Skip to main content
Research Article
Originally Published 24 March 2011
Free Access

Disrupting the EMMPRIN (CD147)–Cyclophilin A Interaction Reduces Infarct Size and Preserves Systolic Function After Myocardial Ischemia and Reperfusion

Arteriosclerosis, Thrombosis, and Vascular Biology

Abstract

Objective—

Inflammation and proteolysis crucially contribute to myocardial ischemia and reperfusion injury. The extracellular matrix metalloproteinase inducer EMMPRIN (CD147) and its ligand cyclophilin A (CyPA) may be involved in both processes. The aim of the study was to characterize the role of the CD147 and CyPA interplay in myocardial ischemia/reperfusion (I/R) injury.

Methods and Results—

Immunohistochemistry showed enhanced expression of CD147 and CyPA in myocardial sections from human autopsies of patients who had died from acute myocardial infarction and from mice at 24 hours after I/R. At 24 hours and 7 days after I/R, the infarct size was reduced in CD147+/− mice vs CD147+/+ mice (C57Bl/6), in mice (C57Bl/6) treated with monoclonal antibody anti-CD147 vs control monoclonal antibody, and in CyPA−/− mice vs CyPA+/+ mice (129S6/SvEv), all of which are associated with reduced monocyte and neutrophil recruitment at 24 hours and with a preserved systolic function at 7 days. The combination of CyPA−/− mice with anti-CD147 treatment did not yield further protection compared with either inhibition strategy alone. In vitro, treatment with CyPA induced monocyte chemotaxis in a CD147- and phosphatidylinositol 3-kinase–dependent manner and induced monocyte rolling and adhesion to endothelium (human umbilical vein endothelial cells) under flow in a CD147-dependent manner.

Conclusion—

CD147 and its ligand CyPA are inflammatory mediators after myocardial ischemia and reperfusion and represent potential targets to prevent myocardial I/R injury.
The most effective treatment for acute myocardial infarction is early coronary reperfusion, which limits infarct size and preserves cardiac function.13 Various processes, including the mitochondrial permeability transition pore (mPTP) and oxidative burst of reactive oxygen species production, contribute to myocardial injury.47 Ischemia/reperfusion (I/R) is accompanied by an extensive inflammatory response, with leukocyte recruitment, induction of a complex inflammatory chemokine network, and generation of matrix metalloproteinases (MMP).810 Monocyte infiltration and reorganization of the extracellular matrix by matrix metalloproteinases (MMPs) is critical for repair mechanisms of the infarcted myocardium; however, the exact role of leukocyte infiltration and MMPs in these remodeling processes is not clear yet.912 Although optimal healing requires a balanced MMP-mediated extracellular matrix turnover,13 inadequate MMP activity is thought to trigger an intensified remodeling process leading to fibrosis and cardiac rupture.14,15
Cyclophilin A (CyPA) and its receptor, the extracellular MMP inducer (EMMPRIN; synonyms: CD147, basigin) represent a ligand/receptor pair that critically regulates both leukocyte recruitment and MMP activity.1618 CD147 is involved in regulating MMPs in various cell types (eg tumor cells, monocytes, endothelial cells, smooth muscle cells), growth factors such as vascular endothelial growth factor (tumor cells, endothelial cells), and nuclear factor-κB–regulated cytokines such as interleukin-6 or macrophage–colony-stimulating factor (monocytes, macrophages, foam cells).1823 We have recently found that CD147 is upregulated on circulating monocytes in patients with acute myocardial infarction and that this regulator modulates MMP-9 and MT1-MMP activity in monocytes.20 Only recently, Kandalam et al showed that an inhibitor of MMPs, tissue inhibitor of metalloproteinase-2, reduces myocardial remodeling postinfarction, primarily via inhibition of MT1-MMP.24 CD147 serves as a surface receptor for CyPA.16,17 CyPA is originally known as a chaperone and as an intracellular ligand for the immunosuppressive drug cyclosporine.25 Upon cell activation or cell damage CyPA is released and binds to its extracellular receptor CD147 on various cell types.2628 Binding of CyPA to CD147 results in outside-in signaling and induces nuclear factor-κB–regulated inflammatory early response genes, including MMP-2 and MMP-9, in various cell types, including monocytes/macrophages and smooth muscle cells.20,29,30 However, to the best of our knowledge, a functional in vivo relevance for CyPA/CD147 has not been described in myocardial infarction so far. In this study we show that CyPA/CD147 interaction is a critical mechanism in the pathophysiology of infarcted myocardium, mediates monocyte chemotaxis, and represents a promising therapeutic target to preserve myocardial function.

Materials and Methods

Hematoxylin/Eosin and Immunostaining

Paraffin-embedded cardiac sections were stained using monoclonal antibodies (mAbs) anti-CyPA (Abcam, Cambridge, MA), anti-CD147 (Abcam), anti-Mac-3 (BD Biosciences, San Jose, CA), anti-CD3 (Neomarkers, Fremont, CA), anti-polymorph nuclear (NIMP-R14, GeneTex, Inc, Irvine, CA), and isotype control mAb according to standard protocols. Rabbit anti-rat (Dako, Heverlee, Belgium), goat anti-rabbit (Dako), and donkey anti-rabbit mAbs (Amersham) were used as secondary antibodies depending on the respective primary antibody. Tissue sections were stained with a streptavidin-biotin-immunoperoxidase method (StreptABComplex/HRP, liquid diaminobenzidine, Substrate-Chromogen, Dako). Staining with hematoxylin and eosin was performed according to a standard protocol. Human cardiac tissue sections were obtained from autopsies of patients with acute myocardial infarction. The study was approved by the local ethics committee (Project No. 253/2009BO2).

Myocardial Ischemia and Reperfusion in Mice

Ten- to 12-week-old mice CyPA+/+ and CyPA−/− (129S6/SvEv, The Jackson Laboratory), C57Bl/6 mice, and the appropriate littermates were used. C57Bl/6 CD147+/− and CD147+/+ mice were a kind gift from Dr Takashi Muramatsu (Nagoya, Japan).31 After anesthesia of the mice and preparation of the left anterior descending artery (LAD), the LAD was ligated for 30 minutes. Successful occlusion was confirmed by visual inspection of color in the apex. After 24 hours or 7 days of reperfusion, the mice were euthanized, and the ischemic area (area at risk) was quantified by negative staining with 4% Evans Blue (after religation of the left coronary artery at the level marked by the suture left in place) and the size of the infarcted area was detected by triphenyltetrazolium staining (Sigma-Aldrich, St Louis, MO).32 Areas were digitally quantified by videoplanimetry. For in vivo inhibition experiments, blocking mAb anti-CD147 (RL 73.2) or its isotype control mAb (HB-189) was injected (1 μg per g of body weight) at the time of LAD ligation, 6 and 18 hours postischemia, and then daily. Rat anti-mouse mAb anti-CD147 was purified from the RL73.2 hybridoma33 by Genovac (Freiburg, Germany). Isotype control mAb obtained from American Type Culture Collection16 was purified from rat IgG2a hybridoma (HB-189) (Genovac).

Gelatin Zymography and Immunoblotting

For extraction of heart samples, the free wall of the left ventricle was minced into 1-mm3 pieces and incubated with PBS containing 0.5% Triton X-100 (Sigma) and 0.01% sodium azide in 4°C for 18 hours. The samples were centrifuged (14 000 rpm, 10 minutes, 4°C), and the supernatants were harvested. After determining concentration for each sample with the BioRad DC Protein Assay kit, 100 μg were subjected to electrophoresis as described.18 For Western blot analysis, tissue of the free wall of the left ventricle was homogenized and lysated in lysis buffer containing protease inhibitors (Roche, Freiburg, Germany). We used rabbit anti-CyPA (Abcam) and anti-N-tyrosine (Santa Cruz Biotechnology), followed by a secondary fluorescence labeled antibody. Phospho-Akt (Ser473) antibody (Cell Signaling Technology) was used at a 1:1000 dilution with a fluorescence-labeled secondary antibody (Licor). Gelatin zymography and Western blots were quantified using the ImageJ software.
Echocardiography was performed using a VEVO 770 ultrasound machine and a 30-Mhz linear transducer as described recently.34

Chemotaxis and Flow Chamber Assay

Monocytes were isolated as described.18 Cell migration was performed using 48-well-modified Boyden chamber (Neuro Probe Inc) with 2 compartments separated by a 5-μm polycarbonate membrane. Monocytes were added to the upper compartments, and medium containing cardiac homogenates (1:200) from CyPA+/+ mice and CyPA−/− mice or medium alone was added to the lower chamber. A chemotactic index was calculated for each well by dividing the number of migrating cells within each test well by the number of cells migrating to recombinant CyPA (R&D Systems GmbH). Heart homogenates were obtained from hearts of CyPA+/+ and CyPA−/− mice by using a glass homogenizer and PBS containing a cocktail of protease inhibitors (Roche). Protein content was equilibrated in every sample (10 μg/μL). For blocking studies anti-EMMPRIN (clone UM-8D6, Ancell) and appropriate isotype control were used. LY294002 (25 μmol/L, Calbiochem) and wortmannin (100 nmol/L, Calbiochem) were used for inhibition of phosphatidylinositol 3-kinase. For flow chamber assay human umbilical vein endothelial cells were allowed to adhere on glass coverslips until they were confluent and then activated with tumor necrosis factor-α (50 ng/mL, Peprotech) and interferon-γ (20 ng/mL, Peprotech) overnight (18 hours). Monocytes (2×105/mL) were perfused over glass coverslips using arterial shear rates (2000 second−1) as described.35

Statistical Analysis

Data are represented as mean±SEM. Difference of means was analyzed by unpaired Student t test. Values of P<0.05 were considered significant.
An expanded Methods section is available online at http://atvb.ahajournals.org.

Results

Expression of CyPA and CD147 Is Enhanced in Infarcted Myocardium Obtained From Humans and in Mice

Cardiac sections from human autopsies of patients who had died from acute myocardial infarction (n=5) were immunostained with anti-CyPA and anti-CD147 and compared with specimen from autopsies of 3 patients with a virtually normal myocardial histology (hematoxylin/eosin), who had died of noncardiac causes (Figure 1A, top). Both CyPA and its receptor CD147 were found to be highly expressed in myocardium derived from patients with acute myocardial infarction. Whereas CD147 was predominantly expressed on cardiomyocytes, CyPA appeared to be highly expressed on infiltrating leukocytes adjunct to the infarcted area (Figure 1A, bottom).
Figure 1. CD147 and CyPA in acute myocardial infarction and I/R injury. A, Cardiac sections from human autopsies of patients who had died of acute myocardial infarction (n=5) were stained with hematoxylin and eosin (HE) and immunostained for CyPA and CD147 and compared with specimen from autopsies of 3 patients, who had died of noncardiac reasons and who had a “normal” histology. B, Serial cardiac sections of mice after I/R injury at days 1 and 7 were stained with HE and immunostained for CD147, CyPA, macrophages (anti-Mac3), neutrophils (anti-polymorph nuclear [PMN]), and MMP-9. Representative images of n≥3 are shown.
Similar results were obtained in mice with myocardial I/R injury (Figure 1B): myocardial expression of CyPA and CD147 was substantially enhanced 24 hours after a transient 30-minute ligation of the LAD and was sustained for at least 7 days after I/R (Figure 1B). Similar to human autopsies (Figure 1A), CD147 was predominantly present on cardiomyocytes, whereas CyPA was highly expressed on infiltrating leukocytes consisting mainly of neutrophils (anti-polymorph nuclear) and macrophages (anti-MAC-3) (Figure 1B). At 7 days, CyPA was still present in the scar mainly associated with macrophages (Figure 1B). Interestingly, MMP-9 expression was found primarily in areas of infiltrating leukocytes both in the acute (day 1) and subacute (day 7) phase (Figure 1B).
These data indicate that both the receptor CD147 and its ligand CyPA are physically present in substantial amounts in acute myocardial infarction, allowing functional activities.

CyPA−/− Mice Are Protected From Myocardial I/R Injury

Based on the findings that CD147 and its ligand CyPA are upregulated in myocardial ischemia we analyzed CyPA-deficient mice (CyPA−/−)36 in the model of myocardial ischemia and reperfusion (Figure 2). After a 30-minute ligation of the left coronary artery (LAD), infarct size was determined in triphenyltetrazolium-stained myocardium on days 1 and 7 of reperfusion. Compared with wild-type (WT) mice, the infarct size was significantly attenuated in CyPA-deficient mice at day 1 (Figure 2A and 2B) and at 7 days (Figure 2C). These data were confirmed by echocardiography: the systolic left ventricular function was quantified by analysis of the fractional area shortening and was found significantly preserved at day 7 (Figure 2D).
Figure 2. CyPA−/− mice are protected from myocardial I/R injury. I/R injury was initiated in CyPA+/+ and CyPA−/− mice as described in Materials and Methods. After 1 day (d1) or 7 days (d7), mice were euthanized and analyzed for area at risk and infarct size. A, Evans Blue staining showed a comparable area at risk (related to the left ventricle in percent) after I/R injury at day 1 (d1). B and C, Quantitative analysis of infarct size (related to area at risk in percent) at day 1 (d1, B) and day 7 (d7, C) (n=7). D, Echocardiographic analysis of fractional area shortening as an indicator for left ventricular systolic function after 7 days (d7) (n=5). *P<0.05 between groups.

CD147+/− Mice and mAb Anti-CD147-Treated WT Mice Are Largely Protected From Myocardial I/R Injury

These data encouraged us to further evaluate the role of the cell-membrane-associated CyPA-receptor, CD147. We studied 2 additional animal models: (1) CD147+/− mice were compared with CD147+/+ mice, and (2) WT mice were treated with the mAb anti-CD147,37 which is known to solely inhibit the chemotactic activity of CD147.38 This antibody has been well characterized and used in various animal models to inhibit the interaction between extracellular CyPA and CD147.3739
Because our breeding did not generate any viable complete CD147−/− mice, we decided to combine the analysis of heterozygote CD147+/− mice with antibody blockade of CD147 in WT mice. In fact, CD147+/− mice showed a decreased expression of CD147 in splenocytes (data not shown).
Figure 3B shows that infarct size at day 1 and at day 7 after the I/R is reduced to a similar extent in both animal models—in mice with reduced CD147 expression (CD147+/−) and in WT mice treated with anti-CD147 compared with their respective control mice (day 1: 24% reduction in CD147+/−, 52% reduction in anti-CD147-treated mice; day 7: 36% reduction in CD147+/−, 46% reduction in anti-CD147-treated mice; Figure 3A and 3B). Moreover, decreased expression of CD147 (CD147+/−) or antibody blockade of CD147 both resulted in a preserved left ventricular systolic function as analyzed by echocardiography (Figure 3C). Interestingly, the use of mAb RL73.2 (anti-CD147), which predominantly blocks the chemotactic activity of CD147 rather than MMP induction,38 yielded results similar to those achieved in CyPA−/− or CD147+/− mice, suggesting that CD147-mediated chemotaxis is an important pathway in this model of ischemia and reperfusion. In addition, these data suggest that a pharmacological inhibitory strategy directed against CD147 may have beneficial effects after myocardial ischemia. In fact, the application of the mAb anti-CD147 directly before opening the LAD also resulted in a significant reduction of infarct size (Figure 3D), which was found to persist after a period of 28 days (Figure 3E).
Figure 3. CD147+/− mice and mAb anti-CD147-treated mice are protected from myocardial I/R injury. I/R injury was initiated in CD147+/+ and CD147+/− mice (C57BL/6) as well as in C57BL/6 mice with treatment with mAb anti-CD147 or isotype control mAb (1 μg/g IP each) as described in Materials and Methods. After 1 day (d1) or 7 days (d7), mice were euthanized and analyzed for area at risk and infarct size (n≥5 each). A, Evans Blue staining showed a comparable area at risk (in percentage of the respective control) after I/R injury in all groups of mice. B, Quantitative analysis of infarct size in percentage of the respective control. C, Echocardiographic quantitative analysis of fractional area shortening at day 7 (n≥5). D and E, C57BL/6 mice were treated with mAb anti-CD147 or an isotype control mAb (1 μg/g IP each) in a more “therapeutic setting.” First application of the antibody was performed directly before reopening of the LAD. Shown is quantitative analysis of infarct size at day 1 (n=7) (D) and day 28 (n=5) (E). *P<0.05.

Anti-CD147 Treatment Does Not Yield Further Protection From I/R in CyPA−/− Mice

To further analyze the respective contribution of CyPA and CD147 in this model of I/R, we have treated CyPA−/− mice with the mAb anti-CD147. Interestingly, this additional treatment did not further decrease infarct size compared with CyPA−/− treated with control mAb (Figure 4). These data suggest that CyPA and CD147 act through a common pathway in mediating I/R injury.
Figure 4. Anti-CD147 treatment does not yield further protection from I/R in CyPA−/− mice. I/R injury was initiated in CyPA−/− mice that were treated with mAb anti-CD147 or isotype control mAb (1 μg/g IP each) as described in Materials and Methods. After 1 day (d1), mice were euthanized and analyzed for area at risk and infarct size (n=6 each). A, Evans Blue staining showed a comparable area at risk (in percentage of the respective control) after I/R injury. B, Quantitative analysis of infarct size in percentage of area at risk. n.s. indicates not significant (P>0.05).

CyPA and CD147 Regulate Macrophage and Neutrophil Recruitment in I/R Injury

Previous data indicated that CyPA and CD147 regulate monocyte and also neutrophil recruitment in vivo.37,40 Based on our findings that CyPA and CD147 expression is upregulated in myocardial infarction (Figure 1) and that inhibition of the chemotactic activity of CD147 by a specific mAb largely protects mice from I/R injury over a 28-day period, we decided to study the influence of CyPA and CD147 on chemotactic activity in more detail.
In fact, the immunohistological analysis of cardiac sections revealed that monocyte and neutrophil recruitment was largely and to a comparable extent inhibited in the 3 mouse models: neutrophil recruitment at day 1 into the infarct border zone was reduced by 65% in CyPA−/− mice compared with CyPA+/+ mice, by 59% in CD147+/− mice compared with CD147+/+ mice, and by 52% in mice treated with anti-CD147 mAb compared with mice treated with control mAb (P<0.01, Figure 5). Similarly, macrophage recruitment was reduced by 58% in CyPA−/− mice compared with CyPA+/+ mice, by 69% (P<0.01) in CD147+/− mice compared with CD147+/+ mice, and by 55% (P<0.01) in mice treated with anti-CD147 mAb compared with mice treated with control mAb (P<0.01, Figure 5).
Figure 5. CyPA and CD147 regulate macrophage and neutrophil recruitment in I/R injury. I/R injury was initiated as described in Materials and Methods. After 1 day of reperfusion, cardiac sections of mice were analyzed for neutrophil (anti-polymorph nuclear [PMN]) and monocyte (anti-Mac3) infiltration into the infarct border zones. A, CyPA+/+ vs CyPA−/− (n=4). B, C57BL/6 CD147+/+ vs CD147+/− (n=4). C, Anti-CD147 treated mice vs isotype-treated C57BL/6 mice (n=5). *P<0.01.
It has been reported that the anti-CD147 antibody RL73.2 does not directly influence MMP activity.37,38 Interestingly, the analysis of the free wall of the left ventricle by zymography revealed no significant difference in MMP-2 and MMP-9 activity at 24 hours after I/R in anti-CD147-treated mice compared with isotype mAb–treated mice (Figure 6A and 6B).
Figure 6. Myocardial MMP activity and oxidative stress in anti-CD147 treated mice after I/R. Mice were treated with mAb anti-CD147 or isotype IgG and underwent I/R injury as described in Materials and Methods. After 24 hours, mice were euthanized, and left ventricular myocardial tissue (free wall) was analyzed. Representative zymography (n=4) (A) and analysis by optical density (O.D.) (B) showed no difference for MMP-2 and MMP-9. C and D, Representative Western blot analysis (n=3) for 3-nitrotyrosine residues and quantification by optical density. *P<0.05.
NADPH oxidase–derived reactive oxygen species play a major role for I/R injury.41 We hypothesized that a reduced presence of monocytes and neutrophils may be accompanied by a reduced oxidative activity. By analyzing the left free wall of the infarcted ventricles after 24 hours of I/R, we found a decreased presence of 3-nitrotyrosine residues as a measure for oxidative stress in anti-CD147-treated mice compared with their respective controls (Figure 6C and 6D).

CyPA Is a Cardiac Chemoattractant for Monocytes and Can Induce Monocyte Adhesion

CyPA has previously been described as a potent chemokine for monocytes.17 We now studied the relative contribution and relevance of cardiac CyPA for monocyte chemoattraction. Hearts from WT (CyPA+/+) and CyPA−/− mice were explanted and homogenized as described in Materials and Methods. In a modified Boyden chamber, CyPA+/+ cardiac homogenates exhibited a strong chemotactic activity for monocytes (Figure 7B). In contrast, CyPA−/− cardiac homogenates exhibited only a weak chemotactic activity (Figure 7B). We hypothesized that if CyPA is a relevant factor in the WT homogenates, the migration should be inhibited by the presence of recombinant CyPA in the upper chamber because of an alignment of the CyPA gradient. Indeed, adding 100 nmol/L recombinant CyPA into the upper chamber completely abolished monocytic migration toward CyPA+/+ homogenates. Similar inhibition was achieved by the presence of anti-CD147 into the upper compartment (Figure 7B). These findings suggest that cardiac CyPA can function as a relevant chemokine in a CD147-dependent manner.
Figure 7. Cardiac-derived CyPA induces chemoattraction and adhesion of monocytes. A, Expression of CyPA in cardiac homogenates of CyPA+/+ vs CyPA−/− mice. One representative immunoblot (of 3) is shown. Actin was used as internal loading control. B, Chemotaxis of human monocytes from the upper compartment (top) containing medium, recombinant CyPA (100 nmol/L), isotype-control mAb, or anti-CD147 (20 μg/mL each), as indicated, toward the lower compartment (bottom) containing medium (negative control, black bar), recombinant CyPA (100 nmol/L, dark gray bars), medium containing homogenates from CyPA+/+ (light gray bars), or CyPA−/− hearts (white bars) (n=6). C, Human isolated monocytes were pretreated with medium, dimethyl sulfoxide (DMSO), wortmannin (100 nmol/L), or LY294002 (25 μmol/L) as indicated. Then cells were treated with medium, lipopolysaccharide (positive control, 100 ng/mL), or CyPA (200 nmol/L) as indicated overnight. Cells were lysated, and phospho-Akt expression was analyzed by Western blotting. D, Chemotaxis of human monocytes from the upper compartment (top) containing medium, wortmannin (100 nmol/L), or Ly294002 (25 μmol/L), as indicated, toward the lower compartment (bottom) containing medium (negative control, black bar), recombinant CyPA (100 nmol/L, dark gray bars), or medium containing homogenates from CyPA+/+ hearts (light gray bars) (n=6). E, After stimulation with medium or CyPA (100 nmol/L, 12 hours) human monocytes (2×105 cells/mL) were perfused over activated (tumor necrosis factor-α and interferon-γ as described in Materials and Methods) human umbilical vein endothelial cells under arterial shear conditions. Cells were pretreated with medium, isotype, or anti-CD147 mAb (20 mg/mL each), as indicated. Monocyte rolling and firm adhesion were counted for 1 minute (n=5). *P<0.05 between groups.
It has been reported that CD147 mediates phosphatidylinositol 3-kinase in tumor cells and smooth muscle cells.42,43 Therefore, we analyzed the expression of phospho-Akt expression in monocytes after CyPA-treatment. Indeed, CyPA induced phospho-Akt. In addition, induction of phospho-Akt and monocytic migration were abrogated by phosphatidylinositol 3-kinase inhibitors wortmannin and LY294002 (Figure 7C and 7D).
Finally, we investigated whether CyPA-activated monocytes show enhanced adhesion under flow conditions. We perfused monocytes over activated human umbilical vein endothelial cells in a flow chamber under arterial shear stress and analyzed rolling and adherent monocytes. CyPA stimulation strongly induced monocyte rolling and adherence, which could be inhibited by preincubation with anti-CD147 antibody (Figure 7E).

Discussion

This study provides evidence that EMMPRIN (CD147) and its ligand CyPA play a pathophysiological role in the extent of myocardial injury after I/R in mice. By using various types of mouse models, we showed that (1) expression of CyPA and CD147 is enhanced in infarcted myocardium obtained from humans and on I/R injury in mice, (2) the absence of CyPA in CyPA−/− mice or decreased expression of CD147 in CD147+/− mice protects mice from myocardial injury on I/R with respect to infarct size and systolic function at 7 days, and (3) pharmacological inhibition of the chemotactic function of CD147 by mAb anti-CD147 decreases monocyte/macrophage and neutrophil recruitment and infarct size and preserves left ventricular function. Interestingly, treatment of CyPA−/− mice with mAb anti-CD147 yielded no additional benefit compared with otherwise untreated CyPA−/− mice or mAb anti-CD147-treated WT mice, which suggests a common pathway. The fact that the application of the mAb anti-CD147 at the time of reperfusion reduced myocardial injury to a comparable extent as achieved by treatment before induction of ischemia makes a future therapeutic strategy in humans at the time of reperfusion (primary peptidylprolylisomerase) feasible.3 The protective effect of mAb anti-CD147 treatment persisted over at least 28 days.
In all mouse models, monocyte/macrophage and neutrophil recruitment were reduced at 24 hours. In addition, oxidative stress as measured by the expression of 3-nitrotyrosine residues was reduced in mice treated with mAb anti-CD147 compared with control IgG. This study cannot definitely (and was not intended to) clarify the general role of leukocyte recruitment in I/R or of distinct (sub)types of leukocytes. Currently, it is well established that CD147 ligation by its extracellular ligand CyPA activates leukocyte chemotaxis.44 Constant and coworkers have characterized an anti-mouse CD147 mAb, which specifically reduced leukocyte accumulation and reduced collagen-induced arthritis and bronchial hyperreactivity in 2 mouse models.37,38 In our study, the application of this mAb, which specifically inhibits the chemotactic activity of CD147 rather than MMP induction,38 severely reduced monocyte/macrophage and neutrophil infiltration into the infarct border zone, reduced the extent of infarct size, and improved left ventricular systolic function. This specific inhibition of the chemotactic function of CD147 yielded effects similar to those found in mice with decreased expression of CD147 (CD147+/−). Given that different mouse strains were used, the extent of the inhibition of leukocyte recruitment, infarct size, and improvement of systolic function appear to be similar in CyPA−/− mice. Notably we did not find any differences in MMP activity in myocardial samples of CD147+/− mice versus CD147+/+ (not shown) or in mice treated either with an anti-CD147 mAb or isotype mAb (Figure 6A and 6B). The decreased monocyte and neutrophil recruitment may account at least in part for the observed reduced oxidative stress as assessed by 3-nitrotyrosine expression.
In vitro experiments further support the concept that leukocyte recruitment is a relevant consequence of CyPA/CD147 interaction: (1) Monocytic chemotaxis toward homogenates from CyPA+/+ hearts was inhibited in the presence of mAb anti-CD147 or by application of CyPA into the upper compartment and was reduced toward CyPA−/− cardiac homogenates (Figure 7B). (2) CyPA induces phospho-Akt in monocytes, and inhibition of phosphatidylinositol 3-kinase by wortmannin or Ly294002 inhibits CyPA-induced monocyte chemotaxis (Figure 7C and 7D). (3) Apart from chemotaxis, CyPA induces monocyte rolling and adhesion to endothelial cells (human umbilical vein endothelial cell) under arterial flow conditions in a CD147-dependent manner (Figure 7E). Notably, an involvement of signaling by reperfusion injury salvage kinases in cardioprotection was found in rodents but not in pigs.45
Our data are supported by the work of Berk and coworkers using CyPA−/− mice in 2 cardiovascular models: the absence of CyPA resulted in decreased angiotensin II-induced aneurysm formation40 and in reduced atherosclerosis in ApoE/CyPA−/− mice.46 In both models, the benefit was associated with reduced leukocyte recruitment. These 2 studies did not discriminate between intra- and extracellular effects of CyPA, nor did they investigate the role of CD147.
Here we provide evidence that CyPA and CD147 represent important factors for the recruitment of neutrophils and monocytes and the resulting myocardial injury on I/R. Although we do not provide clear evidence, the link between CyPA and CD147 is supported by 2 observations. (1) The extent of the effects on leukocyte recruitment, infarct size, and systolic function on I/R was comparable in the 2 models that interfere with CyPA (CyPA−/− versus CyPA+/+) and CD147 (mAb anti-CD147 versus isotype IgG). (2) The combined inhibitory strategy using mAb anti-CD147 in CyPA−/− mice did not yield any additional benefit, suggesting a common pathway for CyPA and CD147.
There exist some established inhibitors of CyPA, such as cyclosporine A and its analog NIM811. However, in addition to the blockade of extracellular CyPA interactions with CD147, these drugs interfere with various intracellular CyPA activities, such as inhibition of the calcium-activated protein phosphatase calcineurin (cyclosporine A) and protein folding (PPIase activity), and inhibit intracellular CyPD-mediated processes involving the mPTP.5,6 Specifically, CyPD-mediated inhibition of the mPTP provides a powerful antiischemic protection.4 Treatment with cyclosporine A or NIM811 has been shown to effectively protect from myocardial injury in various animal models of I/R,5,6,47 and cyclosporine A decreases infarct size in patients with acute myocardial infarction.48 It has been suggested that the CyPD-mediated inhibition of the mPTP mainly accounts for the protective effects.5,6
The focus of our study was to elucidate the role of the extracellular receptor of CyPA, CD147 (EMMPRIN) by avoiding potential intracellular crossreactions, specifically CyPD-mediated mitochondrial effects. To our knowledge, there is no known direct interaction between CyPA and CyPD, namely no effects of CyPA on, eg, the mPTP. Therefore, we have decided to pharmacologically interfere with the extracellular counterpart of CyPA, CD147, in addition to studying CD147+/− mice and CyPA−/− mice. The experiments using CyPA−/− mice clearly demonstrate the relevance of the entire CyPA system and should not directly influence the CyPD-dependent mPTP. The data using anti-CD147 mAb or the CD147+/− mice provide clear evidence that the CD147-mediated pathway is of relevance.
In conclusion our data demonstrate that CD147 and its ligand CyPA are crucially involved in mediating I/R injury and may represent therapeutic targets for myocardial salvage.

Acknowledgments

The technical assistance of Klaudia Posavec is highly appreciated. We thank Harald Langer for critical reading of the manuscript.

Supplemental Material

File (atv200949_supplemental_material.pdf)

Sources of Funding

This study was supported by the Deutsche Stiftung für Herzforschung (to P.S.), the Fortüne Program (to P.S.) and the Deutsche Forschungsgesellschaft (DFG) (Sonderforschungsbereich [SFB] TR19.9 to A.E.M. and M.G.) DFG Li 683/2-1 to M.G. and by National Institutes of Health Specialized Cooperative Centers Program in Reproduction Research U54 Grant HD40093 (to R.A.N.).

References

1.
Cannon CP, Gibson CM, Lambrew CT, Shoultz DA, Levy D, French WJ, Gore JM, Weaver WD, Rogers WJ, Tiefenbrunn AJ. Relationship of symptom-onset-to-balloon time and door-to-balloon time with mortality in patients undergoing angioplasty for acute myocardial infarction. JAMA.2000;283:2941–2947.
2.
Ovize M, Baxter GF, Di LF, Ferdinandy P, Garcia-Dorado D, Hausenloy DJ, Heusch G, Vinten-Johansen J, Yellon DM, Schulz R. 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. Cardiovasc Res. 2010;87:406–423.
3.
Hausenloy DJ, Baxter G, Bell R, Botker HE, Davidson SM, Downey J, Heusch G, Kitakaze M, Lecour S, Mentzer R, Mocanu MM, Ovize M, Schulz R, Shannon R, Walker M, Walkinshaw G, Yellon DM. Translating novel strategies for cardioprotection: the Hatter Workshop Recommendations. Basic Res Cardiol.2010;105:677–686.
4.
Heusch G, Boengler K, Schulz R. Inhibition of mitochondrial permeability transition pore opening: the Holy Grail of cardioprotection. Basic Res Cardiol.2010;105:151–154.
5.
Argaud L, Gateau-Roesch O, Muntean D, Chalabreysse L, Loufouat J, Robert D, Ovize M. Specific inhibition of the mitochondrial permeability transition prevents lethal reperfusion injury. J Mol Cell Cardiol.2005;38:367–374.
6.
Argaud L, Gateau-Roesch O, Raisky O, Loufouat J, Robert D, Ovize M. Postconditioning inhibits mitochondrial permeability transition. Circulation.2005;111:194–197.
7.
Heusch G, Boengler K, Schulz R. Cardioprotection: nitric oxide, protein kinases, and mitochondria. Circulation.2008;118:1915–1919.
8.
Frangogiannis NG, Smith CW, Entman ML. The inflammatory response in myocardial infarction. Cardiovasc Res.2002;53:31–47.
9.
Liehn EA, Piccinini AM, Koenen RR, Soehnlein O, Adage T, Fatu R, Curaj A, Popescu A, Zernecke A, Kungl AJ, Weber C. A new monocyte chemotactic protein-1/chemokine CC motif ligand-2 competitor limiting neointima formation and myocardial ischemia/reperfusion injury in mice. J Am Coll Cardiol.2010;56:1847–1857.
10.
Kaikita K, Hayasaki T, Okuma T, Kuziel WA, Ogawa H, Takeya M. Targeted deletion of CC chemokine receptor 2 attenuates left ventricular remodeling after experimental myocardial infarction. Am J Pathol.2004;165:439–447.
11.
Creemers E, Cleutjens J, Smits J, Heymans S, Moons L, Collen D, Daemen M, Carmeliet P. Disruption of the plasminogen gene in mice abolishes wound healing after myocardial infarction. Am J Pathol.2000;156:1865–1873.
12.
Arslan F, Smeets MB, O'Neill LA, Keogh B, McGuirk P, Timmers L, Tersteeg C, Hoefer IE, Doevendans PA, Pasterkamp G, de Kleijn DP. Myocardial ischemia/reperfusion injury is mediated by leukocytic toll-like receptor-2 and reduced by systemic administration of a novel anti-toll-like receptor-2 antibody. Circulation.2010;121:80–90.
13.
Yarbrough WM, Mukherjee R, Escobar GP, Mingoia JT, Sample JA, Hendrick JW, Dowdy KB, McLean JE, Lowry AS, O'Neill TP, Spinale FG. Selective targeting and timing of matrix metalloproteinase inhibition in post-myocardial infarction remodeling. Circulation.2003;108:1753–1759.
14.
Heymans S, Luttun A, Nuyens D, Theilmeier G, Creemers E, Moons L, Dyspersin GD, Cleutjens JP, Shipley M, Angellilo A, Levi M, Nube O, Baker A, Keshet E, Lupu F, Herbert JM, Smits JF, Shapiro SD, Baes M, Borgers M, Collen D, Daemen MJ, Carmeliet P. Inhibition of plasminogen activators or matrix metalloproteinases prevents cardiac rupture but impairs therapeutic angiogenesis and causes cardiac failure. Nat Med.1999;5:1135–1142.
15.
Matsumura S, Iwanaga S, Mochizuki S, Okamoto H, Ogawa S, Okada Y. Targeted deletion or pharmacological inhibition of MMP-2 prevents cardiac rupture after myocardial infarction in mice. J Clin Invest.2005;115:599–609.
16.
Arora K, Gwinn WM, Bower MA, Watson A, Okwumabua I, MacDonald HR, Bukrinsky MI, Constant SL. Extracellular cyclophilins contribute to the regulation of inflammatory responses. J Immunol.2005;175:517–522.
17.
Yurchenko V, Constant S, Bukrinsky M. Dealing with the family: CD147 interactions with cyclophilins. Immunology.2006;117:301–309.
18.
Seizer P, Schonberger T, Schott M, Lang MR, Langer HF, Bigalke B, Kramer BF, Borst O, Daub K, Heidenreich O, Schmidt R, Lindemann S, Herouy Y, Gawaz M, May AE. EMMPRIN and its ligand cyclophilin A regulate MT1-MMP, MMP-9 and M-CSF during foam cell formation. Atherosclerosis.2010;209:51–57.
19.
Schmidt R, Bultmann A, Fischel S, Gillitzer A, Cullen P, Walch A, Jost P, Ungerer M, Tolley ND, Lindemann S, Gawaz M, Schomig A, May AE. Extracellular matrix metalloproteinase inducer (CD147) is a novel receptor on platelets, activates platelets, and augments nuclear factor κB-dependent inflammation in monocytes. Circ Res.2008;102:302–309.
20.
Schmidt R, Bultmann A, Ungerer M, Joghetaei N, Bulbul O, Thieme S, Chavakis T, Toole BP, Gawaz M, Schomig A, May AE. Extracellular matrix metalloproteinase inducer regulates matrix metalloproteinase activity in cardiovascular cells: implications in acute myocardial infarction. Circulation.2006;113:834–841.
21.
Schmidt R, Redecke V, Breitfeld Y, Wantia N, Miethke T, Massberg S, Fischel S, Neumann FJ, Schomig A, May AE. EMMPRIN (CD 147) is a central activator of extracellular matrix degradation by Chlamydia pneumoniae-infected monocytes. Implications for plaque rupture. Thromb Haemost. 2006;95:151–158.
22.
Tang Y, Nakada MT, Kesavan P, McCabe F, Millar H, Rafferty P, Bugelski P, Yan L. Extracellular matrix metalloproteinase inducer stimulates tumor angiogenesis by elevating vascular endothelial cell growth factor and matrix metalloproteinases. Cancer Res.2005;65:3193–3199.
23.
Bougatef F, Quemener C, Kellouche S, Naimi B, Podgorniak MP, Millot G, Gabison EE, Calvo F, Dosquet C, Lebbe C, Menashi S, Mourah S. EMMPRIN promotes angiogenesis through hypoxia-inducible factor-2α-mediated regulation of soluble VEGF isoforms and their receptor VEGFR-2. Blood.2009;114:5547–5556.
24.
Kandalam V, Basu R, Abraham T, Wang X, Soloway PD, Jaworski DM, Oudit GY, Kassiri Z. TIMP2 deficiency accelerates adverse post-myocardial infarction remodeling because of enhanced MT1-MMP activity despite lack of MMP2 activation. Circ Res.2010;106:796–808.
25.
Marks AR. Cellular functions of immunophilins. Physiol Rev.1996;76:631–649.
26.
Sherry B, Yarlett N, Strupp A, Cerami A. Identification of cyclophilin as a proinflammatory secretory product of lipopolysaccharide-activated macrophages. Proc Natl Acad Sci U S A.1992;89:3511–3515.
27.
Jin ZG, Melaragno MG, Liao DF, Yan C, Haendeler J, Suh YA, Lambeth JD, Berk BC. Cyclophilin A is a secreted growth factor induced by oxidative stress. Circ Res.2000;87:789–796.
28.
Seko Y, Fujimura T, Taka H, Mineki R, Murayama K, Nagai R. Hypoxia followed by reoxygenation induces secretion of cyclophilin A from cultured rat cardiac myocytes. Biochem Biophys Res Commun.2004;317:162–168.
29.
Wang L, Wang CH, Jia JF, Ma XK, Li Y, Zhu HB, Tang H, Chen ZN, Zhu P. Contribution of cyclophilin A to the regulation of inflammatory processes in rheumatoid arthritis. J Clin Immunol.2010;30:24–33.
30.
Yang Y, Lu N, Zhou J, Chen ZN, Zhu P. Cyclophilin A up-regulates MMP-9 expression and adhesion of monocytes/macrophages via CD147 signalling pathway in rheumatoid arthritis. Rheumatology (Oxford).2008;47:1299–1310.
31.
Igakura T, Kadomatsu K, Kaname T, Muramatsu H, Fan QW, Miyauchi T, Toyama Y, Kuno N, Yuasa S, Takahashi M, Senda T, Taguchi O, Yamamura K, Arimura K, Muramatsu T. A null mutation in basigin, an immunoglobulin superfamily member, indicates its important roles in peri-implantation development and spermatogenesis. Dev Biol.1998;194:152–165.
32.
Weig HJ, Bott-Flugel L, Stadele C, Winter K, Schmidt R, Gawaz M, Laugwitz KL, Seyfarth M. Impaired platelet function reduces myocardial infarct size in Gαq knock-out mice in vivo. J Mol Cell Cardiol.2008;44:143–150.
33.
Renno T, Wilson A, Dunkel C, Coste I, Maisnier-Patin K, Benoit de Coignac A, Aubry JP, Lees RK, Bonnefoy JY, MacDonald HR, Gauchat JF. A role for CD147 in thymic development. J Immunol.2002;168:4946–4950.
34.
Thum T, Gross C, Fiedler J, Fischer T, Kissler S, Bussen M, Galuppo P, Just S, Rottbauer W, Frantz S, Castoldi M, Soutschek J, Koteliansky V, Rosenwald A, Basson MA, Licht JD, Pena JT, Rouhanifard SH, Muckenthaler MU, Tuschl T, Martin GR, Bauersachs J, Engelhardt S. MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature.2008;456:980–984.
35.
Seizer P, Borst O, Langer HF, Bultmann A, Munch G, Herouy Y, Stellos K, Kramer B, Bigalke B, Buchele B, Bachem MG, Vestweber D, Simmet T, Gawaz M, May AE. EMMPRIN (CD147) is a novel receptor for platelet GPVI and mediates platelet rolling via GPVI-EMMPRIN interaction. Thromb Haemost.2009;101:682–686.
36.
Colgan J, Asmal M, Yu B, Luban J. Cyclophilin A-deficient mice are resistant to immunosuppression by cyclosporine. J Immunol.2005;174:6030–6038.
37.
Gwinn WM, Damsker JM, Falahati R, Okwumabua I, Kelly-Welch A, Keegan AD, Vanpouille C, Lee JJ, Dent LA, Leitenberg D, Bukrinsky MI, Constant SL. Novel approach to inhibit asthma-mediated lung inflammation using anti-CD147 intervention. J Immunol.2006;177:4870–4879.
38.
Damsker JM, Okwumabua I, Pushkarsky T, Arora K, Bukrinsky MI, Constant SL. Targeting the chemotactic function of CD147 reduces collagen-induced arthritis. Immunology.2009;126:55–62.
39.
Agrawal SM, Silva C, Tourtellotte WW, Yong VW. EMMPRIN: a novel regulator of leukocyte transmigration into the CNS in multiple sclerosis and experimental autoimmune encephalomyelitis. J Neurosci.2011;31:669–677.
40.
Satoh K, Nigro P, Matoba T, O'Dell MR, Cui Z, Shi X, Mohan A, Yan C, Abe J, Illig KA, Berk BC. Cyclophilin A enhances vascular oxidative stress and the development of angiotensin II-induced aortic aneurysms. Nat Med.2009;15:649–656.
41.
Doerries C, Grote K, Hilfiker-Kleiner D, Luchtefeld M, Schaefer A, Holland SM, Sorrentino S, Manes C, Schieffer B, Drexler H, Landmesser U. Critical role of the NAD(P)H oxidase subunit p47phox for left ventricular remodeling/dysfunction and survival after myocardial infarction. Circ Res.2007;100:894–903.
42.
Venkatesan B, Valente AJ, Reddy VS, Siwik DA, Chandrasekar B. Resveratrol blocks interleukin-18-EMMPRIN cross-regulation and smooth muscle cell migration. Am J Physiol Heart Circ Physiol.2009;297:H874–H886.
43.
Tang Y, Nakada MT, Rafferty P, Laraio J, McCabe FL, Millar H, Cunningham M, Snyder LA, Bugelski P, Yan L. Regulation of vascular endothelial growth factor expression by EMMPRIN via the PI3K-Akt signaling pathway. Mol Cancer Res.2006;4:371–377.
44.
Bukrinsky MI. Cyclophilins: unexpected messengers in intercellular communications. Trends Immunol.2002;23:323–325.
45.
Skyschally A, van CP, Boengler K, Gres P, Musiolik J, Schilawa D, Schulz R, Heusch G. Ischemic postconditioning in pigs: no causal role for RISK activation. Circ Res.2009;104:15–18.
46.
Nigro P, Satoh K, O'Dell MR, Soe NN, Cui Z, Mohan A, Abe J, Alexis JD, Sparks JD, Berk BC. Cyclophilin A is an inflammatory mediator that promotes atherosclerosis in apolipoprotein E-deficient mice. J Exp Med.2011;208:53–66.
47.
Skyschally A, Schulz R, Heusch G. Cyclosporine A at reperfusion reduces infarct size in pigs. Cardiovasc Drugs Ther.2010;24:85–87.
48.
Piot C, Croisille P, Staat P, Thibault H, Rioufol G, Mewton N, Elbelghiti R, Cung TT, Bonnefoy E, Angoulvant D, Macia C, Raczka F, Sportouch C, Gahide G, Finet G, Andre-Fouet X, Revel D, Kirkorian G, Monassier JP, Derumeaux G, Ovize M. Effect of cyclosporine on reperfusion injury in acute myocardial infarction. N Engl J Med.2008;359:473–481.

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Arteriosclerosis, Thrombosis, and Vascular Biology
Arteriosclerosis, Thrombosis, and Vascular Biology
Pages: 1377 - 1386
PubMed: 21441138

Versions

You are viewing the most recent version of this article.

History

Received: 8 September 2010
Accepted: 9 March 2011
Published online: 24 March 2011
Published in print: June 2011

Permissions

Request permissions for this article.

Keywords

  1. coronary artery disease
  2. leukocytes
  3. reperfusion injury
  4. EMMPRIN cyclophilin A

Subjects

Authors

Affiliations

Peter Seizer
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Carmen Ochmann
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Tanja Schönberger
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Sebastian Zach
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Melanie Rose
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Oliver Borst
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Karin Klingel
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Reinhard Kandolf
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
H. Robson MacDonald
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Romana A. Nowak
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Stefan Engelhardt
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Florian Lang
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Meinrad Gawaz
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).
Andreas E. May
From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (P.S., C.O., T.S., S.Z., M.R., O.B., M.G., A.E.M.), Institut für Molekulare Pathologie (K.K., R.K.), Institute of Physiology (O.B., F.L.), Eberhard Karls-Universität Tübingen, Germany; Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland (H.R.M.); Department of Animal Sciences, University of Illinois, Urbana, IL (R.A.N.); Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany (S.E.).

Notes

Correspondence to Andreas E. May, MD, Medizinische Klinik III, Universitätsklinikum Tübingen, Otfried-Müller Str 10, 72076 Tübingen, Germany. E-mail [email protected]

Disclosures

None.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. The effect of overexpression of CyPA on gene expression in human umbilical vein endothelial cells, Medicine, 103, 29, (e38886), (2024).https://doi.org/10.1097/MD.0000000000038886
    Crossref
  2. Potential Alternative Receptors for SARS-CoV-2-Induced Kidney Damage: TLR-4, KIM-1/TIM-1, and CD147, Frontiers in Bioscience-Landmark, 29, 1, (2024).https://doi.org/10.31083/j.fbl2901008
    Crossref
  3. Cyclophilin A is a ligand for RAGE in thrombo-inflammation, Cardiovascular Research, 120, 4, (385-402), (2024).https://doi.org/10.1093/cvr/cvad189
    Crossref
  4. CD147 and MMPs as key factors in physiological and pathological processes, Biomedicine & Pharmacotherapy, 157, (113983), (2023).https://doi.org/10.1016/j.biopha.2022.113983
    Crossref
  5. Non‐Immunosuppressive Cyclophilin Inhibitors, Angewandte Chemie International Edition, 61, 39, (2022).https://doi.org/10.1002/anie.202201597
    Crossref
  6. Nicht‐immunsuppressive Cyclophilin‐Inhibitoren, Angewandte Chemie, 134, 39, (2022).https://doi.org/10.1002/ange.202201597
    Crossref
  7. Theranostic Contribution of Extracellular Matrix Metalloprotease Inducer-Paramagnetic Nanoparticles Against Acute Myocardial Infarction in a Pig Model of Coronary Ischemia-Reperfusion, Circulation: Cardiovascular Imaging, 15, 6, (e013379), (2022)./doi/10.1161/CIRCIMAGING.121.013379
    Abstract
  8. Exploratory Investigation of the Plasma Proteome Associated with the Endotheliopathy of Trauma, International Journal of Molecular Sciences, 23, 11, (6213), (2022).https://doi.org/10.3390/ijms23116213
    Crossref
  9. EMMPRIN Promotes the Expression of MMP-9 and Exacerbates Neurological Dysfunction in a Mouse Model of Intracerebral Hemorrhage, Neurochemical Research, 47, 8, (2383-2395), (2022).https://doi.org/10.1007/s11064-022-03630-z
    Crossref
  10. The Effects of EMMPRIN/CD147 on Late Function and Histopathological Lesions of the Renal Graft, Biology, 11, 2, (232), (2022).https://doi.org/10.3390/biology11020232
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Disrupting the EMMPRIN (CD147)–Cyclophilin A Interaction Reduces Infarct Size and Preserves Systolic Function After Myocardial Ischemia and Reperfusion
Arteriosclerosis, Thrombosis, and Vascular Biology
  • Vol. 31
  • No. 6

Purchase access to this journal for 24 hours

Arteriosclerosis, Thrombosis, and Vascular Biology
  • Vol. 31
  • No. 6
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response