Abstract
The pathophysiology of myocardial injury that results from cardiac ischemia and reperfusion (I/R) is incompletely understood. Experimental evidence from murine models indicates that innate immune mechanisms including complement activation via the classical and lectin pathways are crucial. Whether factor B (fB), a component of the alternative complement pathway required for amplification of complement cascade activation, participates in the pathophysiology of myocardial I/R injury has not been addressed. We induced regional myocardial I/R injury by transient coronary ligation in WT C57BL/6 mice, a manipulation that resulted in marked myocardial necrosis associated with activation of fB protein and myocardial deposition of C3 activation products. In contrast, in fB-/- mice, the same procedure resulted in significantly reduced myocardial necrosis (% ventricular tissue necrotic; fB-/- mice, 20 ± 4%; WT mice, 45 ± 3%; P < 0.05) and diminished deposition of C3 activation products in the myocardial tissue (fB-/- mice, 0 ± 0%; WT mice, 31 ± 6%; P<0.05). Reconstitution of fB-/- mice with WT serum followed by cardiac I/R restored the myocardial necrosis and activated C3 deposition in the myocardium. In translational human studies we measured levels of activated fB (Bb) in intracoronary blood samples obtained during cardio-pulmonary bypass surgery before and after aortic cross clamping (AXCL), during which global heart ischemia was induced. Intracoronary Bb increased immediately after AXCL, and the levels were directly correlated with peripheral blood levels of cardiac troponin I, an established biomarker of myocardial necrosis (Spearman coefficient = 0.465, P < 0.01). Taken together, our results support the conclusion that circulating fB is a crucial pathophysiological amplifier of I/R-induced, complement-dependent myocardial necrosis and identify fB as a potential therapeutic target for prevention of human myocardial I/R injury.
Introduction
Myocardial ischemia from inadequate coronary perfusion can occur regionally (e.g. due to coronary atherosclerosis) or globally (e.g. following aortic cross clamping (AXCL) during bypass surgery) and leads to myocardial necrosis. Prolonged regional myocardial ischemia manifests clinically as myocardial infarction (MI) while global cardiac ischemia is commonly subclinical and manifests as an increase in the peripheral blood of cardiac troponin I (cTnI) [1–9]. Emerging evidence suggests that small post-surgical elevations in cTnI can negatively impact long-term outcomes [10, 11], underscoring the importance of such subclinical injury. Reperfusion of the ischemic heart tissue with thrombolytic therapy, percutaneous coronary intervention, or following release of aortic cross clamping can acutely limit the necrotic damage but paradoxically may elicit an inflammatory response that contributes to further tissue damage. The induced reperfusion injury can stun the myocardium (limiting contractile function) and induce non-necrotic forms of cell death [12–15]. Understanding the molecular mechanisms underlying myocardial I/R injury is vital for future design of therapeutic interventions aimed at improving survival and reducing long term morbidity.
The discovery of a protective effect of “ischemic pre-conditioning” in a canine cardiac I/R model by Murry et al. offered hope for nonpharmacological interventions in I/R injury [16]. Since then, various cardioprotective strategies have been proposed to condition the heart directly or indirectly through brief episodes of ischemia and reperfusion. These include ischemic preconditioning, ischemic post-conditioning and remote ischemic conditioning, all of which showed success in animal models and small clinical studies (reviewed in-depth elsewhere) [17–22]. However, two recent large clinical trials investigating the role of remote ischemic preconditioning in cardiac surgery (RIPHeart [23] and ERICCA [24]) had negative outcomes.
Similarly, a number of pharmacological interventions to reduce myocardial damage have also been studied in clinical trials without encouraging results. One recent trial with the CypD inhibitor Cyclosporine-A (the CIRCUS Trial) showed no benefit in long term clinical outcome [25] and while β-blockers greatly improve overall clinical outcomes [26–28], their effect on infarct size in STEMI patients is still debatable [18]. There are other promising cardioprotective agents yet to be evaluated in clinical trials, e.g. glucose modulators, cyclooxygenase (COX)- and lipoxygenase (LOX)-directed lipid mediators [29, 30] and Exendin-4 [31], but the current absence of effective cardioprotective therapies supports the need for identifying novel therapeutic targets potentially capable of limiting cardiac ischemic injury
Current concepts of mechanisms leading to I/R injury [18, 32–35] are that after reperfusion, activation of a number of intracellular signaling pathways leads to calcium influx [36–39], mitochondrial dysfunction [40–45], production of reactive oxygen species [46–49], and activation of proteases [50]. These intracellular changes can directly cause cell death and can activate the vascular endothelium to express adhesion molecules, release proinflammatory cytokines and chemokines, and upregulate production of complement components [51–55]. Complement activation synergizes with toll-like receptor (TLR) signals induced by ligation of damage-associated molecular patterns (DAMPs), including HMGB1, [56–60] to upregulate NF-κB [61, 62] and amplifies local inflammation [63–65] and recruitment of inflammatory cells [66–68].
Complement activation can be initiated by the classical, mannose binding lectin (MBL), and alternative pathways that converge at the generation of C3-convertases. A common central factor B (fB)-dependent amplification loop continually generates C3-convertases, resulting in production of the potent yet short-lived anaphylatoxin C3a and the opsonin C3b, and has been shown to be critical in pathogen-induced inflammation [69, 70]. Subsequent common terminal pathway (C5-9) activation generates another anaphylatoxin, C5a, and causes formation and deposition of the membrane-attack complex (MAC) on cell surfaces [71–75] which can lead to NF-kB-dependent inflammatory responses [69, 70, 76] While prior work by our group among others focused on the role of the classical and MBL pathways as initiators of complement-dependent inflammation in myocardial I/R injury, [69–75, 77–81] the role of fB, required for amplification of the complement cascade, has not been clearly delineated. To test the hypothesis that fB is a key mediator of complement-dependent myocardial I/R injury, we studied surgically-induced cardiac I/R using fB-/- mice and serum samples obtained from patients undergoing global heart ischemia during cardiac surgery. Together our new translational data provide evidence that fB is a key mediator of myocardial I/R injury.
Materials and methods
Mouse model of surgically-induced myocardial I/R injury
Complement fB knockout (fB-/-) mice and wild type (WT) littermates were generated by co-author Dr. Marcella Pekna [82] and maintained at the SUNY Downstate Medical Center Department of Laboratory Animal Resources. Genotyping was provided by GeneTyper (New York, NY) using established PCR protocols [82]. Male mice were used at 10–12 weeks of age (weights 26-30g) in accordance with the requirements of the NIH and the Institutional Animal Care and Use Committee (IACUC) of SUNY Downstate Medical Center. The protocol was approved by the IACUC of SUNY Downstate Medical Center (Approval#11–10276).
We used an established model of myocardial I/R injury model [77, 83] in which mice were anesthetized using pentobarbital sodium (60 mg/kg, i.p.), intubated and ventilated with a mouse ventilator (Harvard Apparatus, MA). After midline sternotomy, the left anterior descending artery (LAD) was ligated for 1 hour; occlusion of the LAD was confirmed by the color change of myocardial tissue and the ST elevation on ECG. Reperfusion was established and verified by the color change of the left ventricle and the appropriate ECG changes. Postoperative management included fluid replacement with normal saline and pain relief with the analgesic buprenorphine (0.1 mg/kg, intramuscularly). The mice were sacrificed after 24h (euthanized in CO2 chamber at Downstate facility), serum was collected and the hearts were harvested for histopathology analyses.
Evaluation of murine myocardial necrosis by fluorescence using two probes delivered in vivo
A fluorescent method for tracking necrosis (initially developed by others [84, 85] and further refined by us [83]) was used. Shortly before the end of the reperfusion period described above in Materials and Methods, and before tissue harvesting, mice were anesthetized, intubated as described above, and injected i.v. with propidium iodide (PI), which enters damaged cells, intercalates with DNA and fluoresces, thus identifying necrotic tissue. The LAD was then re-occluded before heart harvesting and blue fluorescent microspheres (BFM, ThermoFisher, PA) were injected through the aortic arch to delineate the non-ischemic region of the heart. The heart was harvested and atrium was removed. The ventricle was sectioned into four slices (~1mm thickness), which were weighed and imaged under a fluorescent microscope (Olympus, PA), using the red fluorescent channel for PI, the blue for BFM.
The percentage of the tissue in a heart which was at risk for necrosis (no blue fluorescence) and which became necrotic (had red fluorescence) was determined by computerized planimetry (Image J, MD) and by the following equations:
where A was the percentage of the area of a slice staining for necrosis (red fluorescence) measured by planimetry (average of both sides of a slice) and Wt was the weight of that slice of ventricle.
where R is the percentage of the area of a slice which lacked the blue fluorescence of BFM, determined by planimetry (average of both sides of a slice used). In all cases, the tissue with red fluorescence was within the boundary of the tissue which lacked blue fluorescence.
Power analyses performed using G*Power 3.1 [86] showed >99% power for detection of differences in infarct size with 6–8 animals per group.
Western blotting of murine and patients’ fB and its active fragment, Bb
Following SDS polyacrylamide gel electrophoresis, proteins were transferred to a nitrocellulose membrane. A constant amount of plasma from a healthy individual was run on each gel of human samples to control for inter-gel differences in band staining. The membranes were blocked with bovine serum albumin in 20 mM Tris, 0.9% NaCl and 2% Tween-20 and incubated with polyclonal goat anti-fB antibody (Complement Technology, TX) which detects the fB, Ba and Bb proteins separated by electrophoresis. After washing, the membrane with murine samples was incubated with a donkey anti-goat antibody conjugated with AP (Rockland, PA) and developed with BCIP/NBT Substrate System (KPL, MD). The membranes with human samples were incubated with rabbit anti-goat IgG conjugated with alkaline phosphatase (Abcam, MA) and developed with the BCIP/NBT Substrate System. Quantification of bands of interest was carried out using the ImageJ program (NIH). The intensities of bands in the human samples were normalized to that of the 93 kDa fB band of the normal control lane and expressed as relative intensity.
Quantitative real-time PCR to detect fB mRNA expression
RNA isolation, cDNA synthesis, reverse transcription, and real-time RT-PCR were performed as described previously [87]. Briefly, RNA was isolated from heart tissues of fB-/- and WT mice using Trizol (Life Technologies; CA). cDNA was reverse-transcribed using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, NJ) as per the manufacturers’ instructions. Q-PCR was performed with TaqMan primers (Applied Biosystems) and run on the CFX96 Real-Time System (Bio-Rad Laboratories, CA). PCR products were normalized to the 18S control gene and expressed as fold increase over the mean value of fB-/- heart samples using the ΔΔCt method.
fB primers are obtained as TaqMan probes from Applied Biosystems (Waltham, MA): Mm00433918_g1 and 18s primer: Mm03928990_g1.
Immunohistochemical analysis of complement C3 deposition
Frozen sections were cut from the heart slices described in Section 2, fixed in acetone and stained with an FITC-labeled anti-C3c antibody (Dako, CA). Each section was imaged (2x objective lens) using channel 4 (for all fluorescence) to give total area. The C3 positive area was imaged (10x objective lens) and quantified by Image J software. The percentage of the total area that was C3 positive was determined.
Patient enrollment, perioperative management and blood sampling
The prospective clinical study was approved by the Institutional Review Boards of SUNY Downstate Medical Center and Yale University School of Medicine (Approval#07–106). Adult patients (total 105), undergoing elective cardiac surgery with cardiopulmonary bypass (CPB), were enrolled in the study following their consent. All participants provided their written informed consents to participate in this study.
At the discretion of the attending cardiac surgeons, oral antiplatelet agents were discontinued within 7–10 days before surgery. Patients chronically treated with beta-blocking agents or statins continued these medications until the morning of surgery. Midazolam (1-2mg) was given as soon as the standard monitors (i.e., five-lead electrocardiogram with computerized analysis of repolarization, end tidal CO2 monitor and pulse oximetry) were applied and prior to insertion of the arterial line. Other monitors, including central venous pressure and pulmonary artery occlusion pressure were applied after induction of anesthesia. A smooth induction, aiming to maintain hemodynamic parameters as close as possible to each patient’s baseline, was used. Antifibrinolytic therapy with tranexamic acid (a 10-20mg/kg bolus followed by an infusion of 1-2mg/kg/hr.) was administered. CPB was carried out under hypothermia at approximately 32°C with a pump flow of 2.0–2.4 L/min/m2. The cardioplegic solution was delivered after clamping of the aorta. The hematocrit was maintained at ≥16% while patients were on bypass and >23% postoperatively. General anesthesia was maintained with a combination of opioid-volatile techniques and the depth of anesthesia was titrated to meet the requirements of the varying intensities of surgical stimulation. Boluses of vasoactive agents were given intraoperatively as necessary to maintain mean arterial pressure between 50 and 80 mm Hg. If, after aortic unclamping, sinus rhythm did not resume spontaneously, the heart was defibrillated. After termination of CPB, catecholamines were used at the discretion of the anesthesiologist. Postoperatively, patients were admitted to the CT-ICU under the care of the anesthesiologist and cardiac surgeon on duty.
Coronary sinus blood samples were collected after CPB prior to AXCL and 5 minutes after AXCL termination. Blood samples were centrifuged and the plasma stored at -80°C.
Of the 105 patients who consented to participate, complete sets of coronary sinus blood samples were collected from 56. Data from the remaining patients, where at least 1 blood collection was missed, were not included. Collections were missed due to the exigencies of cardiac surgery which prevent surgeons from taking time to sample blood for research. Relevant demographic and clinical parameters were collected (Table 1). Levels of cTnI were determined by ELISA (Calbiotech, CA) on plasma from peripheral blood samples collected pre-surgery and immediately post-surgery.
Table 1. Demographics and baseline data for the 56 patients in the study who underwent open heart surgery.
Age (years) | 64 ± 13 |
Male | 66% |
Body mass index (kg/m2) | 28 ± 5 |
Left ventricle ejection fraction (EF) | 47 ± 18% |
Diabetes mellitus | 44% |
Hypertension | 94% |
Hyperlipidemia | 77% |
Current smokers | 25% |
Type of cardiac procedure: | |
Coronary artery bypass grafting (CABG) | 36% |
Valvular replacement | 39% |
Combined CABG and valvular replacement | 25% |
CPB time (min) | 110 ± 40 |
AXCL (min) | 72 ± 34 |
ELISA measurement of patients’ Bb
Patients’ plasma levels of Bb were measured using the ELISA kit from Quidel (San Diego, CA) which detects the Bb component of activated fB but not native fB, nor the other fragment of fB activation, Ba.
Statistical analysis
Statistical analyses were performed using IBM SPSS Software version 20 (IBM Corp., NY). For animal studies, an independent t-test with two tails and unequal variances was used to determine the statistical significance between the results for fB-/- and WT mice. Descriptive data were summarized as the mean ± standard error of mean. For clinical studies, the patients’ demographic and relevant clinical data, together with plasma Bb levels, were entered into a Microsoft Excel database. Descriptive data were summarized as the mean ± standard deviation. A paired t-test with two tails and unequal variances was used to determine statistically significant differences in the levels of Bb and fB (from Western blotting) in coronary blood taken before and after AXCL. Post hoc power analyses performed using G*Power 3.1 [86] showed >99% power for detection of differences in the levels of Bb in coronary blood taken before and after AXCL. Statistically significant correlations between the post-AXLC levels of Bb, the post-surgery levels of cTnI and AXCL time were determined using Spearman’s correlation. Box-charts were plotted using SigmaPlot 11 software (Systat Software, CA).
Results
Factor B deficiency limits cardiac I/R-induced myocardial necrosis in mice
To test for a relationship between myocardial I/R injury and activation of the alternative pathway complement, we subjected fB-/- and congenic WT control mice to 1 hour of surgically-induced myocardial ischemia (LAD coronary ligation) followed by 24 h of reperfusion. Assays performed on blood samples from WT mice showed activation of fB (manifested as detection of the activation product Bb) at 24 h, while no fB or Bb was detected in peripheral blood of fB-/- mice (P<0.05, Fig 1a and 1b; S1 Fig; Part A in S1 File). When we examined the heart tissue for deposition of C3 activation products by immunohistochemistry, we observed that the absence of fB fully prevented C3 deposition (C3-positive staining area as a % of total section area: fB-/- mice, 0 ± 0%; WT mice, 31 ± 6%, P<0.05; Fig 1c and 1d; Part B in S1 File). Remarkably, we also observed significantly less myocardial necrosis in the fB-/- mice compared with the WT littermate controls (n = 7/per group; % ventricular tissue which was necrotic: 20 ± 4% versus 45 ± 3%, respectively, P < 0.05; Fig 1e and 1f; S2 Fig; Part C in S1 File). Together with the published literature [73, 77], the data indicates a central role for fB and the alternative pathway in the pathophysiology of myocardial I/R injury.
Fig 1. Factor B knockout mice experienced reduced myocardial necrosis and complement C3 deposition.
fB-/- mice and WT were used in a myocardial I/R model. The left anterior descending (LAD) coronary artery was occluded for 1 hr then reperfused for 24 hrs. Propidium iodide and blue fluorescent microspheres (BFM) (the latter after re-occlusion of the LAD) were injected in vivo just prior to heart harvesting to delineate the necrotic tissue and the tissue lacking circulation and therefore at risk for necrosis, respectively. (a) Circulating fB in the blood was significantly activated in WT mice (n = 4) but not in fB-/- mice (n = 4). Serum obtained from cardiac puncture at the end of reperfusion was analyzed by Western blotting as described in Materials and Methods, Section 3. Each lane in the blot represented a separate mouse. Arrows indicate fB and Bb fragments. (b) The bar chart summarizes the relative intensities of Bb fragments. Bars represent Means ± SEM (* indicates P<0.05 compared with WT controls). (c) Cryosections prepared from the heart slices were stained with a FITC-tagged anti-C3 antibody. (d) Bar graph: bars indicate the percentage of total area that is C3 positive. (e) Necrotic tissue (bright red fluorescence) was visualized under a fluorescent microscope immediately after the harvest of hearts, using a 2x objective lens, in slices obtained by dividing the heart into four (top and bottom of each slice are adjacent in the figure). Non-ischemic tissue was defined by the blue fluorescence of BFM, the non-fluorescing tissue constituting weight of tissue at risk (WAR) (n = 7 per group). (f) Bar graph: Necrotic area expressed as % WAR as defined in the Materials and Methods, Section 2.
Factor B from circulation contributes to myocardial necrosis in cardiac I/R
To determine the source of the fB which contributed to post-I/R myocardial necrosis, fB-/- mice (n = 4) were re-constituted i.v. with WT serum to provide an extracellular source of fB (i.e. no local production) and subjected to myocardial IR as above (1 h ischemia / 24 h reperfusion). WT mice (similarly subjected to myocardial I/R) acted as positive controls (n = 4/group) and sham operated fB-/- and WT mice served as negative controls. Reconstitution of fB-/- mice with WT serum restored the myocardial necrosis (Fig 2a and 2b; S3 Fig; Part D in S1 File) and activated C3 deposition in the myocardium (Fig 2c and 2d; Part E in S1 File) after cardiac I/R.
Fig 2. Factor B from circulation contributed myocardial necrosis in cardiac I/R.
fB-/- mice (n = 4) were re-constituted i.v. with 500ul WT serum (first 250ul i.v. 1 hour prior to surgery; second 250 μl i.v. 40 minutes prior to surgery) to provide an extracellular source of fB. WT mice (n = 4; similarly injected) were used as positive controls. Animals were subjected to myocardial IR (1 h ischemia/24 h reperfusion). Sham operated fB-/- (n = 5) or WT mice (n = 5) were included as negative controls. (a) Myocardial necrosis was determined as done in Fig 1. (b) Bar graph: Necrotic area expressed as % WAR as defined in Fig 1. (c) Heart cryosections were stained with a FITC-tagged anti-C3 antibody. (d) Bar graph: bars indicate the percentage of total area that is C3 positive.
To test whether I/R altered gene expression of fB in WT hearts, we measured myocardial fB mRNA levels by qPCR. These assays showed <2-fold differences (not statistically or physiologically different) between fB levels in hearts from naïve mice vs those that underwent I/R or sham surgeries (Fig 3; Part F in S1 File), further supporting the hypothesis that systemic rather than local derived fB is the key mediator of these effects.
Fig 3. mRNA expression of fB in the WT hearts after IR.
RNA were isolated from WT hearts without surgery (naïve group; n = 3), or sham operation (n = 10), or I/R operation (n = 12). cDNA were synthesized, reverse transcribed, and real-time RT-PCR were performed as described in Method Section.
Activation of fB in patients’ coronary circulation during aortic cross-clamping in cardiac surgery
To determine whether fB is activated by I/R in humans during cardiac surgery, we analyzed the Bb levels in plasma from coronary sinus blood collected prior to aortic cross clamping (AXCL) and immediately after its cessation. Consistent with the findings in mice, global heart ischemia (AXCL) and reperfusion (cessation of AXCL) was associated with higher coronary sinus blood Bb levels compared with those prior to application of AXCL ([Bb] prior to AXCL = 2.4 ± 2.0 μg/ml; [Bb] after AXCL cessation = 4.4 ± 2.9 μg/ml, P < 0.01) (Fig 4a; S4 Fig). Likewise, peripheral blood levels of Bb rose post-surgically ([Bb] prior to AXCL = 2.4 ± 1.8 μg/ml; [Bb] after AXCL = 5.0 ± 3.8 μg/ml, P<0.05. Data not shown). The pre-AXCL levels of Bb were similar between the coronary and peripheral blood but after reperfusion the Bb levels in the peripheral blood were slightly higher (P < 0.05) than in the coronary blood. The clinical relevance of this difference is unclear.
Fig 4. Activation of fB in the coronary circulation after AXCL.
(a) Bb levels increased in the coronary circulation after the AXCL of human cardiac surgery. Coronary sinus blood was collected prior to AXCL and 5 minutes after AXCL termination. Bb levels were determined by ELISA using an antibody which detects the Bb component of activated fB but not native fB, nor the other fragment of fB activation, Ba. Statistical significances were analyzed as described in Materials and Methods, Section 8. Box-charts were plotted using SigmaPlot software. The boundary of the box closest to zero indicates the 25th percentile, while the boundary of the box farthest from zero indicates the 75th percentile. Error bars above and below the box indicate the 90th and 10th percentiles. The two filled circles above and below the box indicate the 95th and 5th percentiles. The solid line within the box marks the median, and the dotted line marks the mean (average). N = 56; * indicates statistical significance (P < 0.05). (b) Plasma from the coronary sinus blood obtained before the start of AXCL and 5 minutes after AXCL cessation was analyzed by Western blotting using a polyclonal antibody that detects fB and Bb. A representative blot is depicted showing fB positive bands from a patient’s plasma. Arrows indicate fB and Bb fragments. Band intensities on scanned images of such blots were quantified and normalized to the 93 kDa fB band of a control plasma and expressed as relative intensity. (c) A bar chart summarizes the relative intensities of Bb fragments.
To determine whether these increases in Bb were caused by an increase in the production of the precursor, fB, and/or an increase in the activation rate of existing fB, we next performed Western blotting to separate and quantify the intact fB (93 kDa) and the cleaved product, active Bb (60 kDa) in coronary sinus blood samples. We reasoned that if fB production were increased during the ischemia caused by cross-clamping, then an increase in the amounts of both the intact fB and the Bb fragment would be expected in the post-AXCL samples. If fB activation to produce Bb used pre-existing fB, then an increase in the amount of the Bb fragment, but not the parent protein, would be expected.
The 60 kDa Bb band showed a significant 20% increase (Fig 4b and 4c) in intensity after AXCL (60 kDa band intensities relative to those of the normal control 93 kDa fB band: pre-AXCL = 0.74 ± 0.39; post-AXCL = 0.89 ± 0.46, P < 0.01). In addition, there was a small non-statistically significant decrease (3%) in the intensity of the 93 kDa fB band after cessation of AXCL (93 kB band intensities relative to those of the normal control 93 kDa band: pre-AXCL = 0.70 ± 0.19; post-AXCL = 0.68 ± 0.19, P = 0.201). Together, these results are consistent with the hypothesis that the increased levels of Bb after AXCL cessation are derived from activation of pre-existing fB.
Activated fB levels correlate significantly with the postoperative increase in myocardial necrosis marker cTnI
To investigate the clinical significance of fB activation in the coronary circulation, we determined whether activated fB correlated with post-surgical levels of the myocardial necrosis marker cTnI. Peripheral blood cTnI levels increased significantly following cardiac surgery compared with the immediate pre-surgery values (Table 2), consistent with previous literature [7, 8, 81]. A univariate analysis showed that coronary Bb levels immediately after release of AXCL directly correlated with post-surgical peripheral blood cTnI levels (Spearman’s rho correlation coefficient = 0.465, P < 0.01; Fig 5a). Peripheral blood cTnI levels correlated with AXCL time (Spearman’s rho correlation coefficient = 0.304, P < 0.05; Fig 5b) but Bb levels did not (P >0.05). To assess whether the AXCL time influenced the relationship between Bb and cTn1 we repeated the analyses using the mean and median ACXL times as cutoffs. These analyses showed that Bb levels correlated with cTn1 above and below the tested thresholds (not shown).
Table 2. Levels of the myocardial necrosis marker cTnI in the peripheral blood increased significantly following cardiac surgery.
Time points | cTnI level (ng/ml) | p-value |
---|---|---|
1. immediately pre-surgery | 0.89 ± 0.44 | |
2. immediately post-surgery | 7.03 ± 0.87 | <0.01* |
* indicates statistical significance between cTnI levels at post-surgery and pre-surgery levels.
Fig 5.
(a) The increases in coronary blood Bb levels immediately after AXCL correlated significantly with postoperative increases in cTn1 levels. (b) The increases in postoperative increases in cTn1 levels correlated significantly with AXCL time. Univariate analyses were carried out as described in Method. * indicates statistical significance.
Discussion
Building upon the emerging knowledge regarding mechanisms underlying acute myocardial I/R injury (reviewed in-depth elsewhere [18, 88–90]), various strategies aimed at preventing myocardial I/R injury since the 1980s have shown promise in animal models and small human trials [16–22]. Nonetheless, to date, none of the large trials targeting various mechanisms thought to be involved in I/R injury have shown benefit in reducing post-MI damage [18, 23–25, 29–31]. New mechanistic insights are required to drive novel therapeutic approaches.
Our new findings demonstrate that activation of circulating systemic fB is central to myocardial damage in a murine model of surgically-induced cardiac I/R. Factor B-/- mice showed a significant reduction in complement C3 deposition and a remarkable abrogation of myocardial necrosis compared to WT littermates (Fig 1). Reconstitution of fB-/- mice with WT serum restored both C3 deposition and myocardial damage (Fig 2). These results add to previous studies describing the critical role of the complement alternative pathway in other models of cardiac ischemic injury (e.g. permanent occlusion of coronary arteries, after heart transplantation) [91–94], and on late post-ischemic organ sequelae such as cardiac hypertrophy [95]. The alternative pathway has also been shown to be important in renal IR injury [96–100] but not gastrointestinal IR damage [101, 102] suggesting that there are important organ specific mechanistic differences that need to be explored further.
The current paradigm of complement activation after myocardial I/R injury developed from prior work by our lab, among many others, is that ischemia of cardiac cells results in exposure of neo-antigens on the cell surface that can be recognized upon reperfusion by naturally occurring circulating IgM antibodies[77, 78, 103, 104]. The lectin pathway can recognize these antibody-neoantigen complexes and appears to be the dominant pathway of C3 activation [73, 81, 105]. C3 activation is then amplified in a fB-alternative pathway-dependent manner. The exact mechanism by which fB amplifies complement activity after cardiac I/R injury (the standard model of C3 hydrolysis[106] [107–110], the properidin-directed model[111, 112, 113, 114, 115], or the IgG-mediated model[116, 117]) needs further study.
The downstream effects of complement activation after ischemia reperfusion injury has been well described and include production of the anaphylatoxins C3a/C5a as well as the membrane attack complex C5b-9 [118, 119]. Multiple experimental models have already shown that targeting these mechanisms effectively reduces tissue damage (reviewed in depth elsewhere) [118, 119], but large randomized trials using blockade of C5 did not alter the incidence or severity of I/R injury [120, 121]. Our new findings show that blocking the upstream complement amplification loop is sufficient to mitigate I/R-injury. Whether and how factor B deficiency affects the distal effector mechanisms of the complement cascade still needs to be formally tested.
Complement activation has been implicated in human I/R injury [122–128], but not without some controversy [129–133]. In clinical trials, blockade of the common terminal complement pathway using the anti-complement C5 drug Pexelizumab failed to provide statistically significant improvements in outcomes for cardiac patients undergoing coronary artery bypass graft surgery [134] or PCI [120]. It is possible that this lack of efficacy relates to the fact that targeting downstream C5 cleavage does not affect production of upstream alternative pathway-amplified complement effector molecules including C3-derived C3a and C3b. Two previous clinical studies of patients post cardiopulmonary bypass showed acute increases in systemic serum activated fB [135, 136] as early as 30mins and 1hr post-procedure and concomitant elevations in C3 and C4 activation products, which are all proximal to complement C5. Building upon these findings, we show for the first time that fB activation, as determined by Bb levels, is also increased in samples drawn directly from the coronary circulation immediately following cessation of global heart ischemia induced by ACXL (Fig 4a) supporting the hypothesis that the systemic activation of the complement alternative pathway originates in part from the reperfused heart. Whether fB participates in the pathogenesis of cardiac necrosis in humans with acute myocardial infarction and whether targeting fB will improve outcomes following acute myocardial infarction remain to be determined through future trials.
We also provide evidence that this early activated fB is derived predominantly from pre-existing fB rather than large de novo fB generation. Western blot analysis showed a rise in Bb and a trend toward less fB in serum isolated from the coronary sinus after AXCL leading to a relative rise in Bb vs fB levels (Fig 4b and 4c). Further, as noted above, the myocardial necrosis and complement deposition that was seen in hearts of WT mice after I/R was able to be fully restored in fB-/- mice after reconstitution with WT serum alone (Fig 2c and 2d). Correspondingly we did not detect increased fB mRNA in WT hearts experiencing I/R relative to sham surgery or naïve mice (Fig 3) supporting the argument that systemic fB is the dominant source involved in the injury.
We also show that the Bb levels in the coronary circulation correlated with immediate postoperative levels of the myocardial necrosis marker cTnI (Fig 5) which has been shown to inversely correlate with clinical outcomes [7, 8, 137, 138]. Given our murine findings suggesting the central role of alternative complement pathway activation and myocardial injury after I/R, it is intriguing to consider that activated fB may serve not only as a predictive biomarker but also a potential therapeutic target. Still, since the postoperative cTnI level correlates with length of AXCL, it is possible that the association between intracoronary fB activation (Bb levels) and cardiac necrosis (cTnI levels) reflects the no-flow period rather than a causative relationship. Further, peripheral levels of Bb rose post-surgically and were statistically significantly higher than coronary sinus samples. This could be due to additional Bb generation occurring distal to the collection point of the coronary sinus blood or may be related to a mechanism of complement activation occurring outside the reperfused organ that warrants further investigation.
In summary our new results from our animal and clinical observations provide the first evidence that fB contributes directly to the myocardial necrosis that occurs early after surgical cardiac I/R injury and provides the foundation for testing fB inhibitors to limit IR injury and improve patient outcomes.
Supporting information
It is of note that Bb levels in WT mice (n = 5) did vary from mouse to mouse, and the highest level was about 5 times more than the lowest one. The lowest Bb level in WT mice was indistinguishable from that of fB-/- mice (n = 4) by the current Western blot method using an anti-human fB antibody and a donkey anti-goat antibody conjugated with HRP (Rockland, PA) (developed with an ECL Western blotting kit from Thermo Scientific, NJ).
(EPS)
White arrows indicate BFM.
(EPS)
White arrows indicate BFM.
(EPS)
Plasma from the coronary sinus blood obtained before the start of AXCL and 5 minutes after AXCL cessation was analyzed by Western blotting using a polyclonal antibody that detects fB and Bb. Each diamond and circle represents one patient. * indicates statistical significance between the groups (P < 0.05).
(EPS)
Part A: Original data set of Fig 1b; Part B: Original data set of Fig 1d. Part C: Original data set of Fig 1f; Part D: Original data set of Fig 2b; Part E: Original data set of Fig 2d; Part F: Original data set of Fig 3.
(XLSX)
Acknowledgments
The authors would like to thank Dr. James Cottrell for his continued support, Drs. Anita Mehrotra, Jeremy Weedon and Feng Dai for helpful advice on biostatistical analyses, and the CT-ICU staff members of University Hospital SUNY Downstate and Yale-New Haven Hospital for their valuable assistance. The research was funded in part by NIH grant 1R21HL088527(MZ) and 1R21AI117695-01 (PSH and MZ) and an ECRIP Award from New York State Department of Health (KS and MZ). Dr. Yunfang Hou is an ECRIP fellow. NC is supported by T32 (5T32DK007757).
Data Availability
Unrestricted data are included in the paper and its Supporting Information files. Some data are restricted to protect participant confidentiality, and are available from the SUNY Downstate Medical Center and Yale University School of Medicine Institutional Data Access / Ethics Committee for researchers who meet the criteria for access to confidential data. Researchers may contact Dr. Ming Zhang at ming.zhang@downstate.edu to request data access. Additional non-author contact: Kevin L. Nellis, MS, CIP, Executive Director, Human Research Protection & Quality Assurance, SUNY Downstate Medical Center, Tel: (718) 613-8461; email: Kevin.Nellis@downstate.edu.
Funding Statement
The research was funded in part by NIH grant 1R21HL088527(MZ) and 1R21AI117695-01 (PSH and MZ) and an ECRIP Award from New York State Department of Health (KS and MZ). Dr. Yunfang Hou is an ECRIP fellow. NC is supported by T32 (5T32DK007757).
References
- 1.Eltzschig HK, Eckle T. Ischemia and reperfusion—from mechanism to translation. Nature medicine. 2011;17(11):1391–401. Epub 2011/11/09. doi: 10.1038/nm.2507 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Millington TM, Madsen JC. Innate immunity and cardiac allograft rejection. Kidney Int Suppl. 2010;(119):S18–21. Epub 2010/12/01. doi: 10.1038/ki.2010.417 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lee JC, Christie JD. Primary graft dysfunction. Clin Chest Med. 2011;32(2):279–93. Epub 2011/04/23. doi: 10.1016/j.ccm.2011.02.007 . [DOI] [PubMed] [Google Scholar]
- 4.Schmauss D, Weis M. Cardiac allograft vasculopathy: recent developments. Circulation. 2008;117(16):2131–41. Epub 2008/04/23. doi: 10.1161/CIRCULATIONAHA.107.711911 . [DOI] [PubMed] [Google Scholar]
- 5.Arcasoy SM, Fisher A, Hachem RR, Scavuzzo M, Ware LB. Report of the ISHLT Working Group on Primary Lung Graft Dysfunction part V: predictors and outcomes. The Journal of heart and lung transplantation: the official publication of the International Society for Heart Transplantation. 2005;24(10):1483–8. Epub 2005/10/08. doi: 10.1016/j.healun.2004.11.314 . [DOI] [PubMed] [Google Scholar]
- 6.Alcock RF, Kouzios D, Naoum C, Hillis GS, Brieger DB. Perioperative myocardial necrosis in patients at high cardiovascular risk undergoing elective non-cardiac surgery. Heart. 2012;98(10):792–8. Epub 2012/05/01. doi: 10.1136/heartjnl-2011-301577 . [DOI] [PubMed] [Google Scholar]
- 7.Vermes E, Mesguich M, Houel R, Soustelle C, Le Besnerais P, Hillion ML, et al. Cardiac troponin I release after open heart surgery: a marker of myocardial protection? The Annals of thoracic surgery. 2000;70(6):2087–90. . [DOI] [PubMed] [Google Scholar]
- 8.Greenson N, Macoviak J, Krishnaswamy P, Morrisey R, James C, Clopton P, et al. Usefulness of cardiac troponin I in patients undergoing open heart surgery. American heart journal. 2001;141(3):447–55. doi: 10.1067/mhj.2001.113071 [DOI] [PubMed] [Google Scholar]
- 9.Landesberg G, Shatz V, Akopnik I, Wolf YG, Mayer M, Berlatzky Y, et al. Association of cardiac troponin, CK-MB, and postoperative myocardial ischemia with long-term survival after major vascular surgery. Journal of the American College of Cardiology. 2003;42(9):1547–54. Epub 2003/11/11. . [DOI] [PubMed] [Google Scholar]
- 10.Domanski MJ, Mahaffey K, Hasselblad V, Brener SJ, Smith PK, Hillis G, et al. Association of myocardial enzyme elevation and survival following coronary artery bypass graft surgery. JAMA: the journal of the American Medical Association. 2011;305(6):585–91. doi: 10.1001/jama.2011.99 . [DOI] [PubMed] [Google Scholar]
- 11.Koolen BB, Labout JA, Mulder PG, Gerritse BM, Rijpstra TA, Bentala M, et al. Association of perioperative troponin and atrial fibrillation after coronary artery bypass grafting. Interactive cardiovascular and thoracic surgery. 2013;17(4):608–14. doi: 10.1093/icvts/ivt259 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Schwartz Longacre L, Kloner RA, Arai AE, Baines CP, Bolli R, Braunwald E, et al. New horizons in cardioprotection: recommendations from the 2010 National Heart, Lung, and Blood Institute Workshop. Circulation. 2011;124(10):1172–9. doi: 10.1161/CIRCULATIONAHA.111.032698 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Frohlich GM, Meier P, White SK, Yellon DM, Hausenloy DJ. Myocardial reperfusion injury: looking beyond primary PCI. European heart journal. 2013;34(23):1714–22. Epub 2013/03/29. doi: 10.1093/eurheartj/eht090 . [DOI] [PubMed] [Google Scholar]
- 14.Bernink FJ, Timmers L, Beek AM, Diamant M, Roos ST, Van Rossum AC, et al. Progression in attenuating myocardial reperfusion injury: an overview. International journal of cardiology. 2014;170(3):261–9. Epub 2013/12/03. doi: 10.1016/j.ijcard.2013.11.007 . [DOI] [PubMed] [Google Scholar]
- 15.Erbel R, Wijns W. The Year in Cardiology 2013: coronary intervention. European heart journal. 2014;35(5):313–20. Epub 2014/01/05. doi: 10.1093/eurheartj/eht550 . [DOI] [PubMed] [Google Scholar]
- 16.Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74(5):1124–36. . [DOI] [PubMed] [Google Scholar]
- 17.Pickard JM, Botker HE, Crimi G, Davidson B, Davidson SM, Dutka D, et al. Remote ischemic conditioning: from experimental observation to clinical application: report from the 8th Biennial Hatter Cardiovascular Institute Workshop. Basic research in cardiology. 2015;110(1):453 Epub 2014/12/03. doi: 10.1007/s00395-014-0453-6 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ibanez B, Heusch G, Ovize M, Van de Werf F. Evolving therapies for myocardial ischemia/reperfusion injury. Journal of the American College of Cardiology. 2015;65(14):1454–71. Epub 2015/04/11. doi: 10.1016/j.jacc.2015.02.032 . [DOI] [PubMed] [Google Scholar]
- 19.Heusch G. Cardioprotection: chances and challenges of its translation to the clinic. Lancet. 2013;381(9861):166–75. doi: 10.1016/S0140-6736(12)60916-7 . [DOI] [PubMed] [Google Scholar]
- 20.Hausenloy DJ, Erik Botker H, Condorelli G, Ferdinandy P, Garcia-Dorado D, Heusch G, et al. Translating cardioprotection for patient benefit: position paper from the Working Group of Cellular Biology of the Heart of the European Society of Cardiology. Cardiovascular research. 2013;98(1):7–27. doi: 10.1093/cvr/cvt004 . [DOI] [PubMed] [Google Scholar]
- 21.Heusch G, Botker HE, Przyklenk K, Redington A, Yellon D. Remote ischemic conditioning. Journal of the American College of Cardiology. 2015;65(2):177–95. Epub 2015/01/17. doi: 10.1016/j.jacc.2014.10.031 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ovize M, Baxter GF, Di Lisa F, Ferdinandy P, Garcia-Dorado D, Hausenloy DJ, et al. Postconditioning and protection from reperfusion injury: where do we stand? Position paper from the Working Group of Cellular Biology of the Heart of the European Society of Cardiology. Cardiovascular research. 2010;87(3):406–23. doi: 10.1093/cvr/cvq129 . [DOI] [PubMed] [Google Scholar]
- 23.Meybohm P, Bein B, Brosteanu O, Cremer J, Gruenewald M, Stoppe C, et al. A Multicenter Trial of Remote Ischemic Preconditioning for Heart Surgery. The New England journal of medicine. 2015;373(15):1397–407. doi: 10.1056/NEJMoa1413579 . [DOI] [PubMed] [Google Scholar]
- 24.Hausenloy DJ, Candilio L, Evans R, Ariti C, Jenkins DP, Kolvekar S, et al. Remote Ischemic Preconditioning and Outcomes of Cardiac Surgery. The New England journal of medicine. 2015;373(15):1408–17. doi: 10.1056/NEJMoa1413534 . [DOI] [PubMed] [Google Scholar]
- 25.Cung TT, Morel O, Cayla G, Rioufol G, Garcia-Dorado D, Angoulvant D, et al. Cyclosporine before PCI in Patients with Acute Myocardial Infarction. New Engl J Med. 2015;373(11):1021–31. doi: 10.1056/NEJMoa1505489 [DOI] [PubMed] [Google Scholar]
- 26.Schouten O, Shaw LJ, Boersma E, Bax JJ, Kertai MD, Feringa HH, et al. A meta-analysis of safety and effectiveness of perioperative beta-blocker use for the prevention of cardiac events in different types of noncardiac surgery. Coron Artery Dis. 2006;17(2):173–9. . [DOI] [PubMed] [Google Scholar]
- 27.Mitchell RG, Stoddard MF, Ben-Yehuda O, Aggarwal KB, Allenby KS, Trillo RA, et al. Esmolol in acute ischemic syndromes. American heart journal. 2002;144(5):E9 . [DOI] [PubMed] [Google Scholar]
- 28.Kuhn-Regnier F, Natour E, Dhein S, Dapunt O, Geissler HJ, LaRose K, et al. Beta-blockade versus Buckberg blood-cardioplegia in coronary bypass operation. European journal of cardio-thoracic surgery: official journal of the European Association for Cardio-thoracic Surgery. 1999;15(1):67–74. . [DOI] [PubMed] [Google Scholar]
- 29.Kain V, Prabhu SD, Halade GV. Inflammation revisited: inflammation versus resolution of inflammation following myocardial infarction. Basic research in cardiology. 2014;109(6):444 doi: 10.1007/s00395-014-0444-7 . [DOI] [PubMed] [Google Scholar]
- 30.Kain V, Ingle KA, Colas RA, Dalli J, Prabhu SD, Serhan CN, et al. Resolvin D1 activates the inflammation resolving response at splenic and ventricular site following myocardial infarction leading to improved ventricular function. Journal of molecular and cellular cardiology. 2015;84:24–35. doi: 10.1016/j.yjmcc.2015.04.003 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Robinson E, Cassidy RS, Tate M, Zhao Y, Lockhart S, Calderwood D, et al. Exendin-4 protects against post-myocardial infarction remodelling via specific actions on inflammation and the extracellular matrix. Basic research in cardiology. 2015;110(2):20 doi: 10.1007/s00395-015-0476-7 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tsokos GC. Target it all right, but do not forget the torchbearer. Circulation. 2015;131(13):1153–5. Epub 2015/04/01. doi: 10.1161/CIRCULATIONAHA.115.015613 . [DOI] [PubMed] [Google Scholar]
- 33.Sacks SH, Zhou W. The role of complement in the early immune response to transplantation. Nature reviews Immunology. 2012;12(6):431–42. Epub 2012/05/26. doi: 10.1038/nri3225 . [DOI] [PubMed] [Google Scholar]
- 34.Fleming SD, Tsokos GC. Complement, natural antibodies, autoantibodies and tissue injury. Autoimmunity reviews. 2006;5(2):89–92. Epub 2006/01/25. doi: 10.1016/j.autrev.2005.09.006 . [DOI] [PubMed] [Google Scholar]
- 35.Cravedi P, Heeger PS. Complement as a multifaceted modulator of kidney transplant injury. The Journal of clinical investigation. 2014;124(6):2348–54. Epub 2014/06/04. doi: 10.1172/JCI72273 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Inserte J, Hernando V, Garcia-Dorado D. Contribution of calpains to myocardial ischaemia/reperfusion injury. Cardiovascular research. 2012;96(1):23–31. Epub 2012/07/13. doi: 10.1093/cvr/cvs232 . [DOI] [PubMed] [Google Scholar]
- 37.Das S, Steenbergen C, Murphy E. Does the voltage dependent anion channel modulate cardiac ischemia-reperfusion injury? Biochimica et biophysica acta. 2012;1818(6):1451–6. Epub 2011/11/22. doi: 10.1016/j.bbamem.2011.11.008 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bell JR, Vila-Petroff M, Delbridge LM. CaMKII-dependent responses to ischemia and reperfusion challenges in the heart. Frontiers in pharmacology. 2014;5:96 Epub 2014/05/17. doi: 10.3389/fphar.2014.00096 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Neuhof C, Neuhof H. Calpain system and its involvement in myocardial ischemia and reperfusion injury. World journal of cardiology. 2014;6(7):638–52. Epub 2014/07/30. doi: 10.4330/wjc.v6.i7.638 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Halestrap AP, Richardson AP. The mitochondrial permeability transition: A current perspective on its identity and role in ischaemia/reperfusion injury. Journal of molecular and cellular cardiology. 2014. Epub 2014/09/03. doi: 10.1016/j.yjmcc.2014.08.018 . [DOI] [PubMed] [Google Scholar]
- 41.Morciano G, Giorgi C, Bonora M, Punzetti S, Pavasini R, Wieckowski MR, et al. Molecular identity of the mitochondrial permeability transition pore and its role in ischemia-reperfusion injury. Journal of molecular and cellular cardiology. 2014. Epub 2014/08/31. doi: 10.1016/j.yjmcc.2014.08.015 . [DOI] [PubMed] [Google Scholar]
- 42.Alam MR, Baetz D, Ovize M. Cyclophilin D and myocardial ischemia-reperfusion injury: A fresh perspective. Journal of molecular and cellular cardiology. 2014. Epub 2014/10/05. doi: 10.1016/j.yjmcc.2014.09.026 . [DOI] [PubMed] [Google Scholar]
- 43.Calo L, Dong Y, Kumar R, Przyklenk K, Sanderson TH. Mitochondrial dynamics: an emerging paradigm in ischemia-reperfusion injury. Current pharmaceutical design. 2013;19(39):6848–57. Epub 2013/04/18. . [DOI] [PubMed] [Google Scholar]
- 44.Silachev DN, Plotnikov EY, Pevzner IB, Zorova LD, Babenko VA, Zorov SD, et al. The mitochondrion as a key regulator of ischaemic tolerance and injury. Heart, lung & circulation. 2014;23(10):897–904. Epub 2014/07/22. doi: 10.1016/j.hlc.2014.05.022 . [DOI] [PubMed] [Google Scholar]
- 45.Miura T, Tanno M. Mitochondria and GSK-3beta in cardioprotection against ischemia/reperfusion injury. Cardiovascular drugs and therapy / sponsored by the International Society of Cardiovascular Pharmacotherapy. 2010;24(3):255–63. Epub 2010/05/22. doi: 10.1007/s10557-010-6234-z . [DOI] [PubMed] [Google Scholar]
- 46.Deng Y, Theken KN, Lee CR. Cytochrome P450 epoxygenases, soluble epoxide hydrolase, and the regulation of cardiovascular inflammation. Journal of molecular and cellular cardiology. 2010;48(2):331–41. Epub 2009/11/07. doi: 10.1016/j.yjmcc.2009.10.022 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ago T, Kuroda J, Kamouchi M, Sadoshima J, Kitazono T. Pathophysiological roles of NADPH oxidase/nox family proteins in the vascular system. -Review and perspective. Circulation journal: official journal of the Japanese Circulation Society. 2011;75(8):1791–800. Epub 2011/06/16. . [DOI] [PubMed] [Google Scholar]
- 48.Braunersreuther V, Jaquet V. Reactive oxygen species in myocardial reperfusion injury: from physiopathology to therapeutic approaches. Current pharmaceutical biotechnology. 2012;13(1):97–114. Epub 2011/04/08. . [DOI] [PubMed] [Google Scholar]
- 49.Roberts BW, Mitchell J, Kilgannon JH, Chansky ME, Trzeciak S. Nitric oxide donor agents for the treatment of ischemia/reperfusion injury in human subjects: a systematic review. Shock. 2013;39(3):229–39. Epub 2013/01/30. doi: 10.1097/SHK.0b013e31827f565b . [DOI] [PubMed] [Google Scholar]
- 50.Muller AL, Hryshko LV, Dhalla NS. Extracellular and intracellular proteases in cardiac dysfunction due to ischemia-reperfusion injury. International journal of cardiology. 2013;164(1):39–47. Epub 2012/02/24. doi: 10.1016/j.ijcard.2012.01.103 . [DOI] [PubMed] [Google Scholar]
- 51.Rassaf T, Weber C, Bernhagen J. Macrophage migration inhibitory factor in myocardial ischaemia/reperfusion injury. Cardiovascular research. 2014;102(2):321–8. Epub 2014/03/29. doi: 10.1093/cvr/cvu071 . [DOI] [PubMed] [Google Scholar]
- 52.Linkermann A, Hackl MJ, Kunzendorf U, Walczak H, Krautwald S, Jevnikar AM. Necroptosis in immunity and ischemia-reperfusion injury. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2013;13(11):2797–804. Epub 2013/10/10. doi: 10.1111/ajt.12448 . [DOI] [PubMed] [Google Scholar]
- 53.Huttemann M, Helling S, Sanderson TH, Sinkler C, Samavati L, Mahapatra G, et al. Regulation of mitochondrial respiration and apoptosis through cell signaling: cytochrome c oxidase and cytochrome c in ischemia/reperfusion injury and inflammation. Biochimica et biophysica acta. 2012;1817(4):598–609. Epub 2011/07/21. doi: 10.1016/j.bbabio.2011.07.001 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Przyklenk K, Dong Y, Undyala VV, Whittaker P. Autophagy as a therapeutic target for ischaemia /reperfusion injury? Concepts, controversies, and challenges. Cardiovascular research. 2012;94(2):197–205. Epub 2012/01/05. doi: 10.1093/cvr/cvr358 . [DOI] [PubMed] [Google Scholar]
- 55.Gottlieb RA. Cell death pathways in acute ischemia/reperfusion injury. Journal of cardiovascular pharmacology and therapeutics. 2011;16(3–4):233–8. Epub 2011/08/09. doi: 10.1177/1074248411409581 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mann DL. The emerging role of innate immunity in the heart and vascular system: for whom the cell tolls. Circulation research. 2011;108(9):1133–45. Epub 2011/04/30. doi: 10.1161/CIRCRESAHA.110.226936 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wang Y, Abarbanell AM, Herrmann JL, Weil BR, Poynter J, Manukyan MC, et al. Toll-like receptor signaling pathways and the evidence linking toll-like receptor signaling to cardiac ischemia/reperfusion injury. Shock. 2010;34(6):548–57. Epub 2010/05/12. doi: 10.1097/SHK.0b013e3181e686f5 . [DOI] [PubMed] [Google Scholar]
- 58.Wang E, Feng Y, Zhang M, Zou L, Li Y, Buys ES, et al. Toll-like receptor 4 signaling confers cardiac protection against ischemic injury via inducible nitric oxide synthase- and soluble guanylate cyclase-dependent mechanisms. Anesthesiology. 2011;114(3):603–13. Epub 2011/01/29. doi: 10.1097/ALN.0b013e31820a4d5b ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ha T, Liu L, Kelley J, Kao R, Williams D, Li C. Toll-like receptors: new players in myocardial ischemia/reperfusion injury. Antioxidants & redox signaling. 2011;15(7):1875–93. Epub 2010/11/26. doi: 10.1089/ars.2010.3723 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Arslan F, de Kleijn DP, Pasterkamp G. Innate immune signaling in cardiac ischemia. Nature reviews Cardiology. 2011;8(5):292–300. Epub 2011/03/31. doi: 10.1038/nrcardio.2011.38 . [DOI] [PubMed] [Google Scholar]
- 61.Gordon JW, Shaw JA, Kirshenbaum LA. Multiple facets of NF-kappaB in the heart: to be or not to NF-kappaB. Circulation research. 2011;108(9):1122–32. Epub 2011/04/30. doi: 10.1161/CIRCRESAHA.110.226928 . [DOI] [PubMed] [Google Scholar]
- 62.Zhong C, Zhou Y, Liu H. Nuclear factor kappaB and anesthetic preconditioning during myocardial ischemia-reperfusion. Anesthesiology. 2004;100(3):540–6. Epub 2004/04/28. . [DOI] [PubMed] [Google Scholar]
- 63.Kruger B, Krick S, Dhillon N, Lerner SM, Ames S, Bromberg JS, et al. Donor Toll-like receptor 4 contributes to ischemia and reperfusion injury following human kidney transplantation. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(9):3390–5. Epub 2009/02/17. doi: 10.1073/pnas.0810169106 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Barratt-Due A, Pischke SE, Brekke OL, Thorgersen EB, Nielsen EW, Espevik T, et al. Bride and groom in systemic inflammation—the bells ring for complement and Toll in cooperation. Immunobiology. 2012;217(11):1047–56. Epub 2012/09/12. doi: 10.1016/j.imbio.2012.07.019 . [DOI] [PubMed] [Google Scholar]
- 65.Zhai Y, Shen XD, O'Connell R, Gao F, Lassman C, Busuttil RW, et al. Cutting edge: TLR4 activation mediates liver ischemia/reperfusion inflammatory response via IFN regulatory factor 3-dependent MyD88-independent pathway. J Immunol. 2004;173(12):7115–9. Epub 2004/12/09. . [DOI] [PubMed] [Google Scholar]
- 66.Schofield ZV, Woodruff TM, Halai R, Wu MC, Cooper MA. Neutrophils—a key component of ischemia-reperfusion injury. Shock. 2013;40(6):463–70. Epub 2013/10/04. doi: 10.1097/SHK.0000000000000044 . [DOI] [PubMed] [Google Scholar]
- 67.Hofmann U, Frantz S. Role of lymphocytes in myocardial injury, healing, and remodeling after myocardial infarction. Circulation research. 2015;116(2):354–67. Epub 2015/01/17. doi: 10.1161/CIRCRESAHA.116.304072 . [DOI] [PubMed] [Google Scholar]
- 68.Rao J, Lu L, Zhai Y. T cells in organ ischemia reperfusion injury. Current opinion in organ transplantation. 2014;19(2):115–20. Epub 2014/03/01. doi: 10.1097/MOT.0000000000000064 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: a key system for immune surveillance and homeostasis. Nature immunology. 2010;11(9):785–97. Epub 2010/08/20. doi: 10.1038/ni.1923 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Harboe M, Mollnes TE. The alternative complement pathway revisited. Journal of cellular and molecular medicine. 2008;12(4):1074–84. Epub 2008/04/19. doi: 10.1111/j.1582-4934.2008.00350.x . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Raedler H, Vieyra MB, Leisman S, Lakhani P, Kwan W, Yang M, et al. Anti-complement component C5 mAb synergizes with CTLA4Ig to inhibit alloreactive T cells and prolong cardiac allograft survival in mice. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2011;11(7):1397–406. Epub 2011/06/15. doi: 10.1111/j.1600-6143.2011.03561.x ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhang M, Takahashi K, Alicot EM, Vorup-Jensen T, Kessler B, Thiel S, et al. Activation of the lectin pathway by natural IgM in a model of ischemia/reperfusion injury. J Immunol. 2006;177(7):4727–34. Epub 2006/09/20. . [DOI] [PubMed] [Google Scholar]
- 73.Walsh MC, Bourcier T, Takahashi K, Shi L, Busche MN, Rother RP, et al. Mannose-binding lectin is a regulator of inflammation that accompanies myocardial ischemia and reperfusion injury. J Immunol. 2005;175(1):541–6. Epub 2005/06/24. . [DOI] [PubMed] [Google Scholar]
- 74.Moller-Kristensen M, Wang W, Ruseva M, Thiel S, Nielsen S, Takahashi K, et al. Mannan-Binding Lectin Recognizes Structures on Ischaemic Reperfused Mouse Kidneys and is Implicated in Tissue Injury. Scandinavian journal of immunology. 2005;61(5):426–34. doi: 10.1111/j.1365-3083.2005.01591.x [DOI] [PubMed] [Google Scholar]
- 75.Csencsits K, Burrell BE, Lu G, Eichwald EJ, Stahl GL, Bishop DK. The classical complement pathway in transplantation: unanticipated protective effects of C1q and role in inductive antibody therapy. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2008;8(8):1622–30. Epub 2008/06/19. doi: 10.1111/j.1600-6143.2008.02295.x ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Jane-wit D, Surovtseva YV, Qin L, Li G, Liu R, Clark P, et al. Complement membrane attack complexes activate noncanonical NF-kappaB by forming an Akt+ NIK+ signalosome on Rab5+ endosomes. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(31):9686–91. doi: 10.1073/pnas.1503535112 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zhang M, Michael LH, Grosjean SA, Kelly RA, Carroll MC, Entman ML. The role of natural IgM in myocardial ischemia-reperfusion injury. Journal of molecular and cellular cardiology. 2006;41(1):62–7. doi: 10.1016/j.yjmcc.2006.02.006 [DOI] [PubMed] [Google Scholar]
- 78.Busche MN, Pavlov V, Takahashi K, Stahl GL. Myocardial ischemia and reperfusion injury is dependent on both IgM and mannose-binding lectin. American journal of physiology Heart and circulatory physiology. 2009;297(5):H1853–9. Epub 2009/09/15. doi: 10.1152/ajpheart.00049.2009 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Pavlov VI, Tan YS, McClure EE, La Bonte LR, Zou C, Gorsuch WB, et al. Human mannose-binding lectin inhibitor prevents myocardial injury and arterial thrombogenesis in a novel animal model. The American journal of pathology. 2015;185(2):347–55. Epub 2014/12/09. doi: 10.1016/j.ajpath.2014.10.015 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Schoos MM, Munthe-Fog L, Skjoedt MO, Ripa RS, Lonborg J, Kastrup J, et al. Association between lectin complement pathway initiators, C-reactive protein and left ventricular remodeling in myocardial infarction-a magnetic resonance study. Molecular immunology. 2013;54(3–4):408–14. Epub 2013/02/13. doi: 10.1016/j.molimm.2013.01.008 . [DOI] [PubMed] [Google Scholar]
- 81.Zhang M, Hou YJ, Cavusoglu E, Lee DC, Steffensen R, Yang L, et al. MASP-2 activation is involved in ischemia-related necrotic myocardial injury in humans. International journal of cardiology. 2013;166(2):499–504. Epub 2011/12/20. doi: 10.1016/j.ijcard.2011.11.032 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Pekna M, Hietala MA, Landin A, Nilsson AK, Lagerberg C, Betsholtz C, et al. Mice deficient for the complement factor B develop and reproduce normally. Scandinavian journal of immunology. 1998;47(4):375–80. Epub 1998/05/26. . [DOI] [PubMed] [Google Scholar]
- 83.Charlagorla P, Liu J, Patel M, Rushbrook JI, Zhang M. Loss of plasma membrane integrity, complement response and formation of reactive oxygen species during early myocardial ischemia/reperfusion. Molecular immunology. 2013;56(4):507–12. Epub 2013/08/06. doi: 10.1016/j.molimm.2013.05.001 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ito WD, Schaarschmidt S, Klask R, Hansen S, Schafer HJ, Mathey D, et al. Infarct size measurement by triphenyltetrazolium chloride staining versus in vivo injection of propidium iodide. Journal of molecular and cellular cardiology. 1997;29(8):2169–75. . [DOI] [PubMed] [Google Scholar]
- 85.Wolff RA, Chien GL, van Winkle DM. Propidium iodide compares favorably with histology and triphenyl tetrazolium chloride in the assessment of experimentally-induced infarct size. Journal of molecular and cellular cardiology. 2000;32(2):225–32. doi: 10.1006/jmcc.1999.1074 [DOI] [PubMed] [Google Scholar]
- 86.Faul F, Erdfelder E, Buchner A, Lang AG. Statistical power analyses using G*Power 3.1: tests for correlation and regression analyses. Behav Res Methods. 2009;41(4):1149–60. Epub 2009/11/10. doi: 10.3758/BRM.41.4.1149 . [DOI] [PubMed] [Google Scholar]
- 87.Strainic MG, Liu J, Huang D, An F, Lalli PN, Muqim N, et al. Locally produced complement fragments C5a and C3a provide both costimulatory and survival signals to naive CD4+ T cells. Immunity. 2008;28(3):425–35. Epub 2008/03/11. doi: 10.1016/j.immuni.2008.02.001 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Cabrera-Fuentes HA, Aragones J, Bernhagen J, Boening A, Boisvert WA, Botker HE, et al. From basic mechanisms to clinical applications in heart protection, new players in cardiovascular diseases and cardiac theranostics: meeting report from the third international symposium on "New frontiers in cardiovascular research". Basic research in cardiology. 2016;111(6):69 doi: 10.1007/s00395-016-0586-x ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Brown DI, Griendling KK. Regulation of signal transduction by reactive oxygen species in the cardiovascular system. Circulation research. 2015;116(3):531–49. doi: 10.1161/CIRCRESAHA.116.303584 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ong SB, Samangouei P, Kalkhoran SB, Hausenloy DJ. The mitochondrial permeability transition pore and its role in myocardial ischemia reperfusion injury. Journal of molecular and cellular cardiology. 2015;78:23–34. doi: 10.1016/j.yjmcc.2014.11.005 . [DOI] [PubMed] [Google Scholar]
- 91.Atkinson C, He S, Morris K, Qiao F, Casey S, Goddard M, et al. Targeted complement inhibitors protect against posttransplant cardiac ischemia and reperfusion injury and reveal an important role for the alternative pathway of complement activation. J Immunol. 2010;185(11):7007–13. Epub 2010/10/22. doi: 10.4049/jimmunol.1001504 . [DOI] [PubMed] [Google Scholar]
- 92.Thurman JM, Holers VM. The central role of the alternative complement pathway in human disease. J Immunol. 2006;176(3):1305–10. . [DOI] [PubMed] [Google Scholar]
- 93.Stahl GL, Xu Y, Hao L, Miller M, Buras JA, Fung M, et al. Role for the alternative complement pathway in ischemia/reperfusion injury. The American journal of pathology. 2003;162(2):449–55. doi: 10.1016/S0002-9440(10)63839-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chen J, Crispin JC, Dalle Lucca J, Tsokos GC. A novel inhibitor of the alternative pathway of complement attenuates intestinal ischemia/reperfusion-induced injury. The Journal of surgical research. 2011;167(2):e131–6. Epub 2009/08/21. doi: 10.1016/j.jss.2009.05.041 . [DOI] [PubMed] [Google Scholar]
- 95.Singh MV, Kapoun A, Higgins L, Kutschke W, Thurman JM, Zhang R, et al. Ca2+/calmodulin-dependent kinase II triggers cell membrane injury by inducing complement factor B gene expression in the mouse heart. The Journal of clinical investigation. 2009;119(4):986–96. doi: 10.1172/JCI35814 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Thurman JM, Ljubanovic D, Edelstein CL, Gilkeson GS, Holers VM. Lack of a functional alternative complement pathway ameliorates ischemic acute renal failure in mice. J Immunol. 2003;170(3):1517–23. . [DOI] [PubMed] [Google Scholar]
- 97.Thurman JM, Ljubanovic D, Royer PA, Kraus DM, Molina H, Barry NP, et al. Altered renal tubular expression of the complement inhibitor Crry permits complement activation after ischemia/reperfusion. The Journal of clinical investigation. 2006;116(2):357–68. Epub 2006/01/31. doi: 10.1172/JCI24521 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Miwa T, Sato S, Gullipalli D, Nangaku M, Song WC. Blocking properdin, the alternative pathway, and anaphylatoxin receptors ameliorates renal ischemia-reperfusion injury in decay-accelerating factor and CD59 double-knockout mice. J Immunol. 2013;190(7):3552–9. Epub 2013/02/22. doi: 10.4049/jimmunol.1202275 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Rubin B, Smith A, Romaschin A, Walker P. Participation of the complement system in ischemia/reperfusion injury. Microcirc Endothelium Lymphatics. 1989;5(3–5):207–21. . [PubMed] [Google Scholar]
- 100.Thurman JM, Royer PA, Ljubanovic D, Dursun B, Lenderink AM, Edelstein CL, et al. Treatment with an inhibitory monoclonal antibody to mouse factor B protects mice from induction of apoptosis and renal ischemia/reperfusion injury. Journal of the American Society of Nephrology: JASN. 2006;17(3):707–15. doi: 10.1681/ASN.2005070698 [DOI] [PubMed] [Google Scholar]
- 101.Hart ML, Ceonzo KA, Shaffer LA, Takahashi K, Rother RP, Reenstra WR, et al. Gastrointestinal ischemia-reperfusion injury is lectin complement pathway dependent without involving C1q. J Immunol. 2005;174(10):6373–80. Epub 2005/05/10. . [DOI] [PubMed] [Google Scholar]
- 102.Lee H, Green DJ, Lai L, Hou YJ, Jensenius JC, Liu D, et al. Early complement factors in the local tissue immunocomplex generated during intestinal ischemia/reperfusion injury. Molecular immunology. 2010;47(5):972–81. Epub 2009/12/17. doi: 10.1016/j.molimm.2009.11.022 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Haas MS, Alicot EM, Schuerpf F, Chiu I, Li J, Moore FD, et al. Blockade of self-reactive IgM significantly reduces injury in a murine model of acute myocardial infarction. Cardiovascular research. 2010;87(4):618–27. doi: 10.1093/cvr/cvq141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zhang M, Carroll MC. Natural IgM-mediated innate autoimmunity: a new target for early intervention of ischemia-reperfusion injury. Expert opinion on biological therapy. 2007;7(10):1575–82. doi: 10.1517/14712598.7.10.1575 [DOI] [PubMed] [Google Scholar]
- 105.Schwaeble WJ, Lynch NJ, Clark JE, Marber M, Samani NJ, Ali YM, et al. Targeting of mannan-binding lectin-associated serine protease-2 confers protection from myocardial and gastrointestinal ischemia/reperfusion injury. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(18):7523–8. Epub 2011/04/20. doi: 10.1073/pnas.1101748108 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Lachmann PJ. The amplification loop of the complement pathways. Adv Immunol. 2009;104:115–49. Epub 2009/01/01. doi: 10.1016/S0065-2776(08)04004-2 . [DOI] [PubMed] [Google Scholar]
- 107.Pekna M, Nilsson L, Nilsson-Ekdahl K, Nilsson UR, Nilsson B. Evidence for iC3 generation during cardiopulmonary bypass as the result of blood-gas interaction. Clinical and experimental immunology. 1993;91(3):404–9. Epub 1993/03/01. ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Meri S, Verkkala K, Miettinen A, Valtonen V, Linder E. Complement levels and C3 breakdown products in open-heart surgery: association of C3 conversion with the postpericardiotomy syndrome. Clinical and experimental immunology. 1985;60(3):597–604. . [PMC free article] [PubMed] [Google Scholar]
- 109.Seghaye MC, Duchateau J, Grabitz RG, Faymonville ML, Messmer BJ, Buro-Rathsmann K, et al. Complement activation during cardiopulmonary bypass in infants and children. Relation to postoperative multiple system organ failure. The Journal of thoracic and cardiovascular surgery. 1993;106(6):978–87. . [PubMed] [Google Scholar]
- 110.Bruins P, te Velthuis H, Yazdanbakhsh AP, Jansen PG, van Hardevelt FW, de Beaumont EM, et al. Activation of the complement system during and after cardiopulmonary bypass surgery: postsurgery activation involves C-reactive protein and is associated with postoperative arrhythmia. Circulation. 1997;96(10):3542–8. . [DOI] [PubMed] [Google Scholar]
- 111.Holt GD, Pangburn MK, Ginsburg V. Properdin binds to sulfatide [Gal(3-SO4)beta 1–1 Cer] and has a sequence homology with other proteins that bind sulfated glycoconjugates. The Journal of biological chemistry. 1990;265(5):2852–5. . [PubMed] [Google Scholar]
- 112.Hourcade DE. The role of properdin in the assembly of the alternative pathway C3 convertases of complement. The Journal of biological chemistry. 2006;281(4):2128–32. Epub 2005/11/23. doi: 10.1074/jbc.M508928200 . [DOI] [PubMed] [Google Scholar]
- 113.Spitzer D, Mitchell LM, Atkinson JP, Hourcade DE. Properdin can initiate complement activation by binding specific target surfaces and providing a platform for de novo convertase assembly. J Immunol. 2007;179(4):2600–8. Epub 2007/08/07. . [DOI] [PubMed] [Google Scholar]
- 114.Xu W, Berger SP, Trouw LA, de Boer HC, Schlagwein N, Mutsaers C, et al. Properdin binds to late apoptotic and necrotic cells independently of C3b and regulates alternative pathway complement activation. J Immunol. 2008;180(11):7613–21. Epub 2008/05/21. . [DOI] [PubMed] [Google Scholar]
- 115.Kimura Y, Miwa T, Zhou L, Song WC. Activator-specific requirement of properdin in the initiation and amplification of the alternative pathway complement. Blood. 2008;111(2):732–40. Epub 2007/10/06. doi: 10.1182/blood-2007-05-089821 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Xiao H, Schreiber A, Heeringa P, Falk RJ, Jennette JC. Alternative complement pathway in the pathogenesis of disease mediated by anti-neutrophil cytoplasmic autoantibodies. The American journal of pathology. 2007;170(1):52–64. Epub 2007/01/04. doi: 10.2353/ajpath.2007.060573 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Banda NK, Wood AK, Takahashi K, Levitt B, Rudd PM, Royle L, et al. Initiation of the alternative pathway of murine complement by immune complexes is dependent on N-glycans in IgG antibodies. Arthritis and rheumatism. 2008;58(10):3081–9. Epub 2008/09/30. doi: 10.1002/art.23865 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Stahl GL, Shernan SK, Smith PK, Levy JH. Complement activation and cardiac surgery: a novel target for improving outcomes. Anesthesia and analgesia. 2012;115(4):759–71. doi: 10.1213/ANE.0b013e3182652b7d ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Riedemann NC, Ward PA. Complement in ischemia reperfusion injury. The American journal of pathology. 2003;162(2):363–7. doi: 10.1016/S0002-9440(10)63830-8 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Armstrong PW, Granger CB, Adams PX, Hamm C, Holmes D Jr., O'Neill WW, et al. Pexelizumab for acute ST-elevation myocardial infarction in patients undergoing primary percutaneous coronary intervention: a randomized controlled trial. JAMA: the journal of the American Medical Association. 2007;297(1):43–51. doi: 10.1001/jama.297.1.43 [DOI] [PubMed] [Google Scholar]
- 121.Smith PK, Shernan SK, Chen JC, Carrier M, Verrier ED, Adams PX, et al. Effects of C5 complement inhibitor pexelizumab on outcome in high-risk coronary artery bypass grafting: Combined results from the PRIMO-CABG I and II trials. The Journal of thoracic and cardiovascular surgery. 2011;142(1):89–98. Epub 2010/10/01. doi: 10.1016/j.jtcvs.2010.08.035 . [DOI] [PubMed] [Google Scholar]
- 122.Ibernon M, Moreso F, Seron D. Innate immunity in renal transplantation: the role of mannose-binding lectin. Transplant Rev (Orlando). 2014;28(1):21–5. Epub 2013/12/11. doi: 10.1016/j.trre.2013.10.006 . [DOI] [PubMed] [Google Scholar]
- 123.Berger SP, Roos A, Mallat MJ, Schaapherder AF, Doxiadis II, van Kooten C, et al. Low pretransplantation mannose-binding lectin levels predict superior patient and graft survival after simultaneous pancreas-kidney transplantation. Journal of the American Society of Nephrology: JASN. 2007;18(8):2416–22. Epub 2007/07/20. doi: 10.1681/ASN.2007030262 . [DOI] [PubMed] [Google Scholar]
- 124.Berger SP, Roos A, Mallat MJ, Fujita T, de Fijter JW, Daha MR. Association between mannose-binding lectin levels and graft survival in kidney transplantation. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2005;5(6):1361–6. Epub 2005/05/13. doi: 10.1111/j.1600-6143.2005.00841.x . [DOI] [PubMed] [Google Scholar]
- 125.Fildes JE, Shaw SM, Walker AH, McAlindon M, Williams SG, Keevil BG, et al. Mannose-binding lectin deficiency offers protection from acute graft rejection after heart transplantation. The Journal of heart and lung transplantation: the official publication of the International Society for Heart Transplantation. 2008;27(12):1353–6. Epub 2008/12/09. doi: 10.1016/j.healun.2008.08.011 . [DOI] [PubMed] [Google Scholar]
- 126.Loupy A, Lefaucheur C, Vernerey D, Prugger C, Duong van Huyen JP, Mooney N, et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. The New England journal of medicine. 2013;369(13):1215–26. Epub 2013/09/27. doi: 10.1056/NEJMoa1302506 . [DOI] [PubMed] [Google Scholar]
- 127.Rodriguez ER, Skojec DV, Tan CD, Zachary AA, Kasper EK, Conte JV, et al. Antibody-mediated rejection in human cardiac allografts: evaluation of immunoglobulins and complement activation products C4d and C3d as markers. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2005;5(11):2778–85. Epub 2005/10/11. doi: 10.1111/j.1600-6143.2005.01074.x ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Fitch JC, Rollins S, Matis L, Alford B, Aranki S, Collard CD, et al. Pharmacology and biological efficacy of a recombinant, humanized, single-chain antibody C5 complement inhibitor in patients undergoing coronary artery bypass graft surgery with cardiopulmonary bypass. Circulation. 1999;100(25):2499–506. . [DOI] [PubMed] [Google Scholar]
- 129.Damman J, Seelen MA. Mannan binding lectin: a two-faced regulator of renal allograft injury? Kidney international. 2013;83(2):191–3. Epub 2013/02/01. doi: 10.1038/ki.2012.397 . [DOI] [PubMed] [Google Scholar]
- 130.Bay JT, Sorensen SS, Hansen JM, Madsen HO, Garred P. Low mannose-binding lectin serum levels are associated with reduced kidney graft survival. Kidney international. 2013;83(2):264–71. Epub 2012/11/23. doi: 10.1038/ki.2012.373 . [DOI] [PubMed] [Google Scholar]
- 131.Fiane AE, Ueland T, Simonsen S, Scott H, Endresen K, Gullestad L, et al. Low mannose-binding lectin and increased complement activation correlate to allograft vasculopathy, ischaemia, and rejection after human heart transplantation. European heart journal. 2005;26(16):1660–5. doi: 10.1093/eurheartj/ehi198 [DOI] [PubMed] [Google Scholar]
- 132.Crespo M, Torio A, Mas V, Redondo D, Perez-Saez MJ, Mir M, et al. Clinical relevance of pretransplant anti-HLA donor-specific antibodies: does C1q-fixation matter? Transplant immunology. 2013;29(1–4):28–33. Epub 2013/08/03. doi: 10.1016/j.trim.2013.07.002 . [DOI] [PubMed] [Google Scholar]
- 133.Magro CM, Pope Harman A, Klinger D, Orosz C, Adams P, Waldman J, et al. Use of C4d as a diagnostic adjunct in lung allograft biopsies. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2003;3(9):1143–54. Epub 2003/08/16. . [DOI] [PubMed] [Google Scholar]
- 134.Verrier ED, Shernan SK, Taylor KM, Van de Werf F, Newman MF, Chen JC, et al. Terminal complement blockade with pexelizumab during coronary artery bypass graft surgery requiring cardiopulmonary bypass: a randomized trial. JAMA: the journal of the American Medical Association. 2004;291(19):2319–27. doi: 10.1001/jama.291.19.2319 [DOI] [PubMed] [Google Scholar]
- 135.Marcheix B, Carrier M, Martel C, Cossette M, Pellerin M, Bouchard D, et al. Effect of pericardial blood processing on postoperative inflammation and the complement pathways. The Annals of thoracic surgery. 2008;85(2):530–5. doi: 10.1016/j.athoracsur.2007.08.050 [DOI] [PubMed] [Google Scholar]
- 136.Hoedemaekers C, van Deuren M, Sprong T, Pickkers P, Mollnes TE, Klasen I, et al. The complement system is activated in a biphasic pattern after coronary artery bypass grafting. The Annals of thoracic surgery. 2010;89(3):710–6. Epub 2010/02/23. doi: 10.1016/j.athoracsur.2009.11.049 . [DOI] [PubMed] [Google Scholar]
- 137.Gollop ND, Dhullipala A, Nagrath N, Myint PK. Is periprocedural CK-MB a better indicator of prognosis after emergency and elective percutaneous coronary intervention compared with post-procedural cardiac troponins? Interactive cardiovascular and thoracic surgery. 2013;17(5):867–71. Epub 2013/07/12. doi: 10.1093/icvts/ivt303 ;. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Januzzi JL. What is the role of biomarker measurement after cardiac surgery? Minerva anestesiologica. 2011;77(3):334–41. Epub 2011/03/29. . [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
It is of note that Bb levels in WT mice (n = 5) did vary from mouse to mouse, and the highest level was about 5 times more than the lowest one. The lowest Bb level in WT mice was indistinguishable from that of fB-/- mice (n = 4) by the current Western blot method using an anti-human fB antibody and a donkey anti-goat antibody conjugated with HRP (Rockland, PA) (developed with an ECL Western blotting kit from Thermo Scientific, NJ).
(EPS)
White arrows indicate BFM.
(EPS)
White arrows indicate BFM.
(EPS)
Plasma from the coronary sinus blood obtained before the start of AXCL and 5 minutes after AXCL cessation was analyzed by Western blotting using a polyclonal antibody that detects fB and Bb. Each diamond and circle represents one patient. * indicates statistical significance between the groups (P < 0.05).
(EPS)
Part A: Original data set of Fig 1b; Part B: Original data set of Fig 1d. Part C: Original data set of Fig 1f; Part D: Original data set of Fig 2b; Part E: Original data set of Fig 2d; Part F: Original data set of Fig 3.
(XLSX)
Data Availability Statement
Unrestricted data are included in the paper and its Supporting Information files. Some data are restricted to protect participant confidentiality, and are available from the SUNY Downstate Medical Center and Yale University School of Medicine Institutional Data Access / Ethics Committee for researchers who meet the criteria for access to confidential data. Researchers may contact Dr. Ming Zhang at ming.zhang@downstate.edu to request data access. Additional non-author contact: Kevin L. Nellis, MS, CIP, Executive Director, Human Research Protection & Quality Assurance, SUNY Downstate Medical Center, Tel: (718) 613-8461; email: Kevin.Nellis@downstate.edu.