151
|
1,3,4-Thiadiazole derivatives as selective inhibitors of iNOS versus nNOS: Synthesis and structure-activity dependence. Eur J Med Chem 2012; 50:129-39. [DOI: 10.1016/j.ejmech.2012.01.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 01/20/2012] [Accepted: 01/24/2012] [Indexed: 11/18/2022]
|
152
|
Yao SY, Natarajan C, Sriram S. nNOS mediated mitochondrial injury in LPS stimulated oligodendrocytes. Mitochondrion 2012; 12:336-44. [DOI: 10.1016/j.mito.2012.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 01/03/2012] [Accepted: 01/12/2012] [Indexed: 02/06/2023]
|
153
|
Barros L, Dueñas M, Carvalho AM, Ferreira ICFR, Santos-Buelga C. Characterization of phenolic compounds in flowers of wild medicinal plants from Northeastern Portugal. Food Chem Toxicol 2012; 50:1576-82. [PMID: 22342808 DOI: 10.1016/j.fct.2012.02.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 02/01/2012] [Accepted: 02/03/2012] [Indexed: 11/30/2022]
Abstract
Crataegus monogyna, Cytisus multiflorus, Malva sylvestris and Sambucus nigra have been used as important medicinal plants in the Iberian Peninsula since a long time ago, and are claimed to have various health benefits. This study aimed to determine the phenolic profile and composition of wild medicinal flowers of those species. The analysis was performed by HPLC-DAD-ESI/MS. Flavonoids, and particularly flavonols and flavones, were the main groups in almost all the studied samples. C. multiflorus sample gave the highest levels of total flavonoids (54.5 mg/gdw), being a chrysin derivative the most abundant flavone found (22.3 mg/gdw). C. monogyna revealed the highest concentration in phenolic acids (5.5 mg/gdw) that were not found in C. multiflorus sample; 5-O-caffeoylquinic acid was the most abundant phenolic acid found in the first species, being a procyanidin trimer also found (1.4 mg/gdw). Kaempferol-3-O-rutinoside (0.84 mg/gdw) and quercetin-3-O-rutinoside (14.9 mg/gdw) were the main flavonols present in M. sylvestris and S. nigra, respectively. Due to the well established antioxidant activity of phenolic compounds, the studied wild medicinal flowers could be selected for processing extracts with health-promoting properties or to be incorporate into functional beverages or products with bioactive properties related to oxidative stress.
Collapse
Affiliation(s)
- Lillian Barros
- CIMO/Escola Superior Agrária, Instituto Politécnico de Bragança, Campus de Santa Apolónia, Apartado 1172, 5301-855 Bragança, Portugal
| | | | | | | | | |
Collapse
|
154
|
Agarwal R, Hennings L, Rafferty TM, Letzig LG, McCullough S, James LP, MacMillan-Crow LA, Hinson JA. Acetaminophen-induced hepatotoxicity and protein nitration in neuronal nitric-oxide synthase knockout mice. J Pharmacol Exp Ther 2012; 340:134-42. [PMID: 22001257 PMCID: PMC3251022 DOI: 10.1124/jpet.111.184192] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 10/13/2011] [Indexed: 12/17/2022] Open
Abstract
In overdose acetaminophen (APAP) is hepatotoxic. Toxicity occurs by metabolism to N-acetyl-p-benzoquinone imine, which depletes GSH and covalently binds to proteins followed by protein nitration. Nitration can occur via the strong oxidant and nitrating agent peroxynitrite, formed from superoxide and nitric oxide (NO). In hepatocyte suspensions we reported that an inhibitor of neuronal nitric-oxide synthase (nNOS; NOS1), which has been reported to be in mitochondria, inhibited toxicity and protein nitration. We recently showed that manganese superoxide dismutase (MnSOD; SOD2) was nitrated and inactivated in APAP-treated mice. To understand the role of nNOS in APAP toxicity and MnSOD nitration, nNOS knockout (KO) and wild-type (WT) mice were administered APAP (300 mg/kg). In WT mice serum alanine aminotransferase (ALT) significantly increased at 6 and 8 h, and serum aspartate aminotransferase (AST) significantly increased at 4, 6 and 8 h; however, in KO mice neither ALT nor AST significantly increased until 8 h. There were no significant differences in hepatic GSH depletion, APAP protein binding, hydroxynonenal covalent binding, or histopathological assessment of toxicity. The activity of hepatic MnSOD was significantly lower at 1 to 2 h in WT mice and subsequently increased at 8 h. MnSOD activity was not altered at 0 to 6 h in KO mice but was significantly decreased at 8 h. There were significant increases in MnSOD nitration at 1 to 8 h in WT mice and 6 to 8 h in KO mice. Significantly more nitration occurred at 1 to 6 h in WT than in KO mice. MnSOD was the only observed nitrated protein after APAP treatment. These data indicate a role for nNOS with inactivation of MnSOD and ALT release during APAP toxicity.
Collapse
Affiliation(s)
- Rakhee Agarwal
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | | | | | | | | | | | | |
Collapse
|
155
|
Sharma V, McNeill JH. Parallel effects of β-adrenoceptor blockade on cardiac function and fatty acid oxidation in the diabetic heart: Confronting the maze. World J Cardiol 2011; 3:281-302. [PMID: 21949571 PMCID: PMC3176897 DOI: 10.4330/wjc.v3.i9.281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 07/18/2011] [Accepted: 07/25/2011] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy is a disease process in which diabetes produces a direct and continuous myocardial insult even in the absence of ischemic, hypertensive or valvular disease. The β-blocking agents bisoprolol, carvedilol and metoprolol have been shown in large-scale randomized controlled trials to reduce heart failure mortality. In this review, we summarize the results of our studies investigating the effects of β-blocking agents on cardiac function and metabolism in diabetic heart failure, and the complex inter-related mechanisms involved. Metoprolol inhibits fatty acid oxidation at the mitochondrial level but does not prevent lipotoxicity; its beneficial effects are more likely to be due to pro-survival effects of chronic treatment. These studies have expanded our understanding of the range of effects produced by β-adrenergic blockade and show how interconnected the signaling pathways of function and metabolism are in the heart. Although our initial hypothesis that inhibition of fatty acid oxidation would be a key mechanism of action was disproved, unexpected results led us to some intriguing regulatory mechanisms of cardiac metabolism. The first was upstream stimulatory factor-2-mediated repression of transcriptional master regulator PGC-1α, most likely occurring as a consequence of the improved function; it is unclear whether this effect is unique to β-blockers, although repression of carnitine palmitoyltransferase (CPT)-1 has not been reported with other drugs which improve function. The second was the identification of a range of covalent modifications which can regulate CPT-1 directly, mediated by a signalome at the level of the mitochondria. We also identified an important interaction between β-adrenergic signaling and caveolins, which may be a key mechanism of action of β-adrenergic blockade. Our experience with this labyrinthine signaling web illustrates that initial hypotheses and anticipated directions do not have to be right in order to open up meaningful directions or reveal new information.
Collapse
Affiliation(s)
- Vijay Sharma
- Vijay Sharma, John H McNeill, Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3.F, Canada
| | | |
Collapse
|
156
|
Klymkowsky M. Mitochondrial activity, embryogenesis, and the dialogue between the big and little brains of the cell. Mitochondrion 2011; 11:814-9. [DOI: 10.1016/j.mito.2010.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 11/02/2010] [Accepted: 11/18/2010] [Indexed: 12/31/2022]
|
157
|
Melatonin combats molecular terrorism at the mitochondrial level. Interdiscip Toxicol 2011; 1:137-49. [PMID: 21218104 PMCID: PMC2993480 DOI: 10.2478/v10102-010-0030-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Revised: 07/28/2008] [Accepted: 07/29/2008] [Indexed: 12/15/2022] Open
Abstract
The intracellular environmental is a hostile one. Free radicals and related oxygen and nitrogen-based oxidizing agents persistently pulverize and damage molecules in the vicinity of where they are formed. The mitochondria especially are subjected to frequent and abundant oxidative abuse. The carnage that is left in the wake of these oxygen and nitrogen-related reactants is referred to as oxidative damage or oxidative stress. When mitochondrial electron transport complex inhibitors are used, e.g., rotenone, 1-methyl-1-phenyl-1,2,3,6-tetrahydropyridine, 3-nitropropionic acid or cyanide, pandemonium breaks loose within mitochondria as electron leakage leads to the generation of massive amounts of free radicals and related toxicants. The resulting oxidative stress initiates a series of events that leads to cellular apoptosis. To alleviate mitochondrial destruction and the associated cellular implosion, the cell has at its disposal a variety of free radical scavengers and antioxidants. Among these are melatonin and its metabolites. While melatonin stimulates several antioxidative enzymes it, as well as its metabolites (cyclic 3-hydroxymelatonin, N1-acetyl-N2-formyl-5-methoxykynuramine and N1-acetyl-5-methoxykynuramine), likewise effectively neutralize free radicals. The resulting cascade of reactions greatly magnifies melatonin's efficacy in reducing oxidative stress and apoptosis even in the presence of mitochondrial electron transport inhibitors. The actions of melatonin at the mitochondrial level are a consequence of melatonin and/or any of its metabolites. Thus, the molecular terrorism meted out by reactive oxygen and nitrogen species is held in check by melatonin and its derivatives.
Collapse
|
158
|
|
159
|
Protective effects of synthetic kynurenines on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinsonism in mice. Brain Res Bull 2011; 85:133-40. [PMID: 21419832 DOI: 10.1016/j.brainresbull.2011.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 03/07/2011] [Accepted: 03/09/2011] [Indexed: 10/18/2022]
Abstract
Mitochondrial complex I inhibition is thought to underlie the neurodegenerative process in Parkinson's disease (PD). Moreover, an overproduction of nitric oxide due to both cytosolic (iNOS) and mitochondrial (i-mtNOS) inducible nitric oxide synthases causes free radicals generation and oxidative/nitrosative stress, contributing to mitochondrial dysfunction and neuronal cell death. Looking for active molecules against mitochondrial dysfunction and inflammatory response in PD, we show here the effects of four synthetic kynurenines in the MPTP model of PD in mice. After MPTP administration, mitochondria from substantia nigra and, in a lesser extent, from striatum showed a significant increase in i-mtNOS activity, nitric oxide production, oxidative stress, and complex I inhibition. The four kynurenines assayed counteracted the effects of MPTP, reducing iNOS/i-mtNOS activity, and restoring the activity of the complex I. Consequently, the cytosolic and mitochondrial oxidative/nitrosative stress returned to control values. The results suggest that the kynurenines here reported represent a family of synthetic compounds with neuroprotective properties against PD, and that they can serve as templates for the design of new drugs able to target the mitochondria.
Collapse
|
160
|
Chan WH. Photodynamic treatment induces an apoptotic pathway involving calcium, nitric oxide, p53, p21-activated kinase 2, and c-Jun N-terminal kinase and inactivates survival signal in human umbilical vein endothelial cells. Int J Mol Sci 2011; 12:1041-59. [PMID: 21541041 PMCID: PMC3083688 DOI: 10.3390/ijms12021041] [Citation(s) in RCA: 23] [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: 12/28/2010] [Revised: 01/25/2011] [Accepted: 02/02/2011] [Indexed: 01/24/2023] Open
Abstract
Photodynamic treatment (PDT) elicits a diverse range of cellular responses, including apoptosis. Previously, we showed that PDT stimulates caspase-3 activity, and subsequent cleavage and activation of p21-activated kinase 2 (PAK2) in human epidermal carcinoma A431 cells. In the current study, pretreatment with nitric oxide (NO) scavengers inhibited PDT-induced mitochondrial membrane potential (MMP) changes, activation of caspase-9, caspase-3, p21-activated protein kinase 2 (PAK2) and c-Jun N-terminal kinase (JNK), and gene expression of p53 and p21 involved in apoptotic signaling. Moreover, PAK2 activity was required for PDT-induced JNK activation and apoptosis. Inhibition of p53 mRNA expression using small interfering RNA (siRNA) additionally blocked activation of PAK2 and apoptosis induced by PDT. Importantly, our data also show that PDT triggers cell death via inactivation of ERK-mediated anti-apoptotic pathway. PDT triggers cell death via inactivation of the HSP90/multi-chaperone complex and subsequent degradation of Ras, further inhibiting anti-apoptotic processes, such as the Ras→ERK signal transduction pathway. Furthermore, we did not observe two-stage JNK activation for regulation of PAK2 activity in the PDT-induced apoptotic pathway in HUVECs, which was reported earlier in A431 cells. Based on the collective results, we have proposed a model for the PDT-triggered inactivation of the survival signal and apoptotic signaling cascade with Rose Bengal (RB), which sequentially involves singlet oxygen, Ca2+, NO, p53, caspase-9, caspase-3, PAK2, and JNK.
Collapse
Affiliation(s)
- Wen-Hsiung Chan
- Department of Bioscience Technology and Center for Nanotechnology, Chung Yuan Christian University, Chung Li 32023, Taiwan; E-Mail: ; Tel.: +886-3-2653515
| |
Collapse
|
161
|
Powers SK, Talbert EE, Adhihetty PJ. Reactive oxygen and nitrogen species as intracellular signals in skeletal muscle. J Physiol 2011; 589:2129-38. [PMID: 21224240 DOI: 10.1113/jphysiol.2010.201327] [Citation(s) in RCA: 444] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
It is well established that contracting skeletal muscles produce free radicals. Given that radicals are known to play a prominent role in the pathogenesis of several diseases, the 1980s-90s dogma was that contraction-induced radical production was detrimental to muscle because of oxidative damage to macromolecules within the fibre. In contrast to this early outlook, it is now clear that both reactive oxygen species (ROS) and reactive nitrogen species (RNS) play important roles in cell signalling pathways involved in muscle adaptation to exercise and the remodelling that occurs in skeletal muscle during periods of prolonged inactivity. This review will highlight two important redox sensitive signalling pathways that contribute to ROS and RNS-induced skeletal muscle adaptation to endurance exercise. We begin with a historical overview of radical production in skeletal muscles followed by a discussion of the intracellular sites for ROS and RNS production in muscle fibres. We will then provide a synopsis of the redox-sensitive NF-B and PGC-1α signalling pathways that contribute to skeletal muscle adaptation in response to exercise training. We will conclude with a discussion of unanswered questions in redox signalling in skeletal muscle in the hope of promoting additional research interest in this field.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA.
| | | | | |
Collapse
|
162
|
|
163
|
Reduction of Nω-hydroxy-L-arginine by the mitochondrial amidoxime reducing component (mARC). Biochem J 2010; 433:383-91. [DOI: 10.1042/bj20100960] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
NOSs (nitric oxide synthases) catalyse the oxidation of L-arginine to L-citrulline and nitric oxide via the intermediate NOHA (Nω-hydroxy-L-arginine). This intermediate is rapidly converted further, but to a small extent can also be liberated from the active site of NOSs and act as a transportable precursor of nitric oxide or potent physiological inhibitor of arginases. Thus its formation is of enormous importance for the nitric-oxide-generating system. It has also been shown that NOHA is reduced by microsomes and mitochondria to L-arginine. In the present study, we show for the first time that both human isoforms of the newly identified mARC (mitochondrial amidoxime reducing component) enhance the rate of reduction of NOHA, in the presence of NADH cytochrome b5 reductase and cytochrome b5, by more than 500-fold. Consequently, these results provide the first hints that mARC might be involved in mitochondrial NOHA reduction and could be of physiological significance in affecting endogenous nitric oxide levels. Possibly, this reduction represents another regulative mechanism in the complex regulation of nitric oxide biosynthesis, considering a mitochondrial NOS has been identified. Moreover, this reduction is not restricted to NOHA since the analogous arginase inhibitor NHAM (Nω-hydroxy-Nδ-methyl-L-arginine) is also reduced by this system.
Collapse
|
164
|
van Wessel T, de Haan A, van der Laarse WJ, Jaspers RT. The muscle fiber type-fiber size paradox: hypertrophy or oxidative metabolism? Eur J Appl Physiol 2010; 110:665-94. [PMID: 20602111 PMCID: PMC2957584 DOI: 10.1007/s00421-010-1545-0] [Citation(s) in RCA: 185] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2010] [Indexed: 12/11/2022]
Abstract
An inverse relationship exists between striated muscle fiber size and its oxidative capacity. This relationship implies that muscle fibers, which are triggered to simultaneously increase their mass/strength (hypertrophy) and fatigue resistance (oxidative capacity), increase these properties (strength or fatigue resistance) to a lesser extent compared to fibers increasing either of these alone. Muscle fiber size and oxidative capacity are determined by the balance between myofibrillar protein synthesis, mitochondrial biosynthesis and degradation. New experimental data and an inventory of critical stimuli and state of activation of the signaling pathways involved in regulating contractile and metabolic protein turnover reveal: (1) higher capacity for protein synthesis in high compared to low oxidative fibers; (2) competition between signaling pathways for synthesis of myofibrillar proteins and proteins associated with oxidative metabolism; i.e., increased mitochondrial biogenesis via AMP-activated protein kinase attenuates the rate of protein synthesis; (3) relatively higher expression levels of E3-ligases and proteasome-mediated protein degradation in high oxidative fibers. These observations could explain the fiber type-fiber size paradox that despite the high capacity for protein synthesis in high oxidative fibers, these fibers remain relatively small. However, it remains challenging to understand the mechanisms by which contractile activity, mechanical loading, cellular energy status and cellular oxygen tension affect regulation of fiber size. Therefore, one needs to know the relative contribution of the signaling pathways to protein turnover in high and low oxidative fibers. The outcome and ideas presented are relevant to optimizing treatment and training in the fields of sports, cardiology, oncology, pulmonology and rehabilitation medicine.
Collapse
Affiliation(s)
- T. van Wessel
- Research Institute MOVE, Faculty of Human Movement Sciences, VU University Amsterdam, Van der Boechorststraat 9, 1081 BT Amsterdam, The Netherlands
| | - A. de Haan
- Research Institute MOVE, Faculty of Human Movement Sciences, VU University Amsterdam, Van der Boechorststraat 9, 1081 BT Amsterdam, The Netherlands
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, UK
| | - W. J. van der Laarse
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands
| | - R. T. Jaspers
- Research Institute MOVE, Faculty of Human Movement Sciences, VU University Amsterdam, Van der Boechorststraat 9, 1081 BT Amsterdam, The Netherlands
| |
Collapse
|
165
|
Abstract
Nitric oxide (NO) serves as a messenger molecule in a variety of physiological systems and also converts into toxic radical species that can damage cells through a process known as nitrosative stress. While the physiological roles of NO in blood vessel dilation, the nervous system and the immune system are well established, recent studies have begun to investigate the role of NO in metabolism and energy expenditure through modulation of mitochondria. NO appears to stimulate mitochondrial biogenesis in certain situations through activation of proteins such as peroxisome proliferator-activated receptor γ (PPARγ) co-activator 1α (PGC1-α). Because of this link between NO and mitochondrial biogenesis, the role of NO in certain aspects of metabolism, including exercise response, obesity, fat cell differentiation and caloric restriction, are the subject of increasing investigation. In addition to its role in mitochondrial biogenesis, NO also stimulates mitochondrial fragmentation, which can be caused by too much mitochondrial fission or inhibition of mitochondrial fusion and can result in bioenergetic failure. While the contribution of NO-mediated mitochondrial fragmentation to neurodegenerative diseases seems clear, the mechanisms by which NO causes fragmentation are uncertain and controversial. In this review, we discuss the role of NO in manipulation of mitochondrial biogenesis and dynamics and how these events contribute to human health- and age-related disease.
Collapse
Affiliation(s)
- Andrew B. Knott
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Ella Bossy-Wetzel
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
- Corresponding Author: Ella Bossy-Wetzel, PhD, Burnett School of Biomedical Sciences, College of Medicine, 4000 Central Florida Blvd, Building 20, Orlando, FL 32816, Tel.: +1 (407) 823-3384, Fax: +1 (407) 823-0956,
| |
Collapse
|
166
|
Rajagopalan MS, Stone B, Rwigema JC, Salimi U, Epperly MW, Goff J, Franicola D, Dixon T, Cao S, Zhang X, Buchholz BM, Bauer AJ, Choi S, Bakkenist C, Wang H, Greenberger JS. Intraesophageal manganese superoxide dismutase-plasmid liposomes ameliorates novel total-body and thoracic radiation sensitivity of NOS1-/- mice. Radiat Res 2010; 174:297-312. [PMID: 20726721 DOI: 10.1667/rr2019.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The effect of deletion of the nitric oxide synthase 1 gene (NOS1(-/-)) on radiosensitivity was determined. In vitro, long-term cultures of bone marrow stromal cells derived from NOS1(-/-) were more radioresistant than cells from C57BL/6NHsd (wild-type), NOS2(-/-) or NOS3(-/-) mice. Mice from each strain received 20 Gy thoracic irradiation or 9.5 Gy total-body irradiation (TBI), and NOS1(-/-) mice were more sensitive to both. To determine the etiology of radiosensitivity, studies of histopathology, lower esophageal contractility, gastrointestinal transit, blood counts, electrolytes and inflammatory markers were performed; no significant differences between irradiated NOS1(-/-) and control mice were found. Video camera surveillance revealed the cause of death in NOS1(-/-) mice to be grand mal seizures; control mice died with fatigue and listlessness associated with low blood counts after TBI. NOS1(-/-) mice were not sensitive to brain-only irradiation. MnSOD-PL therapy delivered to the esophagus of wild-type and NOS1(-/-) mice resulted in equivalent biochemical levels in both; however, in NOS1(-/-) mice, MnSOD-PL significantly increased survival after both thoracic and total-body irradiation. The mechanism of radiosensitivity of NOS1(-/-) mice and its reversal by MnSOD-PL may be related to the developmental esophageal enteric neuronal innervation abnormalities described in these mice.
Collapse
Affiliation(s)
- Malolan S Rajagopalan
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Sas K, Párdutz A, Toldi J, Vécsei L. Dementia, stroke and migraine--some common pathological mechanisms. J Neurol Sci 2010; 299:55-65. [PMID: 20828765 DOI: 10.1016/j.jns.2010.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 03/26/2010] [Accepted: 08/02/2010] [Indexed: 01/26/2023]
Abstract
Dementia, stroke and migraine are very common neurological disorders affecting a large percentage of the population, and leading to a high degree of disability. Often, adequate therapy is not available. Although the symptoms, the progression and the outcome differ in these disorders, to some extent they may share some common pathophysiological mechanisms. The genetic background, an energy deficit, and excitotoxicity, vascular and thrombotic properties can influence all three disorders, resulting in a neuronal dysfunction, increased cellular vulnerability, neurodegeneration and ultimately cell death. All these cellular events occur in dementias and stroke, moreover recent studies suggest that, besides a dysfunction, neuronal damage may be an issue in migraine too. One of the most central events in the multiple mechanisms involved in the pathogenesis of these disorders is a metabolic disturbance of certain brain cells. As mitochondria provide the cells with energy, realization of the importance of these organelles in the aetiopathogenesis of several disorders has emerged in recent years. This review surveys the most important features of the pathogenesis of dementia, stroke and migraine from the aspect of mitochondrial malfunction highlighting some of the considerable connections between these neurological disorders.
Collapse
Affiliation(s)
- Katalin Sas
- Department of Neurology, University of Szeged, H-6725 Szeged, Semmelweis u. 6., Hungary
| | | | | | | |
Collapse
|
168
|
Metals, oxidative stress and neurodegenerative disorders. Mol Cell Biochem 2010; 345:91-104. [PMID: 20730621 DOI: 10.1007/s11010-010-0563-x] [Citation(s) in RCA: 722] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 07/29/2010] [Indexed: 12/22/2022]
|
169
|
Galley HF. Bench-to-bedside review: Targeting antioxidants to mitochondria in sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2010; 14:230. [PMID: 20804578 PMCID: PMC2945094 DOI: 10.1186/cc9098] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Development of organ dysfunction associated with sepsis is now accepted to be due at least in part to oxidative damage to mitochondria. Under normal circumstances, complex interacting antioxidant defense systems control oxidative stress within mitochondria. However, no studies have yet provided conclusive evidence of the beneficial effect of antioxidant supplementation in patients with sepsis. This may be because the antioxidants are not accumulating in the mitochondria, where they are most needed. Antioxidants can be targeted selectively to mitochondria by several means. This review describes the in vitro studies and animal models of several diseases involving oxidative stress, including sepsis, in which antioxidants targeted at mitochondria have shown promise, and the future implications for such approaches in patients.
Collapse
Affiliation(s)
- Helen F Galley
- Academic Unit of Anaesthesia & Intensive Care, School of Medicine & Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
170
|
In vitro and in vivo induction and activation of nNOS by LPS in oligodendrocytes. J Neuroimmunol 2010; 229:146-56. [PMID: 20724006 DOI: 10.1016/j.jneuroim.2010.07.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 07/25/2010] [Accepted: 07/27/2010] [Indexed: 11/20/2022]
Abstract
There are currently four known isoforms of nitric oxide synthase (NOS). Of these, neuronal NOS (nNOS) is known to be present exclusively in neurons, endothelial NOS (eNOS) in vascular endothelium, while the inducible form of NOS (iNOS) is known to be activated in oligodendrocytes, astrocytes and microglia. The fourth isoform, mitochondrial NOS (mtNOS), represents a post-translational modification of nNOS. Using western blotting and real time-PCR, we show induction and activation of nNOS following culture of oligodendrocyte progenitor cells (OPC) with lipopolysaccharide (LPS). Activation of nNOS results in accumulation of peroxynitrite and tyrosine nitration of proteins in oligodendrocytes resulting in reduced cell viability. Injection of LPS in vivo into the corpus callosum of rats leads to the development of extensive demyelination of the white matter tracts. Immunostaining of regions close to the injection site shows the presence of nNOS, but not iNOS, in oligodendrocytes. Neither iNOS nor nNOS was seen in astrocytes in areas of demyelination. These studies suggest that activation of nNOS in oligodendrocytes leads to oligodendrocyte injury resulting in demyelination.
Collapse
|
171
|
Abstract
The mitochondrion is the most important organelle in determining continued cell survival and cell death. Mitochondrial dysfunction leads to many human maladies, including cardiovascular diseases, neurodegenerative disease, and cancer. These mitochondria-related pathologies range from early infancy to senescence. The central premise of this review is that if mitochondrial abnormalities contribute to the pathological state, alleviating the mitochondrial dysfunction would contribute to attenuating the severity or progression of the disease. Therefore, this review will examine the role of mitochondria in the etiology and progression of several diseases and explore potential therapeutic benefits of targeting mitochondria in mitigating the disease processes. Indeed, recent advances in mitochondrial biology have led to selective targeting of drugs designed to modulate and manipulate mitochondrial function and genomics for therapeutic benefit. These approaches to treat mitochondrial dysfunction rationally could lead to selective protection of cells in different tissues and various disease states. However, most of these approaches are in their infancy.
Collapse
|
172
|
Davies NW, Guillemin G, Brew BJ. Tryptophan, Neurodegeneration and HIV-Associated Neurocognitive Disorder. Int J Tryptophan Res 2010; 3:121-40. [PMID: 22084594 PMCID: PMC3195234 DOI: 10.4137/ijtr.s4321] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This review presents an up-to-date assessment of the role of the tryptophan metabolic and catabolic pathways in neurodegenerative disease and HIV-associated neurocognitive disorder. The kynurenine pathway and the effects of each of its enzymes and products are reviewed. The differential expression of the kynurenine pathway in cells within the brain, including inflammatory cells, is explored given the increasing recognition of the importance of inflammation in neurodegenerative disease. An overview of common mechanisms of neurodegeneration is presented before a review and discussion of the evidence for a pathogenetic role of the kynurenine pathway in Alzheimer's disease, HIV-associated neurocognitive disorder, Huntington's disease, motor neurone disease, and Parkinson's disease.
Collapse
Affiliation(s)
- Nicholas W.S. Davies
- Department of Neurology, and
- St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Darlinghurst, Sydney, Australia
| | - Gilles Guillemin
- St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Darlinghurst, Sydney, Australia
| | - Bruce J. Brew
- Department of Neurology, and
- St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Darlinghurst, Sydney, Australia
| |
Collapse
|
173
|
LinWu SW, Wang AHJ, Peng FC. Flavin-containing reductase: new perspective on the detoxification of nitrobenzodiazepine. Expert Opin Drug Metab Toxicol 2010; 6:967-81. [DOI: 10.1517/17425255.2010.482928] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
174
|
Silkstone G, Kapetanaki SM, Husu I, Vos MH, Wilson MT. Nitric oxide binds to the proximal heme coordination site of the ferrocytochrome c/cardiolipin complex: formation mechanism and dynamics. J Biol Chem 2010; 285:19785-92. [PMID: 20395293 DOI: 10.1074/jbc.m109.067736] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian mitochondrial cytochrome c interacts with cardiolipin to form a complex (cyt. c/CL) important in apoptosis. Here we show that this interaction leads to structural changes in ferrocytochrome c that leads to an open coordinate site on the central iron, resulting from the dissociation of the intrinsic methionine residue, where NO can rapidly bind (k = 1.2 x 10(7) m(-1) s(-1)). Accompanying NO binding, the proximal histidine dissociates leaving the heme pentacoordinate, in contrast to the hexacoordinate nitrosyl adducts of native ferrocytochrome c or of the protein in which the coordinating methionine is removed by chemical modification or mutation. We present the results of stopped-flow and photolysis experiments that show that following initial NO binding to the heme, there ensues an unusually complex set of kinetic steps. The spectral changes associated with these kinetic transitions, together with their dependence on NO concentration, have been determined and lead us to conclude that NO binding to cyt. c/CL takes place via an overall scheme comparable to that described for cytochrome c' and guanylate cyclase, the final product being one in which NO resides on the proximal side of the heme. In addition, novel features not observed before in other heme proteins forming pentacoordinate nitrosyl species, include a high yield of NO escape after dissociation, rapid (<1 ms) dissociation of proximal histidine upon NO binding and its very fast binding (60 ps) after NO dissociation, and the formation of a hexacoordinate intermediate. These features all point at a remarkable mobility of the proximal heme environment induced by cardiolipin.
Collapse
Affiliation(s)
- Gary Silkstone
- Department of Biological Sciences, Wivenhoe Park, University of Essex, Colchester CO4 3SQ, United Kingdom
| | | | | | | | | |
Collapse
|
175
|
Abstract
Although considered safe at therapeutic doses, at higher doses, acetaminophen produces a centrilobular hepatic necrosis that can be fatal. Acetaminophen poisoning accounts for approximately one-half of all cases of acute liver failure in the United States and Great Britain today. The mechanism occurs by a complex sequence of events. These events include: (1) CYP metabolism to a reactive metabolite which depletes glutathione and covalently binds to proteins; (2) loss of glutathione with an increased formation of reactive oxygen and nitrogen species in hepatocytes undergoing necrotic changes; (3) increased oxidative stress, associated with alterations in calcium homeostasis and initiation of signal transduction responses, causing mitochondrial permeability transition; (4) mitochondrial permeability transition occurring with additional oxidative stress, loss of mitochondrial membrane potential, and loss of the ability of the mitochondria to synthesize ATP; and (5) loss of ATP which leads to necrosis. Associated with these essential events there appear to be a number of inflammatory mediators such as certain cytokines and chemokines that can modify the toxicity. Some have been shown to alter oxidative stress, but the relationship of these modulators to other critical mechanistic events has not been well delineated. In addition, existing data support the involvement of cytokines, chemokines, and growth factors in the initiation of regenerative processes leading to the reestablishment of hepatic structure and function.
Collapse
Affiliation(s)
- Jack A Hinson
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | | | |
Collapse
|
176
|
Gutierrez FRS, Mineo TWP, Pavanelli WR, Guedes PMM, Silva JS. The effects of nitric oxide on the immune system during Trypanosoma cruzi infection. Mem Inst Oswaldo Cruz 2010; 104 Suppl 1:236-45. [PMID: 19753479 DOI: 10.1590/s0074-02762009000900030] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Accepted: 05/29/2009] [Indexed: 01/08/2023] Open
Abstract
Trypanosoma cruzi infection triggers substantial production of nitric oxide (NO), which has been shown to have protective and toxic effects on the host's immune system. Sensing of trypomastigotes by phagocytes activates the inducible NO-synthase (NOS2) pathway, which produces NO and is largely responsible for macrophage-mediated killing of T. cruzi. NO is also responsible for modulating virtually all steps of innate and adaptive immunity. However, NO can also cause oxidative stress, which is especially damaging to the host due to increased tissue damage. The cytokines IFN-gamma and TNF-alpha, as well as chemokines, are strong inducers of NOS2 and are produced in large amounts during T. cruzi acute infection. Conversely, TGF-beta and IL-10 negatively regulate NO production. Here we discuss the recent evidence describing the mechanisms by which NO is able to exert its antimicrobial and immune regulatory effects, the mechanisms involved in the oxidative stress response during infection and the implications of NO for the development of therapeutic strategies against T. cruzi.
Collapse
Affiliation(s)
- Fredy R S Gutierrez
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | | | | | | | | |
Collapse
|
177
|
Leite ACR, Oliveira HCF, Utino FL, Garcia R, Alberici LC, Fernandes MP, Castilho RF, Vercesi AE. Mitochondria generated nitric oxide protects against permeability transition via formation of membrane protein S-nitrosothiols. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:1210-6. [PMID: 20138021 DOI: 10.1016/j.bbabio.2010.01.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 01/27/2010] [Accepted: 01/27/2010] [Indexed: 10/19/2022]
Abstract
Mitochondria generated nitric oxide (NO) regulates several cell functions including energy metabolism, cell cycling, and cell death. Here we report that the NO synthase inhibitors (L-NAME, L-NNA and L-NMMA) administered either in vitro or in vivo induce Ca2+-dependent mitochondrial permeability transition (MPT) in rat liver mitochondria via a mechanism independent on changes in the energy state of the organelle. MPT was determined by the occurrence of cyclosporin A sensitive mitochondrial membrane potential disruption followed by mitochondrial swelling and Ca2+ release. In in vitro experiments, the effect of NOS inhibitors was dose-dependent (1 to 50 microM). In addition to cyclosporin A, L-NAME-induced MPT was sensitive to Mg2+ plus ATP, EGTA, and to a lower degree, to catalase and dithiothreitol. In contrast to L-NAME, its isomer D-NAME did not induce MPT. L-NAME-induced MPT was associated with a significant decrease in both the rate of NO generation and the content of mitochondrial S-nitrosothiol. Acute and chronic in vivo treatment with L-NAME also promoted MPT and decreased the content of mitochondrial S-nitrosothiol. SNAP (a NO donor) prevented L-NAME mediated MPT and reversed the decrease in the rate of NO generation and in the content of S-nitrosothiol. We propose that S-nitrosylation of critical membrane protein thiols by NO protects against MPT.
Collapse
Affiliation(s)
- Ana Catarina R Leite
- Departamento de Fisiologia e Biofísica, Universidade Estadual de Campinas, UNICAMP, Campinas, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
178
|
Abstract
Collagens are major constituents of connective tissues in the animal kingdom. During aging and inflammatory-related diseases, the collagen network undergoes oxidation that leads to structural and biochemical alterations within the collagen molecule. Collagen oxidation appears to be a key determinant of aging and a critical physiopathologic mechanism of numerous diseases. Further, the detection of oxidized-collagen peptides seems to be a promising approach for the diagnosis and the prognosis of inflammatory diseases. This chapter reviews the structural and biochemical changes to collagen induced by reactive oxygen and nitrogen species and discusses recent data on the use of collagen-derived biomarkers for measuring oxidative damage.
Collapse
|
179
|
Yao S, Pandey P, Ljunggren-Rose A, Sriram S. LPS mediated injury to oligodendrocytes is mediated by the activation of nNOS: relevance to human demyelinating disease. Nitric Oxide 2009; 22:197-204. [PMID: 20005301 DOI: 10.1016/j.niox.2009.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 11/19/2009] [Accepted: 12/06/2009] [Indexed: 10/20/2022]
Abstract
Loss of oligodendrocytes and the destruction of myelin form the core features of inflammatory demyelinating disease. Although many of the inflammatory and cellular mediators of tissue injury are known, recent studies have suggested an important role for nitric oxide NO and other reactive nitrogen species in oligodendrocyte injury. The human transformed oligodendrocyte cell line, MO3.13 cells, express Toll like receptor genes (TLR) genes and are activated by lipopolysaccharide (LPS). We determined the activation and consequences of neuronal nitric oxide synthase (nNOS) following stimulation with LPS in the MO3.13 cell line. Our studies show that MO3.13 cells induce nNOS following stimulation with LPS. Most importantly, these studies show a susceptibility of MO3.13 cells to NO mediated cell death by the activation of nNOS but not of inducible NOS (iNOS). MO3.13 cells show increased susceptibility to peroxynitrite mediated cellular injury to mitochondrial proteins and decreased cell survival in the presence of LPS. Our studies suggest that the presence and activation of nNOS in oligodendrocytes can directly mediate oligodendrocyte (OC) injury and reduce cell viability.
Collapse
Affiliation(s)
- S Yao
- Department of Neurology, Multiple Sclerosis Research Center, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | | | | | | |
Collapse
|
180
|
Puddu P, Puddu GM, Cravero E, De Pascalis S, Muscari A. The emerging role of cardiovascular risk factor-induced mitochondrial dysfunction in atherogenesis. J Biomed Sci 2009; 16:112. [PMID: 20003216 PMCID: PMC2800844 DOI: 10.1186/1423-0127-16-112] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 12/09/2009] [Indexed: 12/23/2022] Open
Abstract
An important role in atherogenesis is played by oxidative stress, which may be induced by common risk factors. Mitochondria are both sources and targets of reactive oxygen species, and there is growing evidence that mitochondrial dysfunction may be a relevant intermediate mechanism by which cardiovascular risk factors lead to the formation of vascular lesions. Mitochondrial DNA is probably the most sensitive cellular target of reactive oxygen species. Damage to mitochondrial DNA correlates with the extent of atherosclerosis. Several cardiovascular risk factors are demonstrated causes of mitochondrial damage. Oxidized low density lipoprotein and hyperglycemia may induce the production of reactive oxygen species in mitochondria of macrophages and endothelial cells. Conversely, reactive oxygen species may favor the development of type 2 diabetes mellitus, mainly through the induction of insulin resistance. Similarly - in addition to being a cause of endothelial dysfunction, reactive oxygen species and subsequent mitochondrial dysfunction - hypertension may develop in the presence of mitochondrial DNA mutations. Finally, other risk factors, such as aging, hyperhomocysteinemia and cigarette smoking, are also associated with mitochondrial damage and an increased production of free radicals. So far clinical studies have been unable to demonstrate that antioxidants have any effect on human atherogenesis. Mitochondrial targeted antioxidants might provide more significant results.
Collapse
Affiliation(s)
- Paolo Puddu
- Department of Internal Medicine, Aging and Nephrological Diseases, University of Bologna and S, Orsola-Malpighi Hospital, Bologna, Italy.
| | | | | | | | | |
Collapse
|
181
|
Kim K, Wang L, Hwang I. Acute inhibition of selected membrane-proximal mouse T cell receptor signaling by mitochondrial antagonists. PLoS One 2009; 4:e7738. [PMID: 19901985 PMCID: PMC2768903 DOI: 10.1371/journal.pone.0007738] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 10/09/2009] [Indexed: 12/20/2022] Open
Abstract
T cells absorb nanometric membrane vesicles, prepared from plasma membrane of antigen presenting cells, via dual receptor/ligand interactions of T cell receptor (TCR) with cognate peptide/major histocompatibility complex (MHC) plus lymphocyte function-associated antigen 1 (LFA-1) with intercellular adhesion molecule 1. TCR-mediated signaling for LFA-1 activation is also required for the vesicle absorption. Exploiting those findings, we had established a high throughput screening (HTS) platform and screened a library for isolation of small molecules inhibiting the vesicle absorption. Follow-up studies confirmed that treatments (1 hour) with various mitochondrial antagonists, including a class of anti-diabetic drugs (i.e., Metformin and Phenformin), resulted in ubiquitous inhibition of the vesicle absorption without compromising viability of T cells. Further studies revealed that the mitochondrial drug treatments caused impairment of specific membrane-proximal TCR signaling event(s). Thus, activation of Akt and PLC-gamma1 and entry of extracellular Ca(2+) following TCR stimulation were attenuated while polymerization of monomeric actins upon TCR triggering progressed normally after the treatments. Dynamic F-actin rearrangement concurring with the vesicle absorption was also found to be impaired by the drug treatments, implying that the inhibition by the drug treatments of downstream signaling events (and the vesicle absorption) could result from lack of directional relocation of signaling and cell surface molecules. We also assessed the potential application of mitochondrial antagonists as immune modulators by probing effects of the long-term drug treatments (24 hours) on viability of resting primary T cells and cell cycle progression of antigen-stimulated T cells. This study unveils a novel regulatory mechanism for T cell immunity in response to environmental factors having effects on mitochondrial function.
Collapse
Affiliation(s)
- Kwangmi Kim
- Department of Chemistry and Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Lin Wang
- Department of Chemistry and Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Inkyu Hwang
- Department of Chemistry and Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
182
|
Poyton RO, Castello PR, Ball KA, Woo DK, Pan N. Mitochondria and hypoxic signaling: a new view. Ann N Y Acad Sci 2009; 1177:48-56. [PMID: 19845606 DOI: 10.1111/j.1749-6632.2009.05046.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Eukaryotic cells respond to low oxygen concentrations by upregulating hypoxic and downregulating aerobic nuclear genes (hypoxic signaling). Most of the oxygen-regulated genes in yeast require the mitochondrial respiratory chain for their up- or downregulation when cells experience hypoxia. Although it was shown previously that the mitochondrial respiratory chain is required for the upregulation of some hypoxic genes in both yeast and mammalian cells, its underlying role in this process has been unclear. Recently, we have reported that mitochondria produce nitric oxide (NO(*)) when oxygen becomes limiting. This NO(*) production is nitrite (NO(2) (-))-dependent, requires an electron donor, and is carried out by cytochrome c oxidase in a pH-dependent fashion. We call this activity Cco/NO(*) and incorporate it into a new model for hypoxic signaling. In addition, we have found that some of the NO(*) produced by Cco/NO(*) is released from cells, raising the possibility that mitochondrially generated NO(*) also functions in extracellular hypoxic signaling pathways.
Collapse
Affiliation(s)
- Robert O Poyton
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA.
| | | | | | | | | |
Collapse
|
183
|
Powers SK, Duarte J, Kavazis AN, Talbert EE. Reactive oxygen species are signalling molecules for skeletal muscle adaptation. Exp Physiol 2009; 95:1-9. [PMID: 19880534 PMCID: PMC2906150 DOI: 10.1113/expphysiol.2009.050526] [Citation(s) in RCA: 288] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Increased reactive oxygen species (ROS) production is crucial to the remodelling that occurs in skeletal muscle in response to both exercise training and prolonged periods of disuse. This review discusses the redox-sensitive signalling pathways that are responsible for this ROS-induced skeletal muscle adaptation. We begin with a discussion of the sites of ROS production in skeletal muscle fibres. This is followed by an overview of the putative redox-sensitive signalling pathways that promote skeletal muscle adaptation. Specifically, this discussion highlights redox-sensitive kinases, phosphatases and the transcription factor nuclear factor-κB. We also discuss the evidence that connects redox signalling to skeletal muscle adaptation in response to increased muscular activity (i.e. exercise training) and during prolonged periods of muscular inactivity (i.e. immobilization). In an effort to stimulate further research, we conclude with a discussion of unanswered questions about redox signalling in skeletal muscle.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Room 25, Florida Gym, Gainesville, FL 32611, USA.
| | | | | | | |
Collapse
|
184
|
Singh PP, Mahadi F, Roy A, Sharma P. Reactive oxygen species, reactive nitrogen species and antioxidants in etiopathogenesis of diabetes mellitus type-2. Indian J Clin Biochem 2009; 24:324-42. [PMID: 23105858 PMCID: PMC3453064 DOI: 10.1007/s12291-009-0062-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus type-2 (DMT-2) is a hyperglycemic syndrome with several characteristic features. It continues to rise unabatedly in all pockets of the world, parallels with affluence and can be controlled but not cured. It has a definite involvement of genetic component but environmental factors play overwhelmingly dominant role in etiopathogenesis. Insulin resistance (IR) and obesity are singular instigators of DMT-2. The various events cause critical defects in insulin signaling cascade followed by beta-cell dysfunction. Over a period of time, numerous other metabolic aberrations develop, resulting in diabetic complications which could be both vascular (cardiovascular complications, nephropathy, neuropathy, retinopathy and embryopathy) or a-vascular (cataract and glaucoma etc). It has been proposed that all these abnormal events are initiated or activated by a common mechanism of superoxide anion, which is accompanied with generation of a variety of reactive oxygen species (ROS), reactive nitrogen specie (RNS) and resultant heightened oxidative stress (OS). Provoked OS causes IR and altered gene expressions. Hyperglycemia induces OS through multiple routes: a)stimulated polyol pathway where in ≤ 30% glucose can be diverted to sorbitol and fructose, b)increased transcription of genes for proinflammatory cytokines and plasminogen activator inhibitor-1 (PAI-1) c) activation of protein kinase-C (PKC) leading to several molecular changes d)increased synthesis of Advanced Glycation End Products (AGEs) e)changes in a receptor far AGEs and f) autooxidation of glucose with formation of ketoimines and AGEs. All these processes are accompanied with alteration in redox status, ROS, RNS and OS which trigger DMT-2 and its complications. Initial hurriedly planned and executed experimental and clinical studies showed promising results of antioxidant therapies, but recent studies indicate that excess intake/supplement may have adverse outcomes including increased mortality. It is advocated that antioxidants should be given only if preexisting deficiency is present. Selection of antioxidant is another important aspect. Lastly but most importantly the impact of OS is not obligatory but facultative. As such only those diabetic patients will be benefited by antioxidant therapies that have impelling punch of prooxidants.
Collapse
Affiliation(s)
- P. P. Singh
- Department of Biochemistry, Era’s Lucknow Medical College, Sarfarazganj, Lucknow, Uttar Pradesh India
- Department of Biochemistry, Era’s Lucknow Medical College, Sarfarazganj, Lucknow, 226003 India
| | - Farzana Mahadi
- Department of Biochemistry, Era’s Lucknow Medical College, Sarfarazganj, Lucknow, Uttar Pradesh India
| | - Ajanta Roy
- Department of Biochemistry, Era’s Lucknow Medical College, Sarfarazganj, Lucknow, Uttar Pradesh India
| | - Praveen Sharma
- Department of Biochemistry, SMS Medical College Jaipur, Rajasthan, India
| |
Collapse
|
185
|
Poyton RO, Ball KA, Castello PR. Mitochondrial generation of free radicals and hypoxic signaling. Trends Endocrinol Metab 2009; 20:332-40. [PMID: 19733481 DOI: 10.1016/j.tem.2009.04.001] [Citation(s) in RCA: 363] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/03/2009] [Accepted: 04/06/2009] [Indexed: 12/17/2022]
Abstract
Most reactive oxygen species (ROS) are generated in cells by the mitochondrial respiratory chain. Mitochondrial ROS production is modulated largely by the rate of electron flow through respiratory chain complexes. Recently, it has become clear that under hypoxic conditions, the mitochondrial respiratory chain also produces nitric oxide (NO), which can generate other reactive nitrogen species (RNS). Although excess ROS and RNS can lead to oxidative and nitrosative stress, moderate to low levels of both function in cellular signaling pathways. Especially important are the roles of these mitochondrially generated free radicals in hypoxic signaling pathways, which have important implications for cancer, inflammation and a variety of other diseases.
Collapse
Affiliation(s)
- Robert O Poyton
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA.
| | | | | |
Collapse
|
186
|
|
187
|
Finley LW, Haigis MC. The coordination of nuclear and mitochondrial communication during aging and calorie restriction. Ageing Res Rev 2009; 8:173-88. [PMID: 19491041 DOI: 10.1016/j.arr.2009.03.003] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 02/27/2009] [Accepted: 03/04/2009] [Indexed: 12/24/2022]
Abstract
Mitochondria are dynamic organelles that integrate environmental signals to regulate energy production, apoptosis and Ca(2+) homeostasis. Not surprisingly, mitochondrial dysfunction is associated with aging and the pathologies observed in age-related diseases. The vast majority of mitochondrial proteins are encoded in the nuclear genome, and so communication between the nucleus and mitochondria is essential for maintenance of appropriate mitochondrial function. Several proteins have emerged as major regulators of mitochondrial gene expression, capable of increasing transcription of mitochondrial genes in response to the physiological demands of the cell. In this review, we will focus on PGC-1alpha, SIRT1, AMPK and mTOR and discuss how these proteins regulate mitochondrial function and their potential involvement in aging, calorie restriction and age-related disease. We will also discuss the pathways through which mitochondria signal to the nucleus. Although such retrograde signaling is not well studied in mammals, there is growing evidence to suggest that it may be an important area for future aging research. Greater understanding of the mechanisms by which mitochondria and the nucleus communicate will facilitate efforts to slow or reverse the mitochondrial dysfunction that occurs during aging.
Collapse
|
188
|
Stowe DF, Camara AKS. Mitochondrial reactive oxygen species production in excitable cells: modulators of mitochondrial and cell function. Antioxid Redox Signal 2009; 11:1373-414. [PMID: 19187004 PMCID: PMC2842133 DOI: 10.1089/ars.2008.2331] [Citation(s) in RCA: 351] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 01/12/2009] [Accepted: 01/13/2009] [Indexed: 12/14/2022]
Abstract
The mitochondrion is a major source of reactive oxygen species (ROS). Superoxide (O(2)(*-)) is generated under specific bioenergetic conditions at several sites within the electron-transport system; most is converted to H(2)O(2) inside and outside the mitochondrial matrix by superoxide dismutases. H(2)O(2) is a major chemical messenger that, in low amounts and with its products, physiologically modulates cell function. The redox state and ROS scavengers largely control the emission (generation scavenging) of O(2)(*-). Cell ischemia, hypoxia, or toxins can result in excess O(2)(*-) production when the redox state is altered and the ROS scavenger systems are overwhelmed. Too much H(2)O(2) can combine with Fe(2+) complexes to form reactive ferryl species (e.g., Fe(IV) = O(*)). In the presence of nitric oxide (NO(*)), O(2)(*-) forms the reactant peroxynitrite (ONOO(-)), and ONOOH-induced nitrosylation of proteins, DNA, and lipids can modify their structure and function. An initial increase in ROS can cause an even greater increase in ROS and allow excess mitochondrial Ca(2+) entry, both of which are factors that induce cell apoptosis and necrosis. Approaches to reduce excess O(2)(*-) emission include selectively boosting the antioxidant capacity, uncoupling of oxidative phosphorylation to reduce generation of O(2)(*-) by inducing proton leak, and reversibly inhibiting electron transport. Mitochondrial cation channels and exchangers function to maintain matrix homeostasis and likely play a role in modulating mitochondrial function, in part by regulating O(2)(*-) generation. Cell-signaling pathways induced physiologically by ROS include effects on thiol groups and disulfide linkages to modify posttranslationally protein structure to activate/inactivate specific kinase/phosphatase pathways. Hypoxia-inducible factors that stimulate a cascade of gene transcription may be mediated physiologically by ROS. Our knowledge of the role played by ROS and their scavenging systems in modulation of cell function and cell death has grown exponentially over the past few years, but we are still limited in how to apply this knowledge to develop its full therapeutic potential.
Collapse
Affiliation(s)
- David F Stowe
- Anesthesiology Research Laboratories, Department of Anesthesiology, The Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | |
Collapse
|
189
|
Landim MBP, Casella Filho A, Chagas ACP. Asymmetric dimethylarginine (ADMA) and endothelial dysfunction: implications for atherogenesis. Clinics (Sao Paulo) 2009; 64:471-8. [PMID: 19488614 PMCID: PMC2694252 DOI: 10.1590/s1807-59322009000500015] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Accepted: 02/20/2009] [Indexed: 02/07/2023] Open
Abstract
Atherosclerotic coronary heart disease is the leading cause of morbidity and mortality in industrialized countries, and endothelial dysfunction is considered a precursor phenomenon. The nitric oxide produced by the endothelium under the action of endothelial nitric oxide synthase has important antiatherogenic functions. Its reduced bioavailabilty is the beginning of the atherosclerotic process. The addition of two methyl radicals to arginine, through the action of methyltransferase nuclear proteins, produces asymmetric dimethylarginine, which competes with L-arginine and promotes a reduction in nitric oxide formation in the vascular wall. The asymmetric dimethylarginine, which is itself considered a mediator of the vascular effects of the several risk factors for atherosclerosis, can be eliminated by renal excretion or by the enzymatic action of the dimethylarginine dimethylaminohydrolases. Several basic science and clinical research studies suggest that the increase in asymmetric dimethylarginine occurs in the context of chronic renal insufficiency, dyslipidemia, high blood pressure, diabetes mellitus, and hyperhomocysteinemy, as well as with other conditions. Therapeutic measures to combat atherosclerosis may reverse these asymmetric dimethylarginine effects or at least reduce the concentration of this chemical in the blood. Such an effect can be achieved with competitor molecules or by increasing the expression or activity of its degradation enzyme. Studies are in development to establish the true role of asymmetric dimethylarginine as a marker and mediator of atherosclerosis, with possible therapeutic applications. The main aspects of the formation and degradation of asymmetric dimethylarginine and its implication in the atherogenic process will be addressed in this article.
Collapse
Affiliation(s)
- Maurício Batista Paes Landim
- Heart Institute, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | | | | |
Collapse
|
190
|
Vardatsikos G, Sahu A, Srivastava AK. The insulin-like growth factor family: molecular mechanisms, redox regulation, and clinical implications. Antioxid Redox Signal 2009; 11:1165-90. [PMID: 19014342 DOI: 10.1089/ars.2008.2161] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Insulin-like growth factor (IGF)-induced signaling networks are vital in modulating multiple fundamental cellular processes, such as cell growth, survival, proliferation, and differentiation. Aberrations in the generation or action of IGF have been suggested to play an important role in several pathological conditions, including metabolic disorders, neurodegenerative diseases, and multiple types of cancer. Yet the exact mechanism involved in the pathogenesis of these diseases by IGFs remains obscure. Redox pathways involving reactive oxygen species (ROS) and reactive nitrogen species (RNS) contribute to the pathogenetic mechanism of various diseases by modifying key signaling pathways involved in cell growth, proliferation, survival, and apoptosis. Furthermore, ROS and RNS have been demonstrated to alter IGF production and/or action, and vice versa, and thereby have the ability to modulate cellular functions, leading to clinical manifestations of diseases. In this review, we provide an overview on the IGF system and discuss the potential role of IGF-1/IGF-1 receptor and redox pathways in the pathophysiology of several diseases.
Collapse
Affiliation(s)
- George Vardatsikos
- Laboratory of Cell Signaling, Montreal Diabetes Research Center, Centre Hospitalier de l'Université de Montréal, Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | | | | |
Collapse
|
191
|
Impact of methylglyoxal and high glucose co-treatment on human mononuclear cells. Int J Mol Sci 2009; 10:1445-1464. [PMID: 19468318 PMCID: PMC2680626 DOI: 10.3390/ijms10041445] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 03/13/2009] [Accepted: 03/26/2009] [Indexed: 12/31/2022] Open
Abstract
Hyperglycemia and elevation of methylglyoxal (MG) are symptoms of diabetes mellitus (DM). In this report, we show that co-treatment of human mononuclear cells (HMNCs) with MG (5 μM) and high glucose (HG; 15 – 30 mM) induces apoptosis or necrosis. HG/MG co-treatment directly enhanced the reactive oxygen species (ROS) content in HMNCs, leading to decreased intracellular ATP levels, which control cell death via apoptosis or necrosis. Concentrations of 5 μM MG and 15 mM glucose significantly increased cytoplasmic free calcium and nitric oxide (NO) levels, loss of mitochondrial membrane potential (MMP), activation of caspases-9 and -3, and cell death. In contrast, no apoptotic biochemical changes were detected in HMNCs treated with 5 μM MG and 25 mM glucose, which appeared to undergo necrosis. Pretreatment with nitric oxide (NO) scavengers inhibited apoptotic biochemical changes induced by 5 μM MG/15 mM glucose, and increased the gene expression levels of p53 and p21 involved in apoptotic signaling. The results collectively suggest that the treatment dosage of MG and glucose determines the mode of cell death (apoptosis vs. necrosis) of HMNCs, and that both ROS and NO play important roles in MG/HG-induced apoptosis.
Collapse
|
192
|
Electrochemical quantification of reactive oxygen and nitrogen: challenges and opportunities. Anal Bioanal Chem 2009; 394:95-105. [DOI: 10.1007/s00216-009-2692-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2008] [Revised: 01/29/2009] [Accepted: 02/09/2009] [Indexed: 01/09/2023]
|
193
|
Abstract
Nitric oxide (NO) is an important messenger molecule in a variety of physiological systems. NO, a gas, is produced from L-arginine by different isoforms of nitric oxide synthase (NOS) and serves many normal physiologic purposes, such as promoting vasodilation of blood vessels and mediating communication between nervous system cells. In addition to its physiologic actions, free radical activity of NO can cause cellular damage through a phenomenon known as nitrosative stress. Here, we review the role of NO in health and disease, focusing on its role in function and dysfunction of the nervous system. Substantial evidence indicates that NO plays a key role in most common neurodegenerative diseases, and, although the mechanism of NO-mediated neurodegeneration remains uncertain, studies suggest several possibilities. NO has been shown to modify protein function by nitrosylation and nitrotyrosination, contribute to glutamate excitotoxicity, inhibit mitochondrial respiratory complexes, participate in organelle fragmentation, and mobilize zinc from internal stores. In this review, we discuss and analyze the evidence for each of these mechanisms in different neurodegenerative diseases and propose future directions for research of the role of NO in neurodegeneration.
Collapse
Affiliation(s)
- Andrew B Knott
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 4000 Central Florida Blvd., Orlando, Florida, USA
| | | |
Collapse
|
194
|
Cardioprotection: a radical view Free radicals in pre and postconditioning. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:781-93. [PMID: 19248760 DOI: 10.1016/j.bbabio.2009.02.008] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 02/12/2009] [Accepted: 02/13/2009] [Indexed: 12/13/2022]
Abstract
A series of brief (a few minutes) ischemia/reperfusion cycles (ischemic preconditioning, IP) limits myocardial injury produced by a subsequent prolonged period of coronary artery occlusion and reperfusion. Postconditioning (PostC), which is a series of brief (a few seconds) reperfusion/ischemia cycles at reperfusion onset, attenuates also ischemia/reperfusion injury. In recent years the main idea has been that reactive oxygen species (ROS) play an essential, though double-edged, role in cardioprotection: they may participate in reperfusion injury or may play a role as signaling elements of protection in the pre-ischemic phase. It has been demonstrated that preconditioning triggering is redox-sensitive, using either ROS scavengers or ROS generators. We have shown that nitroxyl triggers preconditioning via pro-oxidative, and/or nitrosative stress-related mechanism(s). Several metabolites, including acetylcholine, bradykinin, opioids and phenylephrine, trigger preconditioning-like protection via a mitochondrial K(ATP)-ROS-dependent mechanism. Intriguingly, and contradictory to the above mentioned theory of ROS as an obligatory part of reperfusion-induced damage, some studies suggest the possibility that some ROS at low concentrations could protect ischemic hearts against reperfusion injury. Yet, we demonstrated that ischemic PostC is also a cardioprotective phenomenon that requires the intervention of redox signaling to be protective. Emerging evidence suggests that in a preconditioning scenario a redox signal is required during the first few minutes of myocardial reperfusion following the index ischemic period. Intriguingly, the ROS signaling in the early reperfusion appear crucial to both preconditioning- and postconditioning-induced protection. Therefore, our and others' results suggest that the role of ROS in reperfusion may be reconsidered as they are not only deleterious.
Collapse
|
195
|
Kapetanaki SM, Silkstone G, Husu I, Liebl U, Wilson MT, Vos MH. Interaction of Carbon Monoxide with the Apoptosis-Inducing Cytochrome c−Cardiolipin Complex. Biochemistry 2009; 48:1613-9. [DOI: 10.1021/bi801817v] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Sofia M. Kapetanaki
- Laboratoire d’Optique et Biosciences, CNRS, Ecole Polytechnique, and INSERM U696, F-91128 Palaiseau, France, and Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Gary Silkstone
- Laboratoire d’Optique et Biosciences, CNRS, Ecole Polytechnique, and INSERM U696, F-91128 Palaiseau, France, and Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Ivan Husu
- Laboratoire d’Optique et Biosciences, CNRS, Ecole Polytechnique, and INSERM U696, F-91128 Palaiseau, France, and Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Ursula Liebl
- Laboratoire d’Optique et Biosciences, CNRS, Ecole Polytechnique, and INSERM U696, F-91128 Palaiseau, France, and Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Michael T. Wilson
- Laboratoire d’Optique et Biosciences, CNRS, Ecole Polytechnique, and INSERM U696, F-91128 Palaiseau, France, and Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Marten H. Vos
- Laboratoire d’Optique et Biosciences, CNRS, Ecole Polytechnique, and INSERM U696, F-91128 Palaiseau, France, and Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| |
Collapse
|
196
|
Piccoli C, Quarato G, Ripoli M, D'Aprile A, Scrima R, Cela O, Boffoli D, Moradpour D, Capitanio N. HCV infection induces mitochondrial bioenergetic unbalance: causes and effects. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2008; 1787:539-46. [PMID: 19094961 DOI: 10.1016/j.bbabio.2008.11.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Revised: 11/15/2008] [Accepted: 11/18/2008] [Indexed: 12/13/2022]
Abstract
Cells infected by the hepatitis C virus (HCV) are characterized by endoplasmic reticulum stress, deregulation of the calcium homeostasis and unbalance of the oxido-reduction state. In this context, mitochondrial dysfunction proved to be involved and is thought to contribute to the outcome of the HCV-related disease. Here, we propose a temporal sequence of events in the HCV-infected cell whereby the primary alteration consists of a release of Ca(2+) from the endoplasmic reticulum, followed by uptake into mitochondria. This causes successive mitochondrial alterations comprising generation of reactive oxygen and nitrogen species and impairment of the oxidative phosphorylation. A progressive adaptive response results in an enhancement of the glycolytic metabolism sustained by up-regulation of the hypoxia inducible factor. Pathogenetic implications of the model are discussed.
Collapse
Affiliation(s)
- C Piccoli
- Department of Biomedical Sciences, University of Foggia, 71100 Foggia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Benamar A, Rolletschek H, Borisjuk L, Avelange-Macherel MH, Curien G, Mostefai HA, Andriantsitohaina R, Macherel D. Nitrite-nitric oxide control of mitochondrial respiration at the frontier of anoxia. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1777:1268-75. [PMID: 18602886 DOI: 10.1016/j.bbabio.2008.06.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Revised: 05/14/2008] [Accepted: 06/01/2008] [Indexed: 11/16/2022]
Abstract
Actively respiring animal and plant tissues experience hypoxia because of mitochondrial O(2) consumption. Controlling oxygen balance is a critical issue that involves in mammals hypoxia-inducible factor (HIF) mediated transcriptional regulation, cytochrome oxidase (COX) subunit adjustment and nitric oxide (NO) as a mediator in vasodilatation and oxygen homeostasis. In plants, NO, mainly derived from nitrite, is also an important signalling molecule. We describe here a mechanism by which mitochondrial respiration is adjusted to prevent a tissue to reach anoxia. During pea seed germination, the internal atmosphere was strongly hypoxic due to very active mitochondrial respiration. There was no sign of fermentation, suggesting a down-regulation of O(2) consumption near anoxia. Mitochondria were found to finely regulate their surrounding O(2) level through a nitrite-dependent NO production, which was ascertained using electron paramagnetic resonance (EPR) spin trapping of NO within membranes. At low O(2), nitrite is reduced into NO, likely at complex III, and in turn reversibly inhibits COX, provoking a rise to a higher steady state level of oxygen. Since NO can be re-oxidized into nitrite chemically or by COX, a nitrite-NO pool is maintained, preventing mitochondrial anoxia. Such an evolutionarily conserved mechanism should have an important role for oxygen homeostasis in tissues undergoing hypoxia.
Collapse
Affiliation(s)
- Abdelilah Benamar
- UMR 1191 Physiologie Moléculaire des Semences, Université d'Angers/INH/INRA, Angers, France
| | | | | | | | | | | | | | | |
Collapse
|
198
|
Deng A, Tang T, Singh P, Wang C, Satriano J, Thomson SC, Blantz RC. Regulation of oxygen utilization by angiotensin II in chronic kidney disease. Kidney Int 2008; 75:197-204. [PMID: 18818681 DOI: 10.1038/ki.2008.481] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Angiotensin II blockade delays progression of chronic kidney disease by modifying intrarenal hemodynamics, but the effects on metabolic adaptations are unknown. Using the remnant kidney model of chronic kidney disease in rats, we measured the effects of combined angiotensin II blockade with captopril and losartan on renal oxygen consumption (QO(2)) and factors influencing QO(2). Remnant kidneys had proteinuria and reductions in the glomerular filtration rate (GFR), renal blood flow (RBF) and nitric oxide synthase-1 protein expression while QO(2), factored by sodium reabsorption (QO(2)/TNa), was markedly increased. Combined blockade treatment normalized these parameters while increasing sodium reabsorption but, since QO(2) was unchanged, QO(2)/TNa also normalized. Triple antihypertensive therapy, to control blood pressure, and treatment with lysine, to increase GFR and RBF, did not normalize QO(2)/TNa, suggesting a specific effect of angiotensin II in elevating QO(2)/TNa. Inhibition of nitric oxide synthase increased QO(2) in the kidney of sham-operated rats but not in the remnant kidney of untreated rats. Our study shows that combined captopril and losartan treatment normalized QO(2)/TNa and functional nitric oxide activity in the remnant kidney independent of blood pressure and GFR effects, suggesting that other mechanisms in addition to hemodynamics underlie the benefits of angiotensin II blockade.
Collapse
Affiliation(s)
- Aihua Deng
- Division of Nephrology-Hypertension, Department of Medicine, School of Medicine, University of California, San Diego, San Diego, California 92161, USA.
| | | | | | | | | | | | | |
Collapse
|
199
|
Pyne-Geithman GJ, Caudell DN, Cooper M, Clark JF, Shutter LA. Dopamine D2-receptor-mediated increase in vascular and endothelial NOS activity ameliorates cerebral vasospasm after subarachnoid hemorrhage in vitro. Neurocrit Care 2008; 10:225-31. [PMID: 18807216 PMCID: PMC2651409 DOI: 10.1007/s12028-008-9143-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2008] [Accepted: 08/25/2008] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Cerebral vasospasm after subarachnoid hemorrhage (SAH) is a serious complication resulting in delayed neurological deficit, increased morbidity, mortality, longer hospital stays, and rehabilitation time. It afflicts approximately 35 per 100,000 Americans per year, and there is currently no effective therapy. We present in vitro data suggesting that increasing intrinsic nitric oxide relaxation pathways in vascular smooth muscle via dopaminergic agonism ameliorates cerebral vasospasm after SAH. METHODS Cerebrospinal fluid (CSF) from patients with cerebral vasospasm after SAH (CSF(V)) was used to induce vasospasm in porcine carotid artery in vitro. Dopamine was added to test its ability to reverse spasm, and specific dopamine receptor antagonists were used to determine which receptor mediated the protection. Immunohistochemical techniques confirmed the presence of dopamine receptor subtypes and the involvement of NOS in the mechanism of dopamine protection. RESULTS Dopamine receptor 1, 2, and 3 subtypes are all present in porcine carotid artery. Dopamine significantly reversed spasm in vitro (67% relaxation), and this relaxation was prevented by Haloperidol, a D(2)R antagonist (10% relaxation, P < 0.05), but not by D(1) or D(3)-receptor antagonism. Both eNOS and iNOS expression were increased significantly in response to CSF(V) alone, and this was significantly enhanced by addition of dopamine, and blocked by Haloperidol. CONCLUSION Cerebral vasospasm is significantly reversed in a functional measure of vasospasm in vitro by dopamine, via a D(2)R-mediated pathway. The increase in NOS protein seen in both the endothelium and vascular smooth muscle in response to CSF(V) is enhanced by dopamine, also in a D(2)R-dependent mechanism.
Collapse
Affiliation(s)
- Gail J Pyne-Geithman
- Department of Neurology, University of Cincinnati, 3125 Eden Avenue, 2324 Vontz Center, Cincinnati, OH 45267-0536, USA.
| | | | | | | | | |
Collapse
|
200
|
de Cavanagh EMV, Flores I, Ferder M, Inserra F, Ferder L. Renin-angiotensin system inhibitors protect against age-related changes in rat liver mitochondrial DNA content and gene expression. Exp Gerontol 2008; 43:919-28. [PMID: 18765277 DOI: 10.1016/j.exger.2008.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Revised: 07/14/2008] [Accepted: 08/08/2008] [Indexed: 02/07/2023]
Abstract
Chronic renin-angiotensin system inhibition protects against liver fibrosis, ameliorates age-associated mitochondrial dysfunction and increases rodent lifespan. We hypothesized that life-long angiotensin-II-mediated stimulation of oxidant generation might participate in mitochondrial DNA "common deletion" formation, and the resulting impairment of bioenergetic capacity. Enalapril (10 mg/kg/d) or losartan (30 mg/kg/d) administered during 16.5 months were unable to prevent the age-dependent accumulation of rat liver mitochondrial DNA "common deletion", but attenuated the decrease of mitochondrial DNA content. This evidence - together with the enhancement of NRF-1 and PGC-1 mRNA contents - seems to explain why enalapril and losartan improved mitochondrial functioning and lowered oxidant production, since both the absolute number of mtDNA molecules and increased NRF-1 and PGC-1 transcription are positively related to mitochondrial respiratory capacity, and PGC-1 protects against increases in ROS production and damage. Oxidative stress evoked by abnormal respiratory function contributes to the pathophysiology of mitochondrial disease and human aging. If the present mitochondrial actions of renin-angiotensin system inhibitors are confirmed in humans they may modify the therapeutic significance of that strategy.
Collapse
Affiliation(s)
- Elena M V de Cavanagh
- Institute of Cardiovascular Pathophysiology (INFICA), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|