American Journal of Respiratory and Critical Care Medicine

The lungs are directly exposed to higher oxygen concentrations than most other tissues. Increased oxidative stress is a significant part of the pathogenesis of obstructive lung diseases such as asthma and chronic obstructive pulmonary disease, parenchymal lung diseases (e.g., idiopathic pulmonary fibrosis and lung granulomatous diseases), and lung malignancies. Lung tissue is protected against these oxidants by a variety of antioxidant mechanisms among which the superoxide dismutases (SODs) are the only ones converting superoxide radicals to hydrogen peroxide. There are three SODs: cytosolic copper–zinc, mitochondrial manganese, and extracellular SODs. These enzymes have specific distributions and functions. Their importance in protecting lung tissue has been confirmed in transgenic and knockout animal studies. Relatively few studies have been conducted on these enzymes in the normal human lung or in human lung diseases. Most human studies suggest that there is induction of manganese SOD and, possibly, extracellular SOD during inflammatory, but not fibrotic, phases of parenchymal lung diseases and that both copper–zinc SOD and manganese SOD may be downregulated in asthmatic airways. Many previous antioxidant therapies have been disappointing, but newly characterized SOD mimetics are being shown to protect against oxidant-related lung disorders in animal models.

Oxidant Stress of the Lung

General Antioxidant Defense of the Lung

Superoxide Dismutases and Their Distribution in the Lung

Expression of Superoxide Dismutases during Lung Development

Regulation of Superoxide Dismutases

CuZnSOD

MnSOD

ECSOD

Induction of Superoxide Dismutases and Tissue Protection

Gene Manipulation and Lung Protection

CuZnSOD

MnSOD

ECSOD

Exogenous Superoxide Dismutases, Superoxide Dismutase Mimetics and Lung Protection

Free Radicals in the Pathogenesis of Human Lung Diseases

Superoxide Dismutase Polymorphisms and Lung Diseases

Superoxide Dismutases in Human Lung Diseases

Obstructive Lung Diseases

Interstitial Lung Diseases

Respiratory Distress Syndrome and Bronchopulmonary Dysplasia

Carcinogenesis and the Expression of Superoxide Dismutases in Lung Tumors

Conclusions and Future Aspects

Lung represents a unique tissue for oxidant stress among most organs because it is directly exposed to higher oxygen tensions. Thus, local oxygen partial pressure at the alveolar level is much higher than in other vital organs such as heart, liver, and brain. Oxygen pressure in the inhaled air is 20 kPa (150 mm Hg) and is ∼13.3 kPa (100 mm Hg) at the alveolus, but it is ∼6 kPa (45 mm Hg) in v̄ blood and may be as low as 1 mm Hg in some sites within other organs. Another special feature of the lung is its large surface area (∼70 m2 in the adult human lung).

Because of their direct exposure to ambient air, lung cells experience enhanced oxidant stress by environmental irritants and pollutants including oxidants such as cigarette smoke, ozone, and free radical–generating environmental carcinogens. In addition, a typical component in most lung disorders and lung infections is inflammation and activation of inflammatory cells with consequent free radical generation. Oxygen therapy is an important but problematic issue in the treatment of prematurely born neonates and in the respiratory insufficiency associated with the acute respiratory distress syndrome. Several therapies, most importantly chemotherapy and radiation, lead to free radical–induced tissue damage where again the lung is the most commonly involved organ. Thus, lung represents a unique tissue exposed not only directly to higher oxygen tensions and environmental oxidants but also to oxidants produced by a variety of lung diseases and in the course of their therapies.

The first reactive oxygen species produced in the reduction pathway of oxygen to water is the superoxide anion, which participates in the generation of other toxic metabolites, most importantly hydrogen peroxide (H2O2), hydroxyl radical, and peroxynitrite (Figure 1)

. Most living cells, including lung cells, generate free radicals under normal conditions nonenzymatically via auto-oxidation. The primary sites of oxygen radical production are the electron transport chain in the mitochondria, and cytoplasmic and membrane-bound oxidant-generating enzymes (e.g., nicotinamide adenine dinucleotide phosphate reduced oxidase, nitric oxide synthase, and xanthine oxidase) (1). The generation of reactive oxygen species by various lung cells has been reviewed previously (2). Typical sources of oxidant production in inflammatory states, for example in asthma, include the activation of inflammatory cells, activation of nicotinamide adenine dinucleotide phosphate reduced oxidase on cell membranes, and activation of inducible nitric oxide synthase in the cytosolic compartments of cells. Mitochondrial energy metabolism plays a key role in cell survival in situations where oxidant stress is elevated. Oxidant stress enhances the disruption of the mitochondrial electron transport chain, the final effect being either cell injury (necrosis) or programmed cell death (apoptosis). Thus, reactive oxygen species are produced by multiple mechanisms in all cell compartments and play a significant role in the pathogenesis of lung diseases where oxidant stress is increased.

Besides causing cell and DNA toxicity, oxidants participate in the regulation of normal cell homeostasis and modulate signaling pathways associated with cell growth and proliferation (3). Oxidants activate several reactions in the regulation of transcription factors, such as nuclear factor κ B, which then are associated with the induction of antioxidant enzymes in the lung (4). Antioxidant enzymes, by detoxifying reactive oxygen species, constitute the main protective mechanism of lung tissue against free radical–mediated injury. Besides the basic oxidant defense offered by the antioxidant enzymes, individual variability in the levels of these enzymes has been shown to play a role in the development of oxidant-related lung diseases.

General Antioxidant Defense of the Lung

Superoxide dismutases (SODs) are the only enzymatic system-decomposing superoxide radicals to H2O2 and are hypothesized to play a significant role against oxidant stress, especially in the lung (2, 5). There are three different mammalian SODs: intracellular copper–zinc SOD (CuZnSOD), mitochondrial manganese SOD (MnSOD) and extracellular SOD (ECSOD) (68). SODs have been detected in all classes of lung cells but show significant variability in cell-specific localization and expression.

Numerous enzymatic mechanisms participate in H2O2 degradation in the lung (Figure 2)

. The most important H2O2 scavenging enzymes include catalase and the glutathione peroxidases, the latter ones being closely associated with the maintenance of reduced glutathione by glutathione reductase and glutathione synthesis by γ-glutamyl cysteine synthase (glutamate cysteine ligase) and glutathione synthase (1, 9). A typical feature in the antioxidant defense of human lung is the high glutathione content in the epithelial lining fluid (∼140 times higher than in the circulating blood) (10), and based on this, glutathione and enzymes associated with its maintenance have been suggested to constitute one of the basic antioxidant defense mechanisms of human lung. Various lung cells, however, differ profoundly in their resistance to oxidant stress that is at least partly associated with the cell-specific expression of antioxidant enzymes in the cells. For example, alveolar type II epithelial cells highly express CuZnSOD, MnSOD, and catalase (2) and are resistant to oxidant stress. In contrast, alveolar type I epithelial cells have low expression of antioxidant enzymes and are sensitive to injury and death under conditions of enhanced oxidative stress. Alveolar macrophages have high expression of catalase and consume exogenous H2O2 mainly by catalase (11, 12). In addition to these “classic” antioxidant enzymes, human lung has recently been shown to express several other enzymes with H2O2 consuming capacity. These include the thioredoxin–thioredoxin reductase system, thioredoxin peroxidases (peroxiredoxins), and glutaredoxins (1315). The expression of most of these enzymes is concentrated in airways and in alveolar macrophages of human lung (16, 17). Several reviews have been published on the H2O2 scavenging enzymes, such as the reduced glutathione–related mechanisms and catalase in the lung (2, 9, 18).

There are also a number of nonenzymatic antioxidants that are important in maintaining lung redox balance but that are outside the scope of this review. For example, lung extracellular fluids are rich in glutathione, and surfactant proteins such as surfactant protein D have been shown to have a significant antioxidant function. Metals such as iron and copper ions are powerful promoters of oxidative damage, but they are mainly bound to proteins (transferrin, ferritin, ceruloplasmin, lactoferrin, etc.). Several other proteins, for example albumin, have been found to possess efficient free radical scavenging capacity. Other antioxidant defenses include numerous small molecular weight vitamins, either lipid (vitamin E) or water (vitamin C) soluble. The present review concentrates on SODs in the lung with special emphasis on those lung diseases with a likely free radical–related pathogenesis.

SODs and Their Distribution in the Lung

Some of the basic characteristics of SODs are given in Table 1

TABLE 1. Selected characteristics of mammalian superoxide dismutases


SOD

Structure

Metals

Molecular Weight
 (kDa)

Chromosomal
 Localization

References
CuZnSOD Homodimer Cu and Zn 32 21q22 7, 310, 311
MnSOD Homotetramer Mn 86–88 6q25 77, 312, 313
ECSOD
Homotetramer
Cu and Zn
135
4q21
6, 38, 42, 120, 315

Definition of abbreviations: CuZnSOD = copper–zinc superoxide dismutase; ECSOD = extracellular superoxide dismutase; MnSOD = manganese superoxide dismutase; SOD = superoxide dismutase.

. Given the importance of SODs as primary enzymes against superoxide radicals, these enzymes may play a major role not only in the primary defense of human lung against free radicals produced as part of normal metabolism but also are critical in protecting against the progression of oxidant-related lung diseases. SODs not only constitute the basic superoxide consuming mechanism in oxidant-exposed cells, but they also participate in the regulation of normal cell homeostasis. Superoxide radicals are produced continuously as a byproduct of cellular metabolism, being then converted to H2O2 (signaling molecule at low concentrations) by SODs. Superoxide is also known to react with nitric oxide, participating in the regulation of vascular tone in normal situations and in the generation of toxic nitrogen metabolites (such as peroxynitrite) in inflammatory states (19). Thus, SODs have multiple functions in regulating intracellular and extracellular levels of superoxide, H2O2, and nitrogen metabolites.

When analyzed from total lung homogenates CuZnSOD and MnSOD activities have been reported to be lower in the lung than in several other vital organs such as liver, kidney, heart, and brain (Figure 3)

(20). In contrast, ECSOD activity has been reported to be remarkably higher in the lung than in the previously listed vital organs (Figure 4) . A few other tissues show even higher levels of ECSOD than that of lung, including thyroid and uterus (20), but among the major solid organs, lung is unique in its high expression of ECSOD. Given the complex architecture and existence of multiple cell types in the lung, in situ hybridization, immunohistochemistry, and immunoelectron microscopy in combination with morphometric analysis (21, 22) have given important information about the significance of these enzymes in various cell types and cell compartments of the lung.

By immunohistochemistry and/or immunoelectron microscopy using the immunogold technique, CuZnSOD has been shown to be highly expressed in bronchial epithelium, alveolar epithelium, mesenchymal cells, fibroblasts, arterioles, and capillary endothelial cells in rat lung (23, 24). In the bronchial epithelium, CuZnSOD is expressed most prominently in ciliated epithelial cells (2527) (Figures 5A and 5B)

. The subcellular distribution of CuZnSOD has been studied extensively in liver cells, which contain the highest CuZnSOD activity among all tissues investigated (28). CuZnSOD is ultrastructurally located mainly in the cytosol but is also expressed in the nucleus at levels that are 50 to 60% of that of the cytosol and is very highly expressed in some organelles such as lysosomes (23, 2831).

In situ hybridization studies on MnSOD have revealed a consistent pattern in rat lung messenger (mRNA) distribution with the most prominent labeling occurring in the airways, especially in the septal tips of alveolar ducts and arterioles near the airways (32, 33). Immunohistochemical studies have shown that MnSOD is moderately or highly expressed in respiratory epithelium, alveolar type II epithelial cells, alveolar macrophages, interstitial fibroblasts, and visceral pleura of hyperoxia-exposed rats (23, 24). In healthy human lung, MnSOD is expressed in proportion to the mitochondrial content of cells with cells such as bronchial epithelium, alveolar type II epithelial cells, and alveolar macrophages having the highest MnSOD expression (11, 26, 27, 3436) (Figures 5C and 5D). MnSOD decreases in cultured cells when assessed by the immunogold technique and is remarkably lower (∼65%) in SV40 immortalized human bronchial BEAS 2B cells compared with primary cultured cells (26). MnSOD is exclusively localized to the mitochondria (8, 23), a critical cell organelle both in the cellular energy metabolism and cell survival. Overall, MnSOD is most highly expressed in alveolar type II epithelial cells and alveolar macrophages, cell types with relatively high metabolic capacity, and plays a major role in protecting lung tissue against free radicals.

The lung is one of the major tissues that strongly expresses the mRNA and protein of ECSOD (3741). In situ hybridization studies on mouse and human lung have found strong signals of ECSOD synthesis in bronchial epithelium, alveolar epithelium, epithelial cells lining intrapulmonary airways, alveolar macrophages, and endothelial cells lining both arteries and veins. (38, 42). Immunohistochemical studies have revealed that ECSOD is primarily located in the extracellular matrix, but it is also expressed in airway epithelial cell junctions and around the surface of vascular and airway smooth muscle cells (19, 40) (Figures 5E and 5F). ECSOD is the main SOD in pulmonary and systemic vessels (over 70% of the total SOD activity in some vessels) where it is located beneath the endothelium, surrounding smooth muscle cells, and throughout the adventitia of vessels (19, 20, 41) as shown in Figure 4. ECSOD has an affinity for negatively charged molecules such as heparin and the negatively charged molecules in the matrix (43). In the lung interstitium, ECSOD has been shown to be highly localized in areas containing high amounts of type I collagen fibers. The distribution of ECSOD in the interstitial space suggests that it has an important role in protecting matrix proteins against extracellular oxidants, as is the case, for instance, in inflammatory states. A summary of the expressions of MnSOD, CuZnSOD, and ECSOD in normal lung is shown in Table 2

TABLE 2. Summary of the major sites of cellular expression of superoxide dismutases in human lung


MnSOD

Alveolar type II epithelial cells
Alveolar macrophages
Bronchial epithelium
CuZnSOD
Bronchial epithelium
Alveolar macrophages
Alveolar type II epithelial cells
ECSOD
Airway walls
Pulmonary vasculature
Alveolar macrophages

Interstitium (association with collagen fibers)

Definition of abbreviations: CuZnSOD = copper–zinc superoxide dismutase; ECSOD = extracellular superoxide dismutase; MnSOD = manganese superoxide dismutase.

.

It is difficult to directly compare the activities of the three SODs in lung because they are localized in compartments of different sizes and have been evaluated by multiple investigators using assays of different sensitivities and specificities and in which all three enzymes were never simultaneously evaluated. Figure 6

attempts to resolve this by correcting activity assays for known impacts of pH on relative activity of the CuZnSODs and MnSOD and by using morphometrically derived data on the volumes of the primary compartments in which the enzymes are distributed. The assumptions that the enzymes are only found in one of the compartments and are uniformly distributed in that compartment are not absolutely correct. For example, CuZnSOD is found in mitochondria and has very high expression in lysosomes, and ECSOD has a minor distribution within cells. Overall, the assumptions are reasonable and the minor exceptions as noted previously would not substantially change the comparisons shown in Figure 6B. As shown in Figure 6A, the dominant SOD in lung is the CuZnSOD with MnSOD and ECSOD each representing ∼15 to 20% of the SOD activity. Although CuZnSOD is present in lung in lower concentrations than in other tissues (such as liver and kidney) and ECSOD is higher in the lung than in these other tissues, CuZnSOD is still the dominant lung SOD. SOD concentrations are believed to be closely related to the redox status of specific cell compartments. The comparison of lung with other organs suggests that cytoplasmic production of superoxide is generally lower in lung cells than in highly metabolic cells such as those of liver and kidney, thus requiring lower overall CuZnSOD concentrations. The relatively high ECSOD levels in lung likely relate to the dense airway and vascular network in the lung and the higher potential of extracellular inflammatory events resulting from direct exposure of the lung to the external environment.

Figure 6B illustrates two important concepts. MnSOD is the most highly concentrated SOD within its compartment. Mitochondria are known to be the largest source of superoxide production within cells (44) and, as would be expected, require a very high SOD level to maintain homeostasis. ECSOD, in contrast, is distributed in a much larger compartment and, even with its relatively high lung localization, ECSOD is not present in a sufficient concentration to be able to function as a bulk scavenger of superoxide across the entire extracellular space. This enzyme is unique in that it has a positively charged carboxyterminus that leads to binding in specific domains within the extracellular spaces. The localization of ECSOD rather than its average concentration likely determines its major biological impact. Whereas CuZnSOD and MnSOD are generally believed to act as bulk scavengers of superoxide in the cell cytosol and in mitochondria, ECSOD has been postulated to be more involved in redox-mediated signal transduction, particularly in the regulation of nitric oxide–mediated signaling across extracellular spaces (45).

Expression of SODs during Lung Development

Lung cells are exposed to a sudden severalfold increase in oxygen concentration at birth. In addition, newborn premature infants may be ventilated using high oxygen concentrations that would increase the risk of toxicity to lung cells. Developmental expression of antioxidant enzymes is critically important in these events and in the resistance of newborn lung to high oxygen tensions. Studies on human lung have concluded that CuZnSOD mRNA increases toward adulthood (46), but the protein expression and enzymatic activity of CuZnSOD are similar in prematurely born human infants and adults (4649). Human MnSOD mRNA increases toward adulthood, but—as with CuZnSOD—the specific activity of MnSOD is similar in neonatal and adult lung (46). Some studies have suggested that MnSOD increases gradually in the peripheral airways, interstitium, and alveolar macrophages during development, but in these studies the immunoreactivity of MnSOD in the bronchiolar epithelium and cuboidal alveolar epithelium (pre–type II pneumocyte cells) was already positive during Weeks 15 to 17 of gestation (47, 49, 50). ECSOD can be detected in the arterial intima, media, bronchiolar epithelium, alveolar epithelium, and extracellular matrix of human neonatal lung (51). Overall, both premature and mature human lungs appear to be prepared for the high oxygen tension (21% oxygen) that will be experienced at birth.

There may be significant differences in the developmental profile of SODs in human and animal lungs. Experimental studies on rats, rabbits, guinea pigs, and lambs have suggested that antioxidant enzymes increase during gestation and at birth in most species (5260). Part of these differences between human and animal lung may be related to the redox-sensitive mRNA-binding proteins that possibly participate in the enzyme regulation during ontogenesis. These proteins can be developmentally regulated so that adult rat lung, for example, contains lower MnSOD RNA–binding activity than neonatal lung (53, 61). In rabbit lungs, ECSOD distribution and activity increase toward adulthood. This may be partly related to changes in the intersubunit disulphide bonds during lung development (62). The localization of ECSOD also appears to be mainly intracellular in rabbit fetuses but increases in the extracellular space toward adulthood (63). Thus, there may be species differences in the developmental profile of SODs. Further studies need to be directed toward better understanding of the developmental expression of antioxidant enzymes, especially in human lung.

CuZnSOD

CuZnSOD gene contains regulatory elements such as nuclear factor-1, specificity protein-1, activator protein-1 and -2, glucocorticoid response element, heat shock transcription factor, nuclear factor κ B, and the metal responsive element (64, 65). In agreement, the promoter of CuZnSOD has been shown to be induced by oxidants and metal ions (65) and the synthesis of CuZnSOD to increase by heat shock in rat lungs (66) and by shear stress in aortic endothelial cells (67). However, in most studies, lung CuZnSOD mRNA or activity is not induced by cytokines or oxidant stress in vivo or in vitro (2, 23, 27, 36, 6873). Cultured endothelial cells exposed to hyperoxia have shown increased CuZnSOD mRNA with the intensive S growth phase of cells, but again there is no increase in enzyme activity (74). Elevated CuZnSOD activity in isolated alveolar type II epithelial cells after exposure of rats to hyperoxia has been found to be primarily associated with hypertrophy of the cells rather than being due to enzyme induction (75). If anything, immunohistochemical staining of CuZnSOD in the bronchial epithelium of ozone-exposed rats has shown lower reactivity than control lungs (76). It appears that CuZnSOD can be induced by certain stimuli, but in the lung, mitochondrial MnSOD—as will be described later—not CuZnSOD is the primary SOD that is substantially induced by hyperoxia and cytokines.

MnSOD

The MnSOD promoter contains binding sites for several transcription factors such as activator protein-1 and -2, specificity protein-1, and nuclear factor κ B (7779). Accordingly, MnSOD mRNA, protein and/or activity are increased in lung homogenates and in cultured pulmonary epithelial cells, endothelial cells, fibroblasts, alveolar, macrophages, granulocytes, and in malignant lung cells by oxidants, cigarette smoke, asbestos fibers, ozone, tumor necrosis factor α (TNFα), IFNγ, interleukins-1 and -6, endotoxin, redox regulators, and during exposure to drugs and thiol-reducing agents (33, 70, 72, 73, 76, 8090). Cytokine-induced MnSOD activation is rapid and sensitive, commonly requiring only 0.1 to 1 ng/ml concentrations of the cytokine. MnSOD induction can be seen as early as 2 hours, the maximum induction being observed at 24 to 48 hours (72). TNFα, which is the most widely investigated cytokine, increases MnSOD activity and mRNA expression in a dose- and time-dependent manner. The increase at the level of specific activity is 520% in A549 lung epithelial cells after exposure to TNFα (10 ng/ml) for 48 hours, the mRNA level increasing 20-fold after the 48-hour incubation (87, 91). Induction of MnSOD is different in various phases of cell growth, with the highest expression being observed in confluent cells (72). It has been postulated that MnSOD is induced only in nonmalignant cells (92), but at least TNFα leads to MnSOD induction both at the level of mRNA and specific activity in malignant lung adenocarcinoma and pleural mesothelioma cell lines (87, 91). MnSOD mRNA levels do not necessarily correlate with protein expression or enzyme activity, one possible explanation has been suggested to be the MnSOD RNA–binding protein which regulates MnSOD translation (93, 94). As described previously, oxidants and cytokines generally cause MnSOD induction. However, at least peroxynitrite (9598) and dexamethasone (99) appear to inactivate MnSOD. Peroxynitrite-related MnSOD inactivation has been suggested to be related to the phosphorylation of SOD binding proteins and to the induction of dityrosine formation and tyrosine oxidation in MnSOD (95, 96, 100). Taken together MnSOD is highly inducible by oxidants and cytokines but is inactivated at least by nitrosative stress in vitro.

The majority of studies on the regulation of MnSOD in vivo have been conducted with animals exposed to high oxygen tensions (hyperoxia). These studies have given valuable information about the regulation of MnSOD both in lung tissue and in individual cells. Exposure of rats to hyperoxia leads to a transient increase in the mRNA of MnSOD (340% maximally) in lung homogenates (69, 101). By in situ hybridization, MnSOD mRNA can be detected in arterioles, septal tips of alveolar ducts, endothelial cells, and in pleural mesothelium of hyperoxia-exposed rats (32). MnSOD mRNA obtained from hyperoxic lungs shows increased transcription and message stability (68). One to two weeks of exposure to hyperoxia also leads to elevated MnSOD immunoreactivity especially in alveolar type II cells and in interstitial fibroblasts (23, 102). Animal models where repeated exposures have resulted in increased tolerance to hyperoxia show increased MnSOD activity, increased MnSOD protein synthesis, and high translational efficiency (94, 103). Acute exposure to severe hyperoxia does not lead to MnSOD induction in human bronchial epithelium (25). MnSOD has also been observed to be downregulated in severe hyperoxia after a transient MnSOD mRNA elevation, and decreased enzyme activity appears to be related to impairment of the MnSOD translational efficiency (68, 94). The studies described previously with hyperoxia-exposed animals and hyperoxia and/or cytokine-exposed cultured cells (2, 36, 71, 104) suggest the importance of cytokines in MnSOD induction. Overall MnSOD is induced significantly by sublethal hyperoxia, whereas it is usually unchanged or downregulated in severe hyperoxia in vivo.

Acute hypoxia in hypoperfused or nonventilated rabbit lung leads to significantly decreased (32%) MnSOD activity (105, 106). Rats exposed to hypoxia for 7 days develop tolerance to hyperoxia with elevated MnSOD protein and activity (180%) (107). When cultured alveolar type II epithelial cells and lung fibroblasts have been exposed in vitro to air (control cells) or to 5% oxygen (hypoxia) MnSOD mRNA expression has decreased in both types of cells, the decrease being of the order of 20% (108, 109). In agreement, culture of alveolar macrophages under hypoxic conditions has resulted in decreased MnSOD activity (110). The variable results of these studies may be partly associated with the experimental conditions, severity of hypoxia, durations of the exposure, or species investigated. Severe hypoxia, which is often associated with chronic pulmonary diseases, can possibly enhance the inactivation of the most important antioxidant enzymes, such as SODs in the lung, thus predisposing these already injured lungs to further oxidant stress.

ECSOD

The gene for ECSOD contains several regulatory sequences such as activator protein-1, glucocorticoid response element, antioxidant response element, xenobiotic response element, and multiple binding sites for the Ets family of transcription factors (37, 63, 111), but direct oxidant stress does not affect ECSOD expression to the extent it commonly induces MnSOD activity (19). ECSOD, however, is induced by cytokines—at least by TNFα and IFNγ in rat type II pneumocyte (L2) cells (112), by TNFα and IFNγ in fibroblasts (113) and by IFNγ and interleukin-4 in vascular smooth muscle cells (114). The induction of the mRNA for ECSOD occurs within the first hours of oxidant exposure, with ECSOD protein levels rising within 24 hours. ECSOD is downregulated by transforming growth factor-β and interleukin-1 in cultured fibroblasts (113) and by TNFα, epidermal growth factor, platelet-derived growth factor, and granulocyte monocyte-colony stimulating factor in vascular smooth muscle cells (114), suggesting a possibly impaired antioxidant defense in various pathologic conditions in vivo where these cytokines and growth factors are expressed.

ECSOD makes up a significant component of the total SOD activity in the lung (20, 41). In one study, neither the distribution nor expression of ECSOD was changed after exposure of rats to hyperoxia (115), whereas exposure to severe hyperoxia resulted in a significant decrease of ECSOD in the lungs and broncholveolar lavage fluid of mice (116). ECSOD appears to be transiently induced in bleomycin-induced acute lung inflammation (117, 118). Exposure of rats to LPS did not reveal major changes in the distribution of ECSOD mRNA in the lung, but high transient ECSOD expression in invading neutrophils and macrophages was suggested to be associated with the defense of inflammatory cells against superoxide radicals (115). An ECSOD molecule contains two dimers with disulphide bonds linking the heparin-binding domains together (119, 120). One feature in the regulation of ECSOD in tissues is its proteolytic cleavage that leads to the removal of the carboxyterminal heparin–binding domain of ECSOD (119, 121) and thereby loss of the enzyme from the extracellular matrix. This cleavage may function as an additional regulatory mechanism of ECSOD in vivo because the proteolyzed ECSOD protein has been detected in the bronchial lavage fluid of bleomycin-treated mice (117). In summary, the expression of ECSOD is elevated to some extent during acute inflammation of the lung. Proteolytic removal of the heparin-binding domain of ECSOD has been suggested to be an important regulatory mechanism in the distribution of ECSOD within the extracellular space.

Induction of SODs and Tissue Protection

Exposure to sufficient hyperoxia or cytokines to cause MnSOD induction has been associated with increased tolerance to hyperoxia. Animals and/or cells pre-exposed to sublethal hyperoxia or TNFα are more resistant to subsequent exposures, and this resistance correlates with elevated SOD activity (5, 92, 122, 123). The association between SOD activity in the lung and oxidant protection, however, is not necessarily causal as has also been emphasized. Tolerance to hyperoxia is not related only to changes in the SODs because in these circumstances the lungs undergo multiple adaptive phenomena and the levels of several antioxidant enzymes (e.g., catalase, glutathione peroxidase, glutathione reductase, and glucose-6-phosphate dehydrogenase) are simultaneously elevated (69, 124127). Protection can also be associated with the severity of the exposures and may be species and cell specific (94, 128). Moreover, oxidant protection has been suggested to be related to mechanisms other than antioxidant enzymes (36, 129). Thus, lung protection against oxidant stress is complex, and several mechanisms in addition to the SODs are involved. (130).

CuZnSOD

Numerous in vitro studies with CuZnSOD have been conducted with neurons due to the importance of CuZnSOD in degenerative neurologic disorders (131, 132). Transfection of CuZnSOD into the cytosol of cultured cells has shown that protection is not dramatic and that overexpression of CuZnSOD without an increase in H2O2 scavenging mechanisms can be harmful. Simultaneous increases in CuZnSOD and in H2O2-decomposing antioxidant enzymes appear to give better protection than induction of CuZnSOD alone, and isolated CuZnSOD overexpression can enhance cell senescence and apoptosis (133). Shortened survival in cells with isolated CuZnSOD overexpression has been suggested to be due to an increased H2O2 level in the cells (133, 134). The experimental evidence for the elevated H2O2 levels in SOD overexpressing cells with consequent toxicity, however, is controversial. Liochev and Fridovich (135) have suggested that due to competitive reactions SOD overexpression may in fact decrease the overall H2O2 production. In agreement, recent mathematic assessments concluded that SOD overexpression may decrease, increase, or have no effect on H2O2 production, the outcome depending on the ratio between the rates of superoxide reduction and the rates of the non-H2O2–producing reactions at normal levels of SOD expression (136). Thus, the final effects of SOD overexpression on oxidant status and cell protection are complicated and depend on the oxidant and antioxidant reactions of the cell.

Studies with transgenic mice overexpressing CuZnSOD have not shown significant lung protection from hyperoxia (137, 138) or from experimentally induced endotoxemia (139). CuZnSOD may, however, play a role in other stress conditions of the lung. Mice overexpressing CuZnSOD were, for example, resistant to allergen-induced lung toxicity (140). Young transgenic mice overexpressing CuZnSOD were also more resistant to high oxygen tensions than their control littermates, but these mice also had elevated catalase, glutathione peroxidase, and glucose-6-phosphate dehydrogenase activities (141). Overall, the balance of several antioxidant enzymes appears to be more important than the level of isolated CuZnSOD in lung protection against high oxygen tension.

The studies with CuZnSOD knockout mice have not found any significant changes in the lungs or other vital organs (142). Fetal fibroblasts derived from mouse fetuses deficient in CuZnSOD, however, show increased sensitivity to paraquat, a compound that induces superoxide generation mainly in the cytoplasmic compartment (143). These studies with knockout animals and cultured cells confirm previous suggestions that CuZnSOD has major importance in the cytosolic compartment. Evidently, constitutive levels of cytosolic CuZnSOD have a role in lung protection, but protection cannot be substantially improved by genetic manipulation of CuZnSOD.

MnSOD

In contrast to CuZnSOD, there are numerous studies showing that transfection of MnSOD protects various cells against oxidants and oxidant-generating compounds. Resistance is increased at least to hyperoxia (144), oxidants such as paraquat (145), cigarette smoke (146, 147), radiation (148, 149), injury caused by asbestos fibers (150), cytokines such as TNFα (148, 151) and interleukin-1 (152), and drugs such as doxorubicin and mitomycin (148). MnSOD can inhibit apoptosis in lung cells (153), mouse liver cells (154), and malignant cells such as fibrosarcoma (155) and hepatocellular carcinoma cells (156) by controlling mitochondrial reactive oxygen species generation. Besides increasing oxidant resistance, overexpression of MnSOD has antiproliferative effects and tumor suppressor characteristics (157, 158). MnSOD modulates DNA-binding activities and transcriptional activation of redox-sensitive transcription factors and oncoproteins (159, 160). As with CuZnSOD, overexpression of multiple antioxidant enzymes can be more beneficial than isolated overexpression of MnSOD alone. MnSOD protects lung cells against hyperoxia, but simultaneous overexpression of H2O2 scavenging enzymes such as catalase offers additional protection (161). Antisense technology to decrease MnSOD activity has not been systematically investigated in lung cells, but in several others (e.g., nonmalignant and malignant cells) it results in lowered oxidant resistance and increased sensitivity to TNFα and IFNs α and γ (151, 162, 163). In summary, the major findings of MnSOD transfection to airway epithelial and/or pulmonary endothelial cells are significant protection against several oxidant-producing agents in vitro and in vivo.

The importance of MnSOD in lung protection has also been proved in transgenic animals. Transgenic animals created using a surfactant protein C promoter and human MnSOD cDNA exhibit an elevated MnSOD level in alveolar type II epithelial cells and bronchiolar epithelial cells (60% higher total lung MnSOD activity and fourfold higher levels of MnSOD immunoreactive protein in the mitochondria of type II cells). These animals survive significantly longer in 95% oxygen compared with nontransgenic littermates (164), suggesting that MnSOD is critical in modulating pulmonary oxygen toxicity in distal respiratory epithelium. Transgenic mice created using the human β-actin promoter to express the human MnSOD widely in the lung (pneumocyte II cells, endothelial cells, fibroblasts, 300% increase in activity) showed no or marginal advantage in survival after exposure to hyperoxia (137, 165). The reasons for the controversies of these two studies remain unclear but may be related to different expression profiles of the MnSOD transgene in various lung cells, disturbance of the ideal antioxidant/oxidant balance, or genetic background of the mice in these models. Compartmentalization plays an important role in oxidant resistance because, for example, introduction of CuZnSOD in MnSOD knockout animals with consequent cytosolic overexpression of CuZnSOD did not prevent oxygen-related neonatal lethality (166). This finding further supports the importance of mitochondria as the major site of reactive oxygen species production and that mitochondria are early targets of oxidant injury. Results with MnSOD transgenic studies suggest the importance of MnSOD in protecting lung tissue against high oxygen tension, provided the enzyme is targeted to the mitochondria of alveolar type II cells.

The total deficiency of MnSOD is lethal, but the lungs in MnSOD−/− mice are only minimally affected. The most important manifestations in MnSOD knockout mice include degenerative changes in large areas of the central nervous system, especially in the basal ganglia, and cardiotoxicity (138, 167, 168). In studies with a deleted exon (168) or deleted exons 1 and 2 (167) of the MnSOD gene, the main findings are similar (i.e., no apparent lung toxicity but increased susceptibility to superoxide-induced mitochondrial damage in the central nervous system and heart). The minimal changes reported to occur in the lungs of MnSOD−/− mice include alveolar dilatation suggestive of delayed postnatal lung development, but due to the severe lung atelectasis more defined assessment of those abnormalities has remained unclear (169). In agreement, exposure of MnSOD knockout animals to 50% oxygen appears to lead to undefined changes in the alveolar general structure of mouse lung (169).

Studies on MnSOD heterozygous mice having an approximate 50% reduction in the MnSOD activity do not reveal sensitization to hyperoxia or pertussis toxin–induced lung injury (170172). This suggests that at least in mice 50% MnSOD activity is enough for normal oxidant resistance of the lung. MnSOD heterozygous mice have decreased lung glutathione concentration (173), and muscle and cardiac mitochondria show increased oxidative damage and an increased tendency for apoptosis (173, 174). The low lung glutathione levels suggest that decreased MnSOD lead to some degree of oxidative stress in the lungs. Interestingly, aged MnSOD heterozygous mice have increased risk for lung cancer (175) and pulmonary fibrosis (176). The results on MnSOD knockout homozygous and heterozygous animals and cells deficient in MnSOD suggest that MnSOD plays a major role in protecting the most important vital organs, especially brain and heart, against endogenous oxidant stress. Half of the normal MnSOD activity is sufficient to block acute oxidative stress in otherwise normal mouse lungs but may contribute to the development of lung fibrosis and lung cancer.

ECSOD

ECSOD is postulated to be of critical importance in the lung extracellular matrix, including the pulmonary vasculature, by consuming superoxide and thereby diminishing the reaction between nitric oxide and superoxide. ECSOD has been shown to protect lung interstitium against free radicals generated during inflammation (19). A threefold increase of ECSOD in alveolar type II and nonciliated bronchial epithelial cells driven by the specificity protein-C promoter leads to protection against hyperoxia partly by attenuating neutrophilic inflammation (177). In mouse lung, ECSOD has been shown to attenuate inflammation when assessed from the samples of bronchoalveolar lavage (178) and alveolar structure (179). Overexpression of lung ECSOD also attenuates lung injury after hemorrhage when analyzed by lung wet to dry weight, lipid peroxidation, activation of nuclear factor κ B, and myeloperoxidase activity (180). Transgenic mice with elevated brain ECSOD were more susceptible to hyperbaric oxygen than control animals. This apparently paradoxical response is believed to be due to high ECSOD expression in the cerebral vasculature leading to sparing of nitric oxide in those vessels and thus blocking the normal vasoconstrictive response of the cerebral vasculature to hyperoxia. By preserving brain blood flow, the ECSOD transgenic animals actually have a higher cerebral oxygen delivery and thus greater sensitivity to oxygen-mediated toxicity (181). The functions of ECSOD are cell and tissue specific and associated with both regulation of nitric oxide–mediated intracellular signaling and with preservation of the extracellular oxidant–antioxidant balance.

Mice lacking ECSOD develop normally and remain healthy until at least 14 months of age. However, when exposed to more than 99% oxygen, these mice display shortened survival compared with control animals and develop more severe lung damage, including vascular congestion, septal thickening, and an increased number of neutrophils in the lung (182). These findings suggest that under normal physiologic conditions other antioxidant enzymes can balance for the loss of ECSOD, whereas under conditions of inflammatory stress ECSOD plays a critical role in protecting the lung and reducing the amplification of the injury

Exogenous SODs, SOD Mimetics, and Lung Protection

Numerous studies have investigated whether SOD enzyme proteins given either by injection or inhalation can protect lung tissue against oxidant injury. In most models some protection has been obtained, with intratracheal instillation generally providing better protection than intravenous or intraperitoneal injections. These models have mainly used CuZnSOD and concluded that exogenous CuZnSOD by inhalation or instillation provides significant protection against acute lung injury although conflicting results have also been obtained (183185). Protection of the lung against oxidant injury has been improved by liposome encapsulation of CuZnSOD, which is postulated to provide a longer half-life and better penetration of the enzyme to the target cells (185, 186). Lecithinized SOD (phosphatidylcholine-SOD) has characteristics similar to liposome-encapsulated SOD, and it has protected against bleomycin-induced pulmonary fibrosis when analyzed by lung hydroxyproline content, neutrophil and lymphocyte accumulation, and expression of several cytokines (187). Efficacy has further been improved when liposomes containing CuZnSOD have included catalase, which decomposes H2O2 (185, 186, 188). In agreement, polyethylene glycol–attached CuZnSOD + catalase and/or polyethylene glycol catalase provides better protection against hyperoxia and fiber-induced lung injury than polyethylene glycol SOD alone (189191). SOD-conjugated polyethylene glycol attenuates airway responsiveness in rabbits, suggesting a role of free radicals also in airway inflammation and hyper-reactivity (192). Aerosolized delivery of recombinant human MnSOD is even more efficient, protecting baboon lung against 100% oxygen. It decreases pulmonary Q̇s/Q̇t, preserves arterial oxygenation, decreases lung edema and, by quantitative morphometry, shows protection of alveolar epithelium from hyperoxia (193, 194). Recombinant human MnSOD is positively charged and, when administered in this form, is likely to behave more like ECSOD (i.e., an ECSOD whose charge leads to affinity to negatively charged extracellular elements such as glycosaminoglycans.). No corresponding studies have been conducted on ECSOD.

Several classes of synthetic SOD mimetics have recently been developed and shown to possess significant antioxidant capacity. These new antioxidants provide an alternative approach to attenuate oxidant-related lung injury in vivo. These small molecular weight compounds distribute more easily to tissues, potentially reaching high concentrations in intracellular domains and avoiding the problem of antigenicity that is a concern with antioxidant proteins. The three classes of metal-containing substantial SOD mimetics with SOD activity include the salen compounds (195), macrocyclics (196), and metalloporphyrins (197) (Figure 7)

. In addition to these compounds, nitroxide SOD mimetics have been shown to have free radical scavenging capacity (198200). The different forms of SOD mimetics have been found to be efficacious in a variety of in vitro and in vivo animal models, but they have not yet been tested in humans.

The salen class of antioxidant mimetics shares a three-ring aromatic structure containing Mn (195). The prototype of these compounds is EUK-8, which expresses both SOD and catalase-like activities. Newer versions include EUK-134, EUK-178, and EUK-189 with the newer salen compounds reported to exhibit higher catalase activity (201). EUK-8 has been shown to ameliorate acute lung injury assessed by arterial hypoxia, pulmonary hypertension, and lung malondialdehyde content (an indicator of lipid peroxidation) after 4 hours of LPS exposure. It also prevents manifestations of LPS-induced respiratory distress syndrome in pigs by detoxifying reactive oxygen species without affecting the release of other important proinflammatory mediators (202). Besides lung protection, the salen antioxidant compounds also have protective effects on other organs. They prolong the survival of MnSOD knockout mice, decrease motor neuron degeneration and apoptosis (203206), attenuate mitochondrial abnormalities (207), and attenuate injury to the kidney, liver, skeletal muscle, pancreas, and heart (208, 209). Overall, EUK compounds have been shown to protect lung and several other organs in experimental oxidant exposures in vivo.

The macrocyclic class of antioxidant mimetics binds Mn in a five-ring structure so that the metal is available for single electron transfers. They are believed to be relatively specific scavengers for superoxide (196). A prototypic macrocyclic SOD mimetic, M40403, has been suggested to be especially important in inflammation. It has been potent in carrageenan-induced pleurisy in rats. In this model, it decreases pleural fluid, neutrophil accumulation, and the concentration of several inflammatory cytokines (196). Besides, in the lung, macrocyclics have been found to protect against the inflammatory states of several other organs (210).

The metalloporphyrin class of antioxidant mimetics exhibit strong antioxidant properties, including scavenging of superoxide, H2O2, peroxynitrite, and lipid peroxyl radicals (197, 211). They exhibit definite, but low, catalase-like activity that has been attributed to their extensive conjugated ring system and reversible one-electron oxidations. Their capacity to scavenge peroxynitrite apparently involves the formation of an oxo-Mn (IV) complex that can be reduced to the Mn(III) oxidation state by endogenous antioxidants. One of the widely investigated metalloporphyrin antioxidant compounds is Mn(III) meso-tetrakis (4-benzoic acid) porphyrin (212). Mn(III) meso-tetrakis (4-benzoic acid) porphyrin has been shown to protect against paraquat-induced pulmonary endothelial cell injury in vitro (211), paraquat-induced lung injury also in vivo (213), and carrageenan-induced pleurisy (214). In a rat pleurisy model, Mn(III) meso-tetrakis (4-benzoic acid) porphyrin has decreased leukocyte recruitment, myeloperoxidase activity, histologic lung injury, peroxynitrite formation, and DNA damage. Mn(III) meso-tetrakis (4-benzoic acid) porphyrin has been shown to attenuate the severe lung fibrosis occurring in mice exposed to bleomycin (215). Other closely related metalloporphyrins (MnTE-2-PyP and MnTDE-1, 3-ImP) have been recently shown to protect against cigarette smoke–induced lung injury, antigen-induced broncho-obstruction and development of bronchopulmonary dysplasia (216220). Besides preventing lung injury, these antioxidants have been found to protect against liver failure (221), zymosan-induced shock (222), cardiomyopathy (223), and neurologic manifestations (224, 225). Overall, new, targeted SOD mimetics have efficient antioxidant capacity and are efficient in preventing lung injury in animal models of enhanced oxidative stress.

Free Radicals in the Pathogenesis of Human Lung Diseases

Free radicals have an evident role in obstructive lung diseases. Asthma is associated with chronic inflammation and an increased oxidant burden. Inflammatory cells of the airways of patients with asthma are activated and generate increased amounts of free radicals. Exhaled air of patients with asthma contains elevated levels of H2O2, nitric oxide, and organic volatile compounds (226230). The bronchial epithelium of patients with asthma shows elevated expression of inducible nitric oxide synthase, which has been linked to increased nitric oxide levels in the exhaled air (230, 231). Thus multiple reactive oxygen species–producing pathways are simultaneously induced in asthmatic airways as shown in Figure 8

.

There is evidence that free radicals play a central role in chronic obstructive pulmonary disease (232234). Chronic obstructive pulmonary disease is related to cigarette smoking in over 90% of cases. One puff of smoke contains 1014–16 free radicals (235), and the airways of patients with chronic obstructive pulmonary disease contain increased numbers of neutrophils (234) that, when activated, have a high capacity to produce free radicals. In addition to the direct toxic effects of cigarette smoke and neutrophil-derived free radicals, activated neutrophils inactivate the natural protease system of the lung, the most intensively investigated of them being α-1-antitrypsin. This inactivation enhances the progression of emphysema in the lung.

Free radicals participate in the pathogenesis of numerous interstitial lung diseases. Idiopathic interstitial pneumonias (236, 237), especially usual interstitial pneumonia, represent parenchymal lung diseases with a poor prognosis and histopathologic similarities with hyperoxia-induced lung injury. In contrast to hyperoxic lung injury, usual interstitial pneumonia has patchy lung involvement with a variable stage of fibrosis (active fibrogenesis occurring in the so-called fibroblastic foci) and a low grade of inflammation (238). There are numerous studies showing that the oxidant burden in the lungs of the patients with pulmonary fibrosis is increased and that inflammatory cells of these patients generate more radicals than the cells of healthy control subjects (239245). In addition, the inflammatory cells of these disorders show elevated inducible nitric oxide synthase expression that then may cause toxicity to multiple cell types of the lung (246, 247). Inflammatory cells of patients with sarcoidosis generate more radicals than the cells of healthy control subjects (248), and the glutathione content of the airways in another granulomatous lung disease—allergic alveolitis—is decreased (249), suggesting increased oxidant burden in granulomatous lung disorders. Furthermore, the granulomas and inflammatory cells of sarcoidosis and allergic alveolitis express elevated levels of inducible nitric oxide synthase as a marker of the oxidant stress in the lung (246). The most widely investigated environmental oxidant-generating agents are asbestos fibers. Asbestos fibers are associated with reactive oxygen species generation both in vitro and in vivo (250, 251) and induce antioxidant enzymes, most importantly MnSOD in bronchial epithelial and mesothelial cells (252254). The production of reactive oxygen species caused by asbestos fibers is believed to be part of the pathogenesis of the resulting fibrotic lung disease (asbestosis). Extracellular macromolecules, including collagen, cartilage, pulmonary surfactant, and proteinase inhibitors, are sensitive to superoxide- and peroxynitrite-mediated damage (19, 255), which then may have potential implications for the enhancement of parenchymal lung diseases. One of the most important growth factors in the pathogenesis of fibrotic lung diseases is the transforming growth factor-β. Importantly, latent transforming growth factor-β, which is abundantly expressed in healthy lung, is activated by free radicals (radiation, nitric oxide) (256, 257). Overall, free radicals play a central role in the initiation of fibrogenesis by activating transforming growth factor-β and in the progression of lung injury by activated inflammatory cells.

The high concentration of oxidants in cigarette smoke has been postulated to contribute to carcinogenic impact of cigarette smoke in the causation of lung cancer (258260). Besides cigarette smoke, asbestos fibers lead to DNA strand breaks, increase cell proliferation, and cause both lung cancer and mesothelioma (253, 261). During these events, cells will carry multiple genetic alterations including inactivation of tumor suppressor genes and activation of oncogenes that will help the escape of the cells from normal growth control. All these events are at least partly regulated by changes in the cellular redox state. The most important of these reactions that have been shown to be regulated by oxidants include mitochondrial apoptosis, apoptosis caused by p53, and growth and senescence regulated by several other oncogenes such as CKIp21 (Cip1) and PTEN (262266). Recent studies also suggest that hypoxia-associated reactive oxygen species generation at the mitochondrial electron transport chain (ubiquinone) contributes to hypoxia-inducible factor stabilization with consequently increased angiogenesis (267). Thus, the association of reactive oxygen species both in carcinogenesis and in cancer progression has been shown in numerous investigations.

Free radicals are closely associated with a variety of acute lung diseases. These include oxygen-related lung injury of prematurely born neonates, which can lead to bronchopulmonary dysplasia (chronic lung disease) (268). The use of high oxygen concentrations in the treatment of severe lung complications leads to problems of acute oxygen toxicity in intensive care departments. Furthermore, free radicals have a central role in lung diseases such as infections, pleural disorders, and uncommon pulmonary diseases such as cystic fibrosis, primary pulmonary hypertension, and complications associated with lung transplantation (269272). Several drugs are associated with reactive oxygen species generation and lung injury with the most widely investigated of them being bleomycin (187). Bleomycin is a chemotherapeutic agent that causes oxidant-mediated lung injury and lung fibrosis both in laboratory animals and in humans (273, 274). Numerous other chemotherapeutic agents, including carmustine (BCNU), antracyclines, antimetabolites, and antibiotics (nitrofurantoin), cause lung injury by reactive oxygen species–mediated mechanisms (1). Overall, free radicals are involved not only in the pathogenesis of lung diseases but also in lung injury caused by drug therapies and radiation.

SOD Polymorphisms and Lung Diseases

SODs are coded by different genes, and their genetic variation plays a role in the pathogenesis of several free radical–associated disorders (131). Over 90 mutations of CuZnSOD have been characterized, many of which are associated with a reduction in specific enzyme activity to 50 to 80% of that of the wild-type protein (131). CuZnSOD polymorphisms are known to be associated with degenerative neurologic diseases (132), but these polymorphisms have not been investigated in human lung diseases.

MnSOD is synthesized in the cytoplasm as a precursor molecule containing the N-terminal leader sequence, which is removed during the transport of the molecule to the mitochondria (77, 275, 276). A polymorphism of MnSOD has been described, where a single nucleotide change in the N-terminal region of the DNA encodes the signal sequence of either alanine (Ala) or valine (Val) in the mitochondrial targeting sequence (276, 277). In a study of over 1,100 patients with lung cancer, individuals with ValVal and ValAla genotypes had a higher risk for lung cancer than the AlaAla genotype (odds ratio 1.67 and 1.4) (278). A similar tendency was observed with mesothelioma (odds ratio 2.0 and 1.4), but the risk did not reach statistical significance, probably due to the small number of the patients in the study (61 cases with mesothelioma) (279). On the basis of recent findings, MnSOD polymorphism was not associated with asthma (Kinnula VL, Lehtonen S, Koistinen P, Kakko S, Savolainen M, Kere J, Ollikainen V, Laitinen T, unpublished). In addition to the polymorphisms described previously, Ho and Crapo have reported the amino acid substitution Thr58Ile in studies on a human cDNA library (280). The Ile-form of MnSOD exhibits threefold higher activity than the Thr-form in transfected MCF-7 cells (281), possibly due to conformational changes in the protein molecule. This polymorphism has not been described in human lung diseases (131, 282). In summary, signal sequence polymorphisms leading to disturbances where essential proteins are not properly targeted, as is the case with MnSOD, have been shown to be associated with the development of malignant lung diseases.

A single nucleotide substitution resulting in an amino acid change from Arg to Gly at 213 (in the middle of the positively charged sequence of the heparin-binding domain of ECSOD) (283) leads to increased circulating ECSOD activity (10-fold). It causes decreased anchoring of ECSOD to negatively charged elements such as heparin in the interstitium and elevated serum levels of ECSOD in those individuals with the decreased heparin-binding variant. Approximately 94 to 98% of the population exhibits the normal (low) level of serum ECSOD activity (283285). This polymorphism has been described (in three patients) with chronic obstructive lung disease (286). Recent studies, however, have shown that ECSOD polymorphism is not associated with asthma or idiopathic pulmonary fibrosis (Kinnula VL, Lehtonen S, Koistinen P, Kakko S, Savolainen M, Kere J, Ollikainen V, Laitinen T, unpublished). It is likely that the polymorphism of ECSOD, being very rare, cannot explain the risk for the development of common lung diseases. Two additional polymorphisms described in the human ECSOD gene do not have any effects on the heparin-binding capacity or activity of this enzyme (285). These findings do not exclude the possibility that together with other impairments in the patients' oxidant-decomposing mechanisms—genetic or environmental—SOD genes become significant in inflammatory or fibrotic lung diseases where oxidant stress is increased.

Obstructive Lung Diseases

Mice overexpressing CuZnSOD are resistant to allergen-induced lung toxicity (140), suggesting that SOD is important in the pathogenesis of allergic inflammation and asthma. In agreement, in most human studies SOD activity decreases in the bronchial epithelium (∼50%), in the cells of bronchoalveolar lavage (25%), and in bronchial brushings (nearly 50%) in patients with asthma compared with control subjects (287, 288). Not only the airways of patients with asthma but also blood monocytes and neutrophils of patients with asthma show a lower MnSOD activity than corresponding cells in control subjects (289). Acute antigen instillation into the lungs of individuals with atopic asthma also leads to a rapid 50% decrease in SOD activity in the bronchoalveolar lavage cells of these individuals (287). Because of the abundance of ECSOD in the lung, part of a decrease in SOD activity in asthma may reflect ECSOD activity, although this has never been directly assayed. In summary, oxidant stress is a component of allergic airways diseases, and a decreased capacity to scavenge superoxide, at least in asthma, may exaggerate progression of the disease.

Serum of patients with chronic obstructive pulmonary disease has been reported to contain elevated levels of ECSOD (30%) compared with that of control subjects (286). In that study, three patients with highest levels of serum ECSOD displayed a single base pair guanine to cytosine missense mutation in the coding region of ECSOD at position 213, which is the same polymorphism described previously as leading to decreased anchoring of ECSOD to the interstitium (283). No studies have been reported on the expression of SODs in lung tissue of patients with chronic obstructive pulmonary disease.

Interstitial Lung Diseases

Only a few studies have investigated the expression of SODs in granulomatous diseases and idiopathic pneumonias of human lung. In normal lung, CuZnSOD is mainly localized to bronchial epithelium, and its expression is similar in healthy lung and pulmonary sarcoidosis (27). Unchanged reactivity of CuZnSOD in control subjects and in patients with sarcoidosis is in agreement with several previous notions that CuZnSOD is not induced by cytokines or oxidants in the lung. MnSOD is elevated in alveolar macrophages and in the granulomas associated with pulmonary sarcoidosis and allergic alveolitis (27). MnSOD is also at least moderately expressed by immunohistochemistry in alveolar type II epithelial cells and macrophages in inflammatory areas of usual interstitial pneumonia and desquamative interstitial pneumonia, but its reactivity appears to be low in the late fibrotic lung lesions of usual interstitial pneumonia (35). These human studies confirm previous animal studies and show that MnSOD is induced during acute inflammatory stages of the lung parenchyma and suggest that antioxidant defense (at least MnSOD) is impaired during the progression of fibrogenesis.

Respiratory Distress Syndrome and Bronchopulmonary Dysplasia

CuZnSOD is expressed mainly in bronchial epithelium, and its immunoreactivity is similar or decreased in acute respiratory disorders of neonatal lung compared with healthy neonatal lung (47, 48). MnSOD expression has been reported to be similar in respiratory distress syndrome, bronchopulmonary dysplasia, age-matched control subjects, and healthy human newborns (47, 50) with the exception of intensive MnSOD immunoreactivity in proliferating alveolar type II cells in bronchopulmonary dysplasia (47). This latter finding is in agreement with a report of increased MnSOD levels in the baboon model of bronchopulmonary dysplasia (290). Preliminary studies with respiratory distress syndrome have found that ECSOD is expressed in the arterial intima and endothelium, and in the metaplastic alveolar epithelium, chondrocytes and hyaline membranes of respiratory distress syndrome (51). The expression of ECSOD in the metaplastic epithelium of this disorder suggests the induction of ECSOD and an attempt to increase antioxidant capacity by the diseased lung. In a recent preliminary study, alveolar macrophages, alveolar epithelium, and interstitium of patients with bronchopulmonary dysplasia suggested enhanced ECSOD reactivity (51). The neonatal lung evidently tries to adapt to enhanced oxidant stress by induction of MnSOD and possibly of ECSOD.

Carcinogenesis and the Expression of SODs in Lung Tumors

SODs can be hypothesized to play a role in carcinogenesis due to multiple effects of reactive oxygen species on cell growth and survival. Most of these studies, however, have been conducted in vitro. A recent study on CuZnSOD and numerous earlier studies on MnSOD have suggested that CuZnSOD and MnSOD genes are tumor suppressive (291293). The primary findings in these experimental studies, conducted on cultured cells containing isolated overexpression of CuZnSOD or MnSOD (in most of the cases) by transfection, were suppression of neoplastic transformation, decreased proliferation, increased differentiation in vitro, and—if inoculated to nude mice—decreased tumor growth (293300). A low level of SODs in cancer cells has been postulated to be associated with specific oncogene products regulated by the redox state of the cell (159, 301), inhibition of transcription factors such as activator protein-1 and nuclear factor κ B (301, 302), and methylation of the upstream transcriptional regulatory sequence of the MnSOD gene (301, 303). Conclusions obtained from in vitro studies cannot be strictly extrapolated to the in vivo situation where several antioxidant enzymes and related detoxification mechanisms may be simultaneously induced. Highly metabolically active tumor cells also have complex interactions with numerous nonmalignant cells that regulate tumor growth and invasion (i.e., vasculature and angiogenesis) where the oxygen environment may also be highly variable.

A small immunohistochemical study on various lung malignancies (altogether 19 cases) has suggested that CuZnSOD is low in lung cancer in general (34). A recent study where lung tumor samples were assessed for CuZnSOD using several methods (Northern blotting, Western blotting, activity) reported similar levels of CuZnSOD in lung tumor homogenates and in nonmalignant lung (304). No studies on CuZnSOD have been conducted on mesothelioma tissue, but one micro array study on a malignant mesothelioma cell line showed higher expression of CuZnSOD mRNA in malignant mesothelioma cells than in nonmalignant ones (305). Thus, there are only a few studies on CuZnSOD and human lung malignancies with variable findings on CuZnSOD expression and its suggested significance.

The first small immunohistochemical study on lung cancer (19 cases) could not show any consistent increase in MnSOD (34). However, a recent much larger study showed higher total SOD activities (200%) in lung cancer than in nonmalignant control lung. Northern blotting and Western blotting analyses confirmed this being related to MnSOD (304). Thus, MnSOD is possibly elevated in lung cancer.

Several studies have investigated MnSOD in mesothelioma. These studies have demonstrated that mesothelioma contains high levels of MnSOD mRNA, protein, and activities compared with nonmalignant mesothelium or mesothelial cells, increasing threefold to 10-fold (306309). MnSOD has been suggested to be a possible diagnostic marker of mesothelioma due to its very intense activity in mesothelioma compared with the much lower levels commonly found in adenocarcinoma metastasized to pleura (307). Thus, it is known that MnSOD is higher in malignant mesothelioma than in the nonmalignant mesothelium, but its significance for tumor promotion and drug resistance is unresolved.

A summary of the expression of SODs in human lung diseases has been given in Table 3

TABLE 3. Summary of the changes in expression of superoxide dismutases in human lung diseases


MnSOD

Lowered or unchanged in asthmatic airways
Elevated in alveolar type II epithelial cells and macrophages  of interstitial pneumonias and sarcoidosis
Low in fibroblasts and fibromyxoid lesions in usual interstitial  pneumonia (idiopathic pulmonary fibrosis)
High in the proliferating type II epithelial cells in bronchopulmonary dysplasia (chronic lung disease)
High in mesothelioma
Possibly elevated in lung cancer
CuZnSOD
Decreased in asthmatic airways
Elevated mRNA in cultured mesothelioma cells (micro array  study)
ECSOD

Positive in hyaline membranes and in the proliferating type  II epithelial cells in respiratory distress syndrome and bron  chopulmonary dysplasia (chronic lung disease)

For definitions of abbreviations see Table 2.

.

CONCLUSIONS AND FUTURE ASPECTS

A basic understanding about the expression and regulation of antioxidant enzymes in normal lung and the changes that occur in lung diseases is necessary to develop therapeutic interventions to control oxidative stress in the lung. Oxidant/antioxidant balance in the bronchial epithelium, alveolar structures, and interstitium constitute the primary defense against oxidant stress of the lung. Although the expression of SODs has been characterized relatively well in animal and human lung, the specific role of the antioxidant enzymes in the pathogenesis of most lung diseases remains unclear. For instance, individual variability of these enzymes (polymorphisms) leading to lower antioxidant activity in lung cells may be hypothesized to play a role in the development of both nonmalignant and malignant lung diseases. Several polymorphisms have in fact been characterized, but their role in the pathogenesis of various lung disorders has not yet been systematically investigated. Antioxidant enzymes exhibit cell-specific expressions in human lung. MnSOD is upregulated in inflammatory areas of some lung diseases, but whether induction offers any protection in these diseases or whether induction is a secondary reaction with variability in individuals and various cell types also remains to be investigated. ECSOD is uniquely expressed in the lung and holds potential importance in multiple human lung diseases, with special interest being directed toward interstitial lung diseases and diseases of the vasculature. The significance of ECSOD in lung diseases also waits for future investigations. More efforts should be directed not only toward understanding the significance of the variability of these enzymes in individuals and various diseases but also to their potential role in therapeutic approaches. It, however, is clear that oxidants are associated with most lung diseases and that severe ongoing oxidant stress causes downregulation of SODs. These phenomena may further enhance the progression of the lung disease. It may, therefore, be hypothesized that therapeutic antioxidants such as SOD mimetics might provide new approaches to attenuate oxidant-associated lung injury in humans. These new agents show efficacy in oxidant-induced experimental lung models, but their significance has not yet been investigated in human lung.

1. Halliwell B, Gutteridge JMC. Free radicals in biology and medicine. Oxford: Claredon Press; 1996.
2. Kinnula VL, Crapo JD, Raivio KO. Generation and disposal of reactive oxygen metabolites in the lung. Lab Invest 1995;73:3–19.
3. Gutteridge JM, Halliwell B. Free radicals and antioxidants in the year 2000: a historical look to the future. Ann N Y Acad Sci 2000;899:136–147.
4. Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol 2000;279:L1005–L1028.
5. Crapo JD, Tierney DF. Superoxide dismutase and pulmonary oxygen toxicity. Am J Physiol 1974;226:1401–1407.
6. Marklund SL. Human copper-containing superoxide dismutase of high molecular weight. Proc Natl Acad Sci USA 1982;79:7634–7638.
7. McCord JM, Fridovich I. Superoxide dismutase: an enzymic function for erythrocuprein (hemocuprein). J Biol Chem 1969;244:6049–6055.
8. Weisiger RA, Fridovich I. Mitochondrial superoxide simutase: site of synthesis and intramitochondrial localization. J Biol Chem 1973;248:4793–4796.
9. Rahman I, MacNee W. Oxidative stress and regulation of glutathione in lung inflammation 15. Eur Respir J 2000;16:534–554.
10. Cantin AM, North SL, Hubbard RC, Crystal RG. Normal alveolar epithelial lining fluid contains high levels of glutathione. J Appl Physiol 1987;63:152–157.
11. Pietarinen-Runtti P, Lakari E, Raivio KO, Kinnula VL. Expression of antioxidant enzymes in human inflammatory cells. Am J Physiol Cell Physiol 2000;278:C118–C125.
12. Pietarinen P, Raivio K, Devlin RB, Crapo JD, Chang LY, Kinnula VL. Catalase and glutathione reductase protection of human alveolar macrophages during oxidant exposure in vitro. Am J Respir Cell Mol Biol 1995;13:434–441.
13. Holmgren A. Antioxidant function of thioredoxin and glutaredoxin systems. Antioxid Redox Signal 2000;2:811–820.
14. Powis G, Mustacich D, Coon A. The role of the redox protein thioredoxin in cell growth and cancer. Free Radic Biol Med 2000;29:312–322.
15. Rhee SG, Kang SW, Netto LE, Seo MS, Stadtman ER. A family of novel peroxidases, peroxiredoxins. Biofactors 1999;10:207–209.
16. Kinnula VL, Lehtonen S, Kaarteenaho-Wiik R, Lakari E, Paakko P, Kang SW, Rhee SG, Soini Y. Cell specific expression of peroxiredoxins in human lung and pulmonary sarcoidosis. Thorax 2002;57:157–164.
17. Soini Y, Kahlos K, Napankangas U, Kaarteenaho-Wiik R, Saily M, Koistinen P, Paaakko P, Holmgren A, Kinnula VL. Widespread expression of thioredoxin and thioredoxin reductase in non-small cell lung carcinoma. Clin Cancer Res 2001;7:1750–1757.
18. Comhair SA, Erzurum SC. Antioxidant responses to oxidant-mediated lung diseases. Am J Physiol Lung Cell Mol Physiol 2002;283:L246–L255.
19. Oury TD, Day BJ, Crapo JD. Extracellular superoxide dismutase: a regulator of nitric oxide bioavailability. Lab Invest 1996;75:617–636.
20. Marklund SL. Extracellular superoxide dismutase in human tissues and human cell lines. J Clin Invest 1984;74:1398–1403.
21. Stone KC, Mercer RR, Gehr P, Stockstill B, Crapo JD. Allometric relationships of cell numbers and size in the mammalian lung. Am J Respir Cell Mol Biol 1992;6:235–243.
22. Stone KC, Mercer RR, Freeman BA, Chang LY, Crapo JD. Distribution of lung cell numbers and volumes between alveolar and nonalveolar tissue. Am Rev Respir Dis 1992;146:454–456.
23. Chang LY, Kang BH, Slot JW, Vincent R, Crapo JD. Immunocytochemical localization of the sites of superoxide dismutase induction by hyperoxia in rat lungs. Lab Invest 1995;73:29–39.
24. Coursin DB, Cihla HP, Oberley TD, Oberley LW. Immunolocalization of antioxidant enzymes and isozymes of glutathione S-transferase in normal rat lung. Am J Physiol 1992;263:L679–L691.
25. Erzurum SC, Danel C, Gillissen A, Chu CS, Trapnell BC, Crystal RG. In vivo antioxidant gene expression in human airway epithelium of normal individuals exposed to 100% O2. J Appl Physiol 1993;75:1256–1262.
26. Kinnula VL, Yankaskas JR, Chang L, Virtanen I, Linnala A, Kang BH, Crapo JD. Primary and immortalized (BEAS 2B) human bronchial epithelial cells have significant antioxidative capacity in vitro. Am J Respir Cell Mol Biol 1994;11:568–576.
27. Lakari E, Paakko P, Kinnula VL. Manganese superoxide dismutase, but not CuZn superoxide dismutase, is highly expressed in the granulomas of pulmonary sarcoidosis and extrinsic allergic alveolitis. Am J Respir Crit Care Med 1998;158:589–596.
28. Chang LY, Slot JW, Geuze HJ, Crapo JD. Molecular immunocytochemistry of the CuZn superoxide dismutase in rat hepatocytes. J Cell Biol 1988;107:2169–2179.
29. Crapo JD, Oury T, Rabouille C, Slot JW, Chang LY. Copper, zinc superoxide dismutase is primarily a cytosolic protein in human cells. Proc Natl Acad Sci USA 1992;89:10405–10409.
30. Keller GA, Warner TG, Steimer KS, Hallewell RA. Cu, Zn superoxide dismutase is a peroxisomal enzyme in human fibroblasts and hepatoma cells. Proc Natl Acad Sci USA 1991;88:7381–7385.
31. Slot JW, Geuze HJ, Freeman BA, Crapo JD. Intracellular localization of the copper-zinc and manganese superoxide dismutases in rat liver parenchymal cells. Lab Invest 1986;55:363–371.
32. Clyde BL, Chang LY, Auten RL, Ho YS, Crapo JD. Distribution of manganese superoxide dismutase mRNA in normal and hyperoxic rat lung. Am J Respir Cell Mol Biol 1993;8:530–537.
33. Gilks CB, Price K, Wright JL, Churg A. Antioxidant gene expression in rat lung after exposure to cigarette smoke. Am J Pathol 1998;152:269–278.
34. Coursin DB, Cihla HP, Sempf J, Oberley TD, Oberley LW. An immunohistochemical analysis of antioxidant and glutathione S-transferase enzyme levels in normal and neoplastic human lung. Histol Histopathol 1996;11:851–860.
35. Lakari E, Paakko P, Pietarinen-Runtti P, Kinnula VL. Manganese superoxide dismutase and catalase are coordinately expressed in the alveolar region in chronic interstitial pneumonias and granulomatous diseases of the lung. Am J Respir Crit Care Med 2000;161:615–621.
36. Pietarinen-Runtti P, Raivio KO, Saksela M, Asikainen TM, Kinnula VL. Antioxidant enzyme regulation and resistance to oxidants of human bronchial epithelial cells cultured under hyperoxic conditions. Am J Respir Cell Mol Biol 1998;19:286–292.
37. Folz RJ, Crapo JD. Extracellular superoxide dismutase (SOD3): tissue-specific expression, genomic characterization, and computer-assisted sequence analysis of the human EC SOD gene. Genomics 1994;22:162–171.
38. Folz RJ, Guan J, Seldin MF, Oury TD, Enghild JJ, Crapo JD. Mouse extracellular superoxide dismutase: primary structure, tissue-specific gene expression, chromosomal localization, and lung in situ hybridization. Am J Respir Cell Mol Biol 1997;17:393–403.
39. Ookawara T, Imazeki N, Matsubara O, Kizaki T, Oh-Ishi S, Nakao C, Sato Y, Ohno H. Tissue distribution of immunoreactive mouse extracellular superoxide dismutase. Am J Physiol 1998;275:C840–C847.
40. Oury TD, Chang LY, Marklund SL, Day BJ, Crapo JD. Immunocytochemical localization of extracellular superoxide dismutase in human lung. Lab Invest 1994;70:889–898.
41. Oury TD, Day BJ, Crapo JD. Extracellular superoxide dismutase in vessels and airways of humans and baboons. Free Radic Biol Med 1996;20:957–965.
42. Su WY, Folz R, Chen JS, Crapo JD, Chang LY. Extracellular superoxide dismutase mRNA expressions in the human lung by in situ hybridization. Am J Respir Cell Mol Biol 1997;16:162–170.
43. Karlsson K, Lindahl U, Marklund SL. Binding of human extracellular superoxide dismutase C to sulphated glycosaminoglycans. Biochem J 1988;256:29–33.
44. Turrens JF, Freeman BA, Levitt JG, Crapo JD. The effect of hyperoxia on superoxide production by lung submitochondrial particles. Arch Biochem Biophys 1982;217:401–410.
45. Crapo JD, Stamler JS. Signaling by nonreceptor surface-mediated redox-active biomolecules. J Clin Invest 1994;93:2304.
46. Asikainen TM, Raivio KO, Saksela M, Kinnula VL. Expression and developmental profile of antioxidant enzymes in human lung and liver. Am J Respir Cell Mol Biol 1998;19:942–949.
47. Dobashi K, Asayama K, Hayashibe H, Munim A, Kawaoi A, Morikawa M, Nakazawa S. Immunohistochemical study of copper-zinc and manganese superoxide dismutases in the lungs of human fetuses and newborn infants: developmental profile and alterations in hyaline membrane disease and bronchopulmonary dysplasia. Virchows Arch A Pathol Anat Histopathol 1993;423:177–184.
48. Strange RC, Cotton W, Fryer AA, Drew R, Bradwell AR, Marshall T, Collins MF, Bell J, Hume R. Studies on the expression of Cu, Zn superoxide dismutase in human tissues during development. Biochim Biophys Acta 1988;964:260–265.
49. Strange RC, Cotton W, Fryer AA, Jones P, Bell J, Hume R. Lipid peroxidation and expression of copper-zinc and manganese superoxide dismutase in lungs of premature infants with hyaline membrane disease and bronchopulmonary dysplasia. J Lab Clin Med 1990;116:666–673.
50. Asikainen TM, Heikkila P, Kaarteenaho-Wiik R, Kinnula VL, Raivio KO. Cell-specific expression of manganese superoxide dismutase protein in the lungs of patients with respiratory distress syndrome, chronic lung disease, or persistent pulmonary hypertension. Pediatr Pulmonol 2001;32:193–200.
51. Lakari E, Kaarteenaho-Wiik R, Soini Y, Kinnula V. The expression of extracellular superoxide dismutase in newborns with respiratory distress syndrome and bronchopulmonary dysplasia. Am J Respir Crit Care Med 2001;163:A726.
52. Carbone GM, St Clair DK, Xu YA, Rose JC. Expression of manganese superoxide dismutase in ovine kidney cortex during development. Pediatr Res 1994;35:41–44.
53. Clerch LB, Massaro D. Rat lung antioxidant enzymes: differences in perinatal gene expression and regulation. Am J Physiol 1992;263:L466–L470.
54. Frank L, Sosenko IR. Failure of premature rabbits to increase antioxidant enzymes during hyperoxic exposure: increased susceptibility to pulmonary oxygen toxicity compared with term rabbits. Pediatr Res 1991;29:292–296.
55. Frank L. Developmental aspects of experimental pulmonary oxygen toxicity. Free Radic Biol Med 1991;11:463–494.
56. Hass MA, Massaro D. Developmental regulation of rat lung Cu, Zn-superoxide dismutase. Biochem J 1987;246:697–703.
57. Hayashibe H, Asayama K, Dobashi K, Kato K. Prenatal development of antioxidant enzymes in rat lung, kidney, and heart: marked increase in immunoreactive superoxide dismutases, glutathione peroxidase, and catalase in the kidney. Pediatr Res 1990;27:472–475.
58. Walther FJ, Wade AB, Warburton D, Forman HJ. Ontogeny of antioxidant enzymes in the fetal lamb lung. Exp Lung Res 1991;17:39–45.
59. Yam J, Frank L, Roberts RJ. Oxygen toxicity: comparison of lung biochemical responses in neonatal and adult rats. Pediatr Res 1978;12:115–119.
60. Yuan HT, Bingle CD, Kelly FJ. Differential patterns of antioxidant enzyme mRNA expression in guinea pig lung and liver during development. Biochim Biophys Acta 1996;1305:163–171.
61. Clerch LB, Massaro D. Oxidation-reduction-sensitive binding of lung protein to rat catalase mRNA. J Biol Chem 1992;267:2853–2855.
62. Nozik-Grayck E, Dieterle CS, Piantadosi CA, Enghild JJ, Oury TD. Secretion of extracellular superoxide dismutase in neonatal lungs. Am J Physiol Lung Cell Mol Physiol 2000;279:L977–L984.
63. Zelko IN, Mariani TJ, Folz RJ. Superoxide dismutase multigene family: a comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med 2002;33:337–349.
64. Kim HT, Kim YH, Nam JW, Lee HJ, Rho HM, Jung G. Study of 5′-flanking region of human Cu/Zn superoxide dismutase. Biochem Biophys Res Commun 1994;201:1526–1533.
65. Yoo HY, Chang MS, Rho HM. Heavy metal-mediated activation of the rat Cu/Zn superoxide dismutase gene via a metal-responsive element. Mol Gen Genet 1999;262:310–313.
66. Hass MA, Massaro D. Regulation of the synthesis of superoxide dismutases in rat lungs during oxidant and hyperthermic stresses. J Biol Chem 1988;263:776–781.
67. Inoue N, Ramasamy S, Fukai T, Nerem RM, Harrison DG. Shear stress modulates expression of Cu/Zn superoxide dismutase in human aortic endothelial cells. Circ Res 1996;79:32–37.
68. Clerch LB, Massaro D. Tolerance of rats to hyperoxia: lung antioxidant enzyme gene expression. J Clin Invest 1993;91:499–508.
69. Ho YS, Dey MS, Crapo JD. Antioxidant enzyme expression in rat lungs during hyperoxia. Am J Physiol 1996;270:L810–L818.
70. Janssen YM, Marsh JP, Absher MP, Hemenway D, Vacek PM, Leslie KO, Borm PJ, Mossman BT. Expression of antioxidant enzymes in rat lungs after inhalation of asbestos or silica. J Biol Chem 1992;267:10625–10630.
71. Janssen YM, Van Houten B, Borm PJ, Mossman BT. Cell and tissue responses to oxidative damage. Lab Invest 1993;69:261–274.
72. Shull S, Heintz NH, Periasamy M, Manohar M, Janssen YM, Marsh JP, Mossman BT. Differential regulation of antioxidant enzymes in response to oxidants. J Biol Chem 1991;266:24398–24403.
73. Visner GA, Dougall WC, Wilson JM, Burr IA, Nick HS. Regulation of manganese superoxide dismutase by lipopolysaccharide, interleukin-1, and tumor necrosis factor: role in the acute inflammatory response. J Biol Chem 1990;265:2856–2864.
74. Kong XJ, Lee SL, Lanzillo JJ, Fanburg BL. Cu, Zn superoxide dismutase in vascular cells: changes during cell cycling and exposure to hyperoxia. Am J Physiol 1993;264:L365–L375.
75. Freeman BA, Mason RJ, Williams MC, Crapo JD. Antioxidant enzyme activity in alveolar type II cells after exposure of rats to hyperoxia. Exp Lung Res 1986;10:203–222.
76. Weller BL, Crapo JD, Slot J, Posthuma G, Plopper CG, Pinkerton KE. Site- and cell-specific alteration of lung copper/zinc and manganese superoxide dismutases by chronic ozone exposure. Am J Respir Cell Mol Biol 1997;17:552–560.
77. Wan XS, Devalaraja MN, St Clair DK. Molecular structure and organization of the human manganese superoxide dismutase gene. DNA Cell Biol 1994;13:1127–1136.
78. Yeh CC, Wan XS, St Clair DK. Transcriptional regulation of the 5′ proximal promoter of the human manganese superoxide dismutase gene. DNA Cell Biol 1998;17:921–930.
79. Zhang N. Characterization of the 5′ flanking region of the human MnSOD gene. Biochem Biophys Res Commun 1996;220:171–180.
80. Akashi M, Takagi S, Hachiya M. Anti-cancer agent OK432 induces manganese superoxide dismutase in human granulocytes. Int J Cancer 1996; 68:384–390.
81. Das KC, Lewis-Molock Y, White CW. Thiol modulation of TNF alpha and IL-1 induced MnSOD gene expression and activation of NF-kappa B. Mol Cell Biochem 1995;148:45–57.
82. Das KC, Lewis-Molock Y, White CW. Activation of NF-kappa B and elevation of MnSOD gene expression by thiol reducing agents in lung adenocarcinoma (A549) cells. Am J Physiol 1995;269:L588–L602.
83. Das KC, Lewis-Molock Y, White CW. Elevation of manganese superoxide dismutase gene expression by thioredoxin. Am J Respir Cell Mol Biol 1997;17:713–726.
84. Harris CA, Derbin KS, Hunte-McDonough B, Krauss MR, Chen KT, Smith DM, Epstein LB. Manganese superoxide dismutase is induced by IFN-gamma in multiple cell types: synergistic induction by IFN-gamma and tumor necrosis factor or IL-1. J Immunol 1991;147:149–154.
85. Rahman I, Clerch LB, Massaro D. Rat lung antioxidant enzyme induction by ozone. Am J Physiol 1991;260:L412–L418.
86. Visner GA, Block ER, Burr IM, Nick HS. Regulation of manganese superoxide dismutase in porcine pulmonary artery endothelial cells. Am J Physiol 1991;260:L444–L449.
87. Warner BB, Burhans MS, Clark JC, Wispe JR. Tumor necrosis factor-alpha increases Mn-SOD expression: protection against oxidant injury. Am J Physiol 1991;260:L296–L301.
88. Warner BB, Stuart L, Gebb S, Wispe JR. Redox regulation of manganese superoxide dismutase. Am J Physiol 1996;271:L150–L158.
89. White JE, Tsan MF. Induction of pulmonary Mn superoxide dismutase mRNA by interleukin-1. Am J Physiol 1994;266:L664–L671.
90. Wong GH, Goeddel DV. Induction of manganous superoxide dismutase by tumor necrosis factor: possible protective mechanism. Science 1988;242:941–944.
91. Jarvinen K, Pietarinen-Runtti P, Linnainmaa K, Raivio KO, Krejsa CM, Kavanagh T, Kinnula VL. Antioxidant defense mechanisms of human mesothelioma and lung adenocarcinoma cells. Am J Physiol Lung Cell Mol Physiol 2000;278:L696–L702.
92. Wong GH. Protective roles of cytokines against radiation: induction of mitochondrial MnSOD. Biochim Biophys Acta 1995;1271:205–209.
93. Chung DJ, Wright AE, Clerch LB. The 3′ untranslated region of manganese superoxide dismutase RNA contains a translational enhancer element. Biochemistry 1998;37:16298–16306.
94. Clerch LB, Massaro D, Berkovich A. Molecular mechanisms of antioxidant enzyme expression in lung during exposure to and recovery from hyperoxia. Am J Physiol 1998;274:L313–L319.
95. Knirsch L, Clerch LB. Tyrosine phosphorylation regulates manganese superoxide dismutase (MnSOD) RNA-binding protein activity and MnSOD protein expression. Biochemistry 2001;40:7890–7895.
96. Macmillan-Crow LA, Crow JP, Thompson JA. Peroxynitrite-mediated inactivation of manganese superoxide dismutase involves nitration and oxidation of critical tyrosine residues. Biochemistry 1998;37:1613–1622.
97. Macmillan-Crow LA, Cruthirds DL. Invited review: manganese superoxide dismutase in disease. Free Radic Res 2001;34:325–336.
98. Yamakura F, Taka H, Fujimura T, Murayama K. Inactivation of human manganese-superoxide dismutase by peroxynitrite is caused by exclusive nitration of tyrosine 34 to 3-nitrotyrosine. J Biol Chem 1998;273:14085–14089.
99. Briehl MM, Cotgreave IA, Powis G. Downregulation of the antioxidant defence during glucocorticoid mediated apoptosis. Cell Death Diff 1995;2:41–46.
100. Macmillan-Crow LA, Thompson JA. Tyrosine modifications and inactivation of active site manganese superoxide dismutase mutant (Y34F) by peroxynitrite. Arch Biochem Biophys 1999;366:82–88.
101. Forman HJ, Fisher AB. Antioxidant enzymes of rat granular pneumocytes: constitutive levels and effect of hyperoxia. Lab Invest 1981;45:1–6.
102. Vincent R, Chang LY, Slot JW, Crapo JD. Quantitative immunocytochemical analysis of Mn SOD in alveolar type II cells of the hyperoxic rat. Am J Physiol 1994;267:L475–L481.
103. Dirami G, Massaro D, Clerch LB. Regulation of lung manganese superoxide dismutase: species variation in response to lipopolysaccharide. Am J Physiol 1999;276:L705–L708.
104. Jornot L, Junod AF. Response of human endothelial cell antioxidant enzymes to hyperoxia. Am J Respir Cell Mol Biol 1992;6:107–115.
105. Russell WJ, Jackson RM. MnSOD protein content changes in hypoxic/hypoperfused lung tissue. Am J Respir Cell Mol Biol 1993;9:610–616.
106. Russell WJ, Matalon S, Jackson RM. Manganese superoxide dismutase expression in alveolar type II epithelial cells from nonventilated and hypoperfused lungs. Am J Respir Cell Mol Biol 1994;11:366–371.
107. Sjostrom K, Crapo JD. Structural and biochemical adaptive changes in rat lungs after exposure to hypoxia. Lab Invest 1983;48:68–79.
108. Jackson RM, Parish G, Ho YS. Effects of hypoxia on expression of superoxide dismutases in cultured ATII cells and lung fibroblasts. Am J Physiol 1996;271:L955–L962.
109. Ohman T, Parish G, Jackson RM. Hypoxic modulation of manganese superoxide dismutase promoter activity and gene expression in lung epithelial cells. Am J Respir Cell Mol Biol 1999;21:119–127.
110. Liu J, Simon LM, Phillips JR, Robin ED. Superoxide dismutase (SOD) activity in hypoxic mammalian systems. J Appl Physiol 1977;42:107–110.
111. Folz RJ, Peno-Green L, Crapo JD. Identification of a homozygous missense mutation (Arg to Gly) in the critical binding region of the human EC-SOD gene (SOD3) and its association with dramatically increased serum enzyme levels. Hum Mol Genet 1994;3:2251–2254.
112. Brady TC, Chang LY, Day BJ, Crapo JD. Extracellular superoxide dismutase is upregulated with inducible nitric oxide synthase after NF-kappa B activation. Am J Physiol 1997;273:L1002–L1006.
113. Marklund SL. Extracellular superoxide dismutase and other superoxide dismutase isoenzymes in tissues from nine mammalian species. Biochem J 1984;222:649–655.
114. Stralin P, Marklund SL. Multiple cytokines regulate the expression of extracellular superoxide dismutase in human vascular smooth muscle cells. Atherosclerosis 2000;151:433–441.
115. Loenders B, Van Mechelen E, Nicolai S, Buyssens N, Van Osselaer N, Jorens PG, Willems J, Herman AG, Slegers H. Localization of extracellular superoxide dismutase in rat lung: neutrophils and macrophages as carriers of the enzyme. Free Radic Biol Med 1998;24:1097–1106.
116. Oury TD, Schaefer LM, Fattman CL, Choi A, Weck KE, Watkins SC. Depletion of pulmonary EC-SOD after exposure to hyperoxia. Am J Physiol Lung Cell Mol Physiol 2002;283:L777–L784.
117. Fattman CL, Chu CT, Kulich SM, Enghild JJ, Oury TD. Altered expression of extracellular superoxide dismutase in mouse lung after bleomycin treatment. Free Radic Biol Med 2001;31:1198–1207.
118. Bowler RP, Nicks M, Warnick K, Crapo JD. Role of extracellular superoxide dismutase in bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2002;282:L719–L726.
119. Enghild JJ, Thogersen IB, Oury TD, Valnickova Z, Hojrup P, Crapo JD. The heparin-binding domain of extracellular superoxide dismutase is proteolytically processed intracellularly during biosynthesis. J Biol Chem 1999;274:14818–14822.
120. Oury TD, Crapo JD, Valnickova Z, Enghild JJ. Human extracellular superoxide dismutase is a tetramer composed of two disulphide-linked dimers: a simplified, high-yield purification of extracellular superoxide dismutase. Biochem J 1996;317:51–57.
121. Bowler RP, Nicks M, Olsen DA, Thogersen IB, Valnickova Z, Hojrup P, Franzusoff A, Enghild JJ, Crapo JD. Furin proteolytically processes the heparin-binding region of extracellular superoxide dismutase. J Biol Chem 2002;277:16505–16511.
122. Tsan MF, White JE, Santana TA, Lee CY. Tracheal insufflation of tumor necrosis factor protects rats against oxygen toxicity. J Appl Physiol 1990;68:1211–1219.
123. White CW, Ghezzi P. Protection against pulmonary oxygen toxicity by interleukin-1 and tumor necrosis factor: role of antioxidant enzymes and effect of cyclooxygenase inhibitors. Biotherapy 1989;1:361–367.
124. Crapo JD, Sjostrom K, Drew RT. Tolerance and cross-tolerance using NO2 and O2. I: Toxicology and biochemistry. J Appl Physiol 1978;44:364–369.
125. Crapo JD, Barry BE, Foscue HA, Shelburne J. Structural and biochemical changes in rat lungs occurring during exposures to lethal and adaptive doses of oxygen. Am Rev Respir Dis 1980;122:123–143.
126. Crapo JD. Morphologic changes in pulmonary oxygen toxicity. Annu Rev Physiol 1986;48:721–731.
127. Kimball RE, Reddy K, Peirce TH, Schwartz LW, Mustafa MG, Cross CE. Oxygen toxicity: augmentation of antioxidant defense mechanisms in rat lung. Am J Physiol 1976;230:1425–1431.
128. Kinnula VL, Pietarinen P, Aalto K, Virtanen I, Raivio KO. Mitochondrial superoxide dismutase induction does not protect epithelial cells during oxidant exposure in vitro. Am J Physiol 1995;268:L71–L77.
129. Baker RR, Holm BA, Panus PC, Matalon S. Development of O2 tolerance in rabbits with no increase in antioxidant enzymes. J Appl Physiol 1989;66:1679–1684.
130. Sagara Y, Dargusch R, Chambers D, Davis J, Schubert D, Maher P. Cellular mechanisms of resistance to chronic oxidative stress. Free Radic Biol Med 1998;24:1375–1389.
131. Forsberg L, de Faire U, Morgenstern R. Oxidative stress, human genetic variation, and disease. Arch Biochem Biophys 2001;389:84–93.
132. Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O'Regan JP, Deng HX. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993;362:59–62.
133. de Haan JB, Cristiano F, Iannello R, Bladier C, Kelner MJ, Kola I. Elevation in the ratio of Cu/Zn-superoxide dismutase to glutathione peroxidase activity induces features of cellular senescence and this effect is mediated by hydrogen peroxide. Hum Mol Genet 1996;5:283–292.
134. Amstad P, Peskin A, Shah G, Mirault ME, Moret R, Zbinden I, Cerutti P. The balance between Cu, Zn-superoxide dismutase and catalase affects the sensitivity of mouse epidermal cells to oxidative stress. Biochemistry 1991;30:9305–9313.
135. Liochev SI, Fridovich I. The role of O2· in the production of HO : in vitro and in vivo. Free Radic Biol Med 1994;16:29–33.
136. Gardner R, Salvador A, Moradas-Ferreira P. Why does SOD overexpression sometimes enhance, sometimes decrease, hydrogen peroxide production? A minimalist explanation. Free Radic Biol Med 2002;32:1351–1357.
137. Ho YS. Transgenic models for the study of lung biology and disease. Am J Physiol 1994;266:L319–L353.
138. Tsan MF. Superoxide dismutase and pulmonary oxygen toxicity: lessons from transgenic and knockout mice. Int J Mol Med 2001;7:13–19.
139. de Vos S, Epstein CJ, Carlson E, Cho SK, Koeffler HP. Transgenic mice overexpressing human copper/zinc-superoxide dismutase (Cu/Zn SOD) are not resistant to endotoxic shock. Biochem Biophys Res Commun 1995;208:523–531.
140. Larsen GL, White CW, Takeda K, Loader JE, Nguyen DD, Joetham A, Groner Y, Gelfand EW. Mice that overexpress Cu/Zn superoxide dismutase are resistant to allergen-induced changes in airway control. Am J Physiol Lung Cell Mol Physiol 2000;279:L350–L359.
141. White CW, Avraham KB, Shanley PF, Groner Y. Transgenic mice with expression of elevated levels of copper-zinc superoxide dismutase in the lungs are resistant to pulmonary oxygen toxicity. J Clin Invest 1991;87:2162–2168.
142. Ho YS, Gargano M, Cao J, Bronson RT, Heimler I, Hutz RJ. Reduced fertility in female mice lacking copper-zinc superoxide dismutase. J Biol Chem 1998;273:7765–7769.
143. Huang TT, Yasunami M, Carlson EJ, Gillespie AM, Reaume AG, Hoffman EK, Chan PH, Scott RW, Epstein CJ. Superoxide-mediated cytotoxicity in superoxide dismutase-deficient fetal fibroblasts. Arch Biochem Biophys 1997;344:424–432.
144. Lindau-Shepard B, Shaffer JB, Del Vecchio PJ. Overexpression of manganous superoxide dismutase (MnSOD) in pulmonary endothelial cells confers resistance to hyperoxia. J Cell Physiol 1994;161:237–242.
145. St Clair DK, Oberley TD, Ho YS. Overproduction of human Mn-superoxide dismutase modulates paraquat-mediated toxicity in mammalian cells. FEBS Lett 1991;293:199–203.
146. Ilizarov AM, Koo HC, Kazzaz JA, Mantell LL, Li Y, Bhapat R, Pollack S, Horowitz S, Davis JM. Overexpression of manganese superoxide dismutase protects lung epithelial cells against oxidant injury. Am J Respir Cell Mol Biol 2001;24:436–441.
147. St Clair DK, Jordan JA, Wan XS, Gairola CG. Protective role of manganese superoxide dismutase against cigarette smoke-induced cytotoxicity. J Toxicol Environ Health 1994;43:239–249.
148. Hirose K, Longo DL, Oppenheim JJ, Matsushima K. Overexpression of mitochondrial manganese superoxide dismutase promotes the survival of tumor cells exposed to interleukin-1, tumor necrosis factor, selected anticancer drugs, and ionizing radiation. FASEB J 1993;7:361–368.
149. Sun J, Chen Y, Li M, Ge Z. Role of antioxidant enzymes on ionizing radiation resistance. Free Radic Biol Med 1998;24:586–593.
150. Mossman BT, Surinrut P, Brinton BT, Marsh JP, Heintz NH, Lindau-Shepard B, Shaffer JB. Transfection of a manganese-containing superoxide dismutase gene into hamster tracheal epithelial cells ameliorates asbestos-mediated cytotoxicity. Free Radic Biol Med 1996;21:125–131.
151. Wong GH, Elwell JH, Oberley LW, Goeddel DV. Manganous superoxide dismutase is essential for cellular resistance to cytotoxicity of tumor necrosis factor. Cell 1989;58:923–931.
152. Hohmeier HE, Thigpen A, Tran VV, Davis R, Newgard CB. Stable expression of manganese superoxide dismutase (MnSOD) in insulinoma cells prevents IL-1beta-induced cytotoxicity and reduces nitric oxide production. J Clin Invest 1998;101:1811–1820.
153. Zwacka RM, Dudus L, Epperly MW, Greenberger JS, Engelhardt JF. Redox gene therapy protects human IB-3 lung epithelial cells against ionizing radiation-induced apoptosis. Hum Gene Ther 1998;9:1381–1386.
154. Kokoszka JE, Coskun P, Esposito LA, Wallace DC. Increased mitochondrial oxidative stress in the Sod2 (+/−) mouse results in the age-related decline of mitochondrial function culminating in increased apoptosis. Proc Natl Acad Sci USA 2001;98:2278–2283.
155. Zhao Y, Kiningham KK, Lin SM, St Clair DK. Overexpression of MnSOD protects murine fibrosarcoma cells (FSa-II) from apoptosis and promotes a differentiation program upon treatment with 5-azacytidine: involvement of MAPK and NFkappaB pathways. Antioxid Redox Signal 2001;3:375–386.
156. Motoori S, Majima HJ, Ebara M, Kato H, Hirai F, Kakinuma S, Yamaguchi C, Ozawa T, Nagano T, Tsujii H, et al. Overexpression of mitochondrial manganese superoxide dismutase protects against radiation-induced cell death in the human hepatocellular carcinoma cell line HLE. Cancer Res 2001;61:5382–5388.
157. Oberley TD, Oberley LW. Antioxidant enzyme levels in cancer. Histol Histopathol 1997;12:525–535.
158. Oberley TD. Oxidative damage and cancer. Am J Pathol 2002;160:403–408.
159. Kiningham KK, Xu Y, Daosukho C, Popova B, St Clair DK. Nuclear factor kappaB-dependent mechanisms coordinate the synergistic effect of PMA and cytokines on the induction of superoxide dismutase 2. Biochem J 2001;353:147–156.
160. Zhao Y, Xue Y, Oberley TD, Kiningham KK, Lin SM, Yen HC, Majima H, Hines J, St Clair D. Overexpression of manganese superoxide dismutase suppresses tumor formation by modulation of activator protein-1 signaling in a multistage skin carcinogenesis model. Cancer Res 2001;61:6082–6088.
161. Ho YS, Magnenat JL, Gargano M, Cao J. The nature of antioxidant defense mechanisms: a lesson from transgenic studies. Environ Health Perspect 1998;106:1219–1228.
162. Kizaki M, Sakashita A, Karmakar A, Lin CW, Koeffler HP. Regulation of manganese superoxide dismutase and other antioxidant genes in normal and leukemic hematopoietic cells and their relationship to cytotoxicity by tumor necrosis factor. Blood 1993;82:1142–1150.
163. Raineri I, Huang TT, Epstein CJ, Epstein LB. Antisense manganese superoxide dismutase mRNA inhibits the antiviral action of interferon-gamma and interferon-alpha. J Interferon Cytokine Res 1996;16:61–68.
164. Wispe JR, Warner BB, Clark JC, Dey CR, Neuman J, Glasser SW, Crapo JD, Chang LY, Whitsett JA. Human Mn-superoxide dismutase in pulmonary epithelial cells of transgenic mice confers protection from oxygen injury. J Biol Chem 1992;267:23937–23941.
165. Ho YS, Vincent R, Dey MS, Slot JW, Crapo JD. Transgenic models for the study of lung antioxidant defense: enhanced manganese-containing superoxide dismutase activity gives partial protection to B6C3 hybrid mice exposed to hyperoxia. Am J Respir Cell Mol Biol 1998;18:538–547.
166. Copin JC, Gasche Y, Chan PH. Overexpression of copper/zinc superoxide dismutase does not prevent neonatal lethality in mutant mice that lack manganese superoxide dismutase. Free Radic Biol Med 2000;28:1571–1576.
167. Lebovitz RM, Zhang H, Vogel H, Cartwright J Jr, Dionne L, Lu N, Huang S, Matzuk MM. Neurodegeneration, myocardial injury, and perinatal death in mitochondrial superoxide dismutase-deficient mice. Proc Natl Acad Sci USA 1996;93:9782–9787.
168. Li Y, Huang TT, Carlson EJ, Melov S, Ursell PC, Olson JL, Noble LJ, Yoshimura MP, Berger C, Chan PH. Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat Genet 1995;11:376–381.
169. Asikainen TM, Huang TT, Taskinen E, Levonen AL, Carlson E, Lapatto R, Epstein CJ, Raivio KO. Increased sensitivity of homozygous Sod2 mutant mice to oxygen toxicity. Free Radic Biol Med 2002;32:175–186.
170. Jackson RM, Helton ES, Viera L, Ohman T. Survival, lung injury, and lung protein nitration in heterozygous MnSOD knockout mice in hyperoxia. Exp Lung Res 1999;25:631–646.
171. Tsan MF, White JE, Caska B, Epstein CJ, Lee CY. Susceptibility of heterozygous MnSOD gene-knockout mice to oxygen toxicity. Am J Respir Cell Mol Biol 1998;19:114–120.
172. Tsan MF, Cao X, White JE, Sacco J, Lee CY. Pertussis toxin-induced lung edema: role of manganese superoxide dismutase and protein kinase C. Am J Respir Cell Mol Biol 1999;20:465–473.
173. Williams MD, Van Remmen H, Conrad CC, Huang TT, Epstein CJ, Richardson A. Increased oxidative damage is correlated to altered mitochondrial function in heterozygous manganese superoxide dismutase knockout mice. J Biol Chem 1998;273:28510–28515.
174. Van Remmen H, Williams MD, Guo Z, Estlack L, Yang H, Carlson EJ, Epstein CJ, Huang TT, Richardson A. Knockout mice heterozygous for Sod2 show alterations in cardiac mitochondrial function and apoptosis. Am J Physiol Heart Circ Physiol 2001;281:H1422–H1432.
175. Velsor LW, Day B. Increased lung tumor formation in aged manganese superoxide dismutase (SOD2) heterozygous knockout mice [abstract]. Am J Respir Crit Care Med 2001;163:A296.
176. Day BJ, Melov S, Wallace D, Crapo DJ. Development of an interstitial fibrosis phenotype in aged SOD2 heterozygous ko mice [abstract]. Am J Respir Crit Care Med 1999;159:A288.
177. Folz RJ, Abushamaa AM, Suliman HB. Extracellular superoxide dismutase in the airways of transgenic mice reduces inflammation and attenuates lung toxicity following hyperoxia. J Clin Invest 1999;103:1055–1066.
178. Ghio AJ, Suliman HB, Carter JD, Abushamaa AM, Folz RJ. Overexpression of extracellular superoxide dismutase decreases lung injury after exposure to oil fly ash. Am J Physiol Lung Cell Mol Physiol 2002;283:L211–L218.
179. Ahmed MN, Suliman HB, Folz RJ, Nozik-Grayck E, Golson ML, Mason SN, Auten RL. Extracellular superoxide dismutase protects lung development in hyperoxia-exposed newborn mice. Am J Respir Crit Care Med 2003;167:400–405.
180. Bowler RP, Crapo JD. Oxidative stress in allergic respiratory diseases. J Allergy Clin Immunol 2002;110:349–356.
181. Oury TD, Ho YS, Piantadosi CA, Crapo JD. Extracellular superoxide dismutase, nitric oxide, and central nervous system O2 toxicity. Proc Natl Acad Sci USA 1992;89:9715–9719.
182. Carlsson LM, Jonsson J, Edlund T, Marklund SL. Mice lacking extracellular superoxide dismutase are more sensitive to hyperoxia. Proc Natl Acad Sci USA 1995;92:6264–6268.
183. Crapo JD, DeLong DM, Sjostrom K, Hasler GR, Drew RT. The failure of aerosolized superoxide dismutase to modify pulmonary oxygen toxicity. Am Rev Respir Dis 1977;115:1027–1033.
184. Padmanabhan RV, Gudapaty R, Liener IE, Schwartz BA, Hoidal JR. Protection against pulmonary oxygen toxicity in rats by the intratracheal administration of liposome-encapsulated superoxide dismutase or catalase. Am Rev Respir Dis 1985;132:164–167.
185. Turrens JF, Crapo JD, Freeman BA. Protection against oxygen toxicity by intravenous injection of liposome-entrapped catalase and superoxide dismutase. J Clin Invest 1984;73:87–95.
186. Freeman BA, Turrens JF, Mirza Z, Crapo JD, Young SL. Modulation of oxidant lung injury by using liposome-entrapped superoxide dismutase and catalase. Fed Proc 1985;44:2591–2595.
187. Tamagawa K, Taooka Y, Maeda A, Hiyama K, Ishioka S, Yamakido M. Inhibitory effects of a lecithinized superoxide dismutase on bleomycin-induced pulmonary fibrosis in mice. Am J Respir Crit Care Med 2000;161:1279–1284.
188. Buckley BJ, Tanswell AK, Freeman BA. Liposome-mediated augmentation of catalase in alveolar type II cells protects against H2O2 injury. J Appl Physiol 1987;63:359–367.
189. Beckman JS, Minor RL Jr, White CW, Repine JE, Rosen GM, Freeman BA. Superoxide dismutase and catalase conjugated to polyethylene glycol increases endothelial enzyme activity and oxidant resistance. J Biol Chem 1988;263:6884–6892.
190. Mossman BT, Marsh JP, Sesko A, Hill S, Shatos MA, Doherty J, Petruska J, Adler KB, Hemenway D, Mickey R. Inhibition of lung injury, inflammation, and interstitial pulmonary fibrosis by polyethylene glycol-conjugated catalase in a rapid inhalation model of asbestosis. Am Rev Respir Dis 1990;141:1266–1271.
191. White CW, Jackson JH, Abuchowski A, Kazo GM, Mimmack RF, Berger EM, Freeman BA, McCord JM, Repine JE. Polyethylene glycol-attached antioxidant enzymes decrease pulmonary oxygen toxicity in rats. J Appl Physiol 1989;66:584–590.
192. Assa'ad AH, Ballard ET, Sebastian KD, Loven DP, Boivin GP, Lierl MB. Effect of superoxide dismutase on a rabbit model of chronic allergic asthma. Ann Allergy Asthma Immunol 1998;80:215–224.
193. Simonson SG, Welty-Wolf KE, Huang YC, Taylor DE, Kantrow SP, Carraway MS, Crapo JD, Piantadosi CA. Aerosolized manganese SOD decreases hyperoxic pulmonary injury in primates. I: physiology and biochemistry. J Appl Physiol 1997;83:550–558.
194. Welty-Wolf KE, Simonson SG, Huang YC, Kantrow SP, Carraway MS, Chang LY, Crapo JD, Piantadosi CA. Aerosolized manganese SOD decreases hyperoxic pulmonary injury in primates. II: morphometric analysis. J Appl Physiol 1997;83:559–568.
195. Baudry M, Etienne S, Bruce A, Palucki M, Jacobsen E, Malfroy B. Salen-manganese complexes are superoxide dismutase-mimics. Biochem Biophys Res Commun 1993;192:964–968.
196. Salvemini D, Mazzon E, Dugo L, Riley DP, Serraino I, Caputi AP, Cuzzocrea S. Pharmacological manipulation of the inflammatory cascade by the superoxide dismutase mimetic, M40403. Br J Pharmacol 2001;132:815–827.
197. Szabo C, Day BJ, Salzman AL. Evaluation of the relative contribution of nitric oxide and peroxynitrite to the suppression of mitochondrial respiration in immunostimulated macrophages using a manganese mesoporphyrin superoxide dismutase mimetic and peroxynitrite scavenger. FEBS Lett 1996;381:82–86.
198. Hahn SM, Sullivan FJ, DeLuca AM, Bacher JD, Liebmann J, Krishna MC, Coffin D, Mitchell JB. Hemodynamic effect of the nitroxide superoxide dismutase mimics. Free Radic Biol Med 1999;27:529–535.
199. Krishna MC, Russo A, Mitchell JB, Goldstein S, Dafni H, Samuni A. Do nitroxide antioxidants act as scavengers of O2· or as SOD mimics? J Biol Chem 1996;271:26026–26031.
200. Zhang R, Goldstein S, Samuni A. Kinetics of superoxide-induced exchange among nitroxide antioxidants and their oxidized and reduced forms. Free Radic Biol Med 1999;26:1245–1252.
201. Baker K, Marcus CB, Huffman K, Kruk H, Malfroy B, Doctrow SR. Synthetic combined superoxide dismutase/catalase mimetics are protective as a delayed treatment in a rat stroke model: a key role for reactive oxygen species in ischemic brain injury. J Pharmacol Exp Ther 1998;284:215–221.
202. Gonzalez PK, Zhuang J, Doctrow SR, Malfroy B, Benson PF, Menconi MJ, Fink MP. EUK-8, a synthetic superoxide dismutase and catalase mimetic, ameliorates acute lung injury in endotoxemic swine. J Pharmacol Exp Ther 1995;275:798–806.
203. Jung C, Rong Y, Doctrow S, Baudry M, Malfroy B, Xu Z. Synthetic superoxide dismutase/catalase mimetics reduce oxidative stress and prolong survival in a mouse amyotrophic lateral sclerosis model. Neurosci Lett 2001;304:157–160.
204. Melov S, Ravenscroft J, Malik S, Gill MS, Walker DW, Clayton PE, Wallace DC, Malfroy B, Doctrow SR, Lithgow GJ. Extension of life-span with superoxide dismutase/catalase mimetics. Science 2000;289:1567–1569.
205. Pong K, Doctrow SR, Huffman K, Adinolfi CA, Baudry M. Attenuation of staurosporine-induced apoptosis, oxidative stress, and mitochondrial dysfunction by synthetic superoxide dismutase and catalase mimetics, in cultured cortical neurons. Exp Neurol 2001;171:84–97.
206. Rong Y, Doctrow SR, Tocco G, Baudry M. EUK-134, a synthetic superoxide dismutase and catalase mimetic, prevents oxidative stress and attenuates kainate-induced neuropathology. Proc Natl Acad Sci USA 1999;96:9897–9902.
207. Melov S, Doctrow SR, Schneider JA, Haberson J, Patel M, Coskun PE, Huffman K, Wallace DC, Malfroy B. Lifespan extension and rescue of spongiform encephalopathy in superoxide dismutase 2 nullizygous mice treated with superoxide dismutase-catalase mimetics. J Neurosci 2001;21:8348–8353.
208. Bianca RV, Wayman NS, McDonald MC, Pinto A, Shape MA, Chatterjee PK, Thiemermann C. Superoxide dismutase mimetic with catalase activity, EUK-134, attenuates the multiple organ injury and dysfunction caused by endotoxin in the rat. Med Sci Monit 2002;8:BR1–BR7.
209. Gianello P, Saliez A, Bufkens X, Pettinger R, Misseleyn D, Hori S, Malfroy B. EUK-134, a synthetic superoxide dismutase and catalase mimetic, protects rat kidneys from ischemia-reperfusion-induced damage. Transplantation 1996;62:1664–1666.
210. Cuzzocrea S, Mazzon E, Dugo L, Caputi AP, Riley DP, Salvemini D. Protective effects of M40403, a superoxide dismutase mimetic, in a rodent model of colitis. Eur J Pharmacol 2001;432:79–89.
211. Day BJ, Shawen S, Liochev SI, Crapo JD. A metalloporphyrin superoxide dismutase mimetic protects against paraquat-induced endothelial cell injury, in vitro. J Pharmacol Exp Ther 1995;275:1227–1232.
212. Day BJ, Fridovich I, Crapo JD. Manganic porphyrins possess catalase activity and protect endothelial cells against hydrogen peroxide-mediated injury. Arch Biochem Biophys 1997;347:256–262.
213. Day BJ, Crapo JD. A metalloporphyrin superoxide dismutase mimetic protects against paraquat-induced lung injury in vivo. Toxicol Appl Pharmacol 1996;140:94–100.
214. Cuzzocrea S, Zingarelli B, Costantino G, Caputi AP. Beneficial effects of Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP), a superoxide dismutase mimetic, in carrageenan-induced pleurisy. Free Radic Biol Med 1999;26:25–33.
215. Oury TD, Thakker K, Menache M, Chang LY, Crapo JD, Day BJ. Attenuation of bleomycin-induced pulmonary fibrosis by a catalytic antioxidant metalloporphyrin. Am J Respir Cell Mol Biol 2001;25:164–169.
216. Chang LY, Crapo JD. Inhibition of airway inflammation and hyperreactivity by an antioxidant mimetic. Free Radic Biol Med 2002;33:379–386.
217. Chang LY, Subramaniam M, Yoder BA, Day BJ, Ellison MC, Sunday ME, Crapo JD. A catalytic antioxidant attenuates alveolar structural remodeling in bronchopulmonary dysplasia. Am J Respir Crit Care Med 2003;167:57–64.
218. Smith KR, Uyeminami DL, Kodavanti UP, Crapo JD, Chang LY, Pinkerton KE. Inhibition of tobacco smoke-induced lung inflammation by a catalytic antioxidant. Free Radic Biol Med 2002;33:1106–1114.
219. Chang L, Crapo JD. Inhibition of allergen induced airway inflammation by a mimetic of superoxide dismutase [abstract]. Am J Respir Crit Care Med 2000;161:A738.
220. Chang LY, Crapo JD. Inhibition of airway inflammation and hyperreactivity by an antioxidant mimetic. Free Radic Biol Med 2002;33:379–386.
221. Malassagne B, Ferret PJ, Hammoud R, Tulliez M, Bedda S, Trebeden H, Jaffray P, Calmus Y, Weill B, Batteux F. The superoxide dismutase mimetic MnTBAP prevents Fas-induced acute liver failure in the mouse. Gastroenterology 2001;121:1451–1459.
222. Cuzzocrea S, Costantino G, Mazzon E, De Sarro A, Caputi AP. Beneficial effects of Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP), a superoxide dismutase mimetic, in zymosan-induced shock. Br J Pharmacol 1999;128:1241–1251.
223. Melov S, Schneider JA, Day BJ, Hinerfeld D, Coskun P, Mirra SS, Crapo JD, Wallace DC. A novel neurological phenotype in mice lacking mitochondrial manganese superoxide dismutase. Nat Genet 1998;18:159–163.
224. Niwa K, Carlson GA, Iadecola C. Exogenous A beta1–40 reproduces cerebrovascular alterations resulting from amyloid precursor protein overexpression in mice. J Cereb Blood Flow Metab 2000;20:1659–1668.
225. Niwa K, Porter VA, Kazama K, Cornfield D, Carlson GA, Iadecola C. A beta-peptides enhance vasoconstriction in cerebral circulation. Am J Physiol Heart Circ Physiol 2001;281:H2417–H2424.
226. Corradi M, Montuschi P, Donnelly LE, Pesci A, Kharitonov SA, Barnes PJ. Increased nitrosothiols in exhaled breath condensate in inflammatory airway diseases. Am J Respir Crit Care Med 2001;163:854–858.
227. Crapo JD, Day BJ. Modulation of nitric oxide responses in asthma by extracellular antioxidants. J Allergy Clin Immunol 1999;104:743–746.
228. Hanazawa T, Kharitonov SA, Barnes PJ. Increased nitrotyrosine in exhaled breath condensate of patients with asthma. Am J Respir Crit Care Med 2000;162:1273–1276.
229. Horvath I, Donnelly LE, Kiss A, Kharitonov SA, Lim S, Fan CK, Barnes PJ. Combined use of exhaled hydrogen peroxide and nitric oxide in monitoring asthma. Am J Respir Crit Care Med 1998;158:1042–1046.
230. Kharitonov SA, Barnes PJ. Exhaled markers of pulmonary disease. Am J Respir Crit Care Med 2001;163:1693–1722.
231. Barnes PJ, Liew FY. Nitric oxide and asthmatic inflammation. Immunol Today 1995;16:128–130.
232. Ichinose M, Sugiura H, Yamagata S, Koarai A, Shirato K. Increase in reactive nitrogen species production in chronic obstructive pulmonary disease airways. Am J Respir Crit Care Med 2000;162:701–706.
233. Paredi P, Kharitonov SA, Leak D, Ward S, Cramer D, Barnes PJ. Exhaled ethane, a marker of lipid peroxidation, is elevated in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2000;162:369–373.
234. Saetta M, Turato G, Maestrelli P, Mapp CE, Fabbri LM. Cellular and structural bases of chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2001;163:1304–1309.
235. Church DF, Pryor WA. Free-radical chemistry of cigarette smoke and its toxicological implications. Environ Health Perspect 1985;64:111–126.
236. Katzenstein AL, Myers JL. Idiopathic pulmonary fibrosis: clinical relevance of pathologic classification. Am J Respir Crit Care Med 1998;157:1301–1315.
237. American Thoracic Society (ATS) and European Respiratory Society (ERS). American Thoracic Society/European Respiratory Society international multidisciplinary consensus classification of the idiopathic interstitial pneumonias [abstract]. Am J Respir Crit Care Med 2002;165:A277.
238. Gross TJ, Hunninghake GW. Idiopathic pulmonary fibrosis. N Engl J Med 2001;345:517–525.
239. Borok Z, Buhl R, Grimes GJ, Bokser AD, Hubbard RC, Holroyd KJ, Roum JH, Czerski DB, Cantin AM, Crystal RG. Effect of glutathione aerosol on oxidant-antioxidant imbalance in idiopathic pulmonary fibrosis. Lancet 1991;338:215–216.
240. Cantin AM, North SL, Fells GA, Hubbard RC, Crystal RG. Oxidant-mediated epithelial cell injury in idiopathic pulmonary fibrosis. J Clin Invest 1987;79:1665–1673.
241. Nemery B, Bast A, Behr J, Borm PJ, Bourke SJ, Camus PH, De Vuyst P, Jansen HM, Kinnula VL, Lison D, et al. Interstitial lung disease induced by exogenous agents: factors governing susceptibility. Eur Respir J Suppl 2001;32:30s–42s.
242. Schaberg T, Rau M, Stephan H, Lode H. Increased number of alveolar macrophages expressing surface molecules of the CD11/CD18 family in sarcoidosis and idiopathic pulmonary fibrosis is related to the production of superoxide anions by these cells. Am Rev Respir Dis 1993;147:1507–1513.
243. Sherson D, Nielsen H, Frederiksen J, Milman N, Struve-Christensen E, Petersen BN. Superoxide anion release from blood monocytes and alveolar macrophages in patients with diffuse lung fibrosis. APMIS 1992;100:408–414.
244. Strausz J, Muller-Quernheim J, Steppling H, Ferlinz R. Oxygen radical production by alveolar inflammatory cells in idiopathic pulmonary fibrosis. Am Rev Respir Dis 1990;141:124–128.
245. Wallaert B, Lassalle P, Fortin F, Aerts C, Bart F, Fournier E, Voisin C. Superoxide anion generation by alveolar inflammatory cells in simple pneumoconiosis and in progressive massive fibrosis of nonsmoking coal workers. Am Rev Respir Dis 1990;141:129–133.
246. Lakari E, Soini Y, Saily M, Koistinen P, Paakko P, Kinnula VL. Inducible nitric oxide synthase, but not xanthine oxidase, is highly expressed in interstitial pneumonias and granulomatous diseases of human lung. Am J Clin Pathol 2002;117:132–142.
247. Saleh D, Barnes PJ, Giaid A. Increased production of the potent oxidant peroxynitrite in the lungs of patients with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 1997;155:1763–1769.
248. Cassatella MA, Berton G, Agostini C, Zambello R, Trentin L, Cipriani A, Semenzato G. Generation of superoxide anion by alveolar macrophages in sarcoidosis: evidence for the activation of the oxygen metabolism in patients with high-intensity alveolitis. Immunology 1989;66:451–458.
249. Behr J, Degenkolb B, Beinert T, Krombach F, Vogelmeier C. Pulmonary glutathione levels in acute episodes of Farmer's lung. Am J Respir Crit Care Med 2000;161:1968–1971.
250. Kamp DW, Graceffa P, Pryor WA, Weitzman SA. The role of free radicals in asbestos-induced diseases. Free Radic Biol Med 1992;12:293–315.
251. Kinnula VL. Oxidant and antioxidant mechanisms of lung disease caused by asbestos fibres. Eur Respir J 1999;14:706–716.
252. Janssen YM, Marsh JP, Driscoll KE, Borm PJ, Oberdorster G, Mossman BT. Increased expression of manganese-containing superoxide dismutase in rat lungs after inhalation of inflammatory and fibrogenic minerals. Free Radic Biol Med 1994;16:315–322.
253. Janssen YM, Driscoll KE, Timblin CR, Hassenbein D, Mossman BT. Modulation of mitochondrial gene expression in pulmonary epithelial cells exposed to oxidants. Environ Health Perspect 1998;106:1191–1195.
254. Mossman BT, Marsh JP, Shatos MA. Alteration of superoxide dismutase activity in tracheal epithelial cells by asbestos and inhibition of cytotoxicity by antioxidants. Lab Invest 1986;54:204–212.
255. Haddad IY, Ischiropoulos H, Holm BA, Beckman JS, Baker JR, Matalon S. Mechanisms of peroxynitrite-induced injury to pulmonary surfactants. Am J Physiol 1993;265:L555–L564.
256. Bellocq A, Azoulay E, Marullo S, Flahault A, Fouqueray B, Philippe C, Cadranel J, Baud L. Reactive oxygen and nitrogen intermediates increase transforming growth factor-beta1 release from human epithelial alveolar cells through two different mechanisms. Am J Respir Cell Mol Biol 1999;21:128–136.
257. Khalil N. TGF-beta: from latent to active. Microbes Infect 1999;1:1255–1263.
258. Hecht SS. Tobacco smoke carcinogens and lung cancer. J Natl Cancer Inst 1999;91:1194–1210.
259. Pryor WA. Cigarette smoke radicals and the role of free radicals in chemical carcinogenicity. Environ Health Perspect 1997;105:875–882.
260. Wink DA, Vodovotz Y, Laval J, Laval F, Dewhirst MW, Mitchell JB. The multifaceted roles of nitric oxide in cancer. Carcinogenesis 1998;19:711–721.
261. Mossman BT, Gee JB. Asbestos-related diseases. N Engl J Med 1989;320:1721–1730.
262. Chuang YY, Chen Y, Gadisetti, Chandramouli VR, Cook JA, Coffin D, Tsai MH, DeGraff W, Yan H, Zhao S, et al. Gene expression after treatment with hydrogen peroxide, menadione, or t-butyl hydroperoxide in breast cancer cells. Cancer Res 2002;62:6246–6254.
263. Granville DJ, Carthy CM, Hunt DW, McManus BM. Apoptosis: molecular aspects of cell death and disease. Lab Invest 1998;78:893–913.
264. Green DR, Reed JC. Mitochondria and apoptosis. Science 1998;281:1309–1312.
265. Lee SR, Yang KS, Kwon J, Lee C, Jeong W, Rhee SG. Reversible inactivation of the tumor suppressor PTEN by H2O2. J Biol Chem 2002; 277:20336–20342.
266. Pani G, Bedogni B, Anzevino R, Colavitti R, Palazzotti B, Borrello S, Galeotti T. Deregulated manganese superoxide dismutase expression and resistance to oxidative injury in p53-deficient cells. Cancer Res 2000;60:4654–4660.
267. Chandel NS, Schumacker PT. Cellular oxygen sensing by mitochondria: old questions, new insight. J Appl Physiol 2000;88:1880–1889.
268. Jobe AH, Bancalari E. Bronchopulmonary dysplasia. Am J Respir Crit Care Med 2001;163:1723–1729.
269. Archer S, Rich S. Primary pulmonary hypertension: a vascular biology and translational research “work in progress”. Circulation 2000;102:2781–2791.
270. Balint B, Kharitonov SA, Hanazawa T, Donnelly LE, Shah PL, Hodson ME, Barnes PJ. Increased nitrotyrosine in exhaled breath condensate in cystic fibrosis. Eur Respir J 2001;17:1201–1207.
271. Kaneko FT, Arroliga AC, Dweik RA, Comhair SA, Laskowski D, Oppedisano R, Thomassen MJ, Erzurum SC. Biochemical reaction products of nitric oxide as quantitative markers of primary pulmonary hypertension. Am J Respir Crit Care Med 1998;158:917–923.
272. Kinnula VL, Sarnesto A, Heikkila L, Toivonen H, Mattila S, Raivio KO. Assessment of xanthine oxidase in human lung and lung transplantation. Eur Respir J 1997;10:676–680.
273. Luna MA, Bedrossian CW, Lichtiger B, Salem PA. Interstitial pneumonitis associated with bleomycin therapy. Am J Clin Pathol 1972;58:501–510.
274. Szapiel SV, Elson NA, Fulmer JD, Hunninghake GW, Crystal RG. Bleomycin-induced interstitial pulmonary disease in the nude, athymic mouse. Am Rev Respir Dis 1979;120:893–899.
275. Matsuda Y, Higashiyama S, Kijima Y, Suzuki K, Kawano K, Akiyama M, Kawata S, Tarui S, Deutsch HF, Taniguchi N. Human liver manganese superoxide dismutase: purification and crystallization, subunit association and sulfhydryl reactivity. Eur J Biochem 1990;194:713–720.
276. Shimoda-Matsubayashi S, Matsumine H, Kobayashi T, Nakagawa-Hattori Y, Shimizu Y, Mizuno Y. Structural dimorphism in the mitochondrial targeting sequence in the human manganese superoxide dismutase gene. Biochem Biophys Res Commun 1996;226:561–565.
277. Rosenblum JS, Gilula NB, Lerner RA. On signal sequence polymorphisms and diseases of distribution. Proc Natl Acad Sci USA 1996;93:4471–4473.
278. Wang LI, Miller DP, Sai Y, Liu G, Su L, Wain JC, Lynch TJ, Christiani DC. Manganese superoxide dismutase alanine-to-valine polymorphism at codon 16 and lung cancer risk. J Natl Cancer Inst 2001;93:1818–1821.
279. Hirvonen A, Tuimala J, Ollikainen T, Linnainmaa K, Kinnula V. Manganese superoxide dismutase genotypes and asbestos-associated pulmonary disorders. Cancer Lett 2002;178:71–74.
280. Ho YS, Crapo JD. Isolation and characterization of complementary DNAs encoding human manganese-containing superoxide dismutase. FEBS Lett 1988;229:256–260.
281. Zhang HJ, Yan T, Oberley TD, Oberley LW. Comparison of effects of two polymorphic variants of manganese superoxide dismutase on human breast MCF-7 cancer cell phenotype 22. Cancer Res 1999;59:6276–6283.
282. Tomblyn M, Kasarskis EJ, Xu Y, St Clair DK. Distribution of MnSOD polymorphisms in sporadic ALS patients. J Mol Neurosci 1998;10:65–66.
283. Sandstrom J, Nilsson P, Karlsson K, Marklund SL. 10-fold increase in human plasma extracellular superoxide dismutase content caused by a mutation in heparin-binding domain. J Biol Chem 1994;269:19163–19166.
284. Adachi T, Ohta H, Yamada H, Futenma A, Kato K, Hirano K. Quantitative analysis of extracellular-superoxide dismutase in serum and urine by ELISA with monoclonal antibody. Clin Chim Acta 1992;212:89–102.
285. Yamada H, Yamada Y, Adachi T, Goto H, Ogasawara N, Futenma A, Kitano M, Miyai H, Fukatsu A, Hirano K, et al. Polymorphism of extracellular superoxide dismutase (EC-SOD) gene: relation to the mutation responsible for high EC-SOD level in serum. Jpn J Hum Genet 1997;42:353–356.
286. Folz RJ, Crapo JD, Peno-Green LA. Elevated levels of extracellular superoxide dismutase in chronic lung disease and characterization of genetic variants. Chest 1997;111:74S.
287. Comhair SA, Bhathena PR, Dweik RA, Kavuru M, Erzurum SC. Rapid loss of superoxide dismutase activity during antigen-induced asthmatic response. Lancet 2000;355:624.
288. De Raeve HR, Thunnissen FB, Kaneko FT, Guo FH, Lewis M, Kavuru MS, Secic M, Thomassen MJ, Erzurum SC. Decreased Cu, Zn-SOD activity in asthmatic airway epithelium: correction by inhaled corticosteroid in vivo. Am J Physiol 1997;272:L148–L154.
289. Vachier I, Damon M, Le Doucen C, de Paulet AC, Chanez P, Michel FB, Godard P. Increased oxygen species generation in blood monocytes of asthmatic patients. Am Rev Respir Dis 1992;146:1161–1166.
290. Clerch LB, Wright AE, Coalson JJ. Lung manganese superoxide dismutase protein expression increases in the baboon model of bronchopulmonary dysplasia and is regulated at a posttranscriptional level. Pediatr Res 1996;39:253–258.
291. Bravard A, Hoffschir F, Sabatier L, Ricoul M, Pinton A, Cassingena R, Estrade S, Luccioni C, Dutrillaux B. Early superoxide dismutase alterations during SV40-transformation of human fibroblasts. Int J Cancer 1992;52:797–801.
292. Oberley LW, Oberley TD. Role of antioxidant enzymes in the cancer phenotype. In: Clerch LB, Massaro DJ, editors. Oxygen, gene expression and cellular function. Washington, DC: Marcel Dekker, Inc; 1997. p. 279–308.
293. Zhang Y, Zhao W, Zhang HJ, Domann FE, Oberley LW. Overexpression of copper zinc superoxide dismutase suppresses human glioma cell growth. Cancer Res 2002;62:1205–1212.
294. Church SL, Grant JW, Ridnour LA, Oberley LW, Swanson PE, Meltzer PS, Trent JM. Increased manganese superoxide dismutase expression suppresses the malignant phenotype of human melanoma cells. Proc Natl Acad Sci USA 1993;90:3113–3117.
295. Li JJ, Oberley LW, St Clair DK, Ridnour LA, Oberley TD. Phenotypic changes induced in human breast cancer cells by overexpression of manganese-containing superoxide dismutase. Oncogene 1995;10:1989–2000.
296. Li N, Oberley TD, Oberley LW, Zhong W. Overexpression of manganese superoxide dismutase in DU145 human prostate carcinoma cells has multiple effects on cell phenotype. Prostate 1998;35:221–233.
297. Safford SE, Oberley TD, Urano M, St Clair DK. Suppression of fibrosarcoma metastasis by elevated expression of manganese superoxide dismutase. Cancer Res 1994;54:4261–4265.
298. St Clair DK, Wan XS, Oberley TD, Muse KE, St Clair WH. Suppression of radiation-induced neoplastic transformation by overexpression of mitochondrial superoxide dismutase. Mol Carcinog 1992;6:238–242.
299. Yan T, Oberley LW, Zhong W, St Clair DK. Manganese-containing superoxide dismutase overexpression causes phenotypic reversion in SV40-transformed human lung fibroblasts. Cancer Res 1996;56:2864–2871.
300. Zhong W, Yan T, Lim R, Oberley LW. Expression of superoxide dismutases, catalase, and glutathione peroxidase in glioma cells. Free Radic Biol Med 1999;27:1334–1345.
301. Li N, Oberley TD, Oberley LW, Zhong W. Inhibition of cell growth in NIH/3T3 fibroblasts by overexpression of manganese superoxide dismutase: mechanistic studies 78. J Cell Physiol 1998;175:359–369.
302. Manna SK, Zhang HJ, Yan T, Oberley LW, Aggarwal BB. Overexpression of manganese superoxide dismutase suppresses tumor necrosis factor-induced apoptosis and activation of nuclear transcription factor-kappaB and activated protein-1. J Biol Chem 1998;273:13245–13254.
303. Huang Y, Peng J, Oberley LW, Domann FE. Transcriptional inhibition of manganese superoxide dismutase (SOD2) gene expression by DNA methylation of the 5′ CpG island. Free Radic Biol Med 1997;23:314–320.
304. Ho JC, Zheng S, Comhair SA, Farver C, Erzurum SC. Differential expression of manganese superoxide dismutase and catalase in lung cancer 56. Cancer Res 2001;61:8578–8585.
305. Rihn BH, Mohr S, McDowell SA, Binet S, Loubinoux J, Galateau F, Keith G, Leikauf GD. Differential gene expression in mesothelioma. FEBS Lett 2000;480:95–100.
306. Kahlos K, Anttila S, Asikainen T, Kinnula K, Raivio KO, Mattson K, Linnainmaa K, Kinnula VL. Manganese superoxide dismutase in healthy human pleural mesothelium and in malignant pleural mesothelioma. Am J Respir Cell Mol Biol 1998;18:570–580.
307. Kahlos K, Paakko P, Kurttila E, Soini Y, Kinnula VL. Manganese superoxide dismutase as a diagnostic marker for malignant pleural mesothelioma. Br J Cancer 2000;82:1022–1029.
308. Kahlos K, Soini Y, Paakko P, Saily M, Linnainmaa K, Kinnula VL. Proliferation, apoptosis, and manganese superoxide dismutase in malignant mesothelioma. Int J Cancer 2000;88:37–43.
309. Kinnula VL, Pietarinen-Runtti P, Raivio K, Kahlos K, Pelin K, Mattson K, Linnainmaa K. Manganese superoxide dismutase in human pleural mesothelioma cell lines. Free Radic Biol Med 1996;21:527–532.
310. Harris JB, Chang LY, Crapo JD. Rat lung alveolar type I epithelial cell injury and response to hyperoxia. Am J Respir Cell Mol Biol 1991;4:115–125.
311. Huret JL, Delabar JM, Marlhens F, Aurias A, Nicole A, Berthier M, Tanzer J, Sinet PM. Down syndrome with duplication of a region of chromosome 21 containing the CuZn superoxide dismutase gene without detectable karyotypic abnormality. Hum Genet 1987;75:251–257.
312. Sherman L, Levanon D, Lieman-Hurwitz J, Dafni N, Groner Y. Human Cu/Zn superoxide dismutase gene: molecular characterization of its two mRNA species. Nucleic Acids Res 1984;12:9349–9365.
313. Church SL. Manganese superoxide dismutase: nucleotide and deduced amino acid sequence of a cDNA encoding a new human transcript. Biochim Biophys Acta 1990;1087:250–252.
314. Wispe JR, Clark JC, Burhans MS, Kropp KE, Korfhagen TR, Whitsett JA. Synthesis and processing of the precursor for human mangano-superoxide dismutase. Biochim Biophys Acta 1989;994:30–36.
315. Hendrickson DJ, Fisher JH, Jones C, Ho YS. Regional localization of human extracellular superoxide dismutase gene to 4pter-q21. Genomics 1990;8:736–738.
Correspondence and requests for reprints should be addressed to James D. Crapo, M.D., National Jewish Medical and Research Center, Room K701, 1400 Jackson Street, Denver, CO 80206. E-mail:

Related

No related items
American Journal of Respiratory and Critical Care Medicine
167
12

Click to see any corrections or updates and to confirm this is the authentic version of record