal infection model, BAI1-deficient mice exhibited increased susceptibility to death by bacterial challenge because of impaired bacterial clearance. Together, these findings suggest that BAI1 mediates the clearance of Gram-negative bacteria by stimulating both phagocytosis and NADPH oxidase activation, thereby coupling bacterial detection to the cellular microbicidal machinery.
Collapse
Affiliation(s)
- Emily A Billings
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Chang Sup Lee
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Katherine A Owen
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ryan S D'Souza
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - James E Casanova
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA. Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
370
|
Engholm M, Pinilla E, Mogensen S, Matchkov V, Hedegaard ER, Chen H, Mulvany MJ, Simonsen U. Involvement of transglutaminase 2 and voltage-gated potassium channels in cystamine vasodilatation in rat mesenteric small arteries. Br J Pharmacol 2016; 173:839-55. [PMID: 26603619 DOI: 10.1111/bph.13393] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 10/13/2015] [Accepted: 11/10/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Vasodilatation may contribute to the neuroprotective and vascular anti-remodelling effect of the tissue transglutaminase 2 (TG2) inhibitor cystamine. Here, we hypothesized that inhibition of TG2 followed by blockade of smooth muscle calcium entry and/or inhibition of Rho kinase underlies cystamine vasodilatation. EXPERIMENTAL APPROACH We used rat mesenteric small arteries and RT-PCR, immunoblotting, and measurements of isometric wall tension, intracellular Ca(2+) ([Ca(2+)]i ), K(+) currents (patch clamp), and phosphorylation of myosin phosphatase targeting subunit 1 (MYPT1) and myosin regulatory light chain, in our experiments. KEY RESULTS RT-PCR and immunoblotting revealed expression of TG2 in mesenteric small arteries. Cystamine concentration-dependently inhibited responses to phenylephrine, 5-HT and U46619 and for extracellular potassium. Selective inhibitors of TG2, LDN 27129 and T101, also inhibited phenylephrine contraction. An inhibitor of PLC suppressed cystamine relaxation. Cystamine relaxed and reduced [Ca(2+)]i in phenylephrine-contracted arteries. In potassium-contracted arteries, cystamine induced less relaxation without changing [Ca(2+)]i , and these relaxations were blocked by mitochondrial complex inhibitors. Blockers of Kv 7 channels, XE991 and linopirdine, inhibited cystamine relaxation and increases in voltage-dependent smooth muscle currents. Cystamine and the Rho kinase inhibitor Y27632 reduced basal MYPT1-Thr(855) phosphorylation, but only Y27632 reduced phenylephrine-induced increases in MYPT1-Thr(855) and myosin regulatory light chain phosphorylation. CONCLUSIONS AND IMPLICATIONS Cystamine induced vasodilatation by inhibition of receptor-coupled TG2, leading to opening of Kv channels and reduction of intracellular calcium, and by activation of a pathway sensitive to inhibitors of the mitochondrial complexes I and III. Both pathways may contribute to the antihypertensive and neuroprotective effect of cystamine.
Collapse
Affiliation(s)
- Morten Engholm
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Estéfano Pinilla
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Susie Mogensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Vladimir Matchkov
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Elise Røge Hedegaard
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Hua Chen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Michael J Mulvany
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| |
Collapse
|
371
|
Yang CM, Lin CC, Hsieh HL. High-Glucose-Derived Oxidative Stress-Dependent Heme Oxygenase-1 Expression from Astrocytes Contributes to the Neuronal Apoptosis. Mol Neurobiol 2016; 54:470-483. [PMID: 26742524 DOI: 10.1007/s12035-015-9666-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/17/2015] [Indexed: 12/26/2022]
Abstract
An elevated level of glucose has been found in the blood of hyperglycemia and diabetes patients associated with several central nervous system (CNS) complications. These disorders may be due to the up-regulation of many neurotoxic mediators by host cells triggered by high glucose (HG). Moreover, heme oxygenase-1 (HO-1) plays a crucial role in tissue pathological changes such as brain injuries. However, the molecular mechanisms underlying HG-induced HO-1 expression in brain cells remain poorly defined. Thus, we use the rat brain astrocytes (RBA-1) as a model to investigate the signaling mechanisms of HO-1 induction by HG and its effects on neuronal cells. We demonstrated that HG induced HO-1 expression via a reactive oxygen species (ROS)-dependent signaling pathway. NADPH oxidase (Nox)- and mitochondrion-dependent ROS generation led to activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun-N-terminal kinase (JNK) and then activated the downstream transcriptional factors nuclear factor-kappaB (NF-κB) and c-Fos/activator protein 1 (AP-1), respectively. Subsequently, the activated NF-κB and AP-1 turned on transcription of HO-1 gene. These results indicated that in brain astrocytes, activation of MAPK-mediated NF-κB and c-Fos/AP-1 cascades by Nox/ROS and mitoROS-dependent events is essential for HO-1 up-regulation induced by HG. Moreover, we found that HG-induced extracellular ROS increase and HO-1 expression from astrocytes resulted in neuronal apoptosis. These results offers new insights into the mechanisms and effects of the action of HG, supporting that HG may cause brain disorders in the development of diabetes- and hyperglycemia-induced CNS complications such as neurodegenerative diseases.
Collapse
Affiliation(s)
- Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Gui-Shan, Tao-Yuan, Taiwan
| | - Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Gui-Shan, Tao-Yuan, Taiwan
| | - Hsi-Lung Hsieh
- Department of Nursing, Division of Basic Medical Sciences, and Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Gui-Shan, Tao-Yuan, Taiwan.
| |
Collapse
|
372
|
Ni R, Cao T, Xiong S, Ma J, Fan GC, Lacefield JC, Lu Y, Le Tissier S, Peng T. Therapeutic inhibition of mitochondrial reactive oxygen species with mito-TEMPO reduces diabetic cardiomyopathy. Free Radic Biol Med 2016; 90:12-23. [PMID: 26577173 PMCID: PMC5066872 DOI: 10.1016/j.freeradbiomed.2015.11.013] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 10/27/2015] [Accepted: 11/09/2015] [Indexed: 02/08/2023]
Abstract
AIMS The mitochondria are important sources of reactive oxygen species (ROS) in the heart. Mitochondrial ROS production has been implicated in the pathogenesis of diabetic cardiomyopathy, suggesting that therapeutic strategies specifically targeting mitochondrial ROS may have benefit in this disease. We investigated the therapeutic effects of mitochondria-targeted antioxidant mito-TEMPO on diabetic cardiomyopathy. METHODS The mitochondria-targeted antioxidant mito-TEMPO was administrated after diabetes onset in a mouse model of streptozotocin-induced type-1 diabetes and type-2 diabetic db/db mice. Cardiac adverse changes were analyzed and myocardial function assessed. Cultured adult cardiomyocytes were stimulated with high glucose, and mitochondrial superoxide generation and cell death were measured. RESULTS Incubation with high glucose increased mitochondria superoxide generation in cultured cardiomyocytes, which was prevented by mito-TEMPO. Co-incubation with mito-TEMPO abrogated high glucose-induced cell death. Mitochondrial ROS generation, and intracellular oxidative stress levels were induced in both type-1 and type-2 diabetic mouse hearts. Daily injection of mito-TEMPO for 30 days inhibited mitochondrial ROS generation, prevented intracellular oxidative stress levels, decreased apoptosis and reduced myocardial hypertrophy in diabetic hearts, leading to improvement of myocardial function in both type-1 and type-2 diabetic mice. Incubation with mito-TEMPO or inhibition of Nox2-containing NADPH oxidase prevented oxidative stress levels and cell death in high glucose-stimulated cardiomyocytes. Mechanistic study revealed that the protective effects of mito-TEMPO were associated with down-regulation of ERK1/2 phosphorylation. CONCLUSIONS Therapeutic inhibition of mitochondrial ROS by mito-TEMPO reduced adverse cardiac changes and mitigated myocardial dysfunction in diabetic mice. Thus, mitochondria-targeted antioxidants may be an effective therapy for diabetic cardiac complications.
Collapse
Affiliation(s)
- Rui Ni
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, VRL 6th Floor, A6-140, 800 Commissioners Road, London, Ont., Canada N6A 4G5; Departments of Medicine and Pathology, The University of Western Ontario, London, Ont., Canada N6A 4G5
| | - Ting Cao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Jian Ma
- Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, VRL 6th Floor, A6-140, 800 Commissioners Road, London, Ont., Canada N6A 4G5; Departments of Medicine and Pathology, The University of Western Ontario, London, Ont., Canada N6A 4G5
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - James C Lacefield
- Electrical and Computer Engineering, Medical Biophysics, Robarts Research Institute, University of Western Ontario, London, Ont., Canada N6A 4G5
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Sydney Le Tissier
- Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, VRL 6th Floor, A6-140, 800 Commissioners Road, London, Ont., Canada N6A 4G5
| | - Tianqing Peng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, VRL 6th Floor, A6-140, 800 Commissioners Road, London, Ont., Canada N6A 4G5; Departments of Medicine and Pathology, The University of Western Ontario, London, Ont., Canada N6A 4G5.
| |
Collapse
|
373
|
Božić A, Marinković A, Bjelogrlić S, Todorović TR, Cvijetić IN, Novaković I, Muller CD, Filipović NR. Quinoline based mono- and bis-(thio)carbohydrazones: synthesis, anticancer activity in 2D and 3D cancer and cancer stem cell models. RSC Adv 2016. [DOI: 10.1039/c6ra23940d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Study of antitumor activity of mono- and bis-quinoline based (thio)carbohydrazones on THP-1 and AsPC-1 cancer stem cells, revealed that thiocarbohydrazones had superior pro-apoptotic activity than carbohydrazones with multi-target profile activities.
Collapse
Affiliation(s)
- Aleksandra Božić
- Faculty of Technology and Metallurgy
- University of Belgrade
- Belgrade
- Serbia
| | | | | | | | - Ilija N. Cvijetić
- Innovation Center of the Faculty of Chemistry
- University of Belgrade
- Belgrade
- Serbia
| | - Irena Novaković
- Institute of Chemistry, Technology and Metallurgy
- University of Belgrade
- Belgrade
- Serbia
| | - Christian D. Muller
- Institut Pluridisciplinaire Hubert Curien
- UMR 7178
- CNRS
- Université de Strasbourg
- 67401 Illkirch
| | | |
Collapse
|
374
|
Protective effects of mito-TEMPO against doxorubicin cardiotoxicity in mice. Cancer Chemother Pharmacol 2015; 77:659-62. [PMID: 26712129 DOI: 10.1007/s00280-015-2949-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/11/2015] [Indexed: 01/23/2023]
Abstract
PURPOSE Doxorubicin (DOX) is a chemotherapeutic that is widely used for the treatment of many human tumors. However, the development of cardiotoxicity has limited its use. The aim of the present study was to evaluate the possible efficacy of mito-TEMPO (mito-T) as a protective agent against DOX-induced cardiotoxicity in mice. METHODS C57BL/6 mice were treated twice with mito-T at low (5 mg/kg body weight) or high (20 mg/kg body weight) dose and once with DOX (24 mg/kg body weight) or saline (0.1 mL/20 g body weight) by means of intraperitoneal injections. The levels of malondialdehyde (MLDA), a marker of lipid peroxidation, and serum levels of creatine kinase were evaluated 48 h after the injection of DOX. RESULTS DOX induced lipid peroxidation in heart mitochondria (p < 0.001), and DOX-treated mice receiving mito-T at low dose had levels of MLDA significantly lower than the mice that received only DOX (p < 0.01). Furthermore, administration of mito-T alone did not cause any significant changes from control values. Additionally, DOX-treated mice treated with mito-T at high dose showed decrease in serum levels of total CK compared to mice treated with DOX alone (p < 0.05). CONCLUSION Our results indicate that mito-T protects mice against DOX-induced cardiotoxicity.
Collapse
|
375
|
Vendrov AE, Vendrov KC, Smith A, Yuan J, Sumida A, Robidoux J, Runge MS, Madamanchi NR. NOX4 NADPH Oxidase-Dependent Mitochondrial Oxidative Stress in Aging-Associated Cardiovascular Disease. Antioxid Redox Signal 2015; 23:1389-409. [PMID: 26054376 PMCID: PMC4692134 DOI: 10.1089/ars.2014.6221] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIMS Increased oxidative stress and vascular inflammation are implicated in increased cardiovascular disease (CVD) incidence with age. We and others demonstrated that NOX1/2 NADPH oxidase inhibition, by genetic deletion of p47phox, in Apoe(-/-) mice decreases vascular reactive oxygen species (ROS) generation and atherosclerosis in young age. The present study examined whether NOX1/2 NADPH oxidases are also pivotal to aging-associated CVD. RESULTS Both aged (16 months) Apoe(-/-) and Apoe(-/-)/p47phox(-/-) mice had increased atherosclerotic lesion area, aortic stiffness, and systolic dysfunction compared with young (4 months) cohorts. Cellular and mitochondrial ROS (mtROS) levels were significantly higher in aortic wall and vascular smooth muscle cells (VSMCs) from aged wild-type and p47phox(-/-) mice. VSMCs from aged mice had increased mitochondrial protein oxidation and dysfunction and increased vascular cell adhesion molecule 1 expression, which was abrogated with (2-(2,2,6,6-Tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (MitoTEMPO) treatment. NOX4 expression was increased in the vasculature and mitochondria of aged mice and its suppression with shRNA in VSMCs from aged mice decreased mtROS levels and improved function. Increased mtROS levels were associated with enhanced mitochondrial NOX4 expression in aortic VSMCs from aged subjects, and NOX4 expression levels in arterial wall correlated with age and atherosclerotic severity. Aged Apoe(-/-) mice treated with MitoTEMPO and 2-(2-chlorophenyl)-4-methyl-5-(pyridin-2-ylmethyl)-1H-pyrazolo[4,3-c]pyridine-3,6(2H,5H)-dione had decreased vascular ROS levels and atherosclerosis and preserved vascular and cardiac function. INNOVATION AND CONCLUSION These data suggest that NOX4, but not NOX1/2, and mitochondrial oxidative stress are mediators of CVD in aging under hyperlipidemic conditions. Regulating NOX4 activity/expression and using mitochondrial antioxidants are potential approaches to reducing aging-associated CVD.
Collapse
Affiliation(s)
- Aleksandr E Vendrov
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| | - Kimberly C Vendrov
- 2 Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina
| | - Alberto Smith
- 3 Cardiovascular Division, Academic Department of Surgery, National Institute for Health Research Biomedical Research Center at Guy's and St Thomas' National Health Service Foundation Trust , King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Jinling Yuan
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| | - Arihiro Sumida
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| | - Jacques Robidoux
- 4 Department of Pharmacology and Toxicology, The East Carolina Diabetes and Obesity Institute, East Carolina University , Greenville, North Carolina
| | - Marschall S Runge
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| | - Nageswara R Madamanchi
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| |
Collapse
|
376
|
Rochael NC, Guimarães-Costa AB, Nascimento MTC, DeSouza-Vieira TS, Oliveira MP, Garcia e Souza LF, Oliveira MF, Saraiva EM. Classical ROS-dependent and early/rapid ROS-independent release of Neutrophil Extracellular Traps triggered by Leishmania parasites. Sci Rep 2015; 5:18302. [PMID: 26673780 PMCID: PMC4682131 DOI: 10.1038/srep18302] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 11/04/2015] [Indexed: 12/23/2022] Open
Abstract
Neutrophil extracellular traps (NETs) extruded from neutrophils upon activation are composed of chromatin associated with cytosolic and granular proteins, which ensnare and kill microorganisms. This microbicidal mechanism named classical netosis has been shown to dependent on reactive oxygen species (ROS) generation by NADPH oxidase and also chromatin decondensation dependent upon the enzymes (PAD4), neutrophil elastase (NE) and myeloperoxidase (MPO). NET release also occurs through an early/rapid ROS-independent mechanism, named early/rapid vital netosis. Here we analyze the role of ROS, NE, MPO and PAD4 in the netosis stimulated by Leishmania amazonensis promastigotes in human neutrophils. We demonstrate that promastigotes induce a classical netosis, dependent on the cellular redox imbalance, as well as by a chloroamidine sensitive and elastase activity mechanism. Additionally, Leishmania also induces the early/rapid NET release occurring only 10 minutes after neutrophil-parasite interaction. We demonstrate here, that this early/rapid mechanism is dependent on elastase activity, but independent of ROS generation and chloroamidine. A better understanding of both mechanisms of NET release, and the NETs effects on the host immune system modulation, could support the development of new potential therapeutic strategies for leishmaniasis.
Collapse
Affiliation(s)
- Natalia C Rochael
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, RJ, 21941-902. Brazil
| | - Anderson B Guimarães-Costa
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, RJ, 21941-902. Brazil.,Vector Molecular Biology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Michelle T C Nascimento
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, RJ, 21941-902. Brazil
| | - Thiago S DeSouza-Vieira
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, RJ, 21941-902. Brazil
| | - Matheus P Oliveira
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Cidade Universitária, 21941-902, Rio de Janeiro, Brazil.,Laboratório de Inflamação e Metabolismo, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem (INBEB), Universidade Federal do Rio de Janeiro, RJ, 21941-902. Brazil
| | - Luiz F Garcia e Souza
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Cidade Universitária, 21941-902, Rio de Janeiro, Brazil.,Laboratório de Inflamação e Metabolismo, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem (INBEB), Universidade Federal do Rio de Janeiro, RJ, 21941-902. Brazil
| | - Marcus F Oliveira
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Cidade Universitária, 21941-902, Rio de Janeiro, Brazil
| | - Elvira M Saraiva
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, RJ, 21941-902. Brazil
| |
Collapse
|
377
|
Joo HK, Lee YR, Kang G, Choi S, Kim CS, Ryoo S, Park JB, Jeon BH. The 18-kDa Translocator Protein Inhibits Vascular Cell Adhesion Molecule-1 Expression via Inhibition of Mitochondrial Reactive Oxygen Species. Mol Cells 2015; 38:1064-70. [PMID: 26608360 PMCID: PMC4696997 DOI: 10.14348/molcells.2015.0165] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/25/2015] [Accepted: 09/30/2015] [Indexed: 12/31/2022] Open
Abstract
Translocator protein 18 kDa (TSPO) is a mitochondrial outer membrane protein and is abundantly expressed in a variety of organ and tissues. To date, the functional role of TSPO on vascular endothelial cell activation has yet to be fully elucidated. In the present study, the phorbol 12-myristate 13-acetate (PMA, 250 nM), an activator of protein kinase C (PKC), was used to induce vascular endothelial activation. Adenoviral TSPO overexpression (10-100 MOI) inhibited PMA-induced vascular cell adhesion molecule-1 (VCAM-1) and intracellular cell adhesion molecule-1 (ICAM-1) expression in a dose dependent manner. PMA-induced VCAM-1 expressions were inhibited by Mito-TEMPO (0.1-0.5 μM), a specific mitochondrial antioxidants, and cyclosporin A (1-5 μM), a mitochondrial permeability transition pore inhibitor, implying on an important role of mitochondrial reactive oxygen species (ROS) on the endothelial activation. Moreover, adenoviral TSPO overexpression inhibited mitochondrial ROS production and manganese superoxide dismutase expression. On contrasts, gene silencing of TSPO with siRNA increased PMA-induced VCAM-1 expression and mitochondrial ROS production. Midazolam (1-50 μM), TSPO ligands, inhibited PMA-induced VCAM-1 and mitochondrial ROS production in endothelial cells. These results suggest that mitochondrial TSPO can inhibit PMA-induced endothelial inflammation via suppression of VCAM-1 and mitochondrial ROS production in endothelial cells.
Collapse
Affiliation(s)
- Hee Kyoung Joo
- Infectious Signaling Network Research Center and Research Institute for Medical Sciences, Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-747,
Korea
| | - Yu Ran Lee
- Infectious Signaling Network Research Center and Research Institute for Medical Sciences, Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-747,
Korea
| | - Gun Kang
- Infectious Signaling Network Research Center and Research Institute for Medical Sciences, Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-747,
Korea
| | - Sunga Choi
- Infectious Signaling Network Research Center and Research Institute for Medical Sciences, Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-747,
Korea
| | - Cuk-Seong Kim
- Infectious Signaling Network Research Center and Research Institute for Medical Sciences, Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-747,
Korea
| | - Sungwoo Ryoo
- Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chunchon 200-701,
Korea
| | - Jin Bong Park
- Infectious Signaling Network Research Center and Research Institute for Medical Sciences, Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-747,
Korea
| | - Byeong Hwa Jeon
- Infectious Signaling Network Research Center and Research Institute for Medical Sciences, Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301-747,
Korea
| |
Collapse
|
378
|
Granata S, Dalla Gassa A, Tomei P, Lupo A, Zaza G. Mitochondria: a new therapeutic target in chronic kidney disease. Nutr Metab (Lond) 2015; 12:49. [PMID: 26612997 PMCID: PMC4660721 DOI: 10.1186/s12986-015-0044-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 11/18/2015] [Indexed: 12/24/2022] Open
Abstract
Cellular metabolic changes during chronic kidney disease (CKD) may induce higher production of oxygen radicals that play a significant role in the progression of renal damage and in the onset of important comorbidities. This condition seems to be in part related to dysfunctional mitochondria that cause an increased electron "leakage" from the respiratory chain during oxidative phosphorylation with a consequent generation of reactive oxygen species (ROS). ROS are highly active molecules that may oxidize proteins, lipids and nucleic acids with a consequent damage of cells and tissues. To mitigate this mitochondria-related functional impairment, a variety of agents (including endogenous and food derived antioxidants, natural plants extracts, mitochondria-targeted molecules) combined with conventional therapies could be employed. However, although the anti-oxidant properties of these substances are well known, their use in clinical practice has been only partially investigated. Additionally, for their correct utilization is extremely important to understand their effects, to identify the correct target of intervention and to minimize adverse effects. Therefore, in this manuscript, we reviewed the characteristics of the available mitochondria-targeted anti-oxidant compounds that could be employed routinely in our nephrology, internal medicine and renal transplant centers. Nevertheless, large clinical trials are needed to provide more definitive information about their use and to assess their overall efficacy or toxicity.
Collapse
Affiliation(s)
- Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| | - Alessandra Dalla Gassa
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| | - Paola Tomei
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| | - Antonio Lupo
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| |
Collapse
|
379
|
Schmidt HHHW, Stocker R, Vollbracht C, Paulsen G, Riley D, Daiber A, Cuadrado A. Antioxidants in Translational Medicine. Antioxid Redox Signal 2015; 23:1130-43. [PMID: 26154592 PMCID: PMC4657516 DOI: 10.1089/ars.2015.6393] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE It is generally accepted that reactive oxygen species (ROS) scavenging molecules or antioxidants exert health-promoting effects and thus their consumption as food additives and nutraceuticals has been greatly encouraged. Antioxidants may be beneficial in situations of subclinical deficiency and increased demand or acutely upon high-dose infusion. However, to date, there is little clinical evidence for the long-term benefit of most antioxidants. Alarmingly, recent evidence points even to health risks, in particular for supplements of lipophilic antioxidants. RECENT ADVANCES The biological impact of ROS depends not only on their quantities but also on their chemical nature, (sub)cellular and tissue location, and the rates of their formation and degradation. Moreover, ROS serve important physiological functions; thus, inappropriate removal of ROS may cause paradoxical reductive stress and thereby induce or promote disease. CRITICAL ISSUES Any recommendation on antioxidants must be based on solid clinical evidence and patient-relevant outcomes rather than surrogate parameters. FUTURE DIRECTIONS Such evidence-based use may include site-directed application, time-limited high dosing, (functional) pharmacological repair of oxidized biomolecules, and triggers of endogenous antioxidant response systems. Ideally, these approaches need guidance by patient stratification through predictive biomarkers and possibly imaging modalities.
Collapse
Affiliation(s)
- Harald H H W Schmidt
- 1 Department of Pharmacology, CARIM, FHML, MIAS, Maastricht University , Maastricht, The Netherlands
| | - Roland Stocker
- 2 Victor Chang Cardiac Research Institute , Sydney, Australia .,3 University of New South Wales , Sydney, Australia
| | - Claudia Vollbracht
- 4 Hochschule Fresenius, University of Applied Sciences , Idstein, Germany
| | | | - Dennis Riley
- 6 Galera Therapeutics Inc., Malvern, Pennsylvania
| | - Andreas Daiber
- 7 Labor für Molekulare Kardiologie, II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes Gutenberg-Universität , Mainz, Germany
| | - Antonio Cuadrado
- 8 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , ISCIII, Madrid, Spain .,9 Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC , Madrid, Spain .,10 Instituto de Investigación Sanitaria La Paz (IdiPaz) , Madrid, Spain .,11 Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid , Madrid, Spain
| |
Collapse
|
380
|
Springo Z, Tarantini S, Toth P, Tucsek Z, Koller A, Sonntag WE, Csiszar A, Ungvari Z. Aging Exacerbates Pressure-Induced Mitochondrial Oxidative Stress in Mouse Cerebral Arteries. J Gerontol A Biol Sci Med Sci 2015; 70:1355-9. [PMID: 25631392 PMCID: PMC4612385 DOI: 10.1093/gerona/glu244] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 12/08/2014] [Indexed: 12/21/2022] Open
Abstract
Epidemiological studies demonstrate that in addition to the increased prevalence of hypertension in old patients, the deleterious cerebrovascular effects of hypertension (including atherosclerosis, stroke, and vascular cognitive impairment) are also exacerbated in elderly individuals. The cellular mechanisms by which aging and hypertension interact to promote cerebrovascular pathologies are not well understood. To test the hypothesis that aging exacerbates high pressure-induced mitochondrial oxidative stress, we exposed isolated segments of the middle cerebral arteries of young (3 months) and aged (24 months) C57BL/6 mice to 60 or 140 mmHg intraluminal pressure and assessed changes in mitochondrial reactive oxygen species production using a mitochondria-targeted redox-sensitive fluorescent indicator dye (MitoSox) by confocal microscopy. Perinuclear MitoSox fluorescence was significantly stronger in high pressure-exposed middle cerebral arteries compared with middle cerebral arteries of the same animals exposed to 60 mmHg, indicating that high pressure increases mitochondrial reactive oxygen species production in the smooth muscle cells of cerebral arteries. Comparison of young and aged middle cerebral arteries showed that aging exacerbates high pressure-induced mitochondrial reactive oxygen species production in cerebral arteries. We propose that increased mechanosensitive mitochondrial oxidative stress may potentially exacerbate cerebrovascular injury and vascular inflammation in aging.
Collapse
Affiliation(s)
- Zsolt Springo
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center. Department of Pathophysiology and Gerontology, Medical School and Szentágothai Research Center, University of Pecs, Hungary
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center
| | - Peter Toth
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center
| | - Zsuzsanna Tucsek
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center
| | - Akos Koller
- Department of Pathophysiology and Gerontology, Medical School and Szentágothai Research Center, University of Pecs, Hungary
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center. The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center. Department of Pathophysiology and Gerontology, Medical School and Szentágothai Research Center, University of Pecs, Hungary. The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center. Department of Pathophysiology and Gerontology, Medical School and Szentágothai Research Center, University of Pecs, Hungary. The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center.
| |
Collapse
|
381
|
ERK5/HDAC5-mediated, resveratrol-, and pterostilbene-induced expression of MnSOD in human endothelial cells. Mol Nutr Food Res 2015; 60:266-77. [DOI: 10.1002/mnfr.201500466] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/23/2015] [Accepted: 09/28/2015] [Indexed: 11/07/2022]
|
382
|
Hameed LS, Berg DA, Belnoue L, Jensen LD, Cao Y, Simon A. Environmental changes in oxygen tension reveal ROS-dependent neurogenesis and regeneration in the adult newt brain. eLife 2015; 4. [PMID: 26485032 PMCID: PMC4635398 DOI: 10.7554/elife.08422] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 10/19/2015] [Indexed: 01/31/2023] Open
Abstract
Organisms need to adapt to the ecological constraints in their habitat. How specific processes reflect such adaptations are difficult to model experimentally. We tested whether environmental shifts in oxygen tension lead to events in the adult newt brain that share features with processes occurring during neuronal regeneration under normoxia. By experimental simulation of varying oxygen concentrations, we show that hypoxia followed by re-oxygenation lead to neuronal death and hallmarks of an injury response, including activation of neural stem cells ultimately leading to neurogenesis. Neural stem cells accumulate reactive oxygen species (ROS) during re-oxygenation and inhibition of ROS biosynthesis counteracts their proliferation as well as neurogenesis. Importantly, regeneration of dopamine neurons under normoxia also depends on ROS-production. These data demonstrate a role for ROS-production in neurogenesis in newts and suggest that this role may have been recruited to the capacity to replace lost neurons in the brain of an adult vertebrate. DOI:http://dx.doi.org/10.7554/eLife.08422.001 During the winter, red-spotted newts remain active in water that is covered by ice. The oxygen levels under the ice tend to drop and so the newts adjust their metabolism to cope with these conditions. However, when oxygen levels return to normal, this may result in the newts producing larger amounts of chemically reactive molecules called reactive oxygen species (ROS). These molecules form naturally as a by-product of oxygen metabolism, but in high quantities they can damage cells and tissues. It has been proposed that red-spotted newts and other animals that experience periods of low oxygen may have evolved processes to repair such damage. Unlike us, red-spotted newts are able to replace nerve cells in the brain that have died or been injured. This regeneration is fuelled by stem cells called ependymoglia cells, which divide to produce new nerve cells. Here, Hameed et al. investigated whether the return of oxygen to normal levels after a period of low oxygen can damage nerve cells in the newts, and whether this is followed by regeneration. The experiments show that nerve cells in the newt brain do indeed die when oxygen levels return to normal. Also, the brain activates an injury response that triggers the ependymoglia cells to divide. During this process, the ependymoglia cells accumulate ROS and their ability to divide is impaired if the production of ROS is blocked. The replacement of injured brain cells in normal oxygen conditions also depends on increased ROS levels. Together, Hameed et al.'s findings demonstrate a key role for ROS production in controlling the regeneration of damaged nerve cells in the red-spotted newt. A future challenge is to identify the genes that control the survival and activation of ependymoglia cells in response to increased ROS levels in the brain. DOI:http://dx.doi.org/10.7554/eLife.08422.002
Collapse
Affiliation(s)
- L Shahul Hameed
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Daniel A Berg
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Laure Belnoue
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Lasse D Jensen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.,Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, University of Leicester, Leicester, United Kingdom
| | - András Simon
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
383
|
Fike CD, Dikalova A, Kaplowitz MR, Cunningham G, Summar M, Aschner JL. Rescue Treatment with L-Citrulline Inhibits Hypoxia-Induced Pulmonary Hypertension in Newborn Pigs. Am J Respir Cell Mol Biol 2015; 53:255-64. [PMID: 25536367 DOI: 10.1165/rcmb.2014-0351oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Infants with cardiopulmonary disorders associated with hypoxia develop pulmonary hypertension. We previously showed that initiation of oral L-citrulline before and continued throughout hypoxic exposure improves nitric oxide (NO) production and ameliorates pulmonary hypertension in newborn piglets. Rescue treatments, initiated after the onset of pulmonary hypertension, better approximate clinical strategies. Mechanisms by which L-citrulline improves NO production merit elucidation. The objective of this study was to determine whether starting L-citrulline after the onset of pulmonary hypertension inhibits disease progression and improves NO production by recoupling endothelial NO synthase (eNOS). Hypoxic and normoxic (control) piglets were studied. Some hypoxic piglets received oral L-citrulline starting on Day 3 of hypoxia and continuing throughout the remaining 7 days of hypoxic exposure. Catheters were placed for hemodynamic measurements, and pulmonary arteries were dissected to assess NO production and eNOS dimer-to-monomer ratios (a measure of eNOS coupling). Pulmonary vascular resistance was lower in L-citrulline-treated hypoxic piglets than in untreated hypoxic piglets but was higher than in normoxic controls. NO production and eNOS dimer-to-monomer ratios were greater in pulmonary arteries from L-citrulline-treated than from untreated hypoxic animals but were lower than in normoxic controls. When started after disease onset, oral L-citrulline treatment improves NO production by recoupling eNOS and inhibits the further development of chronic hypoxia-induced pulmonary hypertension in newborn piglets. Oral L-citrulline may be a novel strategy to halt or reverse pulmonary hypertension in infants suffering from cardiopulmonary conditions associated with hypoxia.
Collapse
Affiliation(s)
- Candice D Fike
- 1 Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee.,2 Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Anna Dikalova
- 1 Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mark R Kaplowitz
- 1 Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gary Cunningham
- 3 Division of Genetics and Metabolism, Children's National Medical Center, Washington, District of Columbia; and
| | - Marshall Summar
- 3 Division of Genetics and Metabolism, Children's National Medical Center, Washington, District of Columbia; and
| | - Judy L Aschner
- 4 Department of Pediatrics, Albert Einstein College of Medicine and the Children's Hospital at Montefiore, New York, New York
| |
Collapse
|
384
|
Daiber A, Münzel T. Organic Nitrate Therapy, Nitrate Tolerance, and Nitrate-Induced Endothelial Dysfunction: Emphasis on Redox Biology and Oxidative Stress. Antioxid Redox Signal 2015; 23:899-942. [PMID: 26261901 PMCID: PMC4752190 DOI: 10.1089/ars.2015.6376] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Organic nitrates, such as nitroglycerin (GTN), isosorbide-5-mononitrate and isosorbide dinitrate, and pentaerithrityl tetranitrate (PETN), when given acutely, have potent vasodilator effects improving symptoms in patients with acute and chronic congestive heart failure, stable coronary artery disease, acute coronary syndromes, or arterial hypertension. The mechanisms underlying vasodilation include the release of •NO or a related compound in response to intracellular bioactivation (for GTN, the mitochondrial aldehyde dehydrogenase [ALDH-2]) and activation of the enzyme, soluble guanylyl cyclase. Increasing cyclic guanosine-3',-5'-monophosphate (cGMP) levels lead to an activation of the cGMP-dependent kinase I, thereby causing the relaxation of the vascular smooth muscle by decreasing intracellular calcium concentrations. The hemodynamic and anti-ischemic effects of organic nitrates are rapidly lost upon long-term (low-dose) administration due to the rapid development of tolerance and endothelial dysfunction, which is in most cases linked to increased intracellular oxidative stress. Enzymatic sources of reactive oxygen species under nitrate therapy include mitochondria, NADPH oxidases, and an uncoupled •NO synthase. Acute high-dose challenges with organic nitrates cause a similar loss of potency (tachyphylaxis), but with distinct pathomechanism. The differences among organic nitrates are highlighted regarding their potency to induce oxidative stress and subsequent tolerance and endothelial dysfunction. We also address pleiotropic effects of organic nitrates, for example, their capacity to stimulate antioxidant pathways like those demonstrated for PETN, all of which may prevent adverse effects in response to long-term therapy. Based on these considerations, we will discuss and present some preclinical data on how the nitrate of the future should be designed.
Collapse
Affiliation(s)
- Andreas Daiber
- The 2nd Medical Clinic, Medical Center of the Johannes Gutenberg University , Mainz, Germany
| | - Thomas Münzel
- The 2nd Medical Clinic, Medical Center of the Johannes Gutenberg University , Mainz, Germany
| |
Collapse
|
385
|
Mitochondrial ROS Induces Cardiac Inflammation via a Pathway through mtDNA Damage in a Pneumonia-Related Sepsis Model. PLoS One 2015; 10:e0139416. [PMID: 26448624 PMCID: PMC4598156 DOI: 10.1371/journal.pone.0139416] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/14/2015] [Indexed: 12/21/2022] Open
Abstract
We have previously shown that mitochondria-targeted vitamin E (Mito-Vit-E), a mtROS specific antioxidant, improves cardiac performance and attenuates inflammation in a pneumonia-related sepsis model. In this study, we applied the same approaches to decipher the signaling pathway(s) of mtROS-dependent cardiac inflammation after sepsis. Sepsis was induced in Sprague Dawley rats by intratracheal injection of S. pneumoniae. Mito-Vit-E, vitamin E or vehicle was administered 30 minutes later. In myocardium 24 hours post-inoculation, Mito-Vit-E, but not vitamin E, significantly protected mtDNA integrity and decreased mtDNA damage. Mito-Vit-E alleviated sepsis-induced reduction in mitochondria-localized DNA repair enzymes including DNA polymerase γ, AP endonuclease, 8-oxoguanine glycosylase, and uracil-DNA glycosylase. Mito-Vit-E dramatically improved metabolism and membrane integrity in mitochondria, suppressed leakage of mtDNA into the cytoplasm, inhibited up-regulation of Toll-like receptor 9 (TLR9) pathway factors MYD88 and RAGE, and limited RAGE interaction with its ligand TFAM in septic hearts. Mito-Vit-E also deactivated NF-κB and caspase 1, reduced expression of the essential inflammasome component ASC, and decreased inflammatory cytokine IL–1β. In vitro, both Mito-Vit-E and TLR9 inhibitor OND-I suppressed LPS-induced up-regulation in MYD88, RAGE, ASC, active caspase 1, and IL–1β in cardiomyocytes. Since free mtDNA escaped from damaged mitochondria function as a type of DAMPs to stimulate inflammation through TLR9, these data together suggest that sepsis-induced cardiac inflammation is mediated, at least partially, through mtDNA-TLR9-RAGE. At last, Mito-Vit-E reduced the circulation of myocardial injury marker troponin-I, diminished apoptosis and amended morphology in septic hearts, suggesting that mitochondria-targeted antioxidants are a potential cardioprotective approach for sepsis.
Collapse
|
386
|
Granger DN, Kvietys PR. Reperfusion injury and reactive oxygen species: The evolution of a concept. Redox Biol 2015; 6:524-551. [PMID: 26484802 PMCID: PMC4625011 DOI: 10.1016/j.redox.2015.08.020] [Citation(s) in RCA: 974] [Impact Index Per Article: 97.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 12/11/2022] Open
Abstract
Reperfusion injury, the paradoxical tissue response that is manifested by blood flow-deprived and oxygen-starved organs following the restoration of blood flow and tissue oxygenation, has been a focus of basic and clinical research for over 4-decades. While a variety of molecular mechanisms have been proposed to explain this phenomenon, excess production of reactive oxygen species (ROS) continues to receive much attention as a critical factor in the genesis of reperfusion injury. As a consequence, considerable effort has been devoted to identifying the dominant cellular and enzymatic sources of excess ROS production following ischemia-reperfusion (I/R). Of the potential ROS sources described to date, xanthine oxidase, NADPH oxidase (Nox), mitochondria, and uncoupled nitric oxide synthase have gained a status as the most likely contributors to reperfusion-induced oxidative stress and represent priority targets for therapeutic intervention against reperfusion-induced organ dysfunction and tissue damage. Although all four enzymatic sources are present in most tissues and are likely to play some role in reperfusion injury, priority and emphasis has been given to specific ROS sources that are enriched in certain tissues, such as xanthine oxidase in the gastrointestinal tract and mitochondria in the metabolically active heart and brain. The possibility that multiple ROS sources contribute to reperfusion injury in most tissues is supported by evidence demonstrating that redox-signaling enables ROS produced by one enzymatic source (e.g., Nox) to activate and enhance ROS production by a second source (e.g., mitochondria). This review provides a synopsis of the evidence implicating ROS in reperfusion injury, the clinical implications of this phenomenon, and summarizes current understanding of the four most frequently invoked enzymatic sources of ROS production in post-ischemic tissue. Reperfusion injury is implicated in a variety of human diseases and disorders. Evidence implicating ROS in reperfusion injury continues to grow. Several enzymes are candidate sources of ROS in post-ischemic tissue. Inter-enzymatic ROS-dependent signaling enhances the oxidative stress caused by I/R. .
Collapse
Affiliation(s)
- D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, United States.
| | - Peter R Kvietys
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
387
|
Adesina SE, Kang BY, Bijli KM, Ma J, Cheng J, Murphy TC, Michael Hart C, Sutliff RL. Targeting mitochondrial reactive oxygen species to modulate hypoxia-induced pulmonary hypertension. Free Radic Biol Med 2015; 87:36-47. [PMID: 26073127 PMCID: PMC4615392 DOI: 10.1016/j.freeradbiomed.2015.05.042] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 05/08/2015] [Accepted: 05/28/2015] [Indexed: 12/11/2022]
Abstract
Pulmonary hypertension (PH) is characterized by increased pulmonary vascular remodeling, resistance, and pressures. Reactive oxygen species (ROS) contribute to PH-associated vascular dysfunction. NADPH oxidases (Nox) and mitochondria are major sources of superoxide (O(2)(•-)) and hydrogen peroxide (H(2)O(2)) in pulmonary vascular cells. Hypoxia, a common stimulus of PH, increases Nox expression and mitochondrial ROS (mtROS) production. The interactions between these two sources of ROS generation continue to be defined. We hypothesized that mitochondria-derived O(2)(•-) (mtO(2)(•-)) and H(2)O(2) (mtH(2)O(2)) increase Nox expression to promote PH pathogenesis and that mitochondria-targeted antioxidants can reduce mtROS, Nox expression, and hypoxia-induced PH. Exposure of human pulmonary artery endothelial cells to hypoxia for 72 h increased mtO(2)(•-) and mtH(2)O(2). To assess the contribution of mtO(2)(•-) and mtH(2)O(2) to hypoxia-induced PH, mice that overexpress superoxide dismutase 2 (Tg(hSOD2)) or mitochondria-targeted catalase (MCAT) were exposed to normoxia (21% O(2)) or hypoxia (10% O(2)) for three weeks. Compared with hypoxic control mice, MCAT mice developed smaller hypoxia-induced increases in RVSP, α-SMA staining, extracellular H(2)O(2) (Amplex Red), Nox2 and Nox4 (qRT-PCR and Western blot), or cyclinD1 and PCNA (Western blot). In contrast, Tg(hSOD2) mice experienced exacerbated responses to hypoxia. These studies demonstrate that hypoxia increases mtO(2)(•-) and mtH(2)O(2). Targeting mtH(2)O(2) attenuates PH pathogenesis, whereas targeting mtO(2)(•-) exacerbates PH. These differences in PH pathogenesis were mirrored by RVSP, vessel muscularization, levels of Nox2 and Nox4, proliferation, and H(2)O(2) release. These studies suggest that targeted reductions in mtH(2)O(2) generation may be particularly effective in preventing hypoxia-induced PH.
Collapse
Affiliation(s)
- Sherry E Adesina
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Bum-Yong Kang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Kaiser M Bijli
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Jing Ma
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Juan Cheng
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Tamara C Murphy
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - C Michael Hart
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Roy L Sutliff
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA.
| |
Collapse
|
388
|
Chaplin NL, Nieves-Cintrón M, Fresquez AM, Navedo MF, Amberg GC. Arterial Smooth Muscle Mitochondria Amplify Hydrogen Peroxide Microdomains Functionally Coupled to L-Type Calcium Channels. Circ Res 2015; 117:1013-23. [PMID: 26390880 DOI: 10.1161/circresaha.115.306996] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/18/2015] [Indexed: 01/21/2023]
Abstract
RATIONALE Mitochondria are key integrators of convergent intracellular signaling pathways. Two important second messengers modulated by mitochondria are calcium and reactive oxygen species. To date, coherent mechanisms describing mitochondrial integration of calcium and oxidative signaling in arterial smooth muscle are incomplete. OBJECTIVE To address and add clarity to this issue, we tested the hypothesis that mitochondria regulate subplasmalemmal calcium and hydrogen peroxide microdomain signaling in cerebral arterial smooth muscle. METHODS AND RESULTS Using an image-based approach, we investigated the impact of mitochondrial regulation of L-type calcium channels on subcellular calcium and reactive oxygen species signaling microdomains in isolated arterial smooth muscle cells. Our single-cell observations were then related experimentally to intact arterial segments and to living animals. We found that subplasmalemmal mitochondrial amplification of hydrogen peroxide microdomain signaling stimulates L-type calcium channels, and that this mechanism strongly impacts the functional capacity of the vasoconstrictor angiotensin II. Importantly, we also found that disrupting this mitochondrial amplification mechanism in vivo normalized arterial function and attenuated the hypertensive response to systemic endothelial dysfunction. CONCLUSIONS From these observations, we conclude that mitochondrial amplification of subplasmalemmal calcium and hydrogen peroxide microdomain signaling is a fundamental mechanism regulating arterial smooth muscle function. As the principle components involved are fairly ubiquitous and positioning of mitochondria near the plasma membrane is not restricted to arterial smooth muscle, this mechanism could occur in many cell types and contribute to pathological elevations of intracellular calcium and increased oxidative stress associated with many diseases.
Collapse
Affiliation(s)
- Nathan L Chaplin
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.)
| | - Madeline Nieves-Cintrón
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.)
| | - Adriana M Fresquez
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.)
| | - Manuel F Navedo
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.)
| | - Gregory C Amberg
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.).
| |
Collapse
|
389
|
Reactive oxygen species and mitochondria: A nexus of cellular homeostasis. Redox Biol 2015; 6:472-485. [PMID: 26432659 PMCID: PMC4596921 DOI: 10.1016/j.redox.2015.09.005] [Citation(s) in RCA: 718] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are integral components of multiple cellular pathways even though excessive or inappropriately localized ROS damage cells. ROS function as anti-microbial effector molecules and as signaling molecules that regulate such processes as NF-kB transcriptional activity, the production of DNA-based neutrophil extracellular traps (NETs), and autophagy. The main sources of cellular ROS are mitochondria and NADPH oxidases (NOXs). In contrast to NOX-generated ROS, ROS produced in the mitochondria (mtROS) were initially considered to be unwanted by-products of oxidative metabolism. Increasing evidence indicates that mtROS have been incorporated into signaling pathways including those regulating immune responses and autophagy. As metabolic hubs, mitochondria facilitate crosstalk between the metabolic state of the cell with these pathways. Mitochondria and ROS are thus a nexus of multiple pathways that determine the response of cells to disruptions in cellular homeostasis such as infection, sterile damage, and metabolic imbalance. In this review, we discuss the roles of mitochondria in the generation of ROS-derived anti-microbial effectors, the interplay of mitochondria and ROS with autophagy and the formation of DNA extracellular traps, and activation of the NLRP3 inflammasome by ROS and mitochondria.
Collapse
|
390
|
Abstract
Aging results in progressive deteriorations in the structure and function of the heart and is a dominant risk factor for cardiovascular diseases, the leading cause of death in Western populations. Although the phenotypes of cardiac aging have been well characterized, the molecular mechanisms of cardiac aging are just beginning to be revealed. With the continuously growing elderly population, there is a great need for interventions in cardiac aging. This article will provide an overview of the phenotypic changes of cardiac aging, the molecular mechanisms underlying these changes, and will present some of the recent advances in the development of interventions to delay or reverse cardiac aging.
Collapse
Affiliation(s)
- Ying Ann Chiao
- Department of Pathology, University of Washington, Seattle, Washington 98195
| | - Peter S Rabinovitch
- Department of Pathology, University of Washington, Seattle, Washington 98195
| |
Collapse
|
391
|
Quarles EK, Dai DF, Tocchi A, Basisty N, Gitari L, Rabinovitch PS. Quality control systems in cardiac aging. Ageing Res Rev 2015; 23:101-15. [PMID: 25702865 PMCID: PMC4686341 DOI: 10.1016/j.arr.2015.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 02/02/2015] [Accepted: 02/12/2015] [Indexed: 12/31/2022]
Abstract
Cardiac aging is an intrinsic process that results in impaired cardiac function, along with cellular and molecular changes. These degenerative changes are intimately associated with quality control mechanisms. This review provides a general overview of the clinical and cellular changes which manifest in cardiac aging, and the quality control mechanisms involved in maintaining homeostasis and retarding aging. These mechanisms include autophagy, ubiquitin-mediated turnover, apoptosis, mitochondrial quality control and cardiac matrix homeostasis. Finally, we discuss aging interventions that have been observed to impact cardiac health outcomes. These include caloric restriction, rapamycin, resveratrol, GDF11, mitochondrial antioxidants and cardiolipin-targeted therapeutics. A greater understanding of the quality control mechanisms that promote cardiac homeostasis will help to understand the benefits of these interventions, and hopefully lead to further improved therapeutic modalities.
Collapse
Affiliation(s)
- Ellen K Quarles
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Dao-Fu Dai
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Autumn Tocchi
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Nathan Basisty
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Lemuel Gitari
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Peter S Rabinovitch
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| |
Collapse
|
392
|
Steven S, Münzel T, Daiber A. Exploiting the Pleiotropic Antioxidant Effects of Established Drugs in Cardiovascular Disease. Int J Mol Sci 2015; 16:18185-223. [PMID: 26251902 PMCID: PMC4581241 DOI: 10.3390/ijms160818185] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 07/20/2015] [Accepted: 07/27/2015] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease is a leading cause of death and reduced quality of life worldwide. Arterial vessels are a primary target for endothelial dysfunction and atherosclerosis, which is accompanied or even driven by increased oxidative stress. Recent research in this field identified different sources of reactive oxygen and nitrogen species contributing to the pathogenesis of endothelial dysfunction. According to lessons from the past, improvement of endothelial function and prevention of cardiovascular disease by systemic, unspecific, oral antioxidant therapy are obviously too simplistic an approach. Source- and cell organelle-specific antioxidants as well as activators of intrinsic antioxidant defense systems might be more promising. Since basic research demonstrated the contribution of different inflammatory cells to vascular oxidative stress and clinical trials identified chronic inflammatory disorders as risk factors for cardiovascular events, atherosclerosis and cardiovascular disease are closely associated with inflammation. Therefore, modulation of the inflammatory response is a new and promising approach in the therapy of cardiovascular disease. Classical anti-inflammatory therapeutic compounds, but also established drugs with pleiotropic immunomodulatory abilities, demonstrated protective effects in various models of cardiovascular disease. However, results from ongoing clinical trials are needed to further evaluate the value of immunomodulation for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Sebastian Steven
- Medical Clinic, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany.
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany.
| | - Thomas Münzel
- Medical Clinic, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany.
| | - Andreas Daiber
- Medical Clinic, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany.
| |
Collapse
|
393
|
Mitochondrial Oxidative Stress, Mitochondrial DNA Damage and Their Role in Age-Related Vascular Dysfunction. Int J Mol Sci 2015; 16:15918-53. [PMID: 26184181 PMCID: PMC4519931 DOI: 10.3390/ijms160715918] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/17/2015] [Accepted: 06/29/2015] [Indexed: 02/06/2023] Open
Abstract
The prevalence of cardiovascular diseases is significantly increased in the older population. Risk factors and predictors of future cardiovascular events such as hypertension, atherosclerosis, or diabetes are observed with higher frequency in elderly individuals. A major determinant of vascular aging is endothelial dysfunction, characterized by impaired endothelium-dependent signaling processes. Increased production of reactive oxygen species (ROS) leads to oxidative stress, loss of nitric oxide (•NO) signaling, loss of endothelial barrier function and infiltration of leukocytes to the vascular wall, explaining the low-grade inflammation characteristic for the aged vasculature. We here discuss the importance of different sources of ROS for vascular aging and their contribution to the increased cardiovascular risk in the elderly population with special emphasis on mitochondrial ROS formation and oxidative damage of mitochondrial DNA. Also the interaction (crosstalk) of mitochondria with nicotinamide adenosine dinucleotide phosphate (NADPH) oxidases is highlighted. Current concepts of vascular aging, consequences for the development of cardiovascular events and the particular role of ROS are evaluated on the basis of cell culture experiments, animal studies and clinical trials. Present data point to a more important role of oxidative stress for the maximal healthspan (healthy aging) than for the maximal lifespan.
Collapse
|
394
|
Jankovic A, Korac A, Buzadzic B, Otasevic V, Stancic A, Daiber A, Korac B. Redox implications in adipose tissue (dys)function--A new look at old acquaintances. Redox Biol 2015; 6:19-32. [PMID: 26177468 PMCID: PMC4511633 DOI: 10.1016/j.redox.2015.06.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/25/2015] [Accepted: 06/30/2015] [Indexed: 12/15/2022] Open
Abstract
Obesity is an energy balance disorder associated with dyslipidemia, insulin resistance and diabetes type 2, also summarized with the term metabolic syndrome or syndrome X. Increasing evidence points to “adipocyte dysfunction”, rather than fat mass accretion per se, as the key pathophysiological factor for metabolic complications in obesity. The dysfunctional fat tissue in obesity characterizes a failure to safely store metabolic substrates into existing hypertrophied adipocytes and/or into new preadipocytes recruited for differentiation. In this review we briefly summarize the potential of redox imbalance in fat tissue as an instigator of adipocyte dysfunction in obesity. We reveal the challenge of the adipose redox changes, insights in the regulation of healthy expansion of adipose tissue and its reduction, leading to glucose and lipids overflow. Adipose tissue (AT) buffers nutrient excess determining overall metabolic health. Redox insight in lipid storage and adipogenesis of AT is reviewed. Redox modulation of AT as therapeutic target in obesity/syndrome X is considered.
Collapse
Affiliation(s)
- Aleksandra Jankovic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Aleksandra Korac
- University of Belgrade, Faculty of Biology, Center for Electron Microscopy, Belgrade, Serbia
| | - Biljana Buzadzic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Vesna Otasevic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Ana Stancic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Andreas Daiber
- 2nd Medical Department, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Bato Korac
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia.
| |
Collapse
|
395
|
Lee DH, Park HY, Lee US, Lee KJ, Noh EC, Jang JH, Kang DH. The effects of brain wave vibration on oxidative stress response and psychological symptoms. Compr Psychiatry 2015; 60:99-104. [PMID: 25842193 DOI: 10.1016/j.comppsych.2015.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/15/2015] [Accepted: 03/16/2015] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVE Brain Wave Vibration (BWV) training is a simple healing practice, a kind of Mind Body Training. This study was designed to investigate the psycho-endocrine differences between BMV practitioners and naïve controls. METHODS The experimental group included 54 individuals who had participated in BWV. The control group included 58 subjects who had not participated in formal BWV. Levels of plasma NO, reactive oxygen species (ROS), and superoxide dismutase (SOD) were measured, and the modified form of the Stress Response Inventory (SRI-MF), the Positive Affect and Negative Affect Scale (PANAS), the Beck Depression Inventory (BDI), and the Beck Anxiety Inventory (BAI) were administered. RESULTS The BWV group demonstrated significantly higher plasma NO levels (p=0.003), and levels of ROS and SOD did not differ between the two groups. The BWV group showed lower scores in BDI (p=0.009), BAI (p=0.009) and stress level (p<0.001) and higher scores on positive affect (p=0.023) compared with the control group. NO levels were associated with increased positive affect (p = 0.024) only in BWV subjects. CONCLUSION BWV may increase NO, a relaxation-related factor, possibly by improving emotional state.
Collapse
Affiliation(s)
- Do-Hyeong Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Hye Yoon Park
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Ul Soon Lee
- Global Cyber University, Cheonan, Republic of Korea
| | - Kyung-Jun Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Eun Chung Noh
- Interdisciplinary Program of Neuroscience, Seoul National University, Seoul, Republic of Korea
| | - Joon Hwan Jang
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Do-Hyung Kang
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
396
|
Antognelli C, Gambelunghe A, Muzi G, Talesa VN. Peroxynitrite-mediated glyoxalase I epigenetic inhibition drives apoptosis in airway epithelial cells exposed to crystalline silica via a novel mechanism involving argpyrimidine-modified Hsp70, JNK, and NF-κB. Free Radic Biol Med 2015; 84:128-141. [PMID: 25841781 DOI: 10.1016/j.freeradbiomed.2015.03.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 03/18/2015] [Accepted: 03/22/2015] [Indexed: 12/11/2022]
Abstract
Glyoxalase I (Glo1) is a cellular defense enzyme involved in the detoxification of methylglyoxal (MG), a cytotoxic by-product of glycolysis, and MG-derived advanced glycation end products (AGEs). Argpyrimidine (AP), one of the major AGEs coming from MG modification of protein arginines, is a proapoptotic agent. Crystalline silica is a well-known occupational health hazard, responsible for a relevant number of pulmonary diseases. Exposure of cells to crystalline silica results in a number of complex biological responses, including apoptosis. The present study was aimed at investigating whether, and through which mechanism, Glo1 was involved in Min-U-Sil 5 crystalline silica-induced apoptosis. Apoptosis, by TdT-mediated dUTP nick-end labeling assay, and transcript and protein levels or enzymatic activity, by quantitative real-time PCR, Western blot, and spectrophotometric methods, respectively, were evaluated in human bronchial BEAS-2B cells exposed or not (control) to crystalline silica and also in experiments with appropriate inhibitors. Reactive oxygen species were evaluated by coumarin-7-boronic acid or Amplex red hydrogen peroxide/peroxidase methods for peroxynitrite (ONOO(-)) or hydrogen peroxide (H2O2) measurements, respectively. Our results showed that Min-U-Sil 5 crystalline silica induced a dramatic ONOO(-)-mediated inhibition of Glo1, leading to AP-modified Hsp70 protein accumulation that, in a mechanism involving JNK and NF-κB, triggered an apoptotic mitochondrial pathway. Inhibition of Glo1 occurred at both functional and transcriptional levels, the latter occurring via ERK1/2 MAPK and miRNA 101 involvement. Taken together, our data demonstrate that Glo1 is involved in the Min-U-Sil 5 crystalline silica-induced BEAS-2B cell mitochondrial apoptotic pathway via a novel mechanism involving Hsp70, JNK, and NF-κB. Because maintenance of an intact respiratory epithelium is a critically important determinant of normal respiratory function, the knowledge of the mechanisms underlying its disruption may provide insight into the genesis, and possibly the prevention, of a number of pathological conditions commonly occurring in silica dust occupational exposure.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Department of Experimental Medicine University of Perugia, 06129 Perugia, Italy.
| | - Angela Gambelunghe
- Department of Medicine, School of Medicine, University of Perugia, 06129 Perugia, Italy
| | - Giacomo Muzi
- Department of Medicine, School of Medicine, University of Perugia, 06129 Perugia, Italy
| | | |
Collapse
|
397
|
Kim JS, Kim B, Lee H, Thakkar S, Babbitt DM, Eguchi S, Brown MD, Park JY. Shear stress-induced mitochondrial biogenesis decreases the release of microparticles from endothelial cells. Am J Physiol Heart Circ Physiol 2015; 309:H425-33. [PMID: 26024684 DOI: 10.1152/ajpheart.00438.2014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 05/26/2015] [Indexed: 01/21/2023]
Abstract
The concept of enhancing structural integrity of mitochondria has emerged as a novel therapeutic option for cardiovascular disease. Flow-induced increase in laminar shear stress is a potent physiological stimulant associated with exercise, which exerts atheroprotective effects in the vasculature. However, the effect of laminar shear stress on mitochondrial remodeling within the vascular endothelium and its related functional consequences remain largely unknown. Using in vitro and in vivo complementary studies, here, we report that aerobic exercise alleviates the release of endothelial microparticles in prehypertensive individuals and that these salutary effects are, in part, mediated by shear stress-induced mitochondrial biogenesis. Circulating levels of total (CD31(+)/CD42a(-)) and activated (CD62E(+)) microparticles released by endothelial cells were significantly decreased (∼40% for both) after a 6-mo supervised aerobic exercise training program in individuals with prehypertension. In cultured human endothelial cells, laminar shear stress reduced the release of endothelial microparticles, which was accompanied by an increase in mitochondrial biogenesis through a sirtuin 1 (SIRT1)-dependent mechanism. Resveratrol, a SIRT1 activator, treatment showed similar effects. SIRT1 knockdown using small-interfering RNA completely abolished the protective effect of shear stress. Disruption of mitochondrial integrity by either antimycin A or peroxisome proliferator-activated receptor-γ coactivator-1α small-interfering RNA significantly increased the number of total, and activated, released endothelial microparticles, and shear stress restored these back to basal levels. Collectively, these data demonstrate a critical role of endothelial mitochondrial integrity in preserving endothelial homeostasis. Moreover, prolonged laminar shear stress, which is systemically elevated during aerobic exercise in the vessel wall, mitigates endothelial dysfunction by promoting mitochondrial biogenesis.
Collapse
Affiliation(s)
- Ji-Seok Kim
- Department of Kinesiology, Temple University, Philadelphia, Pennsylvania
| | - Boa Kim
- Department of Kinesiology, Temple University, Philadelphia, Pennsylvania; Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Hojun Lee
- Department of Kinesiology, Temple University, Philadelphia, Pennsylvania
| | - Sunny Thakkar
- Department of Kinesiology, Temple University, Philadelphia, Pennsylvania
| | - Dianne M Babbitt
- Department of Kinesiology, Temple University, Philadelphia, Pennsylvania
| | - Satoru Eguchi
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Michael D Brown
- Department of Kinesiology and Nutrition and Integrative Physiology Laboratory, University of Illinois at Chicago, Chicago, Illinois
| | - Joon-Young Park
- Department of Kinesiology, Temple University, Philadelphia, Pennsylvania; Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| |
Collapse
|
398
|
van Thiel BS, van der Pluijm I, te Riet L, Essers J, Danser AHJ. The renin-angiotensin system and its involvement in vascular disease. Eur J Pharmacol 2015; 763:3-14. [PMID: 25987425 DOI: 10.1016/j.ejphar.2015.03.090] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/15/2015] [Accepted: 03/24/2015] [Indexed: 10/24/2022]
Abstract
The renin-angiotensin system (RAS) plays a critical role in the pathogenesis of many types of cardiovascular diseases including cardiomyopathy, valvular heart disease, aneurysms, stroke, coronary artery disease and vascular injury. Besides the classical regulatory effects on blood pressure and sodium homoeostasis, the RAS is involved in the regulation of contractility and remodelling of the vessel wall. Numerous studies have shown beneficial effect of inhibition of this system in the pathogenesis of cardiovascular diseases. However, dysregulation and overexpression of the RAS, through different molecular mechanisms, also induces, the initiation of vascular damage. The key effector peptide of the RAS, angiotensin II (Ang II) promotes cell proliferation, apoptosis, fibrosis, oxidative stress and inflammation, processes known to contribute to remodelling of the vasculature. In this review, we focus on the components that are under the influence of the RAS and contribute to the development and progression of vascular disease; extracellular matrix defects, atherosclerosis and ageing. Furthermore, the beneficial therapeutic effects of inhibition of the RAS on the vasculature are discussed, as well as the need for additive effects on top of RAS inhibition.
Collapse
Affiliation(s)
- Bibi S van Thiel
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC, Rotterdam, The Netherlands; Department of Genetics, Erasmus MC, Rotterdam, The Netherlands; Department of Vascular Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Ingrid van der Pluijm
- Department of Genetics, Erasmus MC, Rotterdam, The Netherlands; Department of Vascular Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Luuk te Riet
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC, Rotterdam, The Netherlands; Department of Vascular Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Jeroen Essers
- Department of Genetics, Erasmus MC, Rotterdam, The Netherlands; Department of Vascular Surgery, Erasmus MC, Rotterdam, The Netherlands; Department of Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
399
|
Szewczyk A, Jarmuszkiewicz W, Koziel A, Sobieraj I, Nobik W, Lukasiak A, Skup A, Bednarczyk P, Drabarek B, Dymkowska D, Wrzosek A, Zablocki K. Mitochondrial mechanisms of endothelial dysfunction. Pharmacol Rep 2015; 67:704-10. [PMID: 26321271 DOI: 10.1016/j.pharep.2015.04.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/10/2015] [Accepted: 04/14/2015] [Indexed: 01/09/2023]
Abstract
Endothelial cells play an important physiological role in vascular homeostasis. They are also the first barrier that separates blood from deeper layers of blood vessels and extravascular tissues. Thus, they are exposed to various physiological blood components as well as challenged by pathological stimuli, which may exert harmful effects on the vascular system by stimulation of excessive generation of reactive oxygen species (ROS). The major sources of ROS are NADPH oxidase and mitochondrial respiratory chain complexes. Modulation of mitochondrial energy metabolism in endothelial cells is thought to be a promising target for therapy in various cardiovascular diseases. Uncoupling protein 2 (UCP2) is a regulator of mitochondrial ROS generation and can antagonise oxidative stress-induced endothelial dysfunction. Several studies have revealed the important role of UCP2 in hyperglycaemia-induced modifications of mitochondrial function in endothelial cells. Additionally, potassium fluxes through the inner mitochondrial membrane, which are involved in ROS synthesis, affect the mitochondrial volume and change both the mitochondrial membrane potential and the transport of calcium into the mitochondria. In this review, we concentrate on the mitochondrial role in the cytoprotection phenomena of endothelial cells.
Collapse
Affiliation(s)
- Adam Szewczyk
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warszawa, Poland
| | | | - Agnieszka Koziel
- Department of Bioenergetics, Adam Mickiewicz University, Poznań, Poland
| | - Izabela Sobieraj
- Department of Bioenergetics, Adam Mickiewicz University, Poznań, Poland
| | - Wioletta Nobik
- Department of Bioenergetics, Adam Mickiewicz University, Poznań, Poland
| | - Agnieszka Lukasiak
- Department of Biophysics, Warsaw University of Life Sciences-SGGW, Warszawa, Poland
| | - Agata Skup
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warszawa, Poland
| | - Piotr Bednarczyk
- Department of Biophysics, Warsaw University of Life Sciences-SGGW, Warszawa, Poland
| | - Beata Drabarek
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Warszawa, Poland
| | - Dorota Dymkowska
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Warszawa, Poland
| | - Antoni Wrzosek
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warszawa, Poland.
| | - Krzysztof Zablocki
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Warszawa, Poland
| |
Collapse
|
400
|
Effect of heat stress-induced production of mitochondrial reactive oxygen species on NADPH oxidase and heme oxygenase-1 mRNA levels in avian muscle cells. J Therm Biol 2015; 52:8-13. [PMID: 26267493 DOI: 10.1016/j.jtherbio.2015.04.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 04/21/2015] [Accepted: 04/29/2015] [Indexed: 11/21/2022]
Abstract
Heat stress is a major factor inducing oxidative disturbance in cells. In the present study, we investigated the mechanism of overproduction of reactive oxygen species (ROS) in cultured avian muscle cells in response to heat stress, and also focused attention on the interaction of mitochondrial superoxide anions with altered NADPH oxidase (NOX), superoxide dismutase (SOD) and heme oxygenase-1 (HO-1) mRNA levels in heat-stressed cells. Exposure of cells to heat stress conditions (41°C, 6h) resulted in increased mitochondrial superoxide and intracellular ROS levels, and increased carbonyl protein content as compared with that of normal cells (37°C). The mitochondrial uncoupler 2,4-dinitrophenol lowered intracellular ROS levels in heat-stressed cells. Heat stress increased NOX4 mRNA and decreased HO-1 mRNA levels, while SOD1 and SOD2 mRNA levels remained relatively stable in heat-stressed cells. Addition of the superoxide scavenger 4-hydroxy TEMPO to the culture medium of heat-stressed cells restored mitochondrial superoxide and intracellular ROS levels as well as NOX4 and HO-1 mRNA levels to near-normal values. We suggest that mitochondrial superoxide production could play an influential role in augmenting oxidative damage to avian muscle cells, possibly via the up-regulation of NOX4 and down-regulation of HO-1 in heat-stressed avian muscle cells.
Collapse
|