51
|
Padmini E, Lavanya S, Uthra V. Preeclamptic placental stress and over expression of mitochondrial HSP70. Clin Chem Lab Med 2009; 47:1073-80. [PMID: 19728848 DOI: 10.1515/cclm.2009.247] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Evidence is accumulating that mitochondrial (Mt) oxidative stress plays a role in the pathogenesis of preeclampsia. The current study analyzes the stress levels, energy status and associated enzymatic alteration in placental mitochondria of preeclamptic (n=30) and normotensive (n=35) subjects. METHODS Total Mt stress was measured using dichlorofluorescin (DCFH) oxidant analysis, malondialdehyde (MDA) concentrations, protein carbonyl (PC) concentrations and measurement of nitrite (NO2(-)) and nitrate (NO3(-)). Activity of antioxidant enzymes including superoxide dismutase (SOD), glutathione peroxidase (GPx) and the glutathione redox ratio (GRR) were measured. The ATP/ADP (adenosine triphosphate/adenosine diphosphate) concentrations and respiratory chain enzyme activities were also analyzed. The expression of heat shock protein 70 (HSP70) was measured in mitochondria. RESULTS The DCFH oxidants, MDA, PC concentrations, and concentrations of NO2(-) and NO3(-) were significantly higher in the preeclamptic group (p<0.01) compared with the control group. The activities of SOD, GPx, GRR [glutathione (GSH)/glutathione disulfate (GSSG)] (p<0.01, p<0.001), ATP/ADP and respiratory chain enzyme activities were reduced significantly (p<0.001) in preeclamptic conditions. The placental mitochondrial HSP70 (mtHSP70) showed significant over expression in the preeclamptic group (p<0.001) compared with the control group. CONCLUSIONS These results provide the first line of evidence for accumulated Mt stress demonstrated by increased stress markers, decreased antioxidants and enhanced mtHSP70. The study illustrates the probable protective mechanism of mtHSP70 against the generated stress. This is primarily to combat the enzymatic and free radical mediated damage produced in preeclampsia.
Collapse
Affiliation(s)
- Ekambaram Padmini
- P.G. Department of Biochemistry, Bharathi Women's College, Affiliated to University of Madras, Chennai, Tamilnadu, India.
| | | | | |
Collapse
|
52
|
Clement HW, Vazquez JF, Sommer O, Heiser P, Morawietz H, Hopt U, Schulz E, von Dobschütz E. Lipopolysaccharide-induced radical formation in the striatum is abolished in Nox2 gp91phox-deficient mice. J Neural Transm (Vienna) 2009; 117:13-22. [DOI: 10.1007/s00702-009-0327-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 09/20/2009] [Indexed: 11/24/2022]
|
53
|
Abstract
The endothelium is an important component of vascular homeostasis that is a target for injury in the setting of vascular disease. One means of promoting a maladaptive endothelial cell phenotype such as that seen in atherosclerosis is excess oxidative stress. Although this term once was almost exclusively used to describe low-density lipoprotein (LDL) and lipid oxidation in the vasculature, we now understand that the intracellular oxidant milieu is an important modulator of vascular cell function. Indeed, considerable data indicate that reactive oxygen species (ROS) are an important means of cellular signaling, although the precise mechanisms whereby ROS accomplish this are still under investigation. In this review, the data linking ROS to kinase activation and cell signaling in the endothelium is discussed, with a particular emphasis on the roles of protein thiol modification.
Collapse
Affiliation(s)
- Kai Chen
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | | |
Collapse
|
54
|
Seppet E, Gruno M, Peetsalu A, Gizatullina Z, Nguyen HP, Vielhaber S, Wussling MH, Trumbeckaite S, Arandarcikaite O, Jerzembeck D, Sonnabend M, Jegorov K, Zierz S, Striggow F, Gellerich FN. Mitochondria and energetic depression in cell pathophysiology. Int J Mol Sci 2009; 10:2252-2303. [PMID: 19564950 PMCID: PMC2695278 DOI: 10.3390/ijms10052252] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 04/25/2009] [Accepted: 05/14/2009] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial dysfunction is a hallmark of almost all diseases. Acquired or inherited mutations of the mitochondrial genome DNA may give rise to mitochondrial diseases. Another class of disorders, in which mitochondrial impairments are initiated by extramitochondrial factors, includes neurodegenerative diseases and syndromes resulting from typical pathological processes, such as hypoxia/ischemia, inflammation, intoxications, and carcinogenesis. Both classes of diseases lead to cellular energetic depression (CED), which is characterized by decreased cytosolic phosphorylation potential that suppresses the cell's ability to do work and control the intracellular Ca(2+) homeostasis and its redox state. If progressing, CED leads to cell death, whose type is linked to the functional status of the mitochondria. In the case of limited deterioration, when some amounts of ATP can still be generated due to oxidative phosphorylation (OXPHOS), mitochondria launch the apoptotic cell death program by release of cytochrome c. Following pronounced CED, cytoplasmic ATP levels fall below the thresholds required for processing the ATP-dependent apoptotic cascade and the cell dies from necrosis. Both types of death can be grouped together as a mitochondrial cell death (MCD). However, there exist multiple adaptive reactions aimed at protecting cells against CED. In this context, a metabolic shift characterized by suppression of OXPHOS combined with activation of aerobic glycolysis as the main pathway for ATP synthesis (Warburg effect) is of central importance. Whereas this type of adaptation is sufficiently effective to avoid CED and to control the cellular redox state, thereby ensuring the cell survival, it also favors the avoidance of apoptotic cell death. This scenario may underlie uncontrolled cellular proliferation and growth, eventually resulting in carcinogenesis.
Collapse
Affiliation(s)
- Enn Seppet
- Department of Pathophysiology, University of Tartu, Tartu, Estonia; E-Mail:
(M.G.)
| | - Marju Gruno
- Department of Pathophysiology, University of Tartu, Tartu, Estonia; E-Mail:
(M.G.)
| | - Ants Peetsalu
- Department of Surgery, University of Tartu, Tartu, Estonia; E-Mail:
(A.P.)
| | - Zemfira Gizatullina
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Huu Phuc Nguyen
- Department of Medical Genetics, University of Tübingen, Tübingen, Germany; E-Mail:
(H.P.N.)
| | - Stefan Vielhaber
- Department of Neurology, Otto von Guericke University, Magdeburg, Germany; E-Mail:
(S.V.)
| | - Manfred H.P. Wussling
- Bernstein Institute for Physiology, Martin-Luther-University Halle-Wittenberg, Germany; E-Mail:
(M.H.P.W.)
| | - Sonata Trumbeckaite
- Institute for Biomedical Research, Kaunas University of Medicine, Kaunas, Lithuania; E-Mails:
(S.T.);
(O.A.)
| | - Odeta Arandarcikaite
- Institute for Biomedical Research, Kaunas University of Medicine, Kaunas, Lithuania; E-Mails:
(S.T.);
(O.A.)
| | - Doreen Jerzembeck
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Maria Sonnabend
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Katharina Jegorov
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Stephan Zierz
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Germany; E-Mail:
(S.Z.)
| | - Frank Striggow
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Frank N. Gellerich
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| |
Collapse
|
55
|
Abstract
Hyperhomocysteinemia (HHcy) is a significant and independent risk factor for cardiovascular diseases. Endothelial dysfunction (ED) is the earliest indicator of atherosclerosis and vascular diseases. We and others have shown that HHcy induced ED in human and in animal models of HHcy induced by either high-methionine load or genetic deficiency. Six mechanisms have been suggested explaining HHcy-induced ED. These include 1) nitric oxide inhibition, 2) prostanoids regulation, 3) endothelium-derived hyperpolarizing factors suppression, 4) angiotensin II receptor-1 activation, 5) endothelin-1 induction, and 6) oxidative stress. The goal of this review is to elaborate these mechanisms and to discuss biological and molecular events related to HHcy-induced ED.
Collapse
Affiliation(s)
- Zhongjian Cheng
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, USA
| | | | | |
Collapse
|
56
|
Abstract
NAFLD (non-alcoholic fatty liver disease), associated with obesity and the cardiometabolic syndrome, is an important medical problem affecting up to 20% of western populations. Evidence indicates that mitochondrial dysfunction plays a critical role in NAFLD initiation and progression to the more serious condition of NASH (non-alcoholic steatohepatitis). Herein we hypothesize that mitochondrial defects induced by exposure to a HFD (high fat diet) contribute to a hypoxic state in liver and this is associated with increased protein modification by RNS (reactive nitrogen species). To test this concept, C57BL/6 mice were pair-fed a control diet and HFD containing 35% and 71% total calories (1 cal≈4.184 J) from fat respectively, for 8 or 16 weeks and liver hypoxia, mitochondrial bioenergetics, NO (nitric oxide)-dependent control of respiration, and 3-NT (3-nitrotyrosine), a marker of protein modification by RNS, were examined. Feeding a HFD for 16 weeks induced NASH-like pathology accompanied by elevated triacylglycerols, increased CYP2E1 (cytochrome P450 2E1) and iNOS (inducible nitric oxide synthase) protein, and significantly enhanced hypoxia in the pericentral region of the liver. Mitochondria from the HFD group showed increased sensitivity to NO-dependent inhibition of respiration compared with controls. In addition, accumulation of 3-NT paralleled the hypoxia gradient in vivo and 3-NT levels were increased in mitochondrial proteins. Liver mitochondria from mice fed the HFD for 16 weeks exhibited depressed state 3 respiration, uncoupled respiration, cytochrome c oxidase activity, and mitochondrial membrane potential. These findings indicate that chronic exposure to a HFD negatively affects the bioenergetics of liver mitochondria and this probably contributes to hypoxic stress and deleterious NO-dependent modification of mitochondrial proteins.
Collapse
|
57
|
Dai S, He Y, Zhang H, Yu L, Wan T, Xu Z, Jones D, Chen H, Min W. Endothelial-specific expression of mitochondrial thioredoxin promotes ischemia-mediated arteriogenesis and angiogenesis. Arterioscler Thromb Vasc Biol 2009; 29:495-502. [PMID: 19150880 DOI: 10.1161/atvbaha.108.180349] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Thioredoxin-2 (Trx2), a major antioxidant protein in mitochondria, enhances nitric oxide bioavailability and inhibits ASK1-dependent apoptosis in endothelial cells (ECs). However, the in vivo role of Trx2 in angiogenesis has not been defined. Here we used EC-specific transgenesis of Trx2 (Trx2-TG) in mice to determine the in vivo function of Trx2 in arteriogenesis and angiogenesis. METHODS AND RESULTS In a femoral artery ligation model, Trx2-TG mice had enhanced capacity in limb perfusion recovery and ischemic reserve capacity compared to the nontransgenic littermates. Ischemia-initiated arteriogenesis in the upper limb was augmented in Trx2-TG mice. Trx2-TG mice also showed significantly enhanced capillary formation and maturation in the lower limb. In nontransgenic limb, ischemia specifically induced a downregulation of Trx2 protein, leading to increased oxidative stress, ASK1 activation, and EC apoptosis. In contrast, Trx2-TG maintained a constitutive level of Trx2, reducing the ischemia-induced deleterious responses. We then defined the mechanism by which Trx2 increases angiogenesis using ECs isolated from Trx2-TG mice. Trx2-TG ECs showed increased NO and NO-dependent migration. In addition, these cells were more resistant to oxidative stress-induced activation of ASK1 signaling and apoptosis. Moreover, Trx2-augmented EC survival is NO-independent. To define the relative contributions of Trx2-increased NO and Trx2-reduced ASK1 apoptotic activity to angiogenesis in vivo, we examined Trx2 effects on ischemia-induced angiogenesis in eNOS-deficient mice. The eNOS deletion caused severe impairment in the functional flow recovery in response to ischemia. Trx2 expression in eNOS-KO mice still dramatically inhibited ischemia-induced ASK1 and EC apoptosis, leading to an enhanced functional flow recovery. CONCLUSIONS These in vivo and in vitro data support that Trx2 maintains EC function by two parallel pathways-scavenging ROS to increase NO bioavailability and inhibiting ASK1 activity to enhance EC survival, facilitating ischemia-mediated arteriogenesis and angiogenesis.
Collapse
Affiliation(s)
- Shengchuan Dai
- Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Thomas SR, Witting PK, Drummond GR. Redox control of endothelial function and dysfunction: molecular mechanisms and therapeutic opportunities. Antioxid Redox Signal 2008; 10:1713-65. [PMID: 18707220 DOI: 10.1089/ars.2008.2027] [Citation(s) in RCA: 289] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The endothelium is essential for the maintenance of vascular homeostasis. Central to this role is the production of endothelium-derived nitric oxide (EDNO), synthesized by the endothelial isoform of nitric oxide synthase (eNOS). Endothelial dysfunction, manifested as impaired EDNO bioactivity, is an important early event in the development of various vascular diseases, including hypertension, diabetes, and atherosclerosis. The degree of impairment of EDNO bioactivity is a determinant of future vascular complications. Accordingly, growing interest exists in defining the pathologic mechanisms involved. Considerable evidence supports a causal role for the enhanced production of reactive oxygen species (ROS) by vascular cells. ROS directly inactivate EDNO, act as cell-signaling molecules, and promote protein dysfunction, events that contribute to the initiation and progression of endothelial dysfunction. Increasing data indicate that strategies designed to limit vascular ROS production can restore endothelial function in humans with vascular complications. The purpose of this review is to outline the various ways in which ROS can influence endothelial function and dysfunction, describe the redox mechanisms involved, and discuss approaches for preventing endothelial dysfunction that may highlight future therapeutic opportunities in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Shane R Thomas
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | |
Collapse
|
59
|
Ren B, Zhang N, Yang J, Ding H. Nitric oxide-induced bacteriostasis and modification of iron-sulphur proteins in Escherichia coli. Mol Microbiol 2008; 70:953-64. [PMID: 18811727 DOI: 10.1111/j.1365-2958.2008.06464.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The nitric oxide (NO) cytotoxicity has been well documented in bacteria and mammalian cells. However, the underlying mechanism is still not fully understood. Here we report that transient NO exposure effectively inhibits cell growth of Escherichia coli in minimal medium under anaerobic growth conditions and that cell growth is restored when the NO-exposed cells are either supplemented with the branched-chain amino acids (BCAA) anaerobically or returned to aerobic growth conditions. The enzyme activity measurements show that dihydroxyacid dehydratase (IlvD), an iron-sulphur enzyme essential for the BCAA biosynthesis, is completely inactivated in cells by NO with the concomitant formation of the IlvD-bound dinitrosyl iron complex (DNIC). Fractionation of the cell extracts prepared from the NO-exposed cells reveals that a large number of different protein-bound DNICs are formed by NO. While the IlvD-bound DNIC and other protein-bound DNICs are stable in cells under anaerobic growth conditions, they are efficiently repaired under aerobic growth conditions even without new protein synthesis. Additional studies indicate that L-cysteine may have an important role in repairing the NO-modified iron-sulphur proteins in aerobically growing E. coli cells. The results suggest that cellular deficiency to repair the NO-modified iron-sulphur proteins may directly contribute to the NO-induced bacteriostasis under anaerobic conditions.
Collapse
Affiliation(s)
- Binbin Ren
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | | | | |
Collapse
|
60
|
Hansson GK. Atherosclerosis--an immune disease: The Anitschkov Lecture 2007. Atherosclerosis 2008; 202:2-10. [PMID: 18951547 DOI: 10.1016/j.atherosclerosis.2008.08.039] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 08/13/2008] [Accepted: 08/21/2008] [Indexed: 01/09/2023]
Abstract
Atherosclerosis is an inflammatory disease. This article reviews the emergence of this concept from studies of patients and their lesions, experimental animal models, and epidemiological cohorts. Immunohistochemical studies identified immune cells and mediators and provided evidence for inflammatory activation in the atherosclerotic lesion. In parallel, cell culture studies demonstrated the capacity of vascular cells to interact with immune cells. Subsequent studies of clinical and epidemiological materials have identified inflammatory markers and immunoregulatory genes as contributors of risk for myocardial infarction and stroke. Finally, experiments using gene-targeted mice have provided mechanistic understanding of the disease process. It is now thought that the atherosclerotic process is initiated when low-density lipoproteins accumulate in the intima, activate the endothelium, and promote recruitment of monocytes and T cells. Monocytes differentiate into macrophages, internalize modified lipoproteins, and end up as foam cells. T cells in lesions recognize local antigens and mount T helper-1 responses that contribute to local inflammation and plaque growth. This atherogenic pathway is counterbalanced by anti-inflammatory signals provided by regulatory immunity. Intensified inflammatory activation may lead to local proteolysis, plaque rupture, thrombus formation, ischemia and infarction. Novel therapeutic opportunities may emerge from understanding the role of inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Göran K Hansson
- Center for Molecular Medicine and Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
61
|
Viel EC, Benkirane K, Javeshghani D, Touyz RM, Schiffrin EL. Xanthine oxidase and mitochondria contribute to vascular superoxide anion generation in DOCA-salt hypertensive rats. Am J Physiol Heart Circ Physiol 2008; 295:H281-8. [PMID: 18487445 PMCID: PMC2494748 DOI: 10.1152/ajpheart.00304.2008] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 05/12/2008] [Indexed: 02/06/2023]
Abstract
Vascular superoxide anion (O(2)(*-)) levels are increased in DOCA-salt hypertensive rats. We hypothesized that the endothelin (ET)-1-induced generation of ROS in the aorta and resistance arteries of DOCA-salt rats originates partly from xanthine oxidase (XO) and mitochondria. Accordingly, we blocked XO and the mitochondrial oxidative phosphorylation chain to investigate their contribution to ROS production in mesenteric resistance arteries and the aorta from DOCA-salt rats. Systolic blood pressure rose in DOCA-salt rats and was reduced after 3 wk by apocynin [NAD(P)H oxidase inhibitor and/or radical scavenger], allopurinol (XO inhibitor), bosentan (ET(A/B) receptor antagonist), BMS-182874 (BMS; ET(A) receptor antagonist), and hydralazine. Plasma uric acid levels in DOCA-salt rats were similar to control unilaterally nephrectomized (UniNx) rats, reduced with allopurinol and bosentan, and increased with BMS. Levels of thiobarbituric acid-reacting substances were increased in DOCA-salt rats versus UniNx rats, and BMS, bosentan, and hydralazine prevented their increase. Dihydroethidium staining showed reduced O(2)(*-) production in mesenteric arteries and the aorta from BMS- and bosentan-treated DOCA-salt rats compared with untreated DOCA-salt rats. Increased O(2)(*-) derived from XO was reduced or prevented by all treatments in mesenteric arteries, whereas bosentan and BMS had no effect on aortas from DOCA-salt rats. O(2)(*-) generation decreased with in situ treatment by tenoyltrifluoroacetone and CCCP, inhibitors of mitochondrial electron transport complexes II and IV, respectively, whereas rotenone (mitochondrial complex I inhibitor) had no effect. Our findings demonstrate the involvement of ET(A) receptor-modulated O(2)(*-) derived from XO and from mitochondrial oxidative enzymes in arteries from DOCA-salt rats.
Collapse
Affiliation(s)
- Emilie C Viel
- Lady Davis Institute for Medical Research, Montreal, QC, Canada H3T 1E2
| | | | | | | | | |
Collapse
|
62
|
Abstract
Accumulating evidence supports the importance of redox signaling in the pathogenesis and progression of hypertension. Redox signaling is implicated in many different physiological and pathological processes in the vasculature. High blood pressure is in part determined by elevated total peripheral vascular resistance, which is ascribed to dysregulation of vasomotor function and structural remodeling of blood vessels. Aberrant redox signaling, usually induced by excessive production of reactive oxygen species (ROS) and/or by decreases in antioxidant activity, can induce alteration of vascular function. ROS increase vascular tone by influencing the regulatory role of endothelium and by direct effects on the contractility of vascular smooth muscle. ROS contribute to vascular remodeling by influencing phenotype modulation of vascular smooth muscle cells, aberrant growth and death of vascular cells, cell migration, and extracellular matrix (ECM) reorganization. Thus, there are diverse roles of the vascular redox system in hypertension, suggesting that the complexity of redox signaling in distinct spatial spectrums should be considered for a better understanding of hypertension.
Collapse
Affiliation(s)
- Moo Yeol Lee
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
63
|
Dahmani Y, Marcuello A, Díez-Sanchez C, Ruiz-Pesini E, Montoya J, López-Pérez MJ. Association of human mitochondrial DNA variants with plasma LDL levels. Mitochondrion 2008; 8:247-53. [DOI: 10.1016/j.mito.2008.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 03/05/2008] [Accepted: 04/15/2008] [Indexed: 10/22/2022]
|
64
|
Oxidative stress in vascular disease: causes, defense mechanisms and potential therapies. ACTA ACUST UNITED AC 2008; 5:338-49. [DOI: 10.1038/ncpcardio1211] [Citation(s) in RCA: 413] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Accepted: 02/01/2008] [Indexed: 02/07/2023]
|
65
|
Burwell LS, Brookes PS. Mitochondria as a target for the cardioprotective effects of nitric oxide in ischemia-reperfusion injury. Antioxid Redox Signal 2008; 10:579-99. [PMID: 18052718 DOI: 10.1089/ars.2007.1845] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During cardiac ischemia-reperfusion (IR) injury, excessive generation of reactive oxygen species (ROS) and overload of Ca(2+) at the mitochondrial level both lead to opening of the mitochondrial permeability transition (PT) pore on reperfusion. This can result in the depletion of ATP, irreversible oxidation of proteins, lipids, and DNA within the cardiomyocyte, and can trigger cell-death pathways. In contrast, mitochondria are also implicated in the cardioprotective signaling processes of ischemic preconditioning (IPC), to prevent IR-related pathology. Nitric oxide (NO*) has emerged as a potent effector molecule for a variety of cardioprotective strategies, including IPC. Whereas NO* is most noted for its activation of the "classic" soluble guanylate cyclase (sGC) signaling pathway, emerging evidence indicates that NO can directly act on mitochondria, independent of the sGC pathway, affording acute cardioprotection against IR injury. These direct effects of NO* on mitochondria are the focus of this review.
Collapse
Affiliation(s)
- Lindsay S Burwell
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | |
Collapse
|
66
|
Puddu P, Puddu GM, Cravero E, De Pascalis S, Muscari A. The putative role of mitochondrial dysfunction in hypertension. Clin Exp Hypertens 2008; 29:427-34. [PMID: 17994352 DOI: 10.1080/10641960701613852] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Hypertension is a condition associated with oxidative stress, endothelial dysfunction, and increased vascular resistance, representing probably both a cause and a consequence of elevated levels of reactive oxygen (ROS) and nitrogen (RNS) species. Mitochondria are important sites of ROS production, and a mitochondrial dysfunction, preceding endothelial dysfunction, might favor the development of hypertension. ROS production may also be induced by RNS, which inhibit the respiratory chain and may be generated through the action of a mitochondrial NO synthase. Mitochondrial uncoupling proteins are involved in both experimental and human hypertension. Finally, an excessive production of ROS may damage mitochondrial DNA, with resultant impairment in the synthesis of some components of the respiratory chain and further ROS production, a vicious cycle that may be implicated in hypertensive states.
Collapse
Affiliation(s)
- Paolo Puddu
- Department of Internal Medicine, Cardioangiology, Hepatology, University of Bologna, Bologna, Italy
| | | | | | | | | |
Collapse
|
67
|
Abstract
Elevated plasma levels of homocysteine are a metabolic risk factor for atherosclerotic vascular disease, as shown in numerous clinical studies that linked elevated homocysteine levels to de novo and recurrent cardiovascular events. High levels of homocysteine promote oxidant stress in vascular cells and tissue because of the formation of reactive oxygen species (ROS), which have been strongly implicated in the development of atherosclerosis. In particular, ROS have been shown to cause endothelial injury, dysfunction, and activation. Elevated homocysteine stimulates proinflammatory pathways in vascular cells, resulting in leukocyte recruitment to the vessel wall, mediated by the expression of adhesion molecules on endothelial cells and circulating monocytes and neutrophils, in the infiltration of leukocytes into the arterial wall mediated by increased secretion of chemokines, and in the differentiation of monocytes into cholesterol-scavenging macrophages. Furthermore, it stimulates the proliferation of vascular smooth muscle cells followed by the production of extracellular matrix. Many of these events involve redox-sensitive signaling events, which are promoted by elevated homocysteine, and result in the formation of atherosclerotic lesions. In this article, we review current knowledge about the role of homocysteine on oxidant stress-mediated vascular inflammation during the development of atherosclerosis.
Collapse
Affiliation(s)
- Louisa Papatheodorou
- Department of Vascular Medicine, Medical Policlinic-City Campus, University of Munich Medical Center, Munich, Germany
| | | |
Collapse
|
68
|
Seppet E, Gizatullina Z, Trumbeckaite S, Zierz S, Striggow F, Gellerich FN. Mitochondrial Medicine: The Central Role of Cellular Energetic Depression and Mitochondria in Cell Pathophysiology. MOLECULAR SYSTEM BIOENERGETICS 2007:479-520. [DOI: 10.1002/9783527621095.ch15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
69
|
de Cavanagh EMV, Inserra F, Ferder M, Ferder L. From mitochondria to disease: role of the renin-angiotensin system. Am J Nephrol 2007; 27:545-53. [PMID: 17785964 DOI: 10.1159/000107757] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Accepted: 07/16/2007] [Indexed: 01/12/2023]
Abstract
Mitochondria are energy-producing organelles that conduct other key cellular tasks. Thus, mitochondrial damage may impair various aspects of tissue functioning. Mitochondria generate oxygen- and nitrogen-derived oxidants, being themselves major oxidation targets. Dysfunctional mitochondria seem to contribute to the pathophysiology of hypertension, cardiac failure, the metabolic syndrome, obesity, diabetes mellitus, renal disease, atherosclerosis, and aging. Mitochondrial proteins and metabolic intermediates participate in various cellular processes, apart from their well-known roles in energy metabolism. This emphasizes the participation of dysfunctional mitochondria in disease, notwithstanding that most evidences supporting this concept come from animal and cultured-cell studies. Mitochondrial oxidant production is altered by several factors related to vascular pathophysiology. Among these, angiotensin-II stimulates mitochondrial oxidant release leading to energy metabolism depression. By lowering mitochondrial oxidant production, angiotensin-II inhibition enhances energy production and protects mitochondrial structure. This seems to be one of the mechanisms underlying the benefits of angiotensin-II inhibition in hypertension, diabetes, and aging rodent models. If some of these findings can be reproduced in humans, they would provide a new perspective on the implications that RAS-blockade can offer as a therapeutic strategy. This review intends to present available information pointing to mitochondria as targets for therapeutic Ang-II blockade in human renal and CV disease.
Collapse
Affiliation(s)
- E M V de Cavanagh
- Laboratory of Experimental Nephrology, Institute for Cardiovascular Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
70
|
McMackin CJ, Widlansky ME, Hamburg NM, Huang AL, Weller S, Holbrook M, Gokce N, Hagen TM, Keaney JF, Vita JA. Effect of combined treatment with alpha-Lipoic acid and acetyl-L-carnitine on vascular function and blood pressure in patients with coronary artery disease. J Clin Hypertens (Greenwich) 2007; 9:249-55. [PMID: 17396066 PMCID: PMC2734271 DOI: 10.1111/j.1524-6175.2007.06052.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mitochondria produce reactive oxygen species that may contribute to vascular dysfunction. alpha-Lipoic acid and acetyl-L-carnitine reduce oxidative stress and improve mitochondrial function. In a double-blind crossover study, the authors examined the effects of combined alpha-lipoic acid/acetyl-L-carnitine treatment and placebo (8 weeks per treatment) on vasodilator function and blood pressure in 36 subjects with coronary artery disease. Active treatment increased brachial artery diameter by 2.3% (P=.008), consistent with reduced arterial tone. Active treatment tended to decrease systolic blood pressure for the whole group (P=.07) and had a significant effect in the subgroup with blood pressure above the median (151+/-20 to 142+/-18 mm Hg; P=.03) and in the subgroup with the metabolic syndrome (139+/-21 to 130+/-18 mm Hg; P=.03). Thus, mitochondrial dysfunction may contribute to the regulation of blood pressure and vascular tone. Further studies are needed to confirm these findings and determine the clinical utility of alpha-lipoic acid/acetyl-L-carnitine as antihypertensive therapy.
Collapse
Affiliation(s)
- Craig J. McMackin
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| | - Michael E. Widlansky
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| | - Naomi M. Hamburg
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| | - Alex L. Huang
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| | - Susan Weller
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| | - Monika Holbrook
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| | - Noyan Gokce
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| | - Tory M. Hagen
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| | - John F. Keaney
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| | - Joseph A. Vita
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA; and the Linus Pauling Institute at Oregon State University, Corvallis, OR
| |
Collapse
|
71
|
Abstract
Disturbances in vascular function contribute to the development of several diseases of increasing prevalence and thereby contribute significantly to human mortality and morbidity. Atherosclerosis, diabetes, heart failure, and ischemia with attendant reperfusion injury share many of the same risk factors, among the most important being oxidative stress and alterations in the blood concentrations of compounds that influence oxidative stress, such as oxidized low-density lipoprotein. In this review, we focus on endothelial cells: cells in the frontline against these disturbances. Because ATP supplies in endothelial cells are relatively independent of mitochondrial oxidative pathways, the mitochondria of endothelial cells have been somewhat neglected. However, they are emerging as agents with diverse roles in modulating the dynamics of intracellular calcium and the generation of reactive oxygen species and nitric oxide. The mitochondria may also constitute critical "targets" of oxidative stress, because survival of endothelial cells can be compromised by opening of the mitochondrial permeability transition pore or by mitochondrial pathways of apoptosis. In addition, evidence suggests that endothelial mitochondria may play a "reconnaissance" role. For example, although the exact mechanism remains obscure, endothelial mitochondria may sense levels of oxygen in the blood and relay this information to cardiac myocytes as well as modulating the vasodilatory response mediated by endothelial nitric oxide.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, Department of Medicine, Royal Free and University College Medical School, London, United Kingdom.
| | | |
Collapse
|
72
|
Zhang H, Luo Y, Zhang W, He Y, Dai S, Zhang R, Huang Y, Bernatchez P, Giordano FJ, Shadel G, Sessa WC, Min W. Endothelial-specific expression of mitochondrial thioredoxin improves endothelial cell function and reduces atherosclerotic lesions. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1108-20. [PMID: 17322393 PMCID: PMC1864879 DOI: 10.2353/ajpath.2007.060960] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The function of the mitochondrial antioxidant system thioredoxin (Trx2) in vasculature is not understood. By using endothelial cell (EC)-specific transgenesis of the mitochondrial form of the thioredoxin gene in mice (Trx2 TG), we show the critical roles of Trx2 in regulating endothelium functions. Trx2 TG mice have increased total antioxidants, reduced oxidative stress, and increased nitric oxide (NO) levels in serum compared with their control littermates. Consistently, aortas from Trx2 TG mice show reduced vasoconstriction and enhanced vasodilation. By using ECs isolated from Trx2 TG mice, we further show that Trx2 increases the capacities of ECs in scavenging reactive oxygen species generated from mitochondria, resulting in increases in NO bioavailability in ECs. More importantly, Trx2 improves EC function and reduces atherosclerotic lesions in the apolipoprotein E-deficient mouse model. Our data provide the first evidence that Trx2 plays a critical role in preserving vascular EC function and prevention of atherosclerosis development, in part by reducing oxidative stress and increasing NO bioavailability.
Collapse
Affiliation(s)
- Haifeng Zhang
- Interdepartmental Program in Vascular Biology and Transplantation, Department of Pathology, Yale University School of Medicine, BCMM 454, 295 Congress Ave., New Haven, CT 06510, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Yang Z, Harrison CM, Chuang GC, Ballinger SW. The role of tobacco smoke induced mitochondrial damage in vascular dysfunction and atherosclerosis. Mutat Res 2007; 621:61-74. [PMID: 17428506 PMCID: PMC2212590 DOI: 10.1016/j.mrfmmm.2007.02.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 12/12/2006] [Accepted: 12/13/2006] [Indexed: 01/08/2023]
Abstract
The majority of individuals chronically exposed to tobacco smoke will eventually succumb to cardiovascular disease (CVD). However, despite the major cardiovascular health implications of tobacco smoke exposure, concepts of how such exposure specifically results in cardiovascular cell dysfunction that leads to CVD development are still being explored. Moreover, surprisingly little is known about the effects of prenatal and childhood tobacco smoke exposure on adult CVD development. Herein, it is proposed that the mitochondrion is a central target for environmental oxidants, including tobacco smoke. By virtue of its multiple, essential roles in cell function including energy production, oxidant signaling, apoptosis, immune response, and thermogenesis, damage to the mitochondrion will likely play an important role in the development of multiple common forms of human disease, including CVD. Specifically, this review will discuss the potential role of tobacco smoke and environmental oxidant exposure in the induction of mitochondrial damage which is related to CVD development. Furthermore, mechanisms of how mitochondrial damage can initiate and/or contribute to CVD are discussed, as are experimental results that are consistent with the hypothesis that mitochondrial damage and dysfunction will increase CVD susceptibility. Aspects of both adult and developmental (fetal and childhood) exposure to tobacco smoke on mitochondrial damage, function and disease development are also discussed, including the future implications and direction of studies involving the role of the mitochondrion in influencing disease susceptibility mediated by environmental factors.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294-001, United States
| | | | | | | |
Collapse
|
74
|
Chan JYH, Cheng HL, Chou JLJ, Li FCH, Dai KY, Chan SHH, Chang AYW. Heat Shock Protein 60 or 70 Activates Nitric-oxide Synthase (NOS) I- and Inhibits NOS II-associated Signaling and Depresses the Mitochondrial Apoptotic Cascade during Brain Stem Death. J Biol Chem 2007; 282:4585-4600. [PMID: 17150954 DOI: 10.1074/jbc.m603394200] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The cellular and molecular basis of brain stem death remains an enigma. As the origin of a "life-and-death" signal that reflects the progression toward brain stem death, the rostral ventrolateral medulla (RVLM) is a suitable neural substrate for mechanistic delineation of this phenomenon. Here, we evaluated the hypothesis that heat shock proteins (HSPs) play a neuroprotective role in the RVLM during brain stem death and delineated the underlying mechanisms, using a clinically relevant animal model that employed the organophosphate pesticide mevinphos (Mev) as the experimental insult. In Sprague-Dawley rats, proteomic, Western blot, and real-time PCR analyses demonstrated that Mev induced de novo synthesis of HSP60 or HSP70 in the RVLM without affecting HSP90 level. Loss-of-function manipulations of HSP60 or HSP70 in the RVLM using anti-serum or antisense oligonucleotide potentiated Mev-elicited cardiovascular depression alongside reduced nitric-oxide synthase (NOS) I/protein kinase G signaling, enhanced NOS II/peroxynitrite cascade, intensified nucleosomal DNA fragmentation, elevated cytoplasmic histone-associated DNA fragments or activated caspase-3, and augmented the cytochrome c/caspase-3 cascade of apoptotic signaling in the RVLM. Co-immunoprecipitation experiments further revealed a progressive increase in the complex formed between HSP60 and mitochondrial or cytosolic Bax or mitochondrial Bcl-2 during Mev intoxication, alongside a dissociation of the cytosolic HSP60-Bcl-2 complex. We conclude that HSP60 and HSP70 confer neuroprotection against Mev intoxication by ameliorating cardiovascular depression via an anti-apoptotic action in the RVLM. The possible underlying intracellular processes include enhancing NOS I/protein kinase G signaling and inhibiting the NOS II/peroxynitrite cascade. In addition, HSP60 exerts its effects against apoptosis by blunting Mev-induced activation of the Bax/cytochrome c/caspase-3 cascade.
Collapse
Affiliation(s)
- Julie Y H Chan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81346
| | - Hsiao-Lei Cheng
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the
| | - Jimmy L J Chou
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the
| | - Faith C H Li
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the
| | - Kuang-Yu Dai
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the
| | - Samuel H H Chan
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the; Center for Gene Regulation and Signal Transduction Research, National Cheng Kung University, Tainan 70101, Taiwan
| | - Alice Y W Chang
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the; Center for Gene Regulation and Signal Transduction Research, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
75
|
Elahi MM, Naseem KM, Matata BM. Nitric oxide in blood. The nitrosative-oxidative disequilibrium hypothesis on the pathogenesis of cardiovascular disease. FEBS J 2007; 274:906-23. [PMID: 17244198 DOI: 10.1111/j.1742-4658.2007.05660.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
There is growing evidence that altered production and/or spatio-temporal distribution of reactive oxidant species and reactive nitrosative species in blood creates oxidative and/or nitrosative stresses in the failing myocardium and endothelium. This contributes to the abnormal cardiac and vascular phenotypes that characterize cardiovascular disease. These derangements at the system level can now be interpreted at the integrated cellular and molecular levels in terms of effects on signaling elements in the heart and vasculature. The end results of nitric oxide/redox disequilibrium have implications for cardiac and vascular homeostasis and may result in the development of atherosclerosis, myocardial tissue remodelling and hypertrophy. Reactive oxygen species/reactive nitrogen species generation is also attributed to the transit from hypertrophic to apoptotic phenotypes, a possible mechanism of myocardial failure. In this review, we highlight the possible roles of altered production and/or spatio-temporal distribution of reactive oxidant species and reactive nitrosative species in blood on the pathogenesis of the failing cardiovascular system.
Collapse
|
76
|
Xu W, Koeck T, Lara AR, Neumann D, DiFilippo FP, Koo M, Janocha AJ, Masri FA, Arroliga AC, Jennings C, Dweik RA, Tuder RM, Stuehr DJ, Erzurum SC. Alterations of cellular bioenergetics in pulmonary artery endothelial cells. Proc Natl Acad Sci U S A 2007; 104:1342-7. [PMID: 17227868 PMCID: PMC1783136 DOI: 10.1073/pnas.0605080104] [Citation(s) in RCA: 290] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is pathogenetically related to low levels of the vasodilator nitric oxide (NO). Because NO regulates cellular respiration and mitochondrial biogenesis, we hypothesized that abnormalities of bioenergetics may be present in IPAH. Evaluation of pulmonary artery endothelial cells from IPAH and control lungs in vitro revealed that oxygen consumption of IPAH cells was decreased, especially in state 3 respiration with substrates glutamate-malate or succinate, and this decrease paralleled reduction in Complex IV activity and IPAH cellular NO synthesis. IPAH pulmonary artery endothelial cells had decreased mitochondrial dehydrogenase activity and lowered mitochondrial numbers per cell and mitochondrial DNA content, all of which increased after exposure to NO donors. Although IPAH/pulmonary artery endothelial cells' ATP content was similar to control under normoxia, cellular ATP did not change significantly in IPAH cells under hypoxia, whereas ATP decreased 35% in control cells, identifying a greater dependence on cellular respiration for energy in control cells. Evidence that glucose metabolism was subserving the primary role for energy requirements of IPAH cells was provided by the approximately 3-fold greater glycolytic rate of IPAH cells. Positron emission tomography scan with [18F]fluoro-deoxy-D-glucose performed on IPAH patients and healthy controls revealed significantly higher uptake in IPAH lungs as compared with controls, confirming that the glycolytic rate was increased in vivo. Thus, there are substantial changes in bioenergetics of IPAH endothelial cells, which may have consequences for pulmonary hypertensive responses and potentially in development of novel imaging modalities for diagnosis and evaluation of treatment.
Collapse
Affiliation(s)
| | | | | | - Donald Neumann
- Nuclear Medicine, Cleveland Clinic, Cleveland, OH 44195; and
| | | | | | | | | | | | | | - Raed A. Dweik
- Departments of *Pathobiology
- Pulmonary and Critical Care Medicine, and
| | - Rubin M. Tuder
- Division of Cardiopulmonary Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | - Serpil C. Erzurum
- Departments of *Pathobiology
- Pulmonary and Critical Care Medicine, and
- To whom correspondence should be addressed at:
Cleveland Clinic, Lerner Research Institute, 9500 Euclid Avenue/NC22, Cleveland, OH 44195. E-mail:
| |
Collapse
|
77
|
Clempus RE, Griendling KK. Reactive oxygen species signaling in vascular smooth muscle cells. Cardiovasc Res 2006; 71:216-25. [PMID: 16616906 PMCID: PMC1934427 DOI: 10.1016/j.cardiores.2006.02.033] [Citation(s) in RCA: 268] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Revised: 02/22/2006] [Accepted: 02/27/2006] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) have been shown to function as important signaling molecules in the cardiovascular system. Vascular smooth muscle cells (VSMCs) contain several sources of ROS, among which the NADPH oxidases are predominant. In VSMCs, ROS mediate many pathophysiological processes, such as growth, migration, apoptosis and secretion of inflammatory cytokines, as well as physiological processes, such as differentiation, by direct and indirect effects at multiple signaling levels. Therefore, it becomes critical to understand the different roles ROS play in the physiology and pathophysiology of VSMCs.
Collapse
Affiliation(s)
- Roza E. Clempus
- Department of Medicine, Division of Cardiology, Emory University, 319 WMB, 1639 Pierce Dr. Atlanta, GA 30322, United States
| | - Kathy K. Griendling
- Department of Medicine, Division of Cardiology, Emory University, 319 WMB, 1639 Pierce Dr. Atlanta, GA 30322, United States
| |
Collapse
|
78
|
de Cavanagh EMV, Toblli JE, Ferder L, Piotrkowski B, Stella I, Inserra F. Renal mitochondrial dysfunction in spontaneously hypertensive rats is attenuated by losartan but not by amlodipine. Am J Physiol Regul Integr Comp Physiol 2006; 290:R1616-25. [PMID: 16410402 DOI: 10.1152/ajpregu.00615.2005] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mitochondrial dysfunction is associated with cardiovascular damage; however, data on a possible association with kidney damage are scarce. Here, we aimed at investigating whether 1) kidney impairment is related to mitochondrial dysfunction; and 2) ANG II blockade, compared with Ca2+ channel blockade, can reverse potential mitochondrial changes in hypertension. Eight-week-old male spontaneously hypertensive rats (SHR) received water containing losartan (40 mg·kg−1·day−1, SHR+Los), amlodipine (3 mg·kg−1·day−1, SHR+Amlo), or no additions (SHR) for 6 mo. Wistar-Kyoto rats (WKY) were normotensive controls. Glomerular and tubulointerstitial damage, systolic blood pressure, and proteinuria were higher, and creatinine clearance was lower in SHR vs. SHR+Los and WKY. In SHR+Amlo, blood pressure was similar to WKY, kidney function was similar to SHR, and renal lesions were lower than in SHR, but higher than in SHR+Los. In kidney mitochondria from SHR and SHR+Amlo, membrane potential, nitric oxide synthase, manganese-superoxide dismutase and cytochrome oxidase activities, and uncoupling protein-2 content were lower than in SHR+Los and WKY. In SHR and SHR+Amlo, mitochondrial H2O2 production was higher than in SHR+Los and WKY. Renal glutathione content was lower in SHR+Amlo relative to SHR, SHR+Los, and WKY. In SHR and SHR+Amlo, glutathione was relatively more oxidized than in SHR+Los and WKY. Tubulointerstitial α-smooth muscle actin labeling was inversely related to manganese-superoxide dismutase activity and uncoupling protein-2 content. These findings suggest that oxidant stress is associated with renal mitochondrial dysfunction in SHR. The mitochondrial-antioxidant actions of losartan may be an additional or alternative way to explain some of the beneficial effects of AT1-receptor antagonists.
Collapse
Affiliation(s)
- Elena M V de Cavanagh
- Physical-Chemistry Department, School of Pharmacy and Biochemistry, University of Buenos Aires, Helguera 2365, Buenos Aires 1417, Argentina
| | | | | | | | | | | |
Collapse
|
79
|
VUONG TRI, MARTIN LUC, MATAR CHANTAL. ANTIOXIDANT ACTIVITY OF FERMENTED BERRY JUICES AND THEIR EFFECTS ON NITRIC OXIDE AND TUMOR NECROSIS FACTOR-ALPHA PRODUCTION IN MACROPHAGES 264.7 GAMMA NO(-) CELL LINE. J Food Biochem 2006. [DOI: 10.1111/j.1745-4514.2006.00054.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
80
|
Chen YR, Chen CL, Yeh A, Liu X, Zweier JL. Direct and Indirect Roles of Cytochrome b in the Mediation of Superoxide Generation and NO Catabolism by Mitochondrial Succinate-Cytochrome c Reductase. J Biol Chem 2006; 281:13159-13168. [PMID: 16531408 DOI: 10.1074/jbc.m513627200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Mitochondrial superoxide (O2*-) production is an important mediator of oxidative cellular injury. Succinate-cytochrome c reductase (SCR) of the electron transport chain has been implicated as an essential part of the mediation of O2*- generation and an alternative target of nitric oxide (NO) in the regulation of mitochondrial respiration. The Q cycle mechanism plays a central role in controlling both events. In the present work, O2*- generation by SCR was measured with the EPR spin-trapping technique using DEPMPO (5-diethoxylphosphoryl-5-methyl-1-pyrroline N-oxide) as the spin trap. In the presence of succinate, O2*- generation from SCR was detected as the spin adduct DEPMPO/*OOH. Inhibitors of the Q(o*-) site only marginally reduced (20-30%) this O2*- production, suggesting a secondary role of Q(o*-) in the mediation of O2*- generation. Addition of cyanide significantly decreased (approximately 70%) O2*- production, indicating the involvement of the heme component. UV-visible spectral analysis revealed that oxidation of ferrocytochrome b was accompanied by cytochrome c(1) reduction, and the reaction was mediated by the formation of an O2*- intermediate, indicating a direct role for cytochrome b in O2*- generation. In the presence of NO, DEPMPO/*OOH production was progressively diminished, implying that NO interacted with SCR or trapped the O2*-. The consumption of NO by SCR was investigated by electrochemical detection using an NO electrode. In the presence of succinate, SCR-mediated NO consumption was observed and inhibited by the addition of superoxide dismutase, suggesting the involvement of O2*-. Under the conditions of argon saturation, the NO consumption rate was not enhanced by succinate, suggesting a direct role for O2*- in the mediation of NO consumption. In the presence of succinate, oxidation of the ferrocytochrome b moiety of SCR was accelerated by the addition of NO, and was inhibited by argon saturation, indicating an indirect role for cytochrome b in the mediation of NO consumption.
Collapse
Affiliation(s)
- Yeong-Renn Chen
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210; Department of Molecular and Cellular Biochemistry, College of Medicine, Ohio State University, Columbus, Ohio 43210.
| | - Chwen-Lih Chen
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Alexander Yeh
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Xiaoping Liu
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Jay L Zweier
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210; Department of Molecular and Cellular Biochemistry, College of Medicine, Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
81
|
Chan JYH, Chang AYW, Chan SHH. New insights on brain stem death: From bedside to bench. Prog Neurobiol 2005; 77:396-425. [PMID: 16376477 DOI: 10.1016/j.pneurobio.2005.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Revised: 10/31/2005] [Accepted: 11/03/2005] [Indexed: 01/07/2023]
Abstract
As much as brain stem death is currently the clinical definition of death in many countries and is a phenomenon of paramount medical importance, there is a dearth of information on its mechanistic underpinnings. A majority of the clinical studies are concerned only with methods to determine brain stem death. Whereas a vast amount of information is available on the cellular and molecular mechanisms of cell death, rarely are these studies directed specifically towards the understanding of brain stem death. This review presents a framework for translational research on brain stem death that is based on systematically coordinated clinical and laboratory efforts that center on this phenomenon. It begins with the identification of a novel clinical marker from patients that is related specifically to brain stem death. After realizing that this "life-and-death" signal is related to the functional integrity of the brain stem, its origin is traced to the rostral ventrolateral medulla (RVLM). Subsequent laboratory studies on this neural substrate in animal models of brain stem death provide credence to the notion that both "pro-life" and "pro-death" programs are at work during the progression towards death. Those programs (mitochondrial functions, nitric oxide, peroxynitrite, superoxide anion, coenzyme Q10, heat shock proteins and ubiquitin-proteasome system) hitherto identified from the RVLM are presented, along with their cellular and molecular mechanisms. It is proposed that outcome of the interplay between the "pro-life" and "pro-death" programs (dying) in this neural substrate determines the final fate of the individual (being dead). Thus, identification of additional programs in the RVLM and delineation of their regulatory mechanisms should shed new lights on future directions for clinical management of life-and-death.
Collapse
Affiliation(s)
- Julie Y H Chan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81346, Taiwan, ROC
| | | | | |
Collapse
|
82
|
Perez-de-Arce K, Foncea R, Leighton F. Reactive oxygen species mediates homocysteine-induced mitochondrial biogenesis in human endothelial cells: Modulation by antioxidants. Biochem Biophys Res Commun 2005; 338:1103-9. [PMID: 16259958 DOI: 10.1016/j.bbrc.2005.10.053] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Accepted: 10/07/2005] [Indexed: 11/20/2022]
Abstract
It has been proposed that homocysteine (Hcy)-induces endothelial dysfunction and atherosclerosis by generation of reactive oxygen species (ROS). A previous report has shown that Hcy promotes mitochondrial damage. Considering that oxidative stress can affect mitochondrial biogenesis, we hypothesized that Hcy-induced ROS in endothelial cells may lead to increased mitochondrial biogenesis. We found that Hcy-induced ROS (1.85-fold), leading to a NF-kappaB activation and increase the formation of 3-nitrotyrosine. Furthermore, expression of the mitochondrial biogenesis factors, nuclear respiratory factor-1 and mitochondrial transcription factor A, was significantly elevated in Hcy-treated cells. These changes were accompanied by increase in mitochondrial mass and higher mRNA and protein expression of the subunit III of cytochrome c oxidase. These effects were significantly prevented by pretreatment with the antioxidants, catechin and trolox. Taken together, our results suggest that ROS is an important mediator of mitochondrial biogenesis induced by Hcy, and that modulation of oxidative stress by antioxidants may protect against the adverse vascular effects of Hcy.
Collapse
Affiliation(s)
- Karen Perez-de-Arce
- Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | |
Collapse
|
83
|
Puddu GM, Cravero E, Arnone G, Muscari A, Puddu P. Molecular aspects of atherogenesis: new insights and unsolved questions. J Biomed Sci 2005; 12:839-53. [PMID: 16328782 DOI: 10.1007/s11373-005-9024-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Accepted: 08/17/2005] [Indexed: 10/25/2022] Open
Abstract
The development of atherosclerotic disease results from the interaction between environment and genetic make up. A key factor in atherogenesis is the oxidative modification of lipids, which is involved in the recruitment of mononuclear leukocytes to the arterial intima--a process regulated by several groups of adhesion molecules and cytokines. Activated leukocytes, as well as endothelial mitochondria, can produce reactive oxygen species (ROS) that are associated with endothelial dysfunction, a cause of reduced nitric oxide (NO) bioactivity and further ROS production. Peroxisome proliferator-activated receptors (PPAR) and liver X receptors (LXR) are nuclear receptors significantly involved in the control of lipid metabolism, inflammation and insulin sensitivity. Also, an emerging role has been suggested for G protein coupled receptors and for the small Ras and Rho GTPases in the regulation of the expression of endothelial NO synthase (eNOS) and of tissue factor, which are involved in thrombus formation and modulation of vascular tone. Further, the interactions among eNOS, cholesterol, oxidated LDL and caveola membranes are probably involved in some molecular changes observed in vascular diseases. Despite the relevance of oxidative processes in atherogenesis, anti-oxidants have failed to significantly improve atherosclerosis (ATS) prevention, while statins have proved to be the most successful drugs.
Collapse
Affiliation(s)
- Giovanni Maria Puddu
- Department of Internal Medicine and Aging, S. Orsola-Malpighi Hospital, Bologna, Italy
| | | | | | | | | |
Collapse
|
84
|
Wilcox CS. Oxidative stress and nitric oxide deficiency in the kidney: a critical link to hypertension? Am J Physiol Regul Integr Comp Physiol 2005; 289:R913-35. [PMID: 16183628 DOI: 10.1152/ajpregu.00250.2005] [Citation(s) in RCA: 357] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is growing evidence that oxidative stress contributes to hypertension. Oxidative stress can precede the development of hypertension. In almost all models of hypertension, there is oxidative stress that, if corrected, lowers BP, whereas creation of oxidative stress in normal animals can cause hypertension. There is overexpression of the p22(phox) and Nox-1 components of NADPH oxidase and reduced expression of extracellular superoxide dismutase (EC-SOD) in the kidneys of ANG II-infused rodents, whereas there is overexpression of p47(phox) and gp91(phox) and reduced expression of intracellular SOD with salt loading. Several mechanisms have been identified that can make oxidative stress self-sustaining. Reactive oxygen species (ROS) can enhance afferent arteriolar tone and reactivity both indirectly via potentiation of tubuloglomerular feedback and directly by microvascular mechanisms that diminish endothelium-derived relaxation factor/nitric oxide responses, generate a cyclooxygenase-2-dependent endothelial-derived contracting factor that activates thromboxane-prostanoid receptors, and enhance vascular smooth muscle cells reactivity. ROS can diminish the efficiency with which the kidney uses O(2) for Na(+) transport and thereby diminish the P(O(2)) within the kidney cortex. This may place a break on further ROS generation yet could further enhance vasculopathy and hypertension. There is a tight relationship between oxidative stress in the kidney and the development and maintenance of hypertension.
Collapse
Affiliation(s)
- Christopher S Wilcox
- Division of Nephrology and Hypertension, Georgetown University Medical Center, 3800 Reservoir Rd., NW, Washington, DC 20007, USA.
| |
Collapse
|
85
|
Schild L, Jaroscakova I, Lendeckel U, Wolf G, Keilhoff G. Neuronal nitric oxide synthase controls enzyme activity pattern of mitochondria and lipid metabolism. FASEB J 2005; 20:145-7. [PMID: 16246868 DOI: 10.1096/fj.05-3898fje] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mitochondria are affected by endogenous nitric oxide (NO). Besides effects of NO on mitochondrial enzymes and the stimulation of mitochondrial H2O2 production, a NO-dependent increase in mitochondrial biogenesis in several tissues has been reported. It is still obscure whether NO generated by one specific or different NO synthase (NOS) isoenzymes determine such effects. Therefore, we analyzed the amount of mitochondria, respiratory chain enzyme complexes, and citrate synthase in the brain, muscle, heart, kidney, and liver by comparing wild-type (WT) mice and mice lacking the neuronal nitric oxide synthase isoform (nNOS-KO). Our results show that the activities of NADH:cytochrome c oxidoreductase and succinate cytochrome c oxidoreductase differ between WT and nNOS-KO mice. However, similar quantities of mitochondria were found in the homogenates of tissues in WT and nNOS-KO animals. Most impressive, higher activities and protein of citrate synthase were found in the brain, muscle, heart, kidney, and liver of nNOS-KO mice. Additionally, higher contents of fatty acid synthase and lipids were determined in the livers of nNOS-KO mice but not in the heart and brain. Furthermore, liver mitochondria from nNOS-KO mice consumed pyruvate at a higher rate and released more citric acid. Our data document a previously unrecognized role of endogenous NO in the regulation of lipid metabolism.
Collapse
Affiliation(s)
- Lorenz Schild
- Institut für Klinische Chemie und Pathologische Biochemie, Bereich Pathologische Biochemie, Medizinische Fakultät der Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany.
| | | | | | | | | |
Collapse
|
86
|
Stirone C, Duckles SP, Krause DN, Procaccio V. Estrogen increases mitochondrial efficiency and reduces oxidative stress in cerebral blood vessels. Mol Pharmacol 2005; 68:959-65. [PMID: 15994367 DOI: 10.1124/mol.105.014662] [Citation(s) in RCA: 217] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We report here that estrogen (E(2)) modulates mitochondrial function in the vasculature. Mitochondrial dysfunction is implicated in the etiology of vascular disease; thus, vasoprotection by estrogen may involve hormonal effects on the mitochondria. To test this hypothesis, mitochondria were isolated from cerebral blood vessels obtained from ovariectomized female rats, with or without E(2) replacement. Estrogen receptor-alpha (ER-alpha) was detected in mitochondria by immunoblot and confocal imaging of intact vessels. E(2) treatment in vivo increased the levels of specific proteins in cerebrovascular mitochondria, such as ER-alpha, cytochrome c, subunit IV of complex IV, and manganese superoxide dismutase, all encoded in the nuclear genome, and subunit I of complex IV, encoded in the mitochondrial genome. Levels of glutathione peroxidase-1 and catalase, however, were not affected. Functional assays of mitochondrial citrate synthase and complex IV, key rate-limiting steps in energy production, showed that E(2) treatment increased enzyme activity. In contrast, mitochondrial production of hydrogen peroxide was decreased in vessels from E(2)-treated animals. In vitro incubation of cerebral vessels with 10 nM 17beta-estradiol for 18 h also elevated levels of mitochondrial cytochrome c. This effect was blocked by the estrogen receptor antagonist fulvestrant (ICI-182,780, Faslodex) but was unaffected by inhibitors of nitric-oxide synthase or phosphoinositide-3-kinase. Nuclear respiratory factor-1 protein, a primary regulator of nuclear gene-encoded mitochondrial genes, was significantly increased by long-term estrogen treatment in vivo. In summary, these novel findings suggest that vascular protection by E(2) is mediated, in part, by modulation of mitochondrial function, resulting in greater energy-producing capacity and decreased reactive oxygen species production.
Collapse
Affiliation(s)
- Chris Stirone
- Department of Pharmacology, University of California Irvine, CA 92697, USA
| | | | | | | |
Collapse
|
87
|
Abstract
Estrogens demonstrate vasoprotective activity in many experimental models. These effects have been attributed to beneficial activity of these steroids on lipid metabolism as well as direct effects on the vasculature via modulation of nitric-oxide synthase and phosphatidylinositol-3 kinase/Akt signaling pathways. In this issue of Molecular Pharmacology, Stirone et al. (p. 959) present evidence suggesting that 17beta-estradiol may also exert vasoprotective effects in cerebral blood vessels via stimulation of mitochondrial energy production capacity and inhibition of reactive oxygen species production. These data indicate not only yet another potential mechanism underlying the vasoprotective effects of estrogens but also that the estrogen receptor may coordinate gene expression in both the nuclear and mitochondrial genomes.
Collapse
Affiliation(s)
- Thomas P Burris
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | |
Collapse
|
88
|
Chan SHH, Wu KLH, Wang LL, Chan JYH. Nitric oxide- and superoxide-dependent mitochondrial signaling in endotoxin-induced apoptosis in the rostral ventrolateral medulla of rats. Free Radic Biol Med 2005; 39:603-18. [PMID: 16085179 DOI: 10.1016/j.freeradbiomed.2005.04.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2004] [Revised: 03/21/2005] [Accepted: 04/16/2005] [Indexed: 11/21/2022]
Abstract
This study evaluated the hypothesis that the repertoire of cellular events that underlie circulatory fatality during endotoxemia may entail mitochondrial respiratory enzyme dysfunction, followed by the release of cytochrome c to the cytosol that triggers the activation of caspase cascades, leading to apoptotic cell death in the rostral ventrolateral medulla (RVLM) where sympathetic premotor neurons responsible for maintaining vasomotor tone are located. In adult Sprague-Dawley rats maintained under propofol anesthesia, nucleosomal DNA fragmentation was detected in the RVLM in a temporal profile that coincided positively with the progression of cardiovascular depression during experimental endotoxemia induced by Escherichia coli lipopolysaccharide (LPS). LPS also induced nitric oxide (NO) and superoxide (O(2)(-)) production, depressed mitochondrial Complex I and IV activity, promoted the release of cytochrome c from mitochondria to cytosol, upregulated the cytosolic expression of activated caspase-9 and -3, or increased caspase-3 enzyme activity in the RVLM. Microinjection bilaterally into the RVLM of an inducible nitric oxide synthase (iNOS) blocker, S-methylisothiourea, or a superoxide dismutase mimetic, Tempol, significantly blunted these apoptotic cellular events and antagonized the cardiovascular depression during endotoxemia. We conclude that caspase-dependent apoptotic cell death that results from NO- and O(2)(-)-associated mitochondrial signaling in the RVLM may underlie fatal cardiovascular depression during endotoxemia.
Collapse
Affiliation(s)
- Samuel H H Chan
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 804, Taiwan, Republic of China
| | | | | | | |
Collapse
|
89
|
Ballinger SW. Mitochondrial dysfunction in cardiovascular disease. Free Radic Biol Med 2005; 38:1278-95. [PMID: 15855047 DOI: 10.1016/j.freeradbiomed.2005.02.014] [Citation(s) in RCA: 271] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Revised: 02/15/2005] [Accepted: 02/15/2005] [Indexed: 12/11/2022]
Abstract
Whereas the pathogenesis of atherosclerosis has been intensively studied and described, the underlying events that initiate cardiovascular disease are not yet fully understood. A substantial number of studies suggest that altered levels of oxidative and nitrosoxidative stress within the cardiovascular environment are essential in the development of cardiovascular disease; however, the impact of such changes on the subcellular or organellar components and their functions that are relevant to cardiovascular disease inception are less understood. In this regard, studies are beginning to show that mitochondria not only appear susceptible to damage mediated by increased oxidative and nitrosoxidative stress, but also play significant roles in the regulation of cardiovascular cell function. In addition, accumulating evidence suggests that a common theme among cardiovascular disease development and cardiovascular disease risk factors is increased mitochondrial damage and dysfunction. This review discusses aspects relating mitochondrial damage and function to cardiovascular disease risk factors and disease development.
Collapse
Affiliation(s)
- Scott W Ballinger
- Division of Molecular and Cellular Pathology, VH G019F, 1530 3rd Avenue South, Birmingham, AL 35294-0019, USA.
| |
Collapse
|
90
|
Landar A, Darley-Usmar VM. Nitric oxide signaling gone awry: nitration of glutamine synthetase and hyperammonemia in sepsis. Hepatology 2005; 41:980-2. [PMID: 15841446 DOI: 10.1002/hep.20699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
91
|
Puddu P, Puddu GM, Galletti L, Cravero E, Muscari A. Mitochondrial Dysfunction as an Initiating Event in Atherogenesis: A Plausible Hypothesis. Cardiology 2005; 103:137-41. [PMID: 15665536 DOI: 10.1159/000083440] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2004] [Accepted: 08/03/2004] [Indexed: 12/30/2022]
Abstract
It is now widely accepted that oxidant stress and the ensuing endothelial dysfunction play a key role in the pathogenesis of atherosclerosis and cardiovascular diseases. The mitochondrial respiratory chain is the major source of reactive oxygen species as byproducts of normal cell respiration. Mitochondria may also be important targets for reactive oxygen species, which may damage mitochondrial lipids, enzymes and DNA with following mitochondrial dysfunction. Free cholesterol, oxidized low-density lipoprotein and glycated high-density lipoprotein are further possible causes of mitochondrial dysfunction and/or apoptosis. Moreover, in patients with mitochondrial diseases, vascular complications are commonly observed at an early age, often in the absence of traditional risk factors for atherosclerosis. We propose that mitochondrial dysfunction, besides endothelial dysfunction, represents an important early step in the chain of events leading to atherosclerotic disease.
Collapse
Affiliation(s)
- Paolo Puddu
- Department of Internal Medicine, Cardioangiology, Hepatology, University of Bologna, Bologna, Italy
| | | | | | | | | |
Collapse
|
92
|
Zhang J, Jin B, Li L, Block ER, Patel JM. Nitric oxide-induced persistent inhibition and nitrosylation of active site cysteine residues of mitochondrial cytochrome-c oxidase in lung endothelial cells. Am J Physiol Cell Physiol 2005; 288:C840-9. [PMID: 15561762 DOI: 10.1152/ajpcell.00325.2004] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Persistent inhibition of cytochrome- c oxidase, a terminal enzyme of the mitochondrial electron transport chain, by excessive nitric oxide (NO) derived from inflammation, polluted air, and tobacco smoke contributes to enhanced oxidant production and programmed cell death or apoptosis of lung cells. We sought to determine whether the long-term exposure of pulmonary artery endothelial cells (PAEC) to pathophysiological concentrations of NO causes persistent inhibition of complex IV through redox modification of its key cysteine residues located in a putative NO-sensitive motif. Prolonged exposure of porcine PAEC to 1 mM 2,2′-(hydroxynitrosohydrazino)-bis-ethanamine (NOC-18; slow-releasing NO donor, equivalent to 1–5 μM NO) resulted in a gradual, persistent inhibition of complex IV concomitant with a reduction in ratios of mitochondrial GSH and GSSG. Overexpression of thioredoxin in mitochondria of PAEC attenuated NO-induced loss of complex IV activities, suggesting redox regulation of complex IV activity. Sequence analysis of complex IV subunits revealed a novel putative NO-sensitive motif in subunit II (S2). There are only two cysteine residues in porcine complex IV S2, located in the putative motif. Immunoprecipitation and Western blot analysis and “biotin switch” assay demonstrated that exposure of PAEC to 1 mM NOC-18 increased S-nitrosylation of complex IV S2 by 200%. Site-directed mutagenesis of these two cysteines of complex IV S2 attenuated NO-increased nitrosylation of complex IV S2. These results demonstrate for the first time that NO nitrosylates active site cysteines of complex IV, which is associated with persistent inhibition of complex IV. NO inhibition of complex IV via nitrosylation of NO-sensitive cysteine residues can be a novel upstream event in NO-complex IV signaling for NO toxicity in lung endothelial cells.
Collapse
Affiliation(s)
- Jianliang Zhang
- Pulmonary Division, MSB M452, Dept. of Medicine, Univ. of Florida College of Medicine, 1600 SW Archer Rd., Gainesville, FL 32610-0225, USA.
| | | | | | | | | |
Collapse
|
93
|
Buchwalow IB, Podzuweit T, Samoilova VE, Wellner M, Haller H, Grote S, Aleth S, Boecker W, Schmitz W, Neumann J. An in situ evidence for autocrine function of NO in the vasculature. Nitric Oxide 2005; 10:203-12. [PMID: 15275866 DOI: 10.1016/j.niox.2004.04.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2004] [Revised: 04/09/2004] [Indexed: 10/26/2022]
Abstract
The concept of endothelium derived relaxing factor (EDRF) implies that nitric oxide (NO) generated by NO synthase in the endothelium diffuses to the underlying vascular smooth muscle cells (VSMC) modulating thereby vascular tone. VSMC were regarded as passive recipients of NO from endothelial cells. However, this paradigm of a paracrine function of NO became currently subject to considerable debate. To address this issue, we examined the localization of enzymes engaged in l-arginine-NO-cGMP signaling in the rat blood vessels. Employing multiple immunocytochemical labeling complemented with signal amplification, electron microscopy, Western blotting, and RT-PCR, we found that NO synthase was differentially expressed in blood vessels depending on the blood vessel type. Moreover, the expression pattern of NO synthase in VSMC showed striking parallels with arginase and soluble guanylyl cyclase. Our findings challenge the commonly accepted view that the expression of NO synthase is restricted to vascular endothelial cells and lends further support to an alternative mechanism, by which constitutive local NOS expression in VSMC may modulate vascular functions in an endothelium-independent manner. Moreover, the co-expression of enzymes engaged in l-arginine-NO-cGMP signaling (NO synthase, arginase, and soluble guanylyl cyclase) in VSMC is indicative of an autocrine fashion of NO signaling in the vasculature in addition to the paracrine role of NO generated in the endothelium.
Collapse
Affiliation(s)
- Igor B Buchwalow
- Interdisciplinary Center of Clinical Research and Gerhard Domagk Institute of Pathology, University of Muenster, D-48149 Muenster, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Mo J. A novel theory: biological processes mostly involve two types of mediators, namely general and specific mediators. Med Hypotheses 2005; 65:728-35. [PMID: 15961255 DOI: 10.1016/j.mehy.2005.03.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2004] [Accepted: 03/16/2005] [Indexed: 01/28/2023]
Abstract
A great number of papers have shown that free radicals as well as bioactive molecules can play a role of mediator in a wide spectrum of biological processes, but the biological actions and chemical reactivity of the free radicals are quite different from that of the bioactive molecules, and that a wide variety of bioactive molecules can be easily modified by free radicals due to having functional groups sensitive to redox, and the significance of the interaction between the free radicals and the bioactive molecules in biological processes has been confirmed by the results of some in vitro and in vivo studies. Based on these evidence, this article presented a novel theory about the mediators of biological processes. The essentials of the theory are: (a) mediators of biological processes can be classified into general and specific mediators; the general mediators include two types of free radicals, namely superoxide and nitric oxide; the specific mediators include a wide variety of bioactive molecules, such as specific enzymes, transcription factors, cytokines and eicosanoids; (b) a general mediator can modify almost any class of the biomolecules, and thus play a role of mediator in nearly every biological process via diverse mechanisms; a specific mediator always acts selectively on certain classes of the biomolecules, and may play a role of mediator in different biological processes via a same mechanism; (c) biological processes are mostly controlled by networks of their mediators, so the free radicals can regulate the last consequence of a biological process by modifying some types of the bioactive molecules, or in cooperation with these bioactive molecules; the biological actions of superoxide and nitric oxide may be synergistic or antagonistic. According to this theory, keeping the integrity of these networks and the balance between the free radicals and the bioactive molecules as well as the balance between the free radicals and the free radical scavengers would be of vital importance for physiological processes, and disturbance of these networks and balances would be a critical factor of pathological processes. Therefore, the investigators who want to get a deep and full understanding of the mechanism of a biological process should pay attention to the roles of both free radical and bioactive molecule species, and the free radical scavengers, which are used for health protection, such a vitamin E and carotenoid, should be taken in a suitable dosage.
Collapse
Affiliation(s)
- Jian Mo
- The Fourth Military Medical University, 621-19, 17 Changlexilu, Xian 710032, PR China.
| |
Collapse
|
95
|
Lin KY, Wang HH, Lai ST, Pan JP, Chiang AN. ?2-glycoprotein I protects J774A.1 macrophages and human coronary artery smooth muscle cells against apoptosis. J Cell Biochem 2005; 94:485-96. [PMID: 15534879 DOI: 10.1002/jcb.20314] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
beta(2)-Glycoprotein I (beta(2)-GPI) is a plasma glycoprotein with multifactorial relevance to clinical consequences. It was previously indicated that beta(2)-GPI can selectively bind to apoptotic cells. This study was designed to determine the role of beta(2)-GPI in apoptosis. Using an immunohistochemical study, we observed that beta(2)-GPI was co-localized with the apoptotic macrophages and smooth muscle cells (SMCs) of human coronary arteries. The contribution of beta(2)-GPI to apoptotic death was then investigated in vascular cells. Two nitric oxide (NO) donors, S-nitrosoglutathione (GSNO) and S-nitroso-N-acetyl penicillamine (SNAP) were used in this study to trigger apoptosis in J774A.1 macrophages and human coronary artery smooth muscle cells (HCASMC). Cell viability was significantly improved in beta(2)-GPI-treated cells. It was also possible to detect a remarkable inhibitory effect by beta(2)-GPI on the NO-induced apoptosis by preventing nuclear shrinkage. Furthermore, the NO-induced apoptosis was associated with increase in caspase-3 activity and in the protein levels of caspase-3, c-Fos, and c-Jun. However, all these apoptosis-related events were inhibited in vascular cells treated with 200 microg/ml beta(2)-GPI. This is the first study to show that beta(2)-GPI may be important in the prevention of apoptosis in vascular cells.
Collapse
Affiliation(s)
- Kae-Yuan Lin
- Institute of Biochemistry, National Yang-Ming University, Taipei 112, Taiwan
| | | | | | | | | |
Collapse
|
96
|
Abstract
This review focuses on the role of oxidative processes in atherosclerosis and its resultant cardiovascular events. There is now a consensus that atherosclerosis represents a state of heightened oxidative stress characterized by lipid and protein oxidation in the vascular wall. The oxidative modification hypothesis of atherosclerosis predicts that low-density lipoprotein (LDL) oxidation is an early event in atherosclerosis and that oxidized LDL contributes to atherogenesis. In support of this hypothesis, oxidized LDL can support foam cell formation in vitro, the lipid in human lesions is substantially oxidized, there is evidence for the presence of oxidized LDL in vivo, oxidized LDL has a number of potentially proatherogenic activities, and several structurally unrelated antioxidants inhibit atherosclerosis in animals. An emerging consensus also underscores the importance in vascular disease of oxidative events in addition to LDL oxidation. These include the production of reactive oxygen and nitrogen species by vascular cells, as well as oxidative modifications contributing to important clinical manifestations of coronary artery disease such as endothelial dysfunction and plaque disruption. Despite these abundant data however, fundamental problems remain with implicating oxidative modification as a (requisite) pathophysiologically important cause for atherosclerosis. These include the poor performance of antioxidant strategies in limiting either atherosclerosis or cardiovascular events from atherosclerosis, and observations in animals that suggest dissociation between atherosclerosis and lipoprotein oxidation. Indeed, it remains to be established that oxidative events are a cause rather than an injurious response to atherogenesis. In this context, inflammation needs to be considered as a primary process of atherosclerosis, and oxidative stress as a secondary event. To address this issue, we have proposed an "oxidative response to inflammation" model as a means of reconciling the response-to-injury and oxidative modification hypotheses of atherosclerosis.
Collapse
Affiliation(s)
- Roland Stocker
- Centre for Vascular Research, University of New South Wales, Sydney, New South Wales, Australia.
| | | |
Collapse
|
97
|
Abstract
The mechanisms of alcohol toxicity as related to mitochondrial dysfunction and the glutathione-dependent protective systems are reviewed. The pathophysiology of ethanol-induced liver damage is defined in terms of an early phase and a late phase. CYP2E1 dependent toxicity appears closely related to oxidative stress injury with possible roles of peroxynitrite, TNFalpha, protein adducts, and enhanced protein expression. Modulation of mitochondrial glutathione affects mitochondrial function and cell survival with superoxide and hydrogen peroxide generation being crucial to mitochondrial membrane permeability transition and apoptosis.
Collapse
Affiliation(s)
- Donald J Reed
- Department of Biochemistry and Biophysics, 2011 ALS, Oregon State University, Corvallis, Oregon 97331, USA.
| |
Collapse
|
98
|
Venkatraman A, Shiva S, Wigley A, Ulasova E, Chhieng D, Bailey SM, Darley-Usmar VM. The role of iNOS in alcohol-dependent hepatotoxicity and mitochondrial dysfunction in mice. Hepatology 2004; 40:565-73. [PMID: 15349894 DOI: 10.1002/hep.20326] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nitric oxide (NO) is now known to control both mitochondrial respiration and organelle biogenesis. Under conditions of ethanol-dependent hepatic dysfunction, steatosis is increased, and this is associated with increased expression of inducible nitric oxide synthase (iNOS). We have previously shown that after chronic exposure to ethanol, the sensitivity of mitochondrial respiration to inhibition by NO is enhanced, and we have proposed that this contributes to ethanol-dependent hypoxia. This study examines the role of iNOS in controlling the NO-dependent modification of mitochondrial function. Mitochondria were isolated from the livers of both wild-type (WT) and iNOS knockout (iNOS-/-) mice that were fed an isocaloric ethanol-containing diet for a period of 5 weeks. All animals that consumed ethanol showed some evidence of fatty liver; however, this was to a lesser extent in the iNOS-/- mice compared to controls. At this early stage in ethanol-dependent hepatic dysfunction, infiltration of inflammatory cells and the formation of nitrated proteins was also decreased in response to ethanol feeding in the iNOS-/- animals. Mitochondria isolated from wild-type ethanol-fed mice showed a significant decrease in respiratory control ratio and an increased sensitivity to NO-dependent inhibition of respiration relative to their pair-fed controls. In contrast, liver mitochondria isolated from iNOS-/- mice fed ethanol showed no change in the sensitivity to NO-dependent inhibition of respiration. In conclusion, the hepatic response to chronic alcohol-dependent cytotoxicity involves a change in mitochondrial function dependent on the induction of iNOS.
Collapse
Affiliation(s)
- Aparna Venkatraman
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
99
|
Isom AL, Barnes S, Wilson L, Kirk M, Coward L, Darley-Usmar V. Modification of Cytochrome c by 4-hydroxy- 2-nonenal: evidence for histidine, lysine, and arginine-aldehyde adducts. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2004; 15:1136-1147. [PMID: 15276160 DOI: 10.1016/j.jasms.2004.03.013] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2003] [Revised: 03/08/2004] [Accepted: 03/12/2004] [Indexed: 05/24/2023]
Abstract
4-Hydroxy-2-nonenal (4HNE), a major secondary product of lipid peroxidation, has been associated with a number of disease states involving oxidative stress. Despite the recognized importance of post-translational modification of proteins by products such as 4HNE, little is known of the modification of cytochrome c by this reagent and its analysis by mass spectrometry. The purpose of this study was to investigate the chemical interaction of 4HNE and cytochrome c, a protein essential to cellular respiration, under in vitro conditions. Isoelectric focusing of native and 4HNE-modified cytochrome c using immobilized pH gradient (IpG) strips showed a decrease in the pI of the 4HNE-modified protein suggesting modification of charged amino acids. Reaction of 4HNE with cytochrome c resulted in increases in molecular weight consistent with the addition of four 4HNE residues as determined by matrix-assisted laser desorption time-of-flight mass spectrometry (MALDI-TOF MS). Samples of both native and 4HNE-modified cytochrome c were enzymatically digested and subjected to peptide mass fingerprinting using MALDI-TOF MS. Analysis of these samples using LC-electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS) provided sequence information that was used to determine specific residues to which the aldehyde adducted. Taken together, the data indicated that H33, K87, and R38 were modified by 4HNE. Mapping these results onto the X-ray crystal structure of native cytochrome c suggest that 4HNE adduction to cytochrome c could have significant effects on tertiary structure, electron transport, and ultimately, mitochondrial dysfunction.
Collapse
Affiliation(s)
- Amanda L Isom
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
100
|
Shiva S, Crawford JH, Ramachandran A, Ceaser EK, Hillson T, Brookes PS, Patel RP, Darley-Usmar VM. Mechanisms of the interaction of nitroxyl with mitochondria. Biochem J 2004; 379:359-66. [PMID: 14723605 PMCID: PMC1224084 DOI: 10.1042/bj20031758] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Revised: 12/15/2003] [Accepted: 12/18/2003] [Indexed: 12/30/2022]
Abstract
It is now thought that NO* (nitric oxide) and its redox congeners may play a role in the physiological regulation of mitochondrial function. The inhibition of cytochrome c oxidase by NO* is characterized as being reversible and oxygen dependent. In contrast, peroxynitrite, the product of the reaction of NO* with superoxide, irreversibly inhibits several of the respiratory complexes. However, little is known about the effects of HNO (nitroxyl) on mitochondrial function. This is especially important, since HNO has been shown to be more cytotoxic than NO*, may potentially be generated in vivo, and elicits biological responses with some of the characteristics of NO and peroxynitrite. In the present study, we present evidence that isolated mitochondria, in the absence or presence of substrate, convert HNO into NO* by a process that is dependent on mitochondrial concentration as well as the concentration of the HNO donor Angeli's salt. In addition, HNO is able to inhibit mitochondrial respiration through the inhibition of complexes I and II, most probably via modification of specific cysteine residues in the proteins. Using a proteomics approach, extensive modification of mitochondrial protein thiols was demonstrated. From these data it is evident that HNO interacts with mitochondria through mechanisms distinct from those of either NO* or peroxynitrite, including the generation of NO*, the modification of thiols and the inhibition of complexes I and II.
Collapse
Affiliation(s)
- Sruti Shiva
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | |
Collapse
|