当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第3期 > 正文
编号:11256324
Absence of Heme Oxygenase-1 Exacerbates Myocardial Ischemia/Reperfusion Injury in Diabetic Mice
     1 Pulmonary and Critical Care Division, Brigham and Women’s Hospital, Boston, Massachusetts

    2 Department of Medicine, Harvard Medical School, Boston, Massachusetts

    ABSTRACT

    Increased production of reactive oxygen species contributes to the etiology of diabetes complications. Pathophysiological stimuli that increase oxidative stress upregulate heme oxygenase (HO)-1, a cytoprotective hemeeCdegrading enzyme. We hypothesized that HO-1 may be important in myocardial injury that is exacerbated by diabetes. To test this hypothesis, the left anterior descending coronary arteries of nondiabetic and diabetic wild-type (HO-1+/+) and HO-1 null (HO-1eC/eC) mice were ligated for 1 h followed by 24 h reperfusion. The absence of HO-1 significantly increased myocardial infarct size (36.4 ± 2.0 vs. 21.4 ± 1.8% in HO-1+/+ mice), while cardiac-specific overexpression of HO-1 protected against myocardial ischemic injury in diabetic mice. Despite similar high blood glucose levels, diabetic HO-1eC/eC mice had fourfold higher oxidative stress and larger infarcts (56.0 ± 2.8%) than diabetic HO-1+/+ mice (30.8 ± 6.1%). Moreover, hyperglycemia increased the mortality of HO-1eC/eC mice (31.3%) after ischemia/reperfusion injury, and 55% of diabetic HO-1eC/eC mice had mural thrombi in the left ventricles. The increased mortality of diabetic HO-1eC/eC mice may be in part due to formation of left ventricular mural thrombi. Our data demonstrate that the absence of HO-1 renders animals more susceptible to myocardial ischemia/reperfusion damage and diabetes worsens the injury.

    Coronary artery disease leading to myocardial infarction and heart failure is a chronic complication of diabetes, accounting for >75% of hospitalizations in diabetic patients (1eC3). In addition, myocardial infarction accounts for nearly 50% of all deaths in patients with diabetes (4). The mortality rate of diabetic patients after acute myocardial infarction resulting from ischemia/reperfusion injury is approximately twice that of nondiabetic patients (5eC7).

    Several reports have suggested that increased oxidative stress contributes to the development and progression of diabetes complications (8eC10). Heme oxygenase (HO)-1, the inducible isoform of the HO family of proteins, is induced by a variety of stimuli that increase oxidative stress (11,12). By degrading the oxidant heme and generating the antioxidant bilirubin and anti-inflammatory molecule carbon monoxide (CO), HO-1 protects cells from death due to pathophysiological stress and oxidative injury (13eC18). In diseases that are initiated by stress, such as atherosclerotic lesion formation and vascular remodeling, lack of endogenous HO-1 exacerbates lesion formation (19). Given its cytoprotective role in many cell types and organs, there is growing interest in the role of HO-1 in diabetes (20eC23). HO-1 protects islet cells from apoptosis and improves in vivo function after transplantation (20). HO-1 also protects diabetic rats from endothelial cell dysfunction by preventing endothelial cell sloughing (24).

    Myocardial ischemia/reperfusion injury results in greater damage in the diabetic (db/db) mouse hearts than nondiabetic controls (25). While Nishio et al. (26) reported HO-1 induction in diabetic rat hearts, the functional role of HO-1 in diabetes complications of myocardial infarction has not been examined. We have shown that in response to hypoxia, which leads to pulmonary hypertension, HO-1 null (HO-1eC/eC) mice develop severe right ventricular dilatation and infarction due to oxidative damage (13). This prior study demonstrated the detrimental consequences of HO-1 deficiency in the heart in response to pathological stress. HO-1 has a critical role in cardiac homeostasis in response to oxidative stresseCinduced injury (16,18,27) and that hyperglycemia increases the production of reactive oxygen species in diabetes (28,29). We hypothesized that in the absence of HO-1, cardiomyocytes may be more susceptible to ischemia/reperfusion damage and diabetes may exacerbate the myocardial injury. To test our hypothesis in vivo, we induced diabetes in wild-type and HO-1eC/eC mice by streptozotocin (STZ) and subjected the nondiabetic control and diabetic animals to a myocardial infarction model to determine if the absence of HO-1 renders mice more prone to myocardial damage.

    RESEARCH DESIGN AND METHODS

    Wild-type (HO-1+/+) and HO-1eC/eC mice were obtained by breeding HO-1 heterozygous mice (129Sv and BALB/c mixed genetic background) (13). Cardiac-specific HO-1 transgenic mice (FVB genetic background) were as previously described (16). This study used the TG.H transgenic line with high HO-1 exogenous expression levels (16). Wild-type littermates were used as controls. Animal experiments were performed in accordance with NIH (National Institutes of Health) guidelines, and protocols were approved by the Harvard Medical Area Standing Committee on Animals.

    STZ-induced diabetes in mice.

    At 6 weeks of age, male mice were treated with STZ (Sigma) at a dose of 50 mg/kg body wt in 20 mmol/l citrate buffer, pH 4.5, by five daily intraperitoneal injections (30). Control mice were injected with citrate buffer alone. Fasting glucose levels were measured from tail vein blood using the ACCU-CHEK blood glucose meter (Roche, Indianapolis, IN). Diabetes was confirmed by the presence of hyperglycemia (>200 mg/dl).

    Northern blot analysis.

    Total RNA was isolated from mouse hearts using RNA-Bee RNA isolation reagent (Tel-Test, Friendswood, TX), and Northern blot analysis was performed as described (13). The filters were hybridized with a random-primed 32P-labeled HO-1 cDNA probe (31). Equal loading was verified by hybridizing the filters to a 32P-labeled oligonucleotide complementary to 28S ribosomal RNA.

    Mouse model of myocardial infarction.

    Four weeks after STZ administration, control and diabetic mice were subjected to myocardial ischemia and reperfusion injury (16). During the operation, the animals were kept warm by using heat lamps. In brief, myocardial ischemia was achieved by ligating the anterior descending branch of the left coronary artery. Regional ischemia was confirmed by visual inspection of pale color in the occluded distal myocardium. After occlusion for 1 h, the ligature was released, allowing reperfusion of the formerly ischemic myocardium. Blood flow was confirmed by visualization of the return of a bright red color in the previously pale region. After the chest wall was closed, the animal was kept warm on a 37°C warming plate until recovery from anesthesia (1eC2 h). Hearts were harvested after 24 h reperfusion.

    Assessment of area at risk and infarct size.

    To assess myocardial infarct size after 1 h ischemia and 24 h reperfusion, the left coronary artery was occluded with a suture at the same site as the initial ligation (16). To demarcate the ischemic area at risk, 1% Evans blue dye was perfused into the aorta and coronary arteries (with distribution throughout the ventricular wall proximal to the site of coronary artery ligation) to stain the nonischemic area blue (32,33). Hearts were excised and sliced into five (1 mm) cross sections below the ligature. Heart sections were incubated in a 1% triphenyltetrazolium chloride solution to stain viable myocardium red and the infarct area pale. The infarct area (pale), the area at risk (not blue), and the total left ventricular area from both sides of each section were measured using NIH Image software and averaged. The thickness of each section was measured using dial calipers. The left ventricular area, area at risk, and infarct area of each section were multiplied by the thickness of the section and then totaled from all five sections. The ratio of area at risk to left ventricular area and the ratio of infarct area to area at risk were calculated and expressed as a percentage, as previously described (16).

    Histological analysis and immunohistochemistry.

    Ventricles from HO-1+/+ and HO-1eC/eC mice were fixed in Methyl Carnoy’s solution (60% methanol, 30% chloroform, and 10% acetic acid) at 4°C for 5 h, then in 70% ethanol overnight, and embedded in paraffin. Sections were stained with hematoxylin and eosin (H&E). To detect HO-1 expression and inflammatory cells, sections were stained with a polyclonal HO-1 antiserum (SPA-895, diluted 1:400; StressGen Biotechnologies, Victoria, BC, Canada) and a mouse CD45 (leukocyte common antigen) antibody (diluted 1:1,000; BD Biosciences PharMingen, San Diego, CA) at 4°C overnight. Sections were counterstained with methyl green. To detect oxidation-specific lipid protein adducts, sections were stained with MAL-2 antibody (diluted 1:100; provided by J. Witztum, Uinversity of California, San Diego, La Jolla, CA) (13,16,34). To assess inflammatory cell and oxidative stress accumulation, the areas of ischemic myocardium (obtained at x100 magnification) were measured by computerized planimetry, and the areas staining positive for CD45 and MAL-2 were measured by colorimetric analysis (13,16,35). The respective positive staining area was divided by the ischemic myocardial area and multiplied by 100.

    Statistical analysis.

    Data are presented as means ± SE. Statistical significance was determined by a Student’s t test. A P value 0.05 was accepted as statistically significant.

    RESULTS

    HO-1 induction by myocardial ischemia/reperfusion in diabetic mice.

    To investigate the myocardial expression of HO-1 in diabetic mice, we used an animal model of diabetes. We induced diabetes in mice by injection of STZ and examined HO-1 expression after 28 days. Northern blot analysis showed that STZ injection (n = 3) did not substantially increase HO-1 expression in the heart relative to vehicle-treated controls (n = 4) (Fig. 1A, 118 ± 14% STZ vs. Cont). Our results are different from the fourfold induction observed in rat hearts after STZ injection (26). It is possible that the kinetics of HO-1 induction by STZ may differ between mouse and rat hearts. To evaluate the combined effects of diabetes and myocardial infarction on HO-1 expression, vehicle-treated control and STZ-induced diabetic mice were subjected to 1 h ischemia and 24 h reperfusion to experimentally induce myocardial infarction. In vehicle-treated mice, myocardial ischemia/reperfusion increased HO-1 expression by fourfold (Fig. 1A). Diabetes further enhanced HO-1 expression in the heart after ischemia/reperfusion (n = 2) to a sixfold higher level than control (Fig. 1A, STZ+I/R). In contrast, HO-2 expression was not changed (data not shown).

    Consistent with Northern analysis (Fig. 1A), immunostaining with an HO-1 antibody revealed that HO-1 protein was barely detectable in the heart from vehicle-treated control mice (Fig. 1B). STZ slightly increased HO-1 levels in the cardiomyocytes 28 days after treatment (Fig. 1C). Ischemia/reperfusion strongly induced HO-1 protein expression in the cardiomyocytes (Fig. 1D, brown, arrows) and infiltrated inflammatory cells (Fig. 1D, arrowheads). Furthermore, intense brown staining in the hearts from diabetic mice after ischemia/reperfusion indicated elevated HO-1 protein levels in the cardiomyocytes and inflammatory cells (Fig. 1E, arrows and arrowheads, respectively). These data suggest that HO-1 induction may protect against myocardial injury.

    STZ-induced hyperglycemia in both wild-type and HO-1 null mice.

    To assess the functional role of HO-1 in diabetic myocardial infarction, we first induced diabetes in HO-1+/+ and HO-1eC/eC mice. Under basal physiological conditions, the fasting blood glucose levels were not different between HO-1+/+ (121 ± 4 mg/dl, n = 19) and HO-1eC/eC mice (117 ± 3 mg/dl, n = 27; P = 0.44 vs. wild type). Fasting blood glucose levels in HO-1+/+ mice increased to 418 ± 30 mg/dl (n = 9) 28 days after STZ, as compared with vehicle-treated controls (112 ± 4, n = 10; P < 0.05) (Fig. 2, black bars). In response to STZ injection, HO-1eC/eC mice also developed elevated fasting blood glucose levels (385 ± 15 mg/dl, n = 16) in comparison with vehicle-treated HO-1eC/eC mice (111 ± 3, n = 11; P < 0.05) (Fig. 2, white bars). STZ induced hyperglycemia to a similar degree in HO-1+/+ and HO-1eC/eC mice (P = 0.34) (Fig. 2).

    Absence of HO-1 increases myocardial infarct size.

    To test whether an absence of HO-1 renders animals more prone to cardiac damage, we subjected HO-1+/+ and HO-1eC/eC mice to ischemia/reperfusion injury. In HO-1+/+ mice, 1 h ischemia and 24 h reperfusion resulted in 21.4 ± 1.8% (n = 7) infarction of the left ventricular area at risk (Figs. 3A and E). In contrast, HO-1eC/eC mice had larger infarcts (Fig. 3B) that were 15% greater (36.4 ± 2.0%, n = 6) than HO-1+/+ mice (P < 0.005) (Fig. 3E). These results indicate that in normoglycemic mice, HO-1 protects against myocardial injury.

    Diabetes exacerbates myocardial infarction in the absence of HO-1.

    To determine the effects of diabetes in the setting of HO-1 deficiency in myocardial infarction, we subjected diabetic HO-1+/+ and HO-1eC/eC mice to ischemia/reperfusion injury. In HO-1+/+ mice, hyperglycemia slightly increased infarct size of HO-1+/+ mice to 30.8 ± 6.1% (n = 7) (Figs. 3C and E) compared with control HO-1+/+ mice, although it did not reach statistical significance (P = 0.08) (Fig. 3E, black bars). An absence of HO-1 in diabetic mice significantly exacerbated myocardial injury (Fig. 3D). Diabetic HO-1eC/eC mice had much larger infarcts (56.0 ± 2.8%, n = 7) than diabetic HO-1+/+ mice (P < 0.005) or control HO-1eC/eC mice (P < 0.005) (Fig. 3E).

    Myocardial ischemia/reperfusion increases mortality in diabetic HO-1eC/eC mice.

    There were no obvious histological differences in the hearts from HO-1+/+ and HO-1eC/eC mice under normal physiological conditions (13) or 28 days after STZ injection (data not shown). In response to myocardial injury (1 h ischemia/24 h reperfusion), vehicle-treated HO-1+/+ and HO-1eC/eC mice had similar mortality (8.3% and 9.1%, respectively; Table 1). Hyperglycemia increased the mortality of HO-1+/+ mice almost twofold to 15.4% after ischemia/reperfusion. In contrast, 5 of the 16 (31.3%) diabetic HO-1eC/eC mice that survived 1 h myocardial ischemia died during the subsequent 24 h reperfusion (Table 1).

    Exacerbation of myocardial injury in diabetic HO-1eC/eC mice.

    To investigate the cause of increased mortality in diabetic HO-1eC/eC mice after ischemia/reperfusion, we examined ventricular sections by histological analysis. H&E staining revealed that ischemia/reperfusion caused cardiomyocyte damage, indicated by disruption of cardiomyocytes, in nondiabetic control HO-1+/+ mice (Fig. 4A, arrow). Hyperglycemia slightly increased cardiomyocyte injury in similar ischemic areas in HO-1+/+ mice (Fig. 4C, arrow). In comparison, an absence of HO-1 exacerbated myocardial damage in the ischemic areas in both control and diabetic HO-1eC/eC mouse hearts (Figs. 4B and D, arrows). In addition to myocyte damage, CD45-positive mononuclear inflammatory cell infiltration was evident within the infarcts of control HO-1+/+ hearts after ischemia/reperfusion (Fig. 4E, brown). In similar ischemic areas, diabetic HO-1+/+ hearts had comparable levels of inflammatory cell infiltration (1.8 ± 0.3%) with control HO-1+/+ hearts (1.6 ± 0.1%). Interestingly, a similar degree of inflammatory cell infiltration was observed in the ischemic myocardium from control HO-1eC/eC (2.0 ± 0.1%) and diabetic HO-1eC/eC (2.1 ± 0.1%) mice subjected to ischemia/reperfusion (Figs. 4F and H). We next assessed oxidative stress in the ischemic hearts by MAL-2 immunostaining, which recognizes oxidation-specific lipid-protein adducts. In HO-1+/+ mice, oxidative stress was 2.1 ± 0.1% of the ischemic area in control compared with 3.3 ± 0.1% in diabetic hearts. Diabetes increased oxidative stress by nearly twofold in HO-1eC/eC mouse hearts (from 8.3 ± 0.7 to 15.2 ± 1.5%). Loss of HO-1 resulted in an approximate fourfold increase in oxidative stress in the settings of either control or diabetes after ischemia/reperfusion injury, which led to the highest oxidative stress levels in diabetic HO-1eC/eC mouse hearts.

    Ischemia/reperfusion induces mural thrombi in diabetic HO-1eC/eC mice.

    Histological analysis revealed extensive myocardial damage and inflammatory cell infiltrates in diabetic HO-1eC/eC mice subjected to ischemia/reperfusion; however, it is likely that additional factors contribute to the higher mortality in these mice. Surprisingly, after ischemia/reperfusion, we observed that three of four hearts from diabetic HO-1eC/eC mice harvested for histological analysis contained large mural thrombi, which adhered to the infarcted left ventricles (Fig. 5D). Additionally, we observed mural thrombi in three of seven hearts evaluated for infarct size. In total, mural thrombi developed in 55% of diabetic HO-1eC/eC mice that survived 1 h ischemia and 24 h reperfusion. In contrast, no mural thrombi were present in the hearts from control and diabetic HO-1+/+ or control HO-1eC/eC mice subjected to ischemia/reperfusion (Figs. 5AeCC). Ventricular sections revealed extensive inflammatory cell infiltration within the thrombi (Figs. 5E and F).

    Cardiac-specific overexpression of HO-1 reduces myocardial infarct size in diabetic mice.

    To investigate the role of HO-1 in protecting against myocardial ischemic injury in diabetes, we induced diabetes in wild-type and cardiac-specific HO-1 transgenic mice (TG.H line, FVB strain [16]) and subjected them to experimental myocardial infarction. In diabetic wild-type mice, 1 h ischemia/24 h reperfusion resulted in 52 ± 5% (n = 7) infarction of the left ventricular area at risk (Fig. 6). In contrast, the infarct size of diabetic HO-1 transgenic mice was 20 ± 5% (n = 6), significantly less than that of diabetic wild-type mice (P < 0.005) (Fig. 6), indicating that HO-1 overexpression lessens the diabetes complication of ischemia/reperfusion injury.

    DISCUSSION

    Hyperglycemia increases oxidative stress, which contributes to the development of diabetes complications (8,9,36). Oxidative stress and inflammation in turn exacerbate myocardial ischemia/reperfusion injury (37,38), leading to myocardial infarction—a major diabetes complication. HO-1 and its reaction products have been shown to have both antioxidative and anti-inflammatory properties (16,39,40), indicating a protective role of HO-1 in diabetes complications of myocardial infarction. Using HO-1 heterozygous mouse hearts in isolated perfused heart preparations, Yoshida et al. (41) showed that a reduction of HO-1 worsens the ischemic injury. By gain-of-function experiments, we have previously demonstrated that cardiac-specific overexpression of HO-1 protects against ischemia/reperfusion injury in vivo (16). However, the effect of an absence of HO-1 in myocardial injury in vivo has not been elucidated in the settings of either normo- or hyperglycemic conditions. By using homozygous HO-1 null mice, we demonstrated that an absence of HO-1 exacerbated myocardial injury, particularly in concert with diabetes.

    Myocardial ischemia/reperfusion induced HO-1 expression in the heart (Fig. 1), suggesting that this induction may function to reduce or limit the myocardial damage and that an absence of HO-1 may exacerbate the injury. Consistent with this hypothesis, the first major finding was that HO-1eC/eC mice had larger infarcts than wild-type mice after experimental myocardial infarction (Figs. 3A and B).

    Diabetes elevated cardiac HO-1 expression in response to ischemia/reperfusion (Fig. 1), thus implicating a protective role of HO-1 in diabetic myocardial infarction. The larger infarct of diabetic than control HO-1+/+ hearts suggested that the extent of HO-1 upregulation may not be sufficient to completely protect against the damage elicited by a combination of diabetes and ischemia/reperfusion; therefore, other factors may also contribute to the myocardial injury. Nevertheless, the second major finding in this study was that despite a similar degree of hyperglycemia after STZ injection in both HO-1+/+ and HO-1eC/eC mice (Fig. 2), diabetic HO-1eC/eC mice had more severe myocardial infarction (Fig. 3). These results demonstrate that upregulation of HO-1 protects against diabetic myocardial infarction. Supporting our findings, an AA genotype of T(eC413)A polymorphism in the promoter region of the HO-1 gene was found to associate with reduced incidence of myocardial infarction, possibly due to the higher expression level of HO-1 (42). The protective effect of HO-1 in the diabetic heart was further supported by our findings that diabetic HO-1 transgenic mice had reduced infarct size compared with that of diabetic wild-type mice (Fig. 6). It is worth noting that the infarct size of diabetic FVB wild-type mice was larger than that of diabetic 129sv and BALB/c mixed-background wild-type mice, likely due to strain differences.

    The molecular mechanisms by which HO-1 confers myocardial protection are still under investigation. Interestingly, we found that loss of HO-1 did not substantially increase inflammatory infiltration after ischemia/reperfusion (Figs. 4EeCH). This is somewhat unanticipated, given the anti-inflammatory properties of HO-1. One possibility is that accelerated inflammation is a major effect of myocardial reperfusion injury (38); the limited HO-1 induction in the cardiomyocytes (Figs. 1D and E) may not be able to overcome the overwhelming influx of inflammatory cells. Despite a similar degree of inflammatory cell infiltration, the absence of HO-1 increased oxidative stress levels in diabetic mice after myocardial ischemia/reperfusion injury. These data suggest that the antioxidant properties of HO-1 contribute to the protection against myocardial injury.

    In addition to exaggerated infarct size, we observed an additional twofold higher mortality in diabetic HO-1eC/eC than in diabetic HO-1+/+ mice after ischemia/reperfusion injury. Of major significance was the observation that 55% of diabetic HO-1eC/eC mice that survived 1 h ischemia and 24 h reperfusion developed mural thrombi (Fig. 5), despite patent coronary arteries (data not shown). Interestingly, mortality rates of diabetic patients with acute myocardial infarction remain 1.5eC2 times higher than those in nondiabetic patients (7). It was suggested that this increased mortality rate is in part due to the tendency toward thrombosis in diabetic patients (7). It remains to be determined whether long-term diabetes reduces HO-1 expression in the heart, which in turn contributes to the higher mortality after myocardial infarction in diabetic patients.

    Interestingly, the formation of mural thrombi in the left ventricle of diabetic HO-1eC/eC mice after ischemia/reperfusion injury was similar to what we observed in HO-1eC/eC mice that developed right ventricular mural thrombi in response to chronic hypoxia (13). We previously reported that in response to a similar degree of pulmonary hypertension in wild-type and HO-1eC/eC mice after chronic hypoxia (10% oxygen environment for 5eC7 weeks), HO-1eC/eC mice had increased oxidative injury and developed right ventricular infarcts with attached mural thrombi (13). These previous data suggest that in the absence of HO-1, right ventricular cardiomyocytes have a maladaptive response to hypoxia-induced pulmonary hypertension. The myocardial ischemia/reperfusion model we used in the present study does not induce pulmonary hypertension (32). The elevated oxidative stress and larger infarcts with thrombi in the left ventricles of diabetic HO-1eC/eC mice suggest a similar maladaptive response of left ventricular cardiomyocytes to ischemia/reperfusion injury. HO-1 and its reaction product CO have been shown to inhibit platelet aggregation (43,44) and to suppress thrombi growth in vivo under conditions of increased oxidative stress (45). The high incidence of mural thrombi might result from increased susceptibility to thrombus formation in the absence of HO-1. Inhibition of HO-1 activity promotes the formation of platelet-fibrin thrombi (45,46). Alternatively, the left ventricular thrombi formation may be secondary to larger infarcts and the subsequent more significant left ventricular dysfunction and regional wall motion abnormalities. Taken together, it is conceivable that in the absence of HO-1, ischemia/reperfusion in the setting of diabetes resulted in excessive oxidative damage in the left myocardium. The concerted effects of severe myocardial infarction and the susceptibility to thrombosis may eventually lead to increased mortality.

    In summary, we demonstrated in the present study, by loss-of-function experiments, that HO-1 plays an essential protective role in the adaptation of the heart to diabetes complications of myocardial infarction. Taken together, our findings may provide a new target for developing therapeutic strategies to treat/prevent diabetes complications of myocardial infarction.

    ACKNOWLEDGMENTS

    This study was supported in part by an American Diabetes Association Research Award (1-01-RA-26; to S.-F.Y.) and National Institutes of Health Grant HL-57977 (to S.-F.Y.).

    We are grateful to Mark A. Perrella for his enthusiasm toward our work.

    FOOTNOTES

    H&E, hematoxylin and eosin; HO, heme oxygenase; STZ, streptozotocin.

    REFERENCES

    Jacobs J, Sena M, Fox N: The cost of hospitalization for the late complications of diabetes in the United States. Diabet Med8 :S23 eCS29,1991

    Ray NF, Thamer M, Taylor T, Fehrenbach SN, Ratner R: Hospitalization and expenditures for the treatment of general medical conditions among the U.S. diabetic population in 1991. J Clin Endocrinol Metab81 :3671 eC3679,1996

    Rodrigues B, Cam MC, McNeill JH: Metabolic disturbances in diabetic cardiomyopathy. Mol Cell Biochem180 :53 eC57,1998

    Jelesoff NE, Feinglos M, Granger CB, Califf RM: Outcomes of diabetic patients following acute myocardial infarction: a review of the major thrombolytic trials. Coron Artery Dis7 :732 eC743,1996

    Savage MP, Krolewski AS, Kenien GG, Lebeis MP, Christlieb AR, Lewis SM: Acute myocardial infarction in diabetes mellitus and significance of congestive heart failure as a prognostic factor. Am J Cardiol62 :665 eC669,1988

    Woodfield SL, Lundergan CF, Reiner JS, Greenhouse SW, Thompson MA, Rohrbeck SC, Deychak Y, Simoons ML, Califf RM, Topol EJ, Ross AM: Angiographic findings and outcome in diabetic patients treated with thrombolytic therapy for acute myocardial infarction: the GUSTO-I experience. J Am Coll Cardiol28 :1661 eC1669,1996

    Aronson D, Rayfield EJ, Chesebro JH: Mechanisms determining course and outcome of diabetic patients who have had acute myocardial infarction. Ann Intern Med126 :296 eC306,1997

    Giugliano D, Ceriello A, Paolisso G: Oxidative stress and diabetic vascular complications. Diabetes Care19 :257 eC267,1996

    Dandona P, Thusu K, Cook S, Snyder B, Makowski J, Armstrong D, Nicotera T: Oxidative damage to DNA in diabetes mellitus. Lancet347 :444 eC445,1996

    Baynes JW, Thorpe SR: Role of oxidative stress in diabetic complications: a new perspective on an old paradigm. Diabetes48 :1 eC9,1999

    Choi AM, Alam J: Heme oxygenase-1: function, regulation, and implication of a novel stress-inducible protein in oxidant-induced lung injury. Am J Respir Cell Mol Biol15 :9 eC19,1996

    Maines MD: The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmacol Toxicol37 :517 eC554,1997

    Yet S-F, Perrella MA, Layne MD, Hsieh C-M, Maemura K, Kobzik L, Wiesel P, Christou H, Kourembanas S, Lee M-E: Hypoxia induces severe right ventricular dilatation and infarction in heme oxygenase-1 null mice. J Clin Invest103 :R23 eCR29,1999

    Soares MP, Lin Y, Anrather J, Csizmadia E, Takigami K, Sato K, Grey ST, Colvin RB, Choi AM, Poss KD, Bach FH: Expression of heme oxygenase-1 can determine cardiac xenograft survival. Nat Med4 :1073 eC1077,1998

    Clark JE, Foresti R, Green CJ, Motterlini R: Dynamics of haem oxygenase-1 expression and bilirubin production in cellular protection against oxidative stress. Biochem J348 :615 eC619,2000

    Yet S-F, Tian R, Layne MD, Wang ZY, Maemura K, Solovyeva M, Ith B, Melo LG, Zhang L, Ingwall JS, Dzau VJ, Lee M-E, Perrella MA: Cardiac-specific expression of heme oxygenase-1 protects against ischemia and reperfusion injury in transgenic mice. Circ Res89 :168 eC173,2001

    Fujita T, Toda K, Karimova A, Yan SF, Naka Y, Yet S-F, Pinsky DJ: Paradoxical rescue from ischemic lung injury by inhaled carbon monoxide driven by derepression of fibrinolysis. Nat Med7 :598 eC604,2001

    Melo LG, Agrawal R, Zhang L, Rezvani M, Mangi AA, Ehsan A, Griese DP, Dell’Acqua G, Mann MJ, Oyama J, Yet S-F, Layne MD, Perrella MA, Dzau VJ: Gene therapy strategy for long-term myocardial protection using adeno-associated virus-mediated delivery of heme oxygenase gene. Circulation105 :602 eC607,2002

    Yet S-F, Layne MD, Liu X, Chen Y-H, Ith B, Sibinga NES, Perrella MA: Absence of heme oxygenase-1 exacerbates atherosclerotic lesion formation and vascular remodeling. FASEB J17 :1759 eC1761,2003

    Pileggi A, Cattan P, Berney T, Molano RD, Vizzardelli C, Ricordi C, Bach FH, Inverardi L: HO-1 upregulation protects the pancreatic cell line TC3 from cytokines and Fas-induced apoptosis. Transplant Proc33 :266 eC267,2001

    Tobiasch E, Gunther L, Bach FH: Heme oxygenase-1 protects pancreatic cells from apoptosis caused by various stimuli. J Investig Med49 :566 eC571,2001

    Ribeiro MM, Klein D, Pileggi A, Molano RD, Fraker C, Ricordi C, Inverardi L, Pastori RL: Heme oxygenase-1 fused to a TAT peptide transduces and protects pancreatic -cells. Biochem Biophys Res Commun305 :876 eC881,2003

    Abraham NG, Kushida T, McClung J, Weiss M, Quan S, Lafaro R, Darzynkiewicz Z, Wolin M: Heme oxygenase-1 attenuates glucose-mediated cell growth arrest and apoptosis in human microvessel endothelial cells. Circ Res93 :507 eC514,2003

    Quan S, Kaminski PM, Yang L, Morita T, Inaba M, Ikehara S, Goodman AI, Wolin MS, Abraham NG: Heme oxygenase-1 prevents superoxide anion-associated endothelial cell sloughing in diabetic rats. Biochem Biophys Res Commun315 :509 eC516,2004

    Jones SP, Girod WG, Granger DN, Palazzo AJ, Lefer DJ: Reperfusion injury is not affected by blockade of P-selectin in the diabetic mouse heart. Am J Physiol277 :H763 eCH769,1999

    Nishio Y, Kashiwagi A, Taki H, Shinozaki K, Maeno Y, Kojima H, Maegawa H, Haneda M, Hidaka H, Yasuda H, Horiike K, Kikkawa R: Altered activities of transcription factors and their related gene expression in cardiac tissues of diabetic rats. Diabetes47 :1318 eC1325,1998

    Clark JE, Foresti R, Sarathchandra P, Kaur H, Green CJ, Motterlini R: Heme oxygenase-1-derived bilirubin ameliorates postischemic myocardial dysfunction. Am J Physiol278 :H643 eCH651,2000

    Kashiwagi A, Asahina T, Ikebuchi M, Tanaka Y, Takagi Y, Nishio Y, Kikkawa R, Shigeta Y: Abnormal glutathione metabolism and increased cytotoxicity caused by H2O2 in human umbilical vein endothelial cells cultured in high glucose medium. Diabetologia37 :264 eC269,1994

    Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, Kaneda Y, Yorek MA, Beebe D, Oates PJ, Hammes HP, Giardino I, Brownlee M: Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature404 :787 eC790,2000

    Park L, Raman KG, Lee KJ, Lu Y, Ferran LJ, Jr, Chow WS, Stern D, Schmidt AM: Suppression of accelerated diabetic atherosclerosis by the soluble receptor for advanced glycation endproducts. Nat Med4 :1025 eC1031,1998

    Morita T, Kourembanas S: Endothelial cell expression of vasoconstrictors and growth factors is regulated by smooth muscle cell-derived carbon monoxide. J Clin Invest96 :2676 eC2682,1995

    Michael LH, Entman ML, Hartley CJ, Youker KA, Zhu J, Hall SR, Hawkins HK, Berens K, Ballantyne CM: Myocardial ischemia and reperfusion: a murine model. Am J Physiol269 :H2147 eCH2154,1995

    Michael LH, Ballantyne CM, Zachariah JP, Gould KE, Pocius JS, Taffet GE, Hartley CJ, Pham TT, Daniel SL, Funk E, Entman ML: Myocardial infarction and remodeling in mice: effect of reperfusion. Am J Physiol277 :H660 eCH668,1999

    Rosenfeld ME, Palinski W, Yl-Herttuala S, Butler S, Witzum JL: Distribution of oxidation specific lipid-protein adducts and apolipoprotein B in atherosclerotic lesions of varying severity from WHHL rabbits. Arteriosclerosis10 :336 eC349,1990

    Shi C, Lee W-S, Russell ME, Zhang D, Fletcher DL, Newell JB, Haber E: Hypercholesterolemia exacerbates transplant arteriosclerosis via increased neointimal smooth muscle cell accumulation: studies in apolipoprotein E knockout mice. Circulation96 :2722 eC2728,1997

    Baynes JW: Role of oxidative stress in development of complications in diabetes. Diabetes40 :405 eC412,1991

    Entman ML, Youker K, Shoji T, Kukielka G, Shappell SB, Taylor AA, Smith CW: Neutrophil induced oxidative injury of cardiac myocytes: a compartmented system requiring CD11b/CD18-ICAM-1 adherence. J Clin Invest90 :1335 eC1345,1992

    Entman ML, Smith CW: Postreperfusion inflammation: a model for reaction to injury in cardiovascular disease. Cardiovasc Res28 :1301 eC1311,1994

    Otterbein LE, Bach FH, Alam J, Soares M, Lu HT, Wysk M, Davis RJ, Flavell RA, Choi AM: Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway. Nat Med6 :422 eC428,2000

    Minamino T, Christou H, Hsieh C-M, Liu Y, Dhawan V, Abraham NG, Perrella MA, Mitsialis SA, Kourembanas S: Targeted expression of heme oxygenase-1 prevents the pulmonary inflammatory and vascular responses to hypoxia. Proc Natl Acad Sci U S A98 :8798 eC8803,2001

    Yoshida T, Maulik N, Ho YS, Alam J, Das DK: Hmox-1 constitutes an adaptive response to effect antioxidant cardioprotection: A study with transgenic mice heterozygous for targeted disruption of the Heme oxygenase-1 gene. Circulation103 :1695 eC1701,2001

    Ono K, Goto Y, Takagi S, Baba S, Tago N, Nonogi H, Iwai N: A promoter variant of the heme oxygenase-1 gene may reduce the incidence of ischemic heart disease in Japanese. Atherosclerosis173 :315 eC319,2004

    Brune B, Ullrich V: Inhibition of platelet aggregation by carbon monoxide is mediated by activation of guanylate cyclase. Mol Pharmacol32 :497 eC504,1987

    Wagner CT, Durante W, Christodoulides N, Hellums JD, Schafer AI: Hemodynamic forces induce the expression of heme oxygenase in cultured vascular smooth muscle cells. J Clin Invest100 :589 eC596,1997

    Peng L, Mundada L, Stomel JM, Liu JJ, Sun J, Yet S-F, Fay WP: Induction of heme oxygenase-1 expression inhibits platelet-dependent thrombosis. Antioxid Redox Signal6 :729 eC735,2004

    Sato K, Balla J, Otterbein L, Smith RN, Brouard S, Lin Y, Csizmadia E, Sevigny J, Robson SC, Vercellotti G, Choi AM, Bach FH, Soares MP: Carbon monoxide generated by heme oxygenase-1 suppresses the rejection of mouse-to-rat cardiac transplants. J Immunol166 :4185 eC4194,2001(Xiaoli Liu, Jiao Wei, Dav)