1,614
Views
92
CrossRef citations to date
0
Altmetric
Review Article

The connection between C‐reactive protein and atherosclerosis

, , & , PhD
Pages 110-120 | Published online: 08 Jul 2009

Abstract

The connection between C‐reactive protein (CRP) and atherosclerosis lies on three grounds. First, the concentration of CRP in the serum, which is measured by using highly sensitive (a.k.a. ‘hs’) techniques, correlates with the occurrence of cardiovascular disease. Second, although CRP binds only to Fcγ receptor‐bearing cells and, in general, to apoptotic and damaged cells, almost every type of cultured mammalian cells has been shown to respond to CRP treatment. Many of these responses indicate proatherogenic functions of CRP but are being reinvestigated using CRP preparations that are free of endotoxins, sodium azide, and biologically active peptides derived from the protein itself. Third, CRP binds to modified forms of low‐density lipoprotein (LDL), and, when aggregated, CRP can bind to native LDL as well. Accordingly, CRP is seen with LDL and damaged cells at the atherosclerotic lesions and myocardial infarcts. In experimental rats, human CRP was found to increase the infarct size, an effect that could be abrogated by blocking CRP‐mediated complement activation. In the Apob100/100Ldlr‐/‐ murine model of atherosclerosis, human CRP was shown to be atheroprotective, and the importance of CRP‐LDL interactions in this protection was noted. Despite all this, at the end, the question whether CRP can protect humans from developing atherosclerosis remains unanswered.

Abbreviations
CRP=

C‐reactive protein

Mcrp=

monomeric CRP

PCh=

phosphocholine

Pet=

phosphoethanolamine

Apo=

apolipoprotein

LDL=

low‐density lipoprotein

E‐LDL=

enzymatically modified LDL

ox‐LDL=

oxidized LDL

Introduction

C‐reactive protein (CRP) is a pentagonal doughnut‐shaped acute phase protein whose five identical subunits are arranged in cyclic pentameric symmetry Citation1, Citation2. The abbreviation CRP denotes native pentameric CRP whose concentration in biological fluids is measured by using highly sensitive techniques. Although it has never been felt necessary to associate the name of the technique with the name of the protein, CRP is frequently called high‐sensitivity CRP (hsCRP). In this article, we use the abbreviation CRP and not hsCRP. Research in the last many years has contributed greatly to our knowledge regarding the possible involvement of CRP during the development of atherosclerosis. Atherosclerosis is a disease caused by the deposition and modifications of low‐density lipoprotein (LDL) in artery walls. Modified LDL is engulfed by macrophages to form foam cells that contribute to the development of atherosclerosis Citation3, Citation4. CRP was discovered in 1930, and within the next three decades, data on the interaction of CRP with lipids indicating a connection between CRP and atherosclerosis were published Citation5, Citation6. In this review, we begin there and end with the latest reports on the targeting of CRP for the treatment of cardiovascular disease and on the atheroprotective role of CRP seen in a mouse model of human atherosclerosis Citation7, Citation8. Numerous proinflammatory and proatherogenic functions have also been suggested for CRP, but most of them are not universally accepted yet Citation9–11. We discuss this aspect of CRP too and propose a precaution to take for generating reproducible results in experiments using pure preparations of CRP.

Key messages

  • Human C‐reactive protein (CRP) has been shown to be atheroprotective in the Apob100/100Ldlr ‐/‐ mouse model of human‐like hypercholesterolemia.

  • The capacity of aggregated CRP to bind to native low‐density lipoprotein (LDL) may be exploited to capture serum LDL cholesterol.

  • The capacity of native CRP to bind to modified LDL may be exploited to prevent the formation of LDL‐loaded macrophage foam cells.

  • To obtain reproducible data, CRP should be free of endotoxins and sodium azide and should be freshly purified to avoid contamination with the degradation products derived from the protein itself.

Biosynthesis and clinical value of CRP

The life history of CRP begins with its biosynthesis in the liver followed by secretion into the circulation Citation12. The production of CRP is increased in atherosclerosis and other cardiovascular diseases which involve low‐grade systemic inflammation Citation13–16. Using primary human hepatocytes and hepatoma cells, it has been shown that cytokines interleukin (IL)‐6, IL‐1β and IL‐17 induce CRP gene expression Citation12, Citation17–19. The increased production of CRP results in a rise in its serum concentration. The half‐life of CRP in human circulation is about 19 h Citation20. In response to inflammatory mediators, low‐level expression of CRP in cells other than hepatocytes has also been observed Citation21–23. The extrahepatic CRP‐expressing cells include atherosclerotic plaque tissue, monocytes, aortic endothelial cells, and vascular smooth muscle cells Citation21–28.

The median concentration of CRP in the general population of normal healthy individuals is 0.8 mg/L. A concentration of CRP ranging from 3 mg/L to 10 mg/L is considered slightly elevated Citation20, Citation29. Epidemiological studies have shown that the individuals who developed cardiovascular disease had slightly elevated serum CRP levels Citation30, Citation31. Although it is not known whether CRP can afford these patients a longer life, but because of the correlation between serum levels of CRP and occurrence of cardiovascular disease, the American Heart Association and the Center for Disease Control recommended physicians to routinely measure CRP along with cholesterol levels to predict the risk of future atherosclerotic events Citation32–34.

Statins, the inhibitors of a key enzyme in the cholesterol biosynthesis pathway, are used in humans as cholesterol‐lowering drugs Citation35. However, statins also lower CRP levels in humans and in IL‐1β‐treated human CRP‐transgenic mice Citation30, Citation31, Citation36, Citation37. It has been shown that statins act directly on hepatocytes or hepatoma cells and prevent cytokine‐mediated induction of CRP expression Citation37–39. Similarly, fibrates and the nitric oxide donor, sodium nitroprusside, also prevent induction of cytokine‐induced CRP expression in hepatic cells Citation37, Citation38, Citation40. The clinical value of CRP therefore may be diminished in patients taking these agents which inhibit biosynthesis of CRP even if proinflammatory cytokines are present Citation38.

Dissociation of pentameric CRP into monomeric CRP

Although CRP is secreted by hepatocytes in the form of pentameric molecules, it is susceptible to modifications both in vitro and in vivo. In vitro, while stored under nonphysiological conditions such as in the absence of calcium, the monomers in CRP are slowly dissociated by a nonproteolytic and irreversible process and release the monomeric form of CRP (mCRP) Citation41, Citation42. In the absence of calcium, both CRP and mCRP are susceptible to proteolytic degradation Citation41, Citation43, Citation44. Thus, CRP should always be stored in the presence of calcium. However, it should be noted that a concentration of calcium above 2 mM causes aggregation of CRP, and a concentration of calcium above 1 mM causes precipitation of CRP in phosphate buffers Citation45, and unpublished observations. If it becomes necessary to store CRP without calcium, then CRP should be repurified by a quick high‐performance liquid chromatography gel filtration procedure to recover native pentameric CRP just before use in the experiments.

The mCRP can also be generated by treating pentameric CRP with protein denaturants or by genetic engineering of CRP cDNA Citation46, Citation47. In vivo, the presence of mCRP has been demonstrated in both normal vascular tissue and atherosclerotic lesions Citation47, Citation48. A mechanism for the processing of CRP into mCRP in vivo has been proposed: when CRP is associated with a membrane, it is converted to mCRP Citation49, Citation50. In mice, the half‐life of mCRP in the circulation is less than 5 min compared to approximately 4 h for pentameric CRP; mCRP is rapidly cleared from the circulation and migrates to tissues Citation51.

There are many differences between mCRP and CRP in their structure, ligand recognition properties, and effector functions Citation41, Citation42, Citation47, Citation52–57. Many laboratories and most clinics use commercially available CRP measurement kits to determine the concentration of CRP in serum. Some kits use polyclonal anti‐CRP antibodies that recognize CRP irrespective of the different structural states of CRP. Thus, the levels of CRP measured by only polyclonal antibody‐based methods reflect not only pentameric CRP but also mCRP. Because there is no evidence from clinical trials that it is necessary to make a distinction between different forms of CRP in vivo, and because the concentration of mCRP in serum could be negligible, it is not necessary, at present, for the CRP measurement kits to include monoclonal antibodies specific for different forms of CRP to determine the concentration of CRP in serum.

Binding of CRP to native and modified LDL

Preliminary evidence for the interaction of CRP with lipids indicating a possible relationship between CRP and atherosclerosis came from the finding that CRP had a lipid‐flocculating property Citation6, Citation58. The lipid‐flocculating property of CRP was due to the interaction between CRP and cholesterol Citation59. The interaction between CRP and cholesterol was confirmed subsequently Citation60, Citation61. In vitro, native pentameric CRP does not bind to native LDL; however, it does bind to modified forms of LDL such as oxidized LDL (ox‐LDL) and enzymatically modified LDL (E‐LDL) in a Ca2+‐dependent and reversible manner Citation60–67. Aggregated forms of native pentameric CRP have been shown to bind to native LDL as well in whole serum Citation62. In vitro, native CRP and native LDL interact only when either one is immobilized or modified Citation45, Citation63, Citation65, Citation66. The binding of CRP to ox‐LDL has also been demonstrated in vivo in diabetes mellitus patients with atherosclerosis Citation68. The binding of CRP to traces of very low‐density lipoprotein (VLDL) from whole serum has also been shown Citation62, Citation69, Citation70.

The binding site on CRP for LDL has been explored. One of several thoroughly investigated ligand‐binding sites on CRP is the phosphocholine (PCh)‐binding site for its oldest known ligand PCh Citation71–76. There are five PCh‐binding sites in CRP, one on each monomer Citation1. Besides LDL cholesterol, CRP binds to many PCh‐containing substances such as pneumococcal C‐polysaccharide and damaged or altered cell membranes of apoptotic and necrotic cells Citation77–81 and also to many non‐PCh ligands such as phosphoethanolamine (PEt), galactose‐containing substances, extracellular matrix protein fibronectin, and complement factor H Citation44, Citation74, Citation82–90. Some of these reactions of CRP require prior binding of calcium ions to CRP and occur through the PCh‐binding site of CRP. The participation of the PCh‐binding site of CRP in binding to modified LDL has been demonstrated by the inhibition of binding of CRP to modified LDL by PCh Citation60, Citation62–65. The moieties on the LDL molecule that interact with CRP include ApoB, cholesterol and PCh Citation60, Citation61, Citation63, Citation65, Citation91.

Although the binding of CRP to E‐LDL is mediated by the PCh‐binding site in CRP, our mutagenesis studies have revealed that the amino acids in CRP that contact PCh are not critical for binding to E‐LDL, indicating that the PCh groups in E‐LDL are not necessary for CRP–E‐LDL interaction. We also found that the blocking of the PCh‐binding site of CRP with PEt, but not with PCh, changed the LDL‐binding property of CRP. The binding of PEt‐complexed CRP to E‐LDL was dramatically enhanced over that of uncomplexed CRP. PEt‐complexed CRP also acquired a new function: it bound selectively to native LDL in whole serum. The mechanism of action of PEt is not known, but considering the fact that aggregated CRP binds to native LDL it can be assumed that PEt might cause aggregation of CRP. As expected, PEt inhibited the binding of CRP to other PCh‐containing noncholesterol substances (unpublished observations). These findings raise the possibility that the administration of PEt‐based compounds to target endogenous CRP to form PEt‐CRP complexes or the administration of exogenously prepared PEt‐CRP complexes may be useful to capture native and modified LDL in vivo and, at the same time, to prevent binding of CRP to other PCh‐containing substances such as damaged cells at the myocardial infarcts.

The mCRP binds to both native and modified LDL and thus low levels of mCRP contaminant could confer native CRP obvious native LDL‐binding capacity. It has been proposed that mCRP may exert a protective role by facilitating the clearance of retained native LDL from extracellular space, and thus lower the risk of atherogenic LDL derivative formation, and that the interaction of mCRP with LDL may contribute to the regulation of LDL metabolism Citation92.

CRP and the formation of LDL‐loaded macrophage foam cells

Because CRP binds modified forms of LDL, the possibility that CRP might play a role in the uptake of modified LDL by macrophages has been investigated from time to time. In initial studies, the effect of CRP on the uptake of LDL by macrophages was examined using mixtures of CRP and LDL for the treatment of macrophages. These studies, employing antibodies to CRP, antibodies to ApoB, or labeled LDL, revealed that CRP did not prevent uptake of modified LDL by macrophages Citation45, Citation93, Citation94. When measured indirectly by Fcγ receptor IIa (FcγRIIa, CD32) internalization, CRP was found to increase LDL uptake; however, the involvement of FcγRIIa was questioned Citation45, Citation95. Whether CRP is present inside the cytoplasm of macrophage foam cells located in the atherosclerotic lesions is not known Citation96–98.

We recently investigated the effect of CRP on the accumulation of lipid droplets made up of cholesteryl esters in E‐LDL‐treated macrophages, which is a hallmark of foam cell formation. We found that, in contrast to E‐LDL alone, the CRP‐bound E‐LDL was inactive in the formation of foam cells. This function of CRP required that E‐LDL must be bound to CRP. The mere presence of CRP with E‐LDL was not sufficient to prevent foam cell formation. Thus, an inherent function of CRP is to prevent E‐LDL‐induced formation of macrophage foam cells. Because PEt‐CRP complexes bind to E‐LDL more efficiently than uncomplexed CRP, it may be possible to enhance the function of CRP to prevent foam cell formation, by administering PEt‐based compounds to target endogenous CRP to form PEt‐CRP complexes or by administering exogenously prepared PEt‐CRP complexes (99, and unpublished observations).

The mCRP has also been shown to decrease uptake of ox‐LDL by macrophages, and it has been proposed that the interaction of mCRP with ox‐LDL may contribute to retardation of the foam cell formation by reducing the aggressive macrophage response to ox‐LDL Citation92. A preventive role in foam cell formation has also been reported for serum amyloid P (SAP), a protein which in many ways is similar to CRP. SAP binds to ox‐LDL because ox‐LDL contains amyloid structures. The SAP‐bound ox‐LDL was not taken up by macrophages Citation100.

Effects of CRP on cultured mammalian cells

Almost every type of cultured mammalian cells, including vascular cells, has been shown to respond to CRP treatment. Many of the responses of vascular cells to CRP indicate proinflammatory and proatherogenic functions of CRP Citation9–11, Citation101. In addition to modulating the physiology of cultured cells, CRP also affected the growth of cells. For example, in vascular smooth muscle cells, CRP induced apoptosis Citation102 in one study and increased cellular proliferation in another Citation103. In endothelial cells, CRP triggered intracellular signaling to activate transcription factors such as NF‐κB (nuclear factor‐kappa B) and GADD153 (growth arrest and DNA damage‐inducible protein 153) and the p38 MAPK (mitogen‐activated protein kinase) pathway, and increased the expression of several genes Citation102–110. The antiatherosclerosis drugs statins and fibrates reduce proinflammatory functions of CRP in endothelial cells Citation109–112.

The results obtained from cell culture experiments using purified CRP remain a subject of concern because of the presence of sodium azide and endotoxins in CRP preparations. The main reason for the confusion about the role of CRP in atherosclerosis is that in many studies recombinant Escherichia coli‐expressed, azide‐containing CRP was used and that the results observed could be ascribed to the contamination with bacterial products and azide rather than to CRP itself Citation113–117. Thus, it is not established that CRP is a proatherogenic molecule Citation118. Indeed, it has been confirmed that some, if not all, of these in vitro functions of CRP were not due to azide and endotoxins Citation119–121. But because of the controversies, it is best to use CRP or mCRP free of azide and endotoxins for both in vitro and in vivo experiments.

Most preparations of CRP are labeled as pure because they do not contain a protein contaminant. It is important to note that, in addition to nonprotein contaminants, another major contaminant in CRP preparations is derived from CRP itself. Almost all CRP preparations contain degraded CRP and aggregated CRP generated due to storage conditions Citation41, Citation92. CRP dissociates into monomers and degrades into peptides which may form aggregates also. More recent reports have indicated that mCRP was a prerequisite for CRP to exert stimulatory effects on endothelial cells Citation55. Because the peptides and aggregates of CRP also modulate cellular activities and may have profound effects in cell culture Citation56, Citation122–130, it is critical that purification of CRP using a quick gel filtration method be repeated immediately before using CRP for the treatment of cells. The differences in the contents of these contaminants in various preparations of CRP are likely to generate irreproducible results.

Cells respond to CRP‐treatments, but there is lack of data on the binding of CRP to all kinds of responsive cells. The binding of CRP has been described only for the endothelial cells and, in general, for apoptotic cells and FcγR‐bearing cells Citation78–81, Citation131–134. The binding of CRP to FcγR had been debated some years ago Citation135–137, but in the last 5 years or so, several laboratories have reported FcγR‐mediated functions of CRP Citation56, Citation138–147. Apparently, an experimental condition can be set where CRP will exhibit its FcγR‐binding capability. The binding of CRP to endothelial cells is mediated by FcγR present on these cells, and the binding of CRP to apoptotic cells is mediated by PCh groups present on these cells Citation65, Citation106, Citation142, Citation143. It is not known whether the binding of CRP to a very small percentage of apoptotic cells in culture would change the physiology of nonapoptotic cells. So far, comparative studies involving recombinant mutants of CRP incapable of binding to cells through FcγR or through PCh have not been conducted to confirm cellular responses to CRP.

Functions of CRP in atherosclerosis in mice

CRP has been found deposited and, importantly, colocalized with LDL and macrophages in atherosclerotic lesions in humans and experimental animal models Citation64, Citation98, Citation148, Citation149. The presence of CRP at the lesions was not unexpected because of the known interaction between CRP and LDL in vitro. Although local synthesis of CRP from arterial cells and macrophages has been shown, a recent report indicated that CRP present at the atherosclerotic lesions was transported from the circulation Citation98. To determine the role of CRP in the development of atherosclerosis, human CRP has been used in various mouse models of atherosclerosis Citation8, Citation150–158.

Initially, CRP was found to be proatherogenic. There was modest acceleration in aortic atherosclerosis in male ApoE‐/‐ mice expressing high levels of CRP Citation150. That CRP increased atherosclerosis in mice was also shown in another independent study Citation151. CRP was also shown to be proatherogenic in hypercholesterolemic humans Citation152, Citation153.

Subsequently, CRP was found to be neither proatherogenic nor atheroprotective in ApoE‐/‐ mice Citation154–156. A limited amount of passively administered human CRP was not atheroprotective in ApoE‐/‐ mice Citation151. Similarly, transgenic human CRP was also not atheroprotective in ApoE‐/‐ mice Citation154–156. Thus, the cell culture‐based results implicating CRP as a pathogenetic factor in atherosclerosis could not be extended to in vivo situations. Additionally, it was shown that the lowering of CRP was not required to reduce atherosclerosis in mice transgenic for human CRP; just the lowering of cholesterol was sufficient Citation157, Citation158. The presence of CRP did not affect the efficacy of statins in reducing atherosclerosis indicating that CRP was not proatherogenic Citation157, Citation158. Interestingly, mCRP has been shown to be atheroprotective in mice Citation151.

Recently, CRP was found to be atheroprotective in atherosclerosis‐prone Apob100/100Ldlr‐/‐ mice. These mice are rich in LDL and develop hypercholesterolemia in a more human‐like manner Citation8. These findings indicated that the binding property of CRP to LDL may play a role in slowing the development of atherosclerosis in these mice. It was suggested that the ApoE‐/‐ mouse model of atherosclerosis was not the most appropriate model for investigating the functions of CRP because ApoE‐/‐ hypercholesterolemic mice were rich in VLDL and not in LDL Citation8. The use of ApoE‐/‐ mice in determining the role of human CRP in the development of atherosclerosis is not appropriate for one more reason: the differences in the ability of human CRP to activate the complement system in human and mouse serum. In human serum, after complexing with an appropriate ligand, human CRP activates the classical pathway of complement Citation76. However, human CRP does not activate complement in serum from ApoE‐/‐ mice Citation155, Citation159. Indeed, the deposition of complement C3 in the atherosclerotic lesions in these mice, and also in Apob100/100Ldlr‐/‐ mice, was not different in the presence and absence of human CRP Citation8, Citation154.

Since CRP is only a trace serum protein in mice, they are commonly used to investigate the functions of human CRP. But, recent data have shown that CRP exhibits species specificity. In humans, human CRP binds complement C1q to activate the classical complement pathway and binds lectins to activate the lectin complement pathway Citation76, Citation160, Citation161. In mice, although human CRP activates the lectin pathway, it does not bind mouse C1q and therefore cannot activate the classical complement cascade Citation160. Thus, mouse is not suitable for exploring any function of CRP that may involve CRP‐mediated activation of the classical complement pathway. It is important to establish the in vitro properties of CRP using mouse materials so that experiments to determine in vivo functions of human CRP can be interpreted in mice.

CRP and myocardial infarction

Atherosclerosis leads to myocardial infarction. Since CRP binds to injured cells, CRP gets deposited at myocardial infarcts Citation16, Citation162–164. To define the functions of CRP in myocardial infarction, rats have been employed. Rat CRP does not activate its own complement, but human CRP does activate rat complement. Injection of human CRP into rats undergoing experimental myocardial infarction increased the infarct size, and this was due to the complement‐activating property of CRP‐complexes Citation165, Citation166. Although rats have their own CRP and capable of binding to damaged cells, rat CRP did not enhance the infarct size because rat CRP did not activate its own complement Citation166. Possible interaction between human CRP and rat CRP and possible competition between the two proteins for binding to damaged cells in the rats are not known. CRP also increased thrombosis and arterial occlusion in a mouse model of vascular injury Citation167. Although the activation of complement by CRP deposited at the infarcts is not beneficial, the CRP‐mediated modulation of complement activation by E‐LDL could be protective Citation159.

CRP can have both proinflammatory and anti‐inflammatory activities Citation168, Citation169. In myocardial infarction in rats, CRP has proinflammatory activity which is dependent on complement activation and is detrimental. Under conditions of atherosclerotic plaque formation, when low levels of CRP and low levels of complement are present, the activity of CRP will most likely be anti‐inflammatory and is beneficial Citation8, Citation99, Citation159. A PCh‐based compound was recently reported to inhibit the deposition of human CRP at the rat myocardial infarcts and inhibit the subsequent activation of the rat complement Citation7. However, such a compound should only be used in the acute events because these compounds might lessen the beneficial anti‐inflammatory effects of CRP on atherosclerosis. Therefore, we suggest the use of PEt‐based compounds because these compounds will block the binding of CRP to myocardial infarcts and therefore block the proinflammatory activity of CRP. Simultaneously, PEt‐based compounds will enhance the anti‐inflammatory activity of CRP in atherosclerosis by enhancing the binding of CRP to LDL.

Future directions: pharmacologic intervention of CRP to prevent atherosclerosis

Because human CRP has been shown to be atheroprotective in the mouse model rich in LDL but not in the mouse model rich in VLDL, the capability of CRP to bind to LDL could be further exploited therapeutically to enhance the atheroprotective effects of CRP. The capacity of aggregated CRP to bind to native LDL may be exploited to capture serum LDL cholesterol. The capacity of native CRP to bind to modified LDL may be exploited to prevent the formation of LDL‐loaded macrophage foam cells. Pharmacologic intervention of CRP with small molecules such as PEt to partially block the PCh‐binding site of CRP may cause aggregation of CRP and enhance binding of CRP to both native and modified LDL. Although the fate of the CRP‐bound LDL needs to be determined, possibly CRP‐bound LDL would be catabolized or prevented from further modifications Citation170, Citation171. In conclusion, experiments using Apob100/100Ldlr‐/‐ mice should be pursued to investigate whether CRP can protect these mice from developing atherosclerosis under the conditions that enhance interaction of CRP with LDL molecules.

Acknowledgements

This research has been supported by the National Institutes of Health (R01 HL071233), USA.

References

  • Shrive A. K., Cheetham G. M. T., Holden D., Myles D. A. A., Turnell W. G., Volanakis J. E., et al. Three dimensional structure of human C‐reactive protein. Nature Struct Biol 1996; 3: 346–54
  • Lin S., Lee C‐K., Wang Y‐M., Huang L‐S., Lin Y‐H., Lee S‐Y., et al. Measurement of dimensions of pentagonal doughnut‐shaped C‐reactive protein using an atomic force microscope and a dual polarisation interferometric biosensor. Biosens Bioelectron 2006; 22: 323–7
  • Kruth H. S. Macrophage foam cells and atherosclerosis. Front Biosci 2001; 6: D429–55
  • Libby P. Inflammation in atherosclerosis. Nature 2002; 420: 868–74
  • Tillett W. S., Francis T Jr. Serological reactions in pneumonia with a non‐protein somatic fraction of pneumococcus. J Exp Med 1930; 52: 561–71
  • Riley R. F., Hokama Y., Colver V., Coleman M. K., Dowdy A. H. Seroflocculant activity of ethyl choladienate and various alcohols in the Penn test for cancer. Cancer Res 1958; 18: 833–41
  • Pepys M. B., Hirschfield G. M., Tennent G. A., Gallimore J. R., Kahan M. C., Bellotti V., et al. Targeting C‐reactive protein for the treatment of cardiovascular disease. Nature 2006; 440: 1217–21
  • Kovacs A., Tornvall P., Nilsson R., Tegnér J., Hamsten A., Björkegren J. Human C‐reactive protein slows atherosclerosis development in a mouse model with human‐like hypercholesterolemia. Proc Natl Acad Sci U S A 2007; 104: 13768–73
  • Pasceri V., Willerson J. T., Yeh E. T. H. Direct proinflammatory effect of C‐reactive protein on human endothelial cells. Circulation 2000; 102: 2165–8
  • Labarrere C. A., Zaloga G. P. C‐reactive protein: from innocent bystander to pivotal mediator of atherosclerosis. Am J Med 2004; 117: 499–507
  • Verma S., Devaraj S., Jialal I. Is C‐reactive protein an innocent bystander or proatherogenic culprit? C‐reactive protein promotes atherothrombosis. Circulation 2006; 113: 2135–50
  • Samols D., Agrawal A., Kushner I. Acute phase proteins. Cytokine references online, J. J Oppenheim, M Feldman. Academic Press, LondonUK 2002, Available from: www.apnet.com/cytokinereference
  • Libby P., Ridker P. M. Inflammation and atherosclerosis: role of C‐reactive protein in risk assessment. Am J Med 2004; 116: 9S–16
  • Fichtlscherer S., Zeiher A. M. Endothelial dysfunction in acute coronary syndromes: association with elevated C‐reactive protein levels. Ann Med 2000; 32: 515–8
  • Howard‐Alpe G. M., Sear J. W., Foex P. Methods of detecting atherosclerosis in non‐cardiac surgical patients: the role of biochemical markers. Br J Anaesth 2006; 97: 758–69
  • Sakkinen P., Abbott R. D., Curb J. D., Rodriguez B. L., Yano K., Tracy R. P. C‐reactive protein and myocardial infarction. J Clin Epidemiol 2002; 55: 445–51
  • Voleti B., Agrawal A. Regulation of basal and induced expression of C‐reactive protein through an overlapping element for OCT‐1 and NF‐κB on the proximal promoter. J Immunol 2005; 175: 3386–90
  • Singh P. P., Voleti B., Agrawal A. A novel RBP‐Jκ‐dependent switch from C/EBPβ to C/EBPζ at the C/EBP‐binding site on the C‐reactive protein promoter. J Immunol 2007; 178: 7302–9
  • Patel D. N., King C. A., Bailey S. R., Holt J. W., Venkatachalam K., Agrawal A., et al. Interleukin‐17 stimulates C‐reactive protein expression in hepatocytes and smooth muscle cells via p38 MAPK and ERK1/2‐dependent NF‐κB and C/EBPβ activation. J Biol Chem 2007; 282: 27229–38
  • Pepys M. B., Hirschfield G. M. C‐reactive protein: a critical update. J Clin Invest 2003; 111: 1805–12
  • Calabro P., Willerson J. T., Yeh E. T. H. Inflammatory cytokines stimulated C‐reactive protein production by human coronary artery smooth muscle cells. Circulation 2003; 108: 1930–2
  • Venugopal S. K., Devaraj S., Jialal I. Macrophage conditioned medium induces the expression of C‐reactive protein in human aortic endothelial cells: potential for paracrine/autocrine effects. Am J Pathol 2005; 166: 1265–71
  • Kang D‐H., Park S‐K., Lee I‐K., Johnson R. J. Uric acid‐induced C‐reactive protein expression: implication on cell proliferation and nitric oxide production of human vascular cells. J Am Soc Nephrol 2005; 16: 3553–62
  • Reynolds G. D., Vance R. P. C‐reactive protein immunohistochemical localization in normal and atherosclerotic human aortas. Arch Pathol Lab Med 1987; 111: 265–9
  • Yasojima K., Schwab C., McGeer E. G., McGeer P. L. Generation of C‐reactive protein and complement components in atherosclerotic plaques. Am J Pathol 2001; 158: 1039–51
  • Jabs W. J., Theissing E., Nitschke M., Bechtel J. F. M., Duchrow M., Mohamed S., et al. Local generation of C‐reactive protein in diseased coronary artery venous bypass grafts and normal vascular tissue. Circulation 2003; 108: 1428–31
  • Vogel C. F. A., Sciullo E., Wong P., Kuzmicky P., Kado N., Matsumura F. Induction of proinflammatory cytokines and C‐reactive protein in human macrophage cell line U937 exposed to air pollution particulates. Environ Health Perspect 2005; 113: 1536–41
  • Inoue T., Kato T., Uchida T., Sakuma M., Nakajima A., Shibazaki M., et al. Local release of C‐reactive protein from vulnerable plaque or coronary arterial wall injured by stenting. J Am Coll Cardiol 2005; 46: 239–45
  • Black S., Kushner I., Samols D. C‐reactive protein. J Biol Chem 2004; 279: 48487–90
  • Ridker P. M., Cannon C. P., Morrow D., Rifai N., Rose L. M., McCabe C. H., et al. C‐reactive protein levels and outcomes after statin therapy. N Engl J Med 2005; 352: 20–8
  • Nissen S. E., Tuzcu E. M., Schoenhagen P., Crowe T., Sasiela W. J., Tsai J., et al. Statin therapy, LDL cholesterol, C‐reactive protein, and coronary artery disease. N Engl J Med 2005; 352: 29–38
  • Pearson T. A., Mensah G. A., Hong Y., Smith S. c Jr., CDC; AHA. CDC/AHA Workshop on markers of inflammation and cardiovascular disease: application to clinical and public health practice: overview. Circulation 2004; 110: e543–4
  • De Ferranti S., Rifai N. C‐reactive protein and cardiovascular disease: a review of risk prediction and interventions. Clin Chim Acta 2002; 317: 1–15
  • Kushner I., Rzewnicki D., Samols D. What does minor elevation of C‐reactive protein signify?. Am J Med 2006; 119: 166.e17–28
  • Endres M. Statins: potential new indications in inflammatory conditions. Atheroscler Suppl 2006; 7: 31–5
  • Strandberg T. E., Vanhanen H., Tikkanen M. J. Effect of statins on C‐reactive protein in patients with coronary artery disease. Lancet 1999; 353: 118–9
  • Kleemann R., Verschuren L., de Rooij B. J., Lindeman J., de Maat M. M., Szalai A. J., et al. Evidence for anti‐inflammatory activity of statins and PPARα activators in human C‐reactive protein transgenic mice in vivo and in cultured human hepatocytes in vitro. Blood 2004; 103: 4188–94
  • Voleti B., Agrawal A. Statins and nitric oxide reduce C‐reactive protein production while inflammatory conditions persist. Mol Immunol 2006; 43: 891–6
  • Arnaud C., Burger F., Steffens S., Veillard N. R., Nguyen T. H., Trono D., et al. Statins reduce interleukin‐6‐induced C‐reactive protein in human hepatocytes: new evidence for direct antiinflammatory effects of statins. Arterioscler Thromb Vasc Biol 2005; 25: 1231–6
  • Gervois P., Kleemann R., Pilon A., Percevault F., Koenig W., Staels B., et al. Global suppression of IL‐6‐induced acute phase response gene expression after chronic in vivo treatment with the peroxisome proliferator‐activated receptor‐α activator fenofibrate. J Biol Chem 2004; 279: 16154–60
  • Taylor K. E., van den Berg C. W. Structural and functional comparison of native pentameric, denatured monomeric and biotinylated C‐reactive protein. Immunology 2007; 120: 404–11
  • Schwedler S. B., Filep J. G., Galle J., Wanner C., Potempa L. A. C‐reactive protein: a family of proteins to regulate cardiovascular function. Am J Kidney Dis 2006; 47: 212–22
  • Kinoshita C. M., Ying S. C., Hugli T. E., Siegel J. N., Potempa L. A., Jiang H., et al. Elucidation of a protease‐sensitive site involved in the binding of calcium to C‐reactive protein. Biochemistry 1989; 28: 9840–8
  • Suresh M. V., Singh S. K., Agrawal A. Interaction of calcium‐bound C‐reactive protein with fibronectin is controlled by pH: in vivo implications. J Biol Chem 2004; 279: 52552–7
  • Fu T., Borensztajn J. Macrophage uptake of low‐density lipoprotein bound to aggregated C‐reactive protein: possible mechanism of foam‐cell formation in atherosclerotic lesions. Biochem J 2002; 366: 195–201
  • Potempa L. A., Siegel J. N., Fiedel B. A., Potempa R. T., Gewurz H. Expression, detection and assay of a neoantigen (Neo‐CRP) associated with a free, human C‐reactive protein subunit. Mol Immunol 1987; 24: 531–41
  • Ji S. R., Wu Y., Potempa L. A., Liang Y. H., Zhao J. Effect of modified C‐reactive protein on complement activation: a possible complement regulatory role of modified or monomeric C‐reactive protein in atherosclerotic lesions. Arterioscler Thromb Vasc Biol 2006; 26: 935–41
  • Diehl E. E., Haines G. K 3rd., Radosevich J. A., Potempa L. A. Immunohistochemical localization of modified C‐reactive protein antigen in normal vascular tissue. Am J Med Sci 2000; 319: 79–83
  • Ji S. R., Wu Y., Zhu L., Potempa L. A., Sheng F. L., Lu W., et al. Cell membranes and liposomes dissociate C‐reactive protein (CRP) to form a new, biologically active structural intermediate: mCRPm. FASEB J 2007; 21: 284–94
  • Wang H. W., Sui S. F. Dissociation and subunit rearrangement of membrane‐bound human C‐reactive proteins. Biochem Biophys Res Comm 2001; 288: 75–9
  • Motie M., Schaul K. W., Potempa L. A. Biodistribution and clearance of 125I‐labeled C‐reactive protein and 125I‐labeled modified C‐reactive protein in CD‐1 mice. Drug Metab Dispos 1998; 26: 977–81
  • Wang H. W., Wu Y., Chen Y., Sui S. F. Polymorphism of structural forms of C‐reactive protein. Int J Mol Med 2002; 9: 665–71
  • Kresl J. J., Potempa L. A., Anderson B., Radosevich J. A. Inhibition of mouse mammary adenocarcinoma (EMT6) growth and metastases in mice by a modified form of C‐reactive protein. Tumor Biol 1999; 20: 72–87
  • Sjowall C., Wettero J. Pathogenic implications for autoantibodies against C‐reactive protein and other acute phase proteins. Clin Chim Acta 2006; 378: 13–23
  • Khreiss T., József L., Potempa L. A., Filep J. G. Conformational rearrangement in C‐reactive protein is required for proinflammatory actions on human endothelial cells. Circulation 2004; 109: 2016–22
  • Heuertz R. M., Schneider G. P., Potempa L. A., Webster R. O. Native and modified C‐reactive protein bind different receptors on human neutrophils. Int J Biochem Cell Biol 2005; 37: 320–35
  • Boguslawski G., McGlynn P. W., Potempa L. A., Filep J. G., Labarrere C. A. Conduct unbecoming: C‐reactive protein interactions with a broad range of protein molecules. J Heart Lung Transplant 2007; 26: 705–13
  • Aho K. Studies of syphilitic antibodies. IV. Evidence of reactant partner common for C‐reactive protein and certain anti‐lipoidal antibodies. Br J Vener Dis 1969; 45: 13–8
  • Tsujimoto M., Inoue K., Nojima S. C‐reactive protein induced agglutination of lipid suspensions prepared in the presence and absence of phosphatidylcholine. J Biochem 1980; 87: 1531–7
  • Taskinen S., Kovanen P. T., Jarva H., Meri S., Pentikäinen M. O. Binding of C‐reactive protein to modified low‐density‐lipoprotein particles: identification of cholesterol as a novel ligand for C‐reactive protein. Biochem J 2002; 367: 403–12
  • Taskinen S., Hyvönen M., Kovanen P. T., Meri S., Pentikäinen M. O. C‐reactive protein binds to the 3β‐OH group of cholesterol in LDL particles. Biochem Biophys Res Comm 2005; 329: 1208–16
  • De Beer F. C., Soutar A. K., Baltz M. L., Trayner I. M., Feinstein A., Pepys M. B. Low density lipoprotein and very low density lipoprotein are selectively bound by aggregated C‐reactive protein. J Exp Med 1982; 156: 230–42
  • Nunomura W., Hatakeyama M. Binding of low density lipoprotein (LDL) to C‐reactive protein (CRP): a possible binding through apolipoprotein B in LDL at phosphorylcholine‐binding site of CRP. Hokkaido Igaku Zasshi 1990; 65: 474–80
  • Bhakdi S., Torzewski M., Klouche M., Hemmes M. Complement and atherogenesis: binding of CRP to degraded, nonoxidized LDL enhances complement activation. Arterioscler Thromb Vasc Biol 1999; 19: 2348–54
  • Chang M. K., Binder C. J., Torzewski M., Witztum J. L. C‐reactive protein binds to both oxidized LDL and apoptotic cells through recognition of a common ligand: phosphorylcholine of oxidized phospholipids. Proc Natl Acad Sci U S A 2002; 99: 13043–8
  • Van Tits L., de Graff J., Toenhake H., van Heerde W., Stalenhoef A. C‐reactive protein and annexin A5 bind to distinct sites of negatively charged phospholipids present in oxidized low‐density lipoprotein. Arterioscler Thromb Vasc Biol 2005; 25: 717–22
  • Biró A., Thielens N. M., Cervenák L., Prohászka Z., Füst G., Arlaud G. J. Modified low density lipoproteins differentially bind and activate the C1 complex of complement. Mol Immunol 2007; 44: 1169–77
  • Tabuchi M., Inoue K., Usui‐Kataoka H., Kobayashi K., Teramoto M., Takasugi K., et al. The association of C‐reactive protein with an oxidative metabolite of LDL and its implication in atherosclerosis. J Lipid Res 2007; 48: 768–81
  • Cabana V. G., Gewurz H., Siegel J. N. Interaction of very low density lipoproteins (VLDL) with rabbit C‐reactive protein. J Immunol 1982; 128: 2342–8
  • Toh C. H., Samis J., Downey C., Walker J., Becker L., Brufatto N., et al. Biphasic transmittance waveform in the APTT coagulation assay is due to the formation of a Ca++‐dependent complex of C‐reactive protein with very‐low‐density lipoprotein and is a novel marker of impending disseminated intravascular coagulation. Blood 2002; 100: 2522–9
  • Volanakis J. E., Kaplan M. H. Specificity of C‐reactive protein for choline phosphate residues of pneumococcal C‐polysaccharide. Proc Soc Exp Biol Med 1971; 136: 612–4
  • Agrawal A. CRP after 2004. Mol Immunol 2005; 42: 927–30
  • Thompson D., Pepys M. B., Wood S. P. The physiological structure of human C‐reactive protein and its complex with phosphocholine. Structure 1999; 7: 169–77
  • Agrawal A., Simpson M. J., Black S., Carey M. P., Samols D. A C‐reactive protein mutant that does not bind to phosphocholine and pneumococcal C‐polysaccharide. J Immunol 2002; 169: 3217–22
  • Suresh M. V., Singh S. K., Ferguson D. A Jr., Agrawal A. Human C‐reactive protein protects mice from Streptococcus pneumoniae infection without binding to pneumococcal C‐polysaccharide. J Immunol 2007; 178: 1158–63
  • Volanakis J. E. Human C‐reactive protein: expression, structure, and function. Mol Immunol 2001; 38: 189–97
  • Volanakis J. E., Wirtz K. W. A. Interaction of C‐reactive protein with artificial phosphatidylcholine bilayers. Nature 1979; 281: 155–7
  • Nauta A. J., Daha M. R., van Kooten C., Roos A. Recognition and clearance of apoptotic cells: a role for complement and pentraxins. Trends Immunol 2003; 24: 148–54
  • Hart S. P., Alexander K. M., MacCall S. M., Dransfield I. C‐reactive protein does not opsonize early apoptotic human neutrophils, but binds only membrane‐permeable late apoptotic cells and has no effect on their phagocytosis by macrophages. J Inflamm (Lond) 2005; 2: 5
  • Ciurana C. L., Hack C. E. Competitive binding of pentraxins and IgM to newly exposed epitopes on late apoptotic cells. Cell Immunol 2006; 239: 14–21
  • Trouw L. A., Bengtsson A. A., Gelderman K. A., Dahlbäck B., Sturfelt G., Blom A. M. C4b‐binding protein and factor H compensate for the loss of membrane bound complement inhibitors to protect apoptotic cells against excessive complement attack. J Biol Chem 2007; 282: 28540–8
  • Schwalbe R. A., Dahlbäck B., Coe J. E., Nelsestuen G. L. Pentraxin family of proteins interacts specifically with phosphorylcholine and/or phosphorylethanolamine. Biochemistry 1992; 31: 4907–15
  • Culley F. J., Bodman‐Smith K. B., Ferguson M. A., Nikolaev A. V., Shantilal N., Raynes J. G. C‐reactive protein binds to phosphorylated carbohydrates. Glycobiology 2000; 10: 59–65
  • Lee R. T., Lee Y. C. Carbohydrate ligands of human C‐reactive protein: binding of neoglycoproteins containing galactose‐6‐phosphate and galactose‐terminated disaccharide. Glycoconj J 2006; 23: 317–27
  • Sjoberg A. P., Trouw L. A., Clark S. J., Sjolander J., Heinegard D., Sim R. B., et al. The factor H variant associated with age‐related macular degeneration (His‐384) and the non‐disease‐associated form bind differentially to C‐reactive protein, fibromodulin, DNA, and necrotic cells. J Biol Chem 2007; 282: 10894–900
  • Mold C., Gewurz H., Du Clos T. W. Regulation of complement activation by C‐reactive protein. Immunopharmacology 1999; 42: 23–30
  • Biro A., Rovo Z., Papp D., Cervenak L., Varga L., Fust G., et al. Studies on the interactions between C‐reactive protein and complement proteins. Immunology 2007; 121: 40–50
  • Laine M., Jarva H., Seitsonen S., Haapasalo K., Lehtinen M. J., Lindeman N., et al. Y402H polymorphism of complement factor H affects binding affinity to C‐reactive protein. J Immunol 2007; 178: 3831–6
  • Sjowall C., Wettero J., Bengtsson T., Askendal A., Almroth G., Skogh T., et al. Solid‐phase classical complement activation by C‐reactive protein (CRP) is inhibited by fluid‐phase CRP‐C1q interaction. Biochem Biophys Res Commun 2007; 352: 251–8
  • McRae J. L., Duthy T. G., Griggs K. M., Ormsby R. J., Cowan P. J., Cromer B. A., et al. Human factor H‐related protein 5 has cofactor activity, inhibits C3 convertase activity, binds heparin and C‐reactive protein, and associates with lipoprotein. J Immunol 2005; 174: 6250–6
  • Saxena U., Nagpurkar A., Dolphin P. J., Mookerjea S. A study on the selective binding of apoprotein B‐ and E‐containing human plasma lipoproteins to immobilized rat serum phosphorylcholine‐binding protein. J Biol Chem 1987; 262: 3011–6
  • Ji S. R., Wu Y., Potempa L. A., Qiu Q., Zhao J. Interactions of C‐reactive protein with low‐density lipoproteins: implications for an active role of modified C‐reactive protein in atherosclerosis. Int J Biochem Cell Biol 2006; 38: 648–61
  • Mookerjea S., Francis J., Hunt D., Yang C. Y., Nagpurkar A. Rat C‐reactive protein causes a charge modification of LDL and stimulates its degradation by macrophages. Arterioscler Thromb 1994; 14: 282–7
  • Zwaka T. P., Hombach V., Torzewski J. C‐reactive protein‐mediated low density lipoprotein uptake by macrophages: implications for atherosclerosis. Circulation 2001; 103: 1194–7
  • Verma S., Li S‐H., Badiwala M. V., Weisel R. D., Fedak P. W. M., Li R‐K., et al. Endothelin antagonism and interleukin‐6 inhibition attenuate the proatherogenic effects of C‐reactive protein. Circulation 2002; 105: 1890–6
  • Hatanaka K., Li X‐A., Masuda K., Yutani C., Yamamoto A. Immunohistochemical localization of C‐reactive protein‐binding sites in human atherosclerotic aortic lesions by a modified streptavidin‐biotin‐staining method. Pathol Int 1995; 45: 635–41
  • Turk J. R., Carroll J. A., Laughlin M. H., Thomas T. R., Casati J., Bowles D. K., et al. C‐reactive protein correlates with macrophage accumulation in coronary arteries of hypercholesterolemic pigs. J Appl Physiol 2003; 95: 1301–4
  • Sun H., Koike T., Ichikawa T., Hatakeyama K., Shiomi M., Zhang B., et al. C‐reactive protein in atherosclerotic lesions: its origin and pathophysiological significance. Am J Pathol 2005; 167: 1139–48
  • Singh S. K., Suresh M. V., Voleti B., Prayther D. C., Moorman J. P., Han Z., et al. C‐reactive protein may prevent the formation of macrophage foam cells. J Immunol 2006; 176: S78–78
  • Stewart C. R., Tseng A. A., Mok Y. F., Staples M. K., Schiesser C. H., Lawrence L. J., et al. Oxidation of low‐density lipoproteins induces amyloid‐like structures that are recognized by macrophages. Biochemistry 2005; 44: 9108–16
  • Lim M. Y., Wang H., Kapoun A. M., O'Connell M., O'Young G., Brauer H. A., et al. p38 inhibition attenuates the pro‐inflammatory response to C‐reactive protein by human peripheral blood mononuclear cells. J Mol Cell Cardiol 2004; 37: 1111–4
  • Blaschke F., Bruemmer D., Yin F., Takata Y., Wang W., Fishbein M. C., et al. C‐reactive protein induces apoptosis in human coronary vascular smooth muscle cells. Circulation 2004; 110: 579–87
  • Wang C‐H., Li S‐H., Weisel R. D., Fedak P. W. M., Dumont A. S., Szmitko P. C‐reactive protein upregulates angiotensin type 1 receptors in vascular smooth muscle. Circulation 2003; 107: 1783–90
  • Qamirani E., Ren Y., Kuo L., Hein T. W. C‐reactive protein inhibits endothelium‐dependent NO‐mediated dilation in coronary arterioles by activating p38 kinase and NAD(P)H oxidase. Arterioscler Thromb Vasc Biol 2005; 25: 995–1001
  • Montero I., Orbe J., Varo N., Beloqui O., Monreal J. I., Rodriguez J. A., et al. C‐reactive protein induces matrix metalloproteinase‐1 and ‐10 in human endothelial cells: implications for clinical and subclinical atherosclerosis. J Am Coll Cardiol 2006; 47: 1369–78
  • Liang Y‐J., Shyu K‐G., Wang B‐W., Lai L‐P. C‐reactive protein activates the nuclear factor‐κB pathway and induces vascular cell adhesion molecule‐1 expression through CD32 in human umbilical vein endothelial cells and aortic endothelial cells. J Mol Cell Cardiol 2006; 40: 412–20
  • Verma S., Badiwala M. V., Weisel R. D., Li S‐H., Wang C‐H., Fedak P. W. M., et al. C‐reactive protein activates the nuclear factor‐κB signal transduction pathway in saphenous vein endothelial cells: implications for atherosclerosis and restenosis. J Thoracic Cardiovasc Surg 2003; 126: 1886–91
  • Wang Q., Zhu X., Xu Q., Ding X., Chen Y. E., Song Q. Effect of C‐reactive protein on gene expression in vascular endothelial cells. Am J Physiol 2005; 288: H1539–45
  • Kibayashi E., Urakaze M., Kobashi C., Kishida M., Takata M., Sato A., et al. Inhibitory effect of pitavastatin (NK‐104) on the C‐reactive protein‐induced interleukin‐8 production in human aortic endothelial cells. Clin Sci 2005; 108: 515–21
  • Wang H‐R., Li J‐J., Huang C‐X., Jiang H. Fluvastatin inhibits the expression of tumor necrosis factor‐α and activation of nuclear factor‐κB in human endothelial cells stimulated by C‐reactive protein. Clin Chim Acta 2005; 353: 53–60
  • Lin R., Liu J., Gan W., Yang G. C‐reactive protein‐induced expression of CD40‐CD40L and the effect of lovastatin and fenofibrate on it in human vascular endothelial cells. Biol Pharm Bull 2004; 27: 1537–43
  • Pasceri V., Chang J., Willerson J. T., Yeh E. T. H. Modulation of C‐reactive protein‐mediated monocyte chemoattractant protein‐1 induction in human endothelial cells by anti‐atherosclerosis drugs. Circulation 2001; 103: 2531–4
  • Pepys M. B., Hawkins P. N., Kahan M. C., Tennent G. A., Gallimore J. R., Graham D., et al. Proinflammatory effects of bacterial recombinant human C‐reactive protein are caused by contamination with bacterial products, not by C‐reactive protein itself. Circ Res 2005; 97: e97–103
  • Liu C., Wang S., Deb A., Nath K. A., Katusic Z. S., McConnell J. P., et al. Proapoptotic, antimigratory, antiproliferative, and antiangiogenic effects of commercial C‐reactive protein on various human endothelial cell types in vitro: implications of contaminating presence of sodium azide in commercial preparation. Circ Res 2005; 97: 135–43
  • Swafford A. N Jr., Bratz I. N., Knudson J. D., Rogers P. A., Timmerman J. M., Tune J. D., et al. C‐reactive protein does not relax vascular smooth muscle: effects mediated by sodium azide in commercially available preparations. Am J Physiol Heart Circ Physiol 2005; 288: H1786–95
  • Taylor K. E., Giddings J. C., van den Berg C. W. C‐reactive protein‐induced in vitro endothelial cell activation is an artefact caused by azide and lipopolysaccharide. Arterioscler Thromb Vasc Biol 2005; 25: 1225–30
  • Lafuente N., Azcutia V., Matesanz N., Cercas E., Rodriguez‐Manas L., Sanchez‐Ferrer C. F., et al. Evidence for sodium azide as an artifact mediating the modulation of inducible nitric oxide synthase by C‐reactive protein. J Cardiovasc Pharmacol 2005; 45: 193–6
  • Scirica B. M., Morrow D. A. Is C‐reactive protein an innocent bystander or proatherogenic culprit? The verdict is still out. Circulation 2006; 113: 2128–34
  • Dasu M. R., Devaraj S., Du Clos T. W., Jialal I. Biological effects of C‐reactive protein are not due to endotoxin contamination: evidence from toll‐like receptor 4 knock‐down human aortic endothelial cells. J Lipid Res 2007; 48: 509–12
  • Liuzzo G., Santamaria M., Biasucci L. M., Narducci M., Colafrancesco V., Porto A., et al. Persistent activation of nuclear factor kappa‐B signaling pathway in patients with unstable angina and elevated levels of C‐reactive protein: evidence for a direct proinflammatory effect of azide and lipopolysaccharide‐free C‐reactive protein on human monocytes via nuclear factor kappa‐B activation. J Am Coll Cardiol 2007; 49: 185–94
  • Doronzo G., Russo I., Trovati M., Anfossi G. Sodium azide in commercially available C‐reactive protein preparations does not influence matrix metalloproteinase‐2 synthesis and release in cultured human aortic vascular smooth muscle cells. Clin Chem 2006; 52: 1200–1
  • Heuertz R. M., Ahmed N., Webster R. O. Peptides derived from C‐reactive protein inhibit neutrophil alveolitis. J Immunol 1996; 156: 3412–7
  • Robey F. A., Ohura K., Futaki S., Fujii N., Yajima H., Goldman N., et al. Proteolysis of human C‐reactive protein produces peptides with potent immunomodulating activity. J Biol Chem 1987; 262: 7053–7
  • Shephard E. G., Beer S. M., Anderson R., Strachan A. F., Nel A. E., de Beer F. C. Generation of biologically active C‐reactive protein peptides by a neutral protease on the membrane of phorbol myristate acetate‐stimulated neutrophils. J Immunol 1989; 143: 2974–81
  • Yavin E. J., Fridkin M. Peptides derived from human C‐reactive protein inhibit the enzymatic activities of human leukocyte elastase and cathepsin G: use of overlapping peptide sequences to identify a unique inhibitor. J Peptide Res 1998; 51: 282–9
  • Zouki C., Beauchamp M., Baron C., Filep J. G. Prevention of in vitro neutrophil adhesion to endothelial cells through shedding of L‐selectin by C‐reactive protein and peptides derived from C‐reactive protein. J Clin Invest 1997; 100: 522–9
  • Zeller J. M., Landay A. L., Lint T. F., Gewurz H. Enhancement of human peripheral blood monocyte respiratory burst activity by aggregated C‐reactive protein. J Leukoc Biol 1986; 40: 769–83
  • Fernandez M. C., Mullenix M. C., Christner R. B., Mortensen R. F. A cell attachment peptide from human C‐reactive protein. J Cell Biochem 1992; 50: 83–92
  • Fiedel B. A., Gewurz H. Cleaved forms of C‐reactive protein are associated with platelet inhibition. J Immunol 1986; 136: 2551–5
  • Thomassen M. J., Meeker D. P., Deodhar S. D., Wiedemann H. P., Barna B. P. Activation of human monocytes and alveolar macrophages by a synthetic peptide of C‐reactive protein. J Immunother 1993; 13: 1–6
  • Du Clos T. W. Function of C‐reactive protein. Ann Med 2000; 32: 274–8
  • Mortensen R. F., Duszkiewicz J. A. Mediation of CRP‐dependent phagocytosis through mouse macrophage Fc‐receptors. J Immunol 1977; 119: 1611–6
  • Müller H., Fehr J. Binding of C‐reactive protein to human polymorphonuclear leukocytes: evidence for association of binding sites with Fc receptors. J Immunol 1986; 136: 2202–7
  • Bharadwaj D., Stein M‐P., Volzer M., Mold C., Du Clos T. W. The major receptor for C‐reactive protein on leukocytes is Fcγ receptor II. J Exp Med 1999; 190: 585–90
  • Saeland E., Van Royen A., Hendriksen K., Vilé‐Weekhout H., Rijkers G. T., Sanders L. A. M., et al. Human C‐reactive protein does not bind to FcγRIIa on phagocytic cells. J Clin Invest 2001; 107: 641–2
  • Du Clos T. W., Mold C., Edberg J. E., Kimberly R. P. Reply to: human C‐reactive protein does not bind to FcγRIIa on phagocytic cells. J Clin Invest 2001; 107: 643–3
  • Hundt M., Zielinska‐Skowronek M., Schmidt R. E. Lack of specific receptors for C‐reactive protein on white blood cells. Eur J Immunol 2001; 31: 3475–83
  • Chi M., Tridandapani S., Zhong W., Coggeshall K. M., Mortensen R. F. C‐reactive protein induces signaling through FcγRIIa on HL‐60 granulocytes. J Immunol 2002; 168: 1413–8
  • Rodriguez J. A., Bodman‐Smith K. B., Raynes J. G. Neutrophil responses to CRP are not dependent on polymorphism of human FcγRIIa (R131H). Clin Exp Immunol 2004; 138: 271–7
  • Williams T. N., Zhang C. X., Game B. A., He L., Huang Y. C‐reactive protein stimulates MMP‐1 expression in U937 histiocytes through FcγRII and extracellular signal‐regulated kinase pathway: an implication of CRP involvement in plaque destabilization. Arterioscler Thromb Vasc Biol 2004; 24: 61–6
  • Rocker C., Manolov D. E., Kuzmenkina E. V., Tron K., Slatosch H., Torzewski J., et al. Affinity of C‐reactive protein toward FcγRI is strongly enhanced by the γ‐chain. Am J Pathol 2007; 170: 755–63
  • Devaraj S., Du Clos T. W., Jialal I. Binding and internalization of C‐reactive protein by Fcγ receptors on human aortic endothelial cells mediates biological effects. Arterioscler Thromb Vasc Biol 2005; 25: 1359–63
  • Escribano‐Burgos M., Lopez‐Farre A., del Mar Gonzalez M., Macaya C., Garcia‐Mendez A., Mateos‐Caceres P. J., et al. Effect of C‐reactive protein on Fcγ receptor II in cultured bovine endothelial cells. Clin Sci 2005; 108: 85–91
  • Bang R., Marnell L., Mold C., Stein M‐P., Du Clos K. T., Chivington‐Buck C., et al. Analysis of binding sites in human C‐reactive protein for FcγRI, FcγRIIA, and C1q by site‐directed mutagenesis. J Biol Chem 2005; 280: 25095–102
  • Thomas‐Rudolf D., Du Clos T. W., Snapper C. M., Mold C. C‐reactive protein enhances immunity to Streptococcus pneumoniae by targeting uptake to FcγR on dendritic cells. J Immunol 2007; 178: 7283–91
  • Ryu J., Lee C. W., Shin J‐A., Park C‐S., Kim J. J., Park S‐J., et al. FcγRIIa mediates C‐reactive protein‐induced inflammatory responses of human vascular smooth muscle cells by activating NADPH oxidase 4. Cardiovasc Res 2007; 75: 555–65
  • Yang J., Wezeman M., Zhang X., Lin P., Wang M., Qian J., et al. Human C‐reactive protein binds activating Fcγ receptors and protects myeloma tumor cells from apoptosis. Cancer Cell 2007; 12: 252–65
  • Meuwissen M., van der Wal A. C., Niessen H. W. M., Koch K. T., de Winter R. J., van der Loos C. M., et al. Colocalization of intraplaque C reactive protein, complement, oxidised low density lipoprotein, and macrophages in stable and unstable angina and acute myocardial infarction. J Clin Pathol 2006; 59: 196–201
  • Torzewski M., Rist C., Mortensen R. F., Zwaka T. P., Bienek M., Waltenberger J., et al. C‐reactive protein in the arterial intima: role of C‐reactive protein receptor‐dependent monocyte recruitment in atherogenesis. Arterioscler Thromb Vasc Biol 2000; 20: 2094–9
  • Paul A., Ko K. W. S., Li L., Yechoor V., McCrory M. A., Szalai A. J., et al. C‐reactive protein accelerates the progression of atherosclerosis in apolipoprotein E‐deficient mice. Circulation 2004; 109: 647–55
  • Schwedler S. B., Amann K., Wernicke K., Krebs A., Nauck M., Wanner C., et al. Native C‐reactive protein increases whereas modified C‐reactive protein reduces atherosclerosis in apolipoprotein E‐knockout mice. Circulation 2005; 112: 1016–23
  • Bisoendial R. J., Kastelein J. J. P., Peters S. L. M., Levels J. H. M., Birjmohun R., Rotmans J. I., et al. Effects of CRP‐infusion on endothelial function and coagulation in normo‐ and hypercholesterolemic subjects. J Lipid Res 2007; 48: 952–60
  • Bisoendial R., Birjmohun R., Keller T., van Leuven S., Levels H., Levi M., et al. Letter to the editor. Circ Res 2005; 97: 115–6
  • Hirschfield G. M., Gallimore J. R., Kahan M. C., Hutchinson W. L., Sabin C. A., Benson G. M., et al. Transgenic human C‐reactive protein is not proatherogenic in apolipoprotein E‐deficient mice. Proc Natl Acad Sci U S A 2005; 102: 8309–14
  • Reifenberg K., Lehr H‐A., Baskal D., Wiese E., Schaefer S. C., Black S., et al. Role of C‐reactive protein in atherogenesis: can the apolipoprotein E knockout mouse provide the answer?. Arterioscler Thromb Vasc Biol 2005; 25: 1641–6
  • Trion A., de Maat M. P. M., Jukema J. W., van der Laarse A., Maas M. C., Offerman E. H., et al. No effect of C‐reactive protein on early atherosclerosis development in apolipoprotein E* 3‐Leiden /human C‐reactive protein transgenic mice. Arterioscler Thromb Vasc Biol 2005; 25: 1635–40
  • Trion A., de Maat M., Jukema W., Maas A., Offerman E., Havekes L., et al. Anti‐atherosclerotic effect of amlodipine, alone and in combination with atorvastatin, in APOE* 3‐leiden /hCRP transgenic mice. J Cardiovasc Pharmacol 2006; 47: 89–95
  • Verschuren L., Kleemann R., Offerman E. H., Szalai A. J., Emeis S. J., Princen H. M. G., et al. Effect of low dose atorvastatin versus diet‐induced cholesterol lowering on atherosclerotic lesion progression and inflammation in apolipoprotein E* 3‐leiden transgenic mice. Arterioscler Thromb Vasc Biol 2005; 25: 161–7
  • Bhakdi S., Torzewski M., Paprotka K., Schmitt S., Barsoom H., Suriyaphol P., et al. Possible protective role for C‐reactive protein in atherogenesis: complement activation by modified lipoproteins halts before detrimental terminal sequence. Circulation 2004; 109: 1870–6
  • Suresh M. V., Singh S. K., Ferguson D. A Jr., Agrawal A. Role of the property of C‐reactive protein to activate the classical pathway of complement in protecting mice from pneumococcal infection. J Immunol 2006; 176: 4369–74
  • Ng P. M. L., Le Saux A., Lee C. M., Tan N. S., Lu J., Thiel S., et al. C‐reactive protein collaborates with plasma lectins to boost immune response against bacteria. EMBO J 2007; 26: 3431–40
  • Kushner I., Rakita L., Kaplan M. H. Studies of acute‐phase protein. II. Localization of Cx‐reactive protein in heart in induced myocardial infarction in rabbits. J Clin Invest 1963; 42: 286–92
  • Krijnen P. A. J., Ciurana C., Cramer T., Hazes T., Meijer C. J. L. M., Visser C. A., et al. IgM colocalizes with complement and C‐reactive protein in infarcted human myocardium. J Clin Pathol 2005; 58: 382–8
  • Lagrand W. K., Niessen H. W. M., Wolbink G‐J., Jaspars L. H., Visser C. A., Verheugt F. W. A., et al. C‐reactive protein colocalizes with complement in human hearts during acute myocardial infarction. Circulation 1997; 95: 97–103
  • Barrett T. D., Hennan J. K., Marks R. M., Lucchesi B. R. C‐reactive‐protein‐associated increase in myocardial infarct size after ischemia/reperfusion. J Pharmacol Exp Ther 2002; 303: 1007–13
  • Griselli M., Herbert J., Hutchinson W. L., Taylor K. M., Sohail M., Krausz T., et al. C‐reactive protein and complement are important mediators of tissue damage in acute myocardial infarction. J Exp Med 1999; 190: 1733–40
  • Danenberg H. D., Szalai A. J., Swaminathan R. V., Peng L., Chen Z., Seifert P., et al. Increased thrombosis after arterial injury in human C‐reactive protein‐transgenic mice. Circulation 2003; 108: 512–5
  • Van den Berg C. W., Morgan B. P. Letter in response to A. Agrawal: CRP after 2004. Mol Immunol 2006; 43: 292–3
  • Kushner I., Agrawal A. Letter in response to A. Agrawal: CRP after 2004. Mol Immunol 2007; 44: 670–1
  • Rufail M. L., Ramage S. C., van Antwerpen R. C‐reactive protein inhibits in vitro oxidation of low‐density lipoprotein. FEBS Lett 2006; 580: 5155–60
  • Sammalkorpi K. T., Valtonen V. V., Maury C. P. J. Lipoproteins and acute phase response during acute infection: interrelationships between C‐reactive protein and serum amyloid‐A protein and lipoproteins. Ann Med 1990; 22: 397–401

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.