Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jan 16.
Published in final edited form as: Circ Res. 2015 Jan 16;116(2):368–384. doi: 10.1161/CIRCRESAHA.116.302795

Fcγ Receptors and Ligands and Cardiovascular Disease

Keiji Tanigaki 1, Nathan Sundgren 1, Amit Khera 2, Wanpen Vongpatanasin 2, Chieko Mineo 1, Philip W Shaul 1
PMCID: PMC4331353  NIHMSID: NIHMS650247  PMID: 25593280

Abstract

Fcγ receptors (FcγR) classically modulate intracellular signaling upon binding of the Fc region of IgG in immune response cells. How FcγR and their ligands impact cardiovascular health and disease has recently been interrogated in both preclinical and clinical studies. The stimulation of activating FcγR in endothelial cells, vascular smooth muscle cells and monocytes/macrophages causes a variety of cellular responses that may contribute to vascular disease pathogenesis. Stimulation of the lone inhibitory FγcR, FcγRIIB, also has adverse consequences in endothelial cells, antagonizing NO production and reparative mechanisms. In preclinical disease models, activating FcγR promote atherosclerosis whereas FcγRIIB is protective, and activating FcγR also enhance thrombotic and non-thrombotic vascular occlusion. The FcγR ligand C-reactive protein (CRP) has undergone intense study. Although in rodents CRP does not impact atherosclerosis, it causes hypertension and insulin resistance and worsens myocardial infarction. Massive data has accumulated indicating an association between increases in circulating CRP and coronary heart disease in humans. However, Mendelian randomization studies reveal that CRP is not likely a disease mediator. CRP genetics and hypertension warrants further investigation. Studies to date of genetic variants of activating FcγR are insufficient to implicate the receptors in coronary heart disease pathogenesis in humans. However, a link between FcγRIIB and human hypertension may be emerging. Further knowledge of the vascular biology of FcγR and their ligands will potentially enhance our understanding of cardiovascular disorders, particularly in patients whose greater predisposition for disease is not explained by traditional risk factors, such as individuals with autoimmune disorders.

Keywords: atherosclerosis, C-reactive protein, endothelial nitric oxide synthase, Fcγ receptor, hypertension

Introduction

Fcγ receptors (FcγR) are plasma membrane-associated receptors for IgG and the pentraxins C-reactive protein (CRP) and serum amyloid P component (SAP). As a result of recent interest in the role for inflammation in cardiovascular disorders, FcγR and their ligands have been investigated in the context of cardiovascular disease. This review will highlight major observations made in both preclinical and clinical studies in that realm. Following a brief description of FcγR subtypes and their functions, their IgG and pentraxin ligands will be discussed. Investigations performed in cell culture will then be highlighted, followed by queries of the impact of FcγR and ligands in animal models of cardiovascular disorders. Attempts to understand the influence of IgG and pentraxins on cardiovascular disease risk and outcome in humans will then be summarized, including the voluminous studies of CRP as a risk factor and possible disease mediator. How genetic variants in FcγR impact human cardiovascular health will be presented, including a recent interrogation of a variant in the lone inhibitory FcγR, FcγRIIB, that alters receptor function in endothelium. Finally, the current knowledge gaps in this area and the rationale to fill them will be presented. By recognizing what is known and not yet known about the influence of FcγR on cardiovascular health, it is hoped that this review will stimulate further work in this area. Such future efforts are warranted because FcγR biology may underlie a considerable component of vascular disease predisposition that is not currently explained by traditional risk factors.

Fcγ Receptor Subtypes and Functions

Fcγ Receptor Subtypes

The classical function of FcγR is to invoke intracellular signaling upon IgG binding in immune response cells and thereby modulate numerous inflammatory processes. FcγR are categorized into activating receptors and inhibitory receptors (Table 1)1. In humans, the activating Fc receptors are FcγRI (CD64), FcγRIIA (CD32a), FcγRIIC (CD32c), FcγRIIIA (CD16a) and FcγRIIIB (CD16b), and the sole inhibitory receptor is FcγRIIB (CD32b). In mice the activating FcγR are FcγRI, FcγRIII and FcγRIV, and in mice differential splicing of a single gene for inhibitory FcγRIIB yields FcγRIIB1, B2 and B3. FcγR belong to the large immunoglobulin superfamily and they are type I transmembrane glycoproteins with the exception of the human subtype FcγRIIIB, which is GPI-anchored. All Fcγ receptors display a high degree of sequence identity in their extracellular portion (50–96%), but differ significantly in their cytoplasmic domains14.

Table 1.

Activating and inhibitory Fcγ receptors in humans and mice.

Human Mouse
Activating FcγRI (ITAM, γ-chain) CD64 FcγRI (ITAM, γ-chain) CD64
FcγRIIA (ITAM) CD32a
FcγRIIC (ITAM) CD 32c
FcγRIIIA (ITAM, γ-chain) CD16a FcγRIII (ITAM, γ-chain) CD16
FcγRIIIB (GPI-anchored) CD16b
FcγRIV (ITAM, γ-chain) CD16-2
Inhibitory FcγRIIB (ITIM) CD32b FcγRIIB (ITIM) CD32b

Innate immune effector cells, such as monocytes, macrophages, dendritic cells (DCs), basophils and mast cells, express activating and inhibitory FcγR. In human leukocytes, FcγRI and FcγRIIA are abundant in monocytes, macrophages and granulocytes, FcγRIIIA are expressed primarily on macrophages but also in monocytes, and FcγRIIIB are found in granulocytes5, 6. In mice, monocytes and macrophages express all activating and inhibitory FcγR, neutrophils mainly express the activating FcγRIII and FcγRIV and the inhibitory FcγRIIB, whereas the expression of FcγRI, FcγRIIB and FcγRIII dominates on DCs. There are two cell types that do not co-express activating and inhibitory receptors; NK cells solely express the activating receptor FcγRIII, and B cells only express the inhibitory receptor FcγRIIB. In B cells, FcγRIIB functions as a regulator of activating signals transmitted by the B cell receptor (BCR)3. In DC, the balance between activating and inhibitory FcγR activity influences their relative activation status7. In addition to hematopoietic cells, FcγR are expressed by follicular DCs, endothelial cells, microglial cells, osteoclasts and mesangial cells3. Interestingly, ligand binding impacts the abundance of cell surface FcγR. Immune complex binding to FcγRIIA leads to their endocytosis and ubiquitination and degradation, and engaged FcγRIIIA also undergo internalization and degradation8; in contrast, in human endothelial cells C-reactive protein (CRP) upregulates the surface expression of CD32 and CD649.

Activating Fcγ Receptor Function

The activating FcγR, with the exception of the human GPI-anchored FcγRIIIB, activate signaling pathways through immunoreceptor tyrosine-based activation motifs (ITAMs) contained in their cytoplasmic regions. Human FcγRIIA and FcγRIIC are comprised of a single subunit with a cytoplasmic ITAM (Table 1), and they transduce activating signaling pathways autonomously. All other activating FcγR (human FcγRI, FcγRIIIA and mouse FcγRI, FcγRIII, FcγRIV) consist of a ligand-binding α-chain and a signal-transducing γ chain that contains an ITAM in its cytoplasmic domain. In some instances the signal transducing subunits for an FcγR can differ between cell types. Whereas human FcγRIIIA is associated with an ITAM-containing γ-chain in monocytes and macrophages, it associates with the ITAM-containing CD2 ζ chain in NK cells. Along with its signaling function, the γ-chain is important for the assembly and cell-surface transport of the respective α chains. Because of their expression in innate immune effector cells, activating FcγR have been proposed to link the specificity of antibodies generated by the adaptive immune system to the potent effector functions of the innate immune system13.

After crosslinking by immune complexes, the signaling pathways initiated by the different activating FcγR are quite similar, beginning with tyrosine phosphorylation of the ITAM by kinases of the SRC family. This leads to the recruitment of SYK-family kinases to the ITAM, followed by the activation of various downstream targets, such as the linker of activation of T cells, multimolecular adaptor complexes and PI3 kinase. By generating PIP3, PI3 kinase creates membrane-docking sites for Bruton's tyrosine kinase (BTK) and phospholipase Cγ (PLCγ). Activation of PLCγ causes an increase in intracellular calcium and triggering of further downstream signaling events. In addition to calcium-dependent pathways, the RAS-RAF-MAPK-pathway is of central importance for cell activation following activating FcγR crosslinking. An additional major function of activating FcγR is to promote the endocytosis or phagocytosis of immune complexes, which include antibody-coated microorganisms and soluble proteins. In the case of granulocytes, monocytes and macrophages, this will mainly result in the rapid degradation of the engulfed material in lysosomal compartments13.

Inhibitory Fγc Receptor Function

The only known inhibitory FcγR, FcγRIIB, is the most broadly expressed FcγR, and it is present on virtually all leukocytes with the exception of NK cells and T cells. Because of the broad expression pattern, global genetic deletion of FcγRIIB can result in complex phenotypic changes affecting either innate or adaptive immune responses. FcγRIIB transmits inhibitory signals through an immunoreceptor tyrosine-based inhibitory motif (ITIM) contained in its cytoplasmic region. FcγRIIB function is best understood in B cells, in which it serves as a checkpoint for humoral immunity, playing a critical role in preventing the generation of autoreactive antibodies. Partnering with BCR, the simultaneous triggering of the ITIM-containing FcγRIIB and the BCR results in the recruitment of phosphatases including SHIP (SH2 domain-containing inositol polyphosphate 5' phosphatase) and SHP1 (SH2-domain -containing protein tyrosine phosphatase 1) that interfere with activating signaling pathways by hydrolyzing phosphoinositide intermediates. This prevents the recruitment of pleckstrin homology-domain-containing kinases, such as BTK or PLCγ, to the cell membrane, thereby diminishing downstream events such as increases in intracellular calcium levels. Processes that disrupt FcγRIIB function in B cells result in a lower threshold for B cell activation and stronger activating signals after BCR crosslinking13.

In addition to its participation in adaptive immunity by tempering antibody production by B cells, FcγRIIB is a regulator of innate immunity via its actions in mast cells, granulocytes and macrophages. As these cells have the capacity to trigger strong proinflammatory responses, their activation needs to be tightly controlled. In the case of antibody-mediated responses, such as phagocytosis, antibody-dependent cell-mediated cytotoxicity, allergic reactions and the release of pro-inflammatory mediators, this is the function of the inhibitory FcγRIIB. FcγRIIB contributes varying levels of negative regulation depending on the specific IgG subclass that is bound to the receptor (see below)13.

Fcγ Receptor Ligands

Immunoglobulins

The complexity of the FcγR family is mirrored by the presence of four different IgG subclasses in humans (IgG1-IgG4) and in mice (IgG1, IgG2a/c, IgG2b and IgG3), which bind with varying affinity and specificity to different FcγR via the Fc portion of the IgG. Overall, in humans IgG1 and IgG3 are the most pro-inflammatory IgG subclasses, and in mice IgG2a and IgG2b are the most pro-inflammatory IgG molecules. FcγR vary in their affinity for IgG. FcγRI is the single high-affinity receptor in humans and in mice, particularly regarding binding of IgG1 and IgG3 in humans or IgG2a in mice. All other FcγR have a 100–1000 fold lower affinity and show a broader IgG subclass specificity. Relative binding affinity of mouse and human IgG subclasses to mouse and human FcγR is shown in Table 210. The low affinity nature of IgG binding to most of the FcγR proteins serves an important function in that it prevents the binding by monomeric antibody molecules that are always present at high levels in the circulation, thereby avoiding the potential non-specific activation of pro-inflammatory responses. In contrast, the high-affinity FcγRI is constantly saturated with ligand. However, as has been described for the binding of IgE to the high-affinity FcεRI, cell activation only ensues after the FcγRI receptors have been crosslinked by antigen. Considering the existence of both activating and inhibiting FcγR, and the varying affinities for IgG subclasses, the summary in vivo actions of IgG-FcγR tandems can be difficult to predict. To deal with this complexity, the ratio of affinities of a given IgG subclass for the activating versus the inhibitory receptors has been termed the A/I-ratio and it has emerged as a helpful predictive value for the activity of a specific IgG subclass in vivo13.

Table 2.

Binding affinity between IgG subclasses and Fcγ receptors10.

Human Mouse
IgG 1 2 3 4 IgG 1 2a 2b 3
Activating FcγRI high none high high FcγRI none high low very low
FcγRIIA low low low low
FcγRIIC low very low low low
FcγRIIIA low very low low low FcγRIII low low low none
FcγRIIIB low none low low
FcγRIV none high high none
Inhibitory FcγRIIB low very low low low FcγRIIB low low low none

Pentraxins

In addition to IgG, the acute phase reactant C-reactive protein (CRP) is a ligand for FcγR. CRP is produced principally by the liver in response to a variety of pathologic conditions including inflammation, infection and trauma. CRP is a member of the pentraxin family of proteins, and it was originally described as a protein which binds to the C-polysaccharide of the cell wall of pneumococci. The protein consists of five identical 23 kDa subunits noncovalently associated in a flat pentameric disk. The primary stimulus for hepatocyte synthesis and secretion of CRP is the proinflammatory cytokine interleukin (IL)-6. Circulating CRP levels can rise 500-fold within 24 to 48h of the initiation of an inflammatory process. CRP was initially found to serve as an opsonin, binding to pathogenetic microorganisms and mediating the phagocytosis of sensitized erythrocytes. CRP also activates complement by binding to C1q, and studies in mice indicate that it is protective against infection and the development of autoimmunity. CRP levels are often used clinically as an indicator of infection and/or inflammation1114.

In addition to the liver being a source of CRP, transcript for the pentraxin has been detected in human atherosclerotic lesions, with mRNA levels 10-fold greater in plaques than in normal arteries15. CRP transcript has also been detected in areas of myointimal hyperplasia in a porcine arteriovenous graft model16. Expression of CRP has been demonstrated in endothelial cells, particularly upon treatment with macrophage conditioned medium17 or electronegative low density lipoprotein cholesterol18. In vascular smooth muscle, angiotensin II, endothelin-1 and homocysteine promote CRP expression1921. Adipose tissue may be an additional important source of CRP since the mRNA is detectable in human adipose tissue, where it is expressed in both adipocytes and stromal cells22,23. CRP mRNA is increased in the adipose tissue of obese individuals compared with controls and it is upregulated in vitro in adipose tissue explants by LPS or IL-624.

Regarding the forms of CRP that impact the cardiovascular system, it is controversial whether the potential actions of the pentraxin of relevance to cardiovascular health are mediated by native, pentameric CRP, or monomeric (m)CRP. In some studies it has been reported that mCRP is detected in normal human blood vessels and in inflamed tissues2527, yet others indicate that mCRP is deposited in human atherosclerotic plaques but not in healthy vessels, and that pentameric CRP is not detectable in either healthy or diseased blood vessels28. It has been reported that mCRP promotes a proinflammatory phenotype in cultured endothelial cells. However, in earlier investigations mCRP actions on endothelial cells were modified by anti-CD16 (FcγRIII) blocking antibody and not by anti-FcγRII antibody29, yet genetic studies in mice indicate that FcγRIIB is critically involved in the actions of endogenous CRP on endothelium30. In addition, there is a recent report that mCRP actually mediates responses in human endothelial cells via plasma membrane insertion rather than by binding to surface FcγRs31. Furthermore, it has been demonstrated that native CRP impairs endothelial function in vivo, whereas mCRP does not32. Activated platelets disassociate pentameric CRP to mCRP via lysophosphatidylcholine which is only present on activated platelets28, suggesting that it may remain challenging to definitively determine which form of CRP impacts the cardiovascular system in vivo.

It was initially determined that CRP binds to the high affinity receptor for IgG, FcγRI, in human monocytes and transfected COS-7 cells33,34. FcγRIIA, which is a low-affinity receptor for IgG, is also a high-affinity CRP receptor35,36. FcγR were additionally identified as CRP receptors in mouse leukocytes using knockout lines lacking FcγR; blood cells from γ chain−/− mice displayed less CRP binding than controls, FcγRIIB−/− mice also had diminished CRP binding, and no CRP binding was observed in leukocytes from double-deficient mice37. A role for FcγRIII in CRP binding is less likely since NK cells from both humans and mice, which express FcγRIII as their sole FcγR, display negligible CRP binding36. Thus, in both humans and mice FcγRI and FcγRII most likely serve as the major receptors for CRP. The structurally similar pentraxin, serum amyloid P-component (SAP), which is the CRP-equivalent acute phase reactant in mice, also binds to FcγR including FcγRII38. Structural studies of pentraxin-FcγR binding have demonstrated that the molecular basis for FcγR recognition is conserved between CRP, SAP and IgG39.

Fcγ Receptor-Ligand Actions in Vascular Cells

Endothelial Cells

Based upon observed associations between chronic modest elevations in CRP and cardiovascular disease in humans (see below), a variety of studies have been performed in cultured endothelial cells evaluating the effects of CRP. One of the first demonstrated that CRP decreases endothelial NO synthase (eNOS) mRNA and protein abundance and enzymatic activity in human aortic endothelial cells, and that there is an associated promotion of monocyte adhesion to the endothelial cells (Figure 1A)40. CRP upregulation of endothelium-derived plasminogen activator inhibitor-1 (PAI-1) was then demonstrated41, and this was followed by studies showing that CRP upregulates interleukin-8 (IL-8) and endothelial cell-monocyte adhesion by activating NF-kB, with these responses as well as CRP binding to endothelial cells being attenuated by anti-CD32 (FcγRII) and anti-CD64 (FγcRI) antibodies9,42,43. Nuclear actions of CRP in endothelial cells were further delineated in studies of cells transfected with human eNOS 5’ flanking sequence fused to luciferase that indicated that CRP decreases eNOS gene transcription44.

Figure 1.

Figure 1

Effects of Fcγ receptors and their ligands on vascular cells (A) and on preclinical models of cardiometabolic disorders (B). ((Ilustration credit: Ben Smith).

Acute effects of CRP on endothelial cell function were also demonstrated in studies that showed that the rapid activation of eNOS by diverse agonists is blocked by the pentraxin. SAP and aggregated IgG used to mimic immune complexes also inhibited eNOS activation. FcγRIIB mRNA expression was demonstrated in endothelial cells, and heterologous expression studies revealed that CRP antagonism of eNOS requires FcγRIIB. Demonstrating in vivo actions of CRP on the endothelium for the first time, it was found that acute CRP administration blunts acetylcholine-induced increases in carotid artery conductance in wild-type mice; in contrast, in FcγRIIB−/− mice CRP actually enhanced the vasodilatory response to acetylcholine45. Long-term actions of CRP impacting the endothelium have also been shown in vivo, with CRP transgenic mice (TG-CRP) displaying attenuated carotid artery reendothelialization following perivascular electric injury. In parallel, CRP blunts cultured endothelial cell migration44. CRP administration to rats for 72 hours resulted in decreased eNOS activity and blunted vasodilatory responses to acetylcholine in isolated, pressurized mesenteric arterioles46. Endothelial actions of CRP have additionally been interrogated in mice harboring an inflammation-sensitive CRP transgene. Turpentine activation of the CRP transgene yielded serum levels of human CRP of 276 µg/ml, with CRP concentrations being <1.0 ug/ml in healthy humans and rarely exceeding 2 ug/ml in mice even in response to inflammatory stimuli14,47. Aortic rings isolated from the mice with elevated CRP displayed impaired endothelium-dependent responses to acetylcholine, and NO release and eNOS-Ser1179 phosphorylation were decreased. In addition, CRP-overexpressing mice had increased perivascular fibrosis, greater endothelial VCAM-1 and MCP-1 staining, and enhanced macrophage infiltration48.

The features of FcγRIIB and related signaling events required for CRP to antagonize eNOS were revealed in studies of eNOS activation by insulin49, which stimulates the enzyme to promote vasodilation and capillary recruitment in the skeletal muscle microvasculature, thereby enhancing skeletal muscle glucose disposal50. In mice the activating phosphorylation of eNOS-Ser1179 and also Akt phosphorylation in response to insulin were decreased by CRP treatment, and reconstitution experiments with wild-type and mutant FcγRIIB in NIH3T3IR cells revealed that these processes require the ITIM of the receptor. It was further shown that endothelial cells express SHIP-1, that CRP induces SHIP-1 stimulatory phosphorylation in endothelium in culture and in vivo, and that SHIP-1 knockdown by small interfering RNA prevents CRP antagonism of insulin-induced eNOS activation49. How CRP causes the critical FcγRIIB ITIM phosphorylation in endothelial cells has additionally been elucidated. It was determined in cultured cells that FγcRI blocking antibodies prevent CRP antagonism of eNOS (Figure 1A), that CRP activates SRC via FcγRI, and that both ITIM phosphorylation and eNOS antagonism caused by CRP are SRC-dependent. Parallel processes were found to mediate the inhibition of eNOS by aggregated IgG used to mimic immune complex. As such, FcγRI-FcγRIIB coupling by SRC kinase underlies both CRP and IgG attenuation of endothelial NO production. The partnership between FcγRI and FcγRIIB in the actions of CRP on endothelium was demonstrated in vivo in studies of carotid artery reendothelialization in TG-CRP mice crossed with either mice lacking the γ subunit of FcγRI (FcRγ−/−) or FcγRIIB−/− mice. Whereas reendothelialization was impaired in TG-CRP vs wild-type mice, it was normal in FcRγ−/−;TG-CRP mice and in FcγRIIB−/−;TG-CRP mice30. Thus, both FcγRI and FcγRIIB have been implicated in the actions of CRP on endothelium in vivo.

Whereas various forms of FcγRIIB loss-of-function have been employed to implicate the receptor in the actions of immune complex and pentraxin on endothelial cells, and FcγRIIB mRNA has been demonstrated in endothelium45, evidence that FcγRIIB potentially impacts cardiovascular health in humans would be strengthened if FcγRIIB protein expression could be demonstrated in human endothelial cells. This has been perplexing due to the lack of specificity of antibodies and the challenges involved in reliable detection of an antibody receptor using antibodies as probes. Fluorescence-activated cell sorting (FACS) was therefore performed with human endothelial cells using an Alexa Fluor 488-conjugated monoclonal Ab (clone 8B5, MacroGenics) to human FcγRIIB generated by immunization of human FcγRIIA transgenic mice to prevent cross-reactivity with FcγRIIA. In 8B5, Asn297 in the Fc domain has been mutated to Gln to prevent Fc-FcγR binding51, thereby minimizing nonspecific binding. Using 8B5 Ab, FcγRIIB protein was readily detected in Raji positive control cells (Fig. 2A), and in human aortic, coronary artery and umbilical vein endothelial cells (Fig. 2B–D, HAEC, HCAEC, HUVEC). Further confirmation of Ab specificity was obtained by siRNA knockdown of FcγRIIB (Fig. 2E, green and red for scrambled and FcγRIIB-targeted siRNA, respectively). Immunoblotting detected FcγRIIB as a 40 kDa protein in plasma membranes from Raji cells (positive control) and HAEC (Fig. 2F). These findings indicate that FcγRIIB protein is expressed in human endothelial cells derived from a variety of vascular beds.

Figure 2.

Figure 2

FcγRIIB protein is expressed in human endothelial cells. A–D. FACS was performed in Raji cells (positive control) (A) and in human aortic endothelial cells (HAEC) (B), human coronary artery endothelial cells (C), and human umbilical vein endothelial cells (HUVEC) (D) using 8B5, which is an Alexa Fluor 488-conjugated monoclonal antibody to human FcγRIIB. Findings in the absence of antibody (Ab) and with 8B5 are shown. E. FACS with 8B5 was performed in HAEC transfected with control siRNA (D-001810–02–05, Dharmacon) or siRNA targeting FcγRIIB (GCUACAGGUUCAAGGCCAA, Dharmacon). F. Immunoblotting was performed with 8B5 on plasma membranes isolated from Raji positive control cells and HAEC.

Vascular Smooth Muscle Cells

As has been the case for studies of FcγR ligand actions on endothelial cells, CRP has been the principal ligand employed in investigations in vascular smooth muscle (VSM) cells. Using human coronary artery VSM cells it was demonstrated that CRP induces caspase-mediated apoptosis (Fig. 1A). Upregulation of the growth arrest- and DNA damage-inducible gene 153 (GADD153) was also observed, the impact of CRP was primarily on GADD153 mRNA stability, and GADD153 silencing decreased the pro-apoptotic effect of CRP. Potentially supportive of a role for GADD153 in processes occurring in atherosclerotic plaques, GADD153 was specifically localized to apoptotic VSM cells in human coronary artery lesions52. It has also been shown that CRP induces intracellular reactive oxygen species (ROS) generation in VSM cells by NADPH oxidase 4 isoform (Nox4), and AP-1/NF-kB activation, and MCP-1, IL-6 and ET-1 production53,54. CRP additionally upregulates VSM cell matrix metalloproteinase-2 (MMP-2) synthesis and activity55, and VSM cell tissue factor expression56. Furthermore, CRP upregulates VSM cell angiotensin II type 1 receptor (AT1-R) expression, and it promotes VSM cell migration and proliferation and enhances the effects of angiotensin II on those processes16,57.

Regarding the potential FcγR operative in the actions of CRP on VSM cells, FcγRIIA expression has been demonstrated in cultured VSM cells isolated form human coronary arteries, and the receptor has been colocalized with α-actin positive VSM cells in atheromatous regions in human coronary artery plaques52. FcγRIIA blocking antibody attenuates the effects of CRP on VSM cell ROS and inflammatory cytokine production, and apoptosis (Fig. 1A)52, FcγRIIA and FcγRIIIA transcripts have been detected in VSM cells, and the silencing of FcγRIIIA attenuates the upregulation of tissue factor expression by CRP56. Thus, in studies limited to cell culture, CRP has direct adverse effects on VSM typical of responses observed with injury or inflammation, and the operative FcγR are FcγRIIA and FcγRIIIA.

Leukocytes and Platelets

Evidence for potential actions of FcγR ligands and receptors in leukocytes has primarily been obtained in studies of LDL cholesterol-containing immune complexes (LDL-IC). Human monocytes/macrophages exposed to LDL-IC display marked cholesterol ester accumulation such that they assume a foam cell phenotype (Fig. 1A), and the LDL-IC cause increased LDL receptor expression. The uptake of LDL-IC is mediated by FcγRI, and it results in the release of cytokines, particularly TNF-alpha, and matrix metalloproteinase-158,59. In addition, the LDL-IC-FcγRI tandem promotes the survival and proliferation of monocytes/macrophages60,61. Since LDL-IC are abundant in atherosclerotic lesions59,6264, these processes mediated by FcγR in monocytes/macrophages may play important roles in atherosclerosis and its consequences (see below).

In human platelets the ITAM-containing FcγR FcγRIIA is expressed, and it participates in platelet activation in a number of immune-mediated thrombocytopenias and thrombosis syndromes. These include disseminated intravascular coagulation and bacterial sepsis-associated thrombocytopenia, and heparin-induced thrombocytopenia and thrombosis6570. FcγRIIA also enables platelets to participate in innate immunity, allowing them to bind to and be activated by antibody-opsonized pathogens, thereby promoting their clearance66,67,71,72. Since there is not a murine homologue for human FcγRIIA, the receptor is studied in vivo in transgenic mice expressing the human receptor65,73. Platelets express two additional ITAM receptors. These are C-type lectin 2, which is the receptor for the snake venom toxin rhodocytin and for podoplanin, and FcγR γ chain, which associates with the glycoprotein IV collagen receptor. Platelet ITAM receptors, their signaling mechanisms and functions have recently been extensively reviewed65, and therefore they will be mentioned in limited fashion in the remainder of this review.

Fcγ Receptors, Ligands and CVD in Animal Models

Atherosclerosis

Atherosclerosis is considered to be a chronic inflammatory disease, and both innate and adaptive immunity play critical roles in its initiation and progression74. FcγR are expressed in virtually all individual cell types participating in atherogenesis (see above), and as such it is logical for FcγR to potentially influence atherosclerosis severity. This has been tested in vivo primarily in four studies in mice. The first entailed experiments in apoE−/− versus apoE−/−;FcγR γ chain−/− mice, with the latter having genetic loss-of-function of activating FcγR. Without affecting plasma lipids, FcγR γ chain deletion resulted in decreased atherosclerotic lesion size (Fig. 1B), and lesion macrophage and T-cell content were cut approximately in half75. In a separate study in mice fed a high fat diet for 10 weeks, the global deletion of the FcγR γ chain caused improved acetylcholine-induced relaxation of isolated aortic rings, and it lowered superoxide abundance in the vascular wall76. These findings suggest that FcγR γ chain-mediated processes may promote atherosclerosis by attenuating endothelial function or by increasing oxygen-derived free radical abundance. Alternatively, since the quantification of FcγR in the aorta in these studies revealed that the activating/inhibitory ratio changed from 2.5 to 0.4 with the deletion of the FcγR γ chain75, it is possible that the decrease in atherosclerosis observed in the absence of the FcγR γ chain reflects an increase in the relative atheroprotective properties of the inhibitory FcγR FcγRIIB. In bone marrow reconstitution experiments, hematopoietic deficiency of FcγR γ chain in apoE−/− mice resulted in decreased atherosclerotic lesion size, with fewer macrophages and T lymphocytes within lesions and also increased plaque stability. The expression of pro-inflammatory genes was attenuated and anti-inflammatory gene expression was increased. In vitro experiments using murine macrophages revealed decreases in foam cell formation, pro-inflammatory gene expression and oxidative stress in cells lacking FcγR γ chain, suggesting that the cell type in which the γ chain influences atherosclerosis severity is the macrophage77 . Other work in a similar model suggested that with hypercholesterolemia, activating FcγR promote atherosclerosis by increasing antigen-presenting cell IL-6 secretion, resulting in an enhanced Th17 response 78. The impact of loss-of-function of an activating FcγR was also evaluated in studies of LDLR−/− versus LDLR−/−;FcγRIII−/− mice. At later stages of atherosclerosis development, FcγRIII silencing resulted in smaller lesions, and there was an associated increase in interferon-γ and IL-10 production by an expansion of CD4+ T cells79. Potential modulation of atherosclerosis severity by FcγRIIB has also been evaluated, with either bone marrow reconstitution with FcγRIIB+/+ versus FcγRIIB−/− marrow in LDLR−/− mice, or global deletion of the receptor in apoE−/− mice. Independent of changes in plasma lipids, FcγRIIB omission by either strategy resulted in exaggerated atherosclerosis. In the study with global FcγRIIB silencing, there were both increased proinflammatory cytokines in the aorta and increased antibody titers to modified LDL. The latter finding may have been related to a loss of suppression of B cell production of antibodies to LDL80,81. Thus, consistent with the inflammatory nature of atherosclerosis, and with impacts that parallel their activating versus inhibitory input into immune responses, FcγR modulate atherosclerosis in mice.

Additional potential evidence of FcγR modulation of atherosclerosis severity comes from studies of the effect of immunoglobulin treatment in apoE−/− mice. The injection of polyclonal intravenous immunoglobulin preparations (IVIg) reduces fatty streak formation, and this is associated with reduced IgM antibodies to oxidized LDL and the inactivation of spleen and lymph node T cells82. Whereas the administration of intact immunoglobulin causes a decrease in lesion size and fewer macrophages within lesions, treatment with F(ab’)2 fragments of human immunoglobulin has no effect, thereby implicating involvement of the Fc region of IgG and FcγR.

The potential contribution of CRP to atherogenesis has also been investigated, using primarily gain-of-function strategies. Using generally similar strategies in male apoE−/− mice, the transgenic expression of human CRP has been found to either accelerate atherosclerosis progression83, or not affect the development or severity of atherosclerosis, with the increased atherosclerosis in the former study being attributed to excessively high CRP levels84,85. In apoE*3-Leiden transgenic mice expressing a human CRP transgene, the CRP did not affect atherosclerosis86. In atherosclerosis-prone ApoB100/100;LDLR−/− mice with human-like hypercholesterolemia, human CRP blunted atherosclerosis development87. In apoE−/− mice administered CRP via osmotic minipump for 4 weeks, atherosclerosis was unchanged88. Compared with wild-type rabbits, transgenic rabbits expressing human CRP developed similar atherosclerotic lesions on a cholesterol-rich diet89. Regarding loss-of-function studies, CRP-deficient mice displayed either equivalent or actually increased atherosclerotic lesions compared with controls90, and whereas CRP antisense oligonucleotides effectively lowered plasma CRP levels in Watanabe heritable hyperlipidemic rabbits, they did not impact atherosclerosis in the rabbits91. Recognizing that SAP is the acute phase reactant and CRP equivalent in mice, SAP concentrations have been measured during the development of atherosclerosis in apoE−/− mice, and they were found to be unchanged84,85. Collectively these observations in mice and rabbits do not support a role for CRP in atherosclerosis pathogenesis. Gain- or loss-of-function studies of potential participation of SAP in modulating atherosclerosis severity in mice have not yet been reported.

Vascular Injury, Occlusion and Thrombosis

The potential participation of FcγR and their ligands in altering the response to vascular injury and degree of thrombosis has been interrogated in mice. Using human CRP transgenic mice, it was demonstrated that the pentraxin causes exaggerated neointima formation following carotid artery ligation (Fig. 1B)92. Followup studies were done crossing the human CRP transgenic mouse with FcγR null mice, and in them neointima development was decreased with either FcγRI or FcγR γ chain deletion. In contrast, the deletion of FcγRIIB or FcγRIII resulted in neointima formation that was equal to or greater than that observed without FcγR manipulation; the findings with FcγRIII excision may have resulted from decreased competition for the γ chain that is common between FcγRI and FcγRIII93. Studies have also been performed in rats, with CRP administration causing increased neointima formation in a carotid artery angioplasty model57. In addition to influencing neointima formation, it has been demonstrated that CRP promotes thrombosis, with CRP transgenic mice displaying exaggerated clot formation in both a transluminal wire injury model and a photochemical arterial injury model of thrombosis (Fig. 1B).94 Furthermore, there is evidence that the impact of FcγR and their ligands on neointima development may relate to their modulation of thrombosis, since in a trans-femoral artery wire injury model in which thrombosis was controlled with aspirin and heparin, CRP transgenic mice displayed less neointima formation.95 The interpretation of findings regarding neointima development with FcγR manipulation may be complex because the FcγR γ chain plays a key role in platelet activation by collagen, and platelet activation may be instrumental to neointima initiation and progression.96 Overall, the animal model data available to date indicate that FcγR impact thrombotic and non-thrombotic vascular occlusion.

Hypertension

Since chronic elevations in CRP are associated with the development of hypertension in humans97, to evaluate a possible causal relationship, blood pressure has been studied in transgenic mice expressing rabbit CRP under the regulation of the phosphoenolpyruvate carboxykinase promoter. Compared with controls, CRP transgenic mice had hypertension that was predominantly systolic, and the severity of hypertension varied in parallel with changes in CRP levels modulated by dietary carbohydrate manipulation (Fig. 1B). The regulated transgene made it possible to study CRP levels as low as 9ug/ml, and such mice were hypertensive, indicating that modest elevations in CRP are sufficient to alter BP in the mouse. The CRP transgenic mice displayed exaggerated BP elevation in response to angiotensin II but not in response to norepinephrine, and there was a reduction in vascular angiotensin II receptor subtype 2 (AT2) expression. In contrast, the decline in BP with angiotensin II receptor subtype 1 (AT1) antagonism and vascular AT1 abundance were unaltered, indicating a selective effect of CRP on AT2. Ex vivo experiments further showed that the CRP-induced decrease in vascular AT2 is a direct effect on the vascular wall not requiring systemic responses, and that it is reversed by an NO donor, suggesting a role for NO deficiency in the in vivo process. In parallel, the chronic inhibition of NO synthase in wild-type mice attenuated vascular AT2 expression without affecting AT1. These findings provided direct evidence for CRP-induced hypertension in mice98. In rats, human CRP expression via an adeno-associated virus similarly resulted in hypertension, and there was also evidence of decreased NO production indicated by a fall in serum NO and urine cGMP and impaired endothelium-dependent relaxation99. In a complementary study, human CRP was expressed in the livers of spontaneously-hypertensive rats using a transgene under the control of the apoE promoter. The rats with elevated CRP displayed both greater systolic and diastolic blood pressure100. These collective findings indicate that CRP causes hypertension in rodents.

Myocardial Infarction

Since the relative rise in plasma CRP provoked by a myocardial infarction in humans is strongly associated with postinfarct morbidity and mortality101103, a potential contribution of CRP to disease severity has been evaluated. In studies in rats, the administration of human CRP following coronary artery ligation caused a 40% increase in infarct size (Fig. 1B)104. A small molecule inhibitor of CRP was then identified, 1,6-bis (phosphocholine)-hexane, which binds to CRP and occludes its ligand-binding B-face and thereby blocks its function. When administered to rats given human CRP following coronary artery ligation, the agent attenuated the increase in infarct size and the cardiac dysfunction caused by CRP.105 Using a rat CRP-specific antisense oligonucleotide, it was recently observed that lowering blood CRP resulted in reduced infarct size and improved cardiac function following myocardial infarction caused by ligation of the left anterior descending coronary artery in rats. In addition, in human CRP transgenic mice, declines in circulating human CRP induced by a human CRP-specific antisense oligonucleotide were associated with decreased neointima formation following carotid artery ligation106. The specific actions of CRP as well as possible isoforms of FcγR that influence myocardial infarction severity or outcome are unknown.

Insulin Resistance

Individuals with insulin resistance have markedly greater risk of developing cardiovascular disease, and there is mechanistic linkage between insulin resistance and endothelial dysfunction and vascular disease50. Since chronic elevations in CRP are associated with increased risk of both cardiovascular disorders and type 2 diabetes in humans107111, the impact of CRP on glucose homeostasis has been queried in mice. Using the mouse transgenic for rabbit CRP or the administration of human recombinant CRP to wild-type mice, it was discovered that elevations in CRP cause insulin resistance (Fig. 1B)112. Paralleling these findings, spontaneously-hypertensive rats expressing CRP driven by an apoE promoter-regulated transgene display glucose intolerance and hyperinsulinemia100. In the CRP transgenic mice, animals lacking FcγRIIB were protected from CRP-induced insulin resistance, and immunohistochemistry revealed that FcγRIIB is expressed in skeletal muscle microvascular endothelium. Consistent with the receptor distribution, the primary mechanism in glucose homeostasis disrupted by CRP was skeletal muscle glucose delivery, and CRP attenuated insulin-induced skeletal muscle blood flow. In contrast, CRP did not impair skeletal muscle glucose delivery in FcγRIIB−/− mice or in eNOS knock-in mice with phosphomimetic modification of eNOS Ser1176 (Ser1179 in human eNOS); eNOS Ser1176/9 is normally phosphorylated by insulin signaling to stimulate NO-mediated skeletal muscle blood flow and glucose delivery, and is it dephosphorylated by CRP/FcγRIIB112 Thus, CRP causes insulin resistance in mice through FcγRIIB-mediated inhibition of skeletal muscle glucose delivery, and this may represent an additional mechanism whereby FcγR and one of its ligands contribute to cardiovascular disease.

FcγR Ligands and CVD in Humans

IgG and Immune Complexes

Specific antigens and their respective IgG antibodies have been detected in the serum of patients with cardiovascular disease. In addition, IgG deposits have been detected in atherosclerotic lesions both in humans and in mice114115. Two identified antigens in humans are oxidized LDL and heat shock protein116,117, and along with their corresponding antibodies they have the potential to form immune complexes which may contribute to cardiovascular disease due to their proinflammatory properties involving either FcγR or complement activation. In an investigation in 52 patients undergoing carotid artery endarterectomy, there was a correlation between the levels of IgG against oxidized LDL at the time of surgery and arterial wall thickness six months later118. In a study of over 500 patients undergoing coronary angiography, IgG oxidized LDL autoantibodies and IgG apolipoprotein B-100 immune complexes were positively associated with angiographically-determined coronary artery disease in logistic regression analyses. However, in this population the relationships were not apparent in multivariate analyses116, and inconsistencies in correlations between IgGs and manifestations of cardiovascular disease have been observed between studies119. A potential explanation for such inconsistencies may be the lack of reproducible antigens used to quantify the IgGs120. Investigations in larger numbers of subjects, for example in the EPIC-Norfolk cohort, have failed to demonstrate a relationship between levels of IgG autoantibodies or immune complexes and coronary artery disease events121. Interestingly, for decades it has been recognized that serum levels of antibodies are elevated in patients with both essential and pregnancy-associated hypertension122125. The antigens targeted by these autoantibodies include the angiotensin II type-1 receptor, L-type voltage gated calcium channels, the alpha-1 adrenergic receptor, and the beta-1 adrenergic receptor126. Recognizing that these proteins regulate blood pressure by governing sodium and water reabsorption by the kidney, vascular tone, and cardiac output, the potential contributions of the autoantibodies to disease pathogenesis have thus far logically been attributed to antibody recognition of their antigen targets, and not to FcγR activation. Systemic autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis and the antiphospholipid syndrome, are characterized by accelerated atherosclerosis and greater risk of coronary artery disease that is not explained by traditional risk factors. The increase in vascular disease risk associated with these autoimmune conditions hypothetically may be related to the presence of autoantibodies and autoantigens and the subsequent formation of immune complexes127. However, evidence for a causal link between immune complex formation and cardiovascular disease risk or severity is currently lacking in patients with autoimmune disorders, as well as in individuals without known complications of autoimmunity.

Pentraxins

Pentraxins and Coronary Heart Disease

Over the past few decades massive data has accumulated indicating an association between modest elevations in circulating CRP and incident coronary heart disease107. This is perhaps best exemplified by two meta-analyses that have been performed, the first entailing 22 studies including over 7000 coronary heart disease cases and a mean follow-up period of 12 years. Comparing individuals in the top one-third versus the bottom one-third for baseline CRP level, the odds ratio for disease was 1.58 (95% CI 1.48–1.68)128. The second analysis of 23 studies comparing subjects with CRP levels of >3.0 mg/l (>3.0 ug/ml) versus <1.0 mg/l (<1.0 ug/ml) yielded a similar odds ratio of 1.58 (1.37–1.83)129.

Another approach that has been used to evaluate potential linkage between CRP and cardiovascular disease incidence or severity is to study an intervention that alters CRP and then determine how outcome is modified. This has been done in studies of statins which lower not only LDL cholesterol but also CRP130. In a study of 502 individuals with angiographically documented coronary disease, the impact of moderate versus intensive statin therapy (40 mg pravastatin versus 80 mg atorvastatin daily, respectively) on atherosclerosis progression was evaluated using intravascular ultrasonography. Decreases in both LDL cholesterol and CRP predictably occurred with statin treatment, and after adjustment for changes in lipids, the declines in CRP were independently correlated with less atherosclerosis progression131. In the JUPITER trial involving almost 18,000 subjects without cardiovascular disease at baseline, LDL cholesterol levels below 130 mg/dl, and CRP levels greater than or equal to 2 mg/l, outcomes with placebo versus 20 mg rosuvastatin treatment daily were compared. The trial was stopped early at a median of 1.9 years of follow-up when a 44% decline in vascular events was observed with rosuvastatin. This was associated with a 50% fall in LDL cholesterol and a 37% decrease in CRP132. The event rate in the placebo group and the clinical impact observed with treatment and the associated fall in CRP suggested that elevated levels of CRP increased vascular disease risk in these subjects, even when the LDL cholesterol level was within acceptable range based on guidelines at the time of the trial. However, recognizing that statins have numerous actions besides lowering CRP, the statin-based intervention trials shed only modest light on whether there is causal linkage between CRP and cardiovascular disease.

In addition to the effects of inflammatory insults and behavioral and environmental factors on CRP levels, 20–50% of the differences in CRP between individuals has been attributed to genetic variability133. Several single nucleotide polymorphisms (SNPs) in the CRP gene influence its abundance, and loci in other genes involved in inflammatory pathways have also been identified to impact CRP133,134. The identification of genetic modifiers of CRP allowed numerous studies to be performed involving Mendelian randomization, in which the genetic variants serve as likely unconfounded proxies for CRP levels to evaluate if CRP has a causal role in coronary heart disease135. The outcomes of the Mendelian randomization queries are exemplified by the collaborative study reported in 2011 involving close to 150,000 control subjects and more than 46,000 individuals with prevalent or incident coronary heart disease. Although four SNPs were demonstrated to influence CRP levels, and increasing CRP concentration was associated with increased risk of disease, no association was found between the SNPs and coronary heart disease136. When combined with the overall neutral outcomes of animal studies of CRP and atherosclerosis, such findings indicate that CRP does not likely contribute to coronary heart disease pathogenesis in humans.

Pentraxins and Hypertension

In a number of reports CRP levels have been shown to predict the development of hypertension in previously normotensive individuals. This was the case in the Women’s Health Study, in which 5365 women out of 20,525 with normal blood pressure at study onset developed hypertension over a median follow-up duration of 8 years137. Even after adjustment for a variety of risk factors, baseline CRP was an independent predictor of the development of incident hypertension. Comparable observations were made in the Framingham Offspring Study, in which 232 subjects out of 1456 initially normotensive individuals developed hypertension over a mean follow-up period of 3 years138. Thus, there is epidemiologic evidence linking CRP levels with incident hypertension.

As has been done in queries of possible linkage of CRP with coronary artery disease but in far fewer studies, the genetics governing CRP level and hypertension prevalence or development have been investigated. In a British Women’s Heart and Health Study investigation of a single CRP SNP, although the SNP was associated with a marked difference in CRP level and CRP was associated with systolic BP, there was no relationship between the SNP and the prevalence of hypertension139. In a study of almost 2000 Turkish subjects, CRP haplotypes were associated with hypertension in both men and women140, and two CRP SNPs were associated with hypertension in a study of 1400 control and 1331 Han Chinese with elevated BP141. However, in a larger study of 2000 Han Chinese in which eight CRP SNPs were investigated, although CRP levels were associated with increasing systolic as well as diastolic BP, none of the SNPs were associated with prevalent or incident hypertension142. In a recent study a weighted genetic risk score was created to evaluate the combined effects of genetic variants associated with changes in CRP levels on blood pressure in almost 750 Korean subjects. Individuals with the highest genetic risk score had CRP levels that were approximately 2.5-fold higher than subjects with the lowest genetic risk score, and an elevated genetic risk score increased the likelihood of hypertension (OR 2.18)143. Since it has been demonstrated that elevations in CRP cause hypertension in rodents (see above), additional human studies of the genetics of CRP regulation and hypertension may be warranted, including the application of the weighted genetic risk score for CRP to other populations.

Fcγ Receptors and CVD in Humans

Activating Fcγ Receptors

Activating FcγR, in particular FcγRI, FcγRIIA and FcγRIIIA, have been detected in human atherosclerotic lesions and in macrophages and other cell types in the medial and adventitial regions of the vascular wall144. The observed functions of activating FcγR in cell types of relevance to vascular health and the findings with receptor deletion in animal models of atherosclerosis (Fig. 1B) provide sound rationale for studies determining if genetic variants in FcγR influence the incidence or severity of vascular disease in humans. In a study of almost 900 subjects undergoing coronary angiography, the polymorphism FcγRIIIA-F158V was related to coronary artery disease, with individuals with FcγRIIIA-158V/V having decreased risk of disease (OR 0.53, CI 0.32–0.90). From a functional perspective, FcRγIIIA-158V/V displays greater IgG1 and IgG3 binding than FcγRIIIA-F/F145.

FcγRIIA is another activating FcγR for which genetic variation has been evaluated in a number of studies of cardiovascular disease. In a query of the polymorphism FcγRIIA-R131H involving over 700 patients with a first acute coronary syndrome (ACS) event compared to almost 500 individuals with stable angina pectoris, the FcγRIIA-131R/R genotype was associated with ACS as the first manifestation of coronary disease (OR 2.86, CI 2.06–3.99)146. In 553 individuals with either stable angina pectoris or unstable angina pectoris, those with FcγRIIA-131R/R were more likely to have unstable angina (OR 4.02, CI 2.52–6.41)147. In an evaluation of 430 individuals with peripheral atherosclerosis and 411 controls in the Rotterdam Study, FcγRIIA-131H heterozygous and homozygous subjects were protected against advanced peripheral atherosclerosis. The age- and gender-adjusted odds ratios were 0.77 (CI 0.54–1.12) and 0.65 (CI 0.44–0.98), respectively148. In 78 hypercholesterolemic subjects, homozygous carriers of the H allele compared with the R allele displayed better endothelial dependent vasodilation as assessed by changes in forearm blood flow in response to intra-arterial acetylcholine infusion149. In contrast to these four studies suggesting a cardiovascular health benefit of the H allele, in 1041 Finnish subjects, FcγRIIA-131H/H homozygotes had more premature atherosclerosis150, and in a number of other investigations, the FcγRIIA-H131R polymorphism was found to have no impact on cardiovascular disease risk145,151154. Regarding impact on receptor function, FcγRIIA-131R/R has increased signal transduction upon CRP binding compared with FcγRIIA-131H/H155. However, FcγRIIA-131H/H has higher binding efficiency for IgG2 and IgG3 than FcγRIIA-131R/R, resulting in decreased internalization of IgG2-opsonized particles by phagocytes in FcγRIIA-R/R individuals156. In addition to these studies of FcγRIIA SNPs, differences in receptor expression have been investigated. An evaluation of FcγRIIA abundance on peripheral monocytes by flow cytometry found that the receptor expression is decreased in subjects with clinical atherosclerosis compared with controls157. Recognizing that FcγRIIA participates with collagen in platelet activation, its abundance on platelets has also been assessed, and it was found to be increased in patients with acute myocardial infarction, unstable angina or ischemic stroke158. Not surprising considering the complex nature of FcγR biology, the cumulative available information about genetic variation in activating FcγR or differences in their expression has not yet added clarity to our understanding of how the receptors may influence cardiovascular health and disease in humans.

Inhibitory Fcγ Receptor FcγRIIB

In an attempt to understand the potential participation of FcγRIIB in cardiovascular disease in humans, advantage has been taken of previous interrogations of genetic variation in the receptor in the context of lupus159. One particular site of known single amino acid variation is amino acid 232, which is threonine (T) or isoleucine (I) depending on basepair 695 in exon 5 being C or T, respectively160. The T allele is less common than the I allele, with the T allele frequency 0.13 in Caucasians and 0.29 in African Americans161. Recognizing that this SNP resides in the transmembrane domain and may thereby affect receptor function, the abilities of human FcγRIIB-T232 versus FcγRIIB-I232 to antagonize eNOS were compared in cultured endothelial cells. Doing so tests the impact of the variant in the cell type in which FcγRIIB actions potently influence vascular health30,44,45. Cell context is critical because whereas prior studies of autoimmunity-related mechanisms showed less receptor function for FcγRIIB-T232 than FcγRIIB-I232 in monocytes, the opposite was found in B cells161,162. Endogenous FcγRIIB was knocked down in bovine aortic endothelial cells (BAEC) by siRNA, followed by sham transfection or introduction of either human FcγRIIB-T232 or FcγRIIB-I232 to equal abundance by transient transfection (Fig. 3A). CRP (25 ug/ml) antagonism of eNOS activation by VEGF (100 ng/ml) was then evaluated. Whereas sham-transfected cells deficient in FcγRIIB displayed no antagonism by CRP, CRP inhibited eNOS activation in cells expressing FcγRIIB-T232 (Fig. 3B). In contrast, in cells expressing FcγRIIB-I232 there was no antagonism of eNOS by CRP. Thus, the identity of amino acid 232 in human FcγRIIB markedly affects the capacity of the receptor to alter endothelial cell function, providing a mechanism-based genetic entry point for the study of FcγRIIB in vascular disease pathogenesis in humans.

Figure 3.

Figure 3

The FcγRIIB-I232 variant has attenuated capacity to mediate CRP action in endothelium. Following siRNA-based knockdown of endogenous FcγRIIB (GAAACCAGCCUCUGAAU, Dharmacon), bovine aortic endothelial cells were transfected with sham plasmid or cDNA encoding human FcγRIIB-T232 or FcγRIIB-I232. A. FcγRIIB abundance was evaluated by immunoblotting, with eNOS detection providing assessment of protein loading. B. CRP (25 ug/ml) antagonism of eNOS activation by VEGF (100 ng/ml) was evaluated by measuring 14C-L-arginine conversion to 14C-L-citrulline conversion by intact cells over 15 min. Values are mean±SEM, n=4, *p<0.05 vs no CRP.

In light of the preclinical observations made regarding FcγRIIB, its ligands, eNOS, endothelial function and hypertension30,44,45,98, FcγRIIB-I232/T232 and its potential influence on blood pressure were then interrogated in the Dallas Heart Study (DHS). DHS is a population-based epidemiologic study of Dallas County residents that has provided a cohort for successful genetic inquiry into processes underlying cardiovascular disorders and their risk factors163168. In the DHS, subjects between the ages of 30 to 65 years old have undergone extensive cardiovascular phenotyping169, including detailed studies of blood pressure163,170. 3493 subjects in DHS were genotyped for FcγRIIB-I232 versus FcγRIIB-T232, and due to high homology between FcγRIIB and FcγRIIC, the strategy entailed PCR followed by SNP genotyping by sequence-specific fluorescent hybridization probing160. The prevalence of genotypes is shown in Table 3, and as previously observed161, the T allele was less common than the I allele, particularly in non-African Americans. The study population then consisted of 2925 subjects without active malignancy, inflammatory illness, or a diagnosis of lupus for which genotype, CRP and BP data were available. The relationship between FcγRIIB-I232/T232 genotype and systolic BP was then evaluated for all subjects with CRP>2.0 mg/L (n=2069), noting that FcγRIIB genotype was not associated with CRP level. The genotype distribution in the study population was similar to that of the total population, and both were within the parameters of Hardy-Weinberg. In DHS subjects with CRP>2.0 mg/L of any race/ethnicity, the IT or TT genotype was associated with higher systolic BP compared with the II genotype (Table 4, upper panel), which encodes an FcγRIIB that is less able to antagonize eNOS (Fig. 3). Recognizing that the IT or TT genotype is more prevalent in African Americans (Table 3), who in general have higher BP than non-African-Americans for a variety of reasons171,172, separate analysis was performed in African Americans, and the association between genotype and systolic BP was again observed. Due to the low prevalence of the IT or TT genotype in Whites and Hispanics (Table 3), statistical power was insufficient to assess the genotype-phenotype link in those groups. Importantly, the 3–4 mmHg difference in systolic BP between genotypes observed for the SNP is similar in degree to the effect sizes for individual variants in known HTN susceptibility genes173,174. Since a number of subjects in the DHS query were on antihypertensive treatment, the comparisons were repeated using the recommended strategy of adding a constant to the observed BP in treated subjects (15 mmHg added to systolic BP)175. It was again observed that systolic BP is greater with IT or TT versus II genotype in the combined racial/ethnic groups and in African Americans (Table 4, lower panel). Importantly, subject age and sex distribution did not differ by genotype. These findings in a single population indicate that the FcγRIIB-I232/T232 variant impacts BP in the setting of elevated CRP in humans. Queries of the FcγRIIB-I232/T232 variant are now warranted in additional populations.

Table 3.

FcγRIIB-I232/T232 genotypes in the Dallas Heart Study population.

II IT TT
Total Population (n=3493) 2297 1058 138
66% 30% 4%
Blacks (n=1792) 948 721 123
53% 40% 7%
Whites (n=1032) 806 218 8
78% 21% 0.8%
Hispanics (n-595) 492 101 2
83% 17% 0.3%
Other (n=74) 51 18 5
69% 24% 7%

Table 4.

Impact of FcγRIIB-I232/T232 genotype on systolic blood pressure in Dallas Heart Study subjects with CRP>2.0 mg/L.

Overall African-American
IT or TT 130.6 ± 20.2 134.0 ± 21
(725) (536)
II 126.0 ± 18.9 131.0 ± 19.7
(1344) (608)
P<0.05 P<0.05
Corrected for Antihypertension Therapy
IT or TT 134.7 ± 22.9 138.2 ± 23.7
(725) (536)
II 129.3 ± 21.4 135.5 ± 22.2
(1344) (608)
P<0.05 P<0.05

(N values)

Conclusions and Current Unknowns

The recent interest in the contribution of inflammation to cardiovascular disorders has led to both preclinical and clinical studies of FcγR and their ligands in the context of cardiovascular disease. The activating FcγR, particularly FcγRI, FcγRIIA and FcγRIIIA, cause a variety of cellular responses in endothelial cells, vascular smooth muscle cells and monocytes/macrophages that may contribute to vascular disease pathogenesis (Fig. 1A). The lone inhibitory FγcR, FcγRIIB, which blunts proinflammatory processes in immune response cells, invokes detrimental processes in endothelial cells, inhibiting NO production, promoting adhesion and attenuating the capacity for endothelial monolayer repair. In animal models, activating FγcR promote atherosclerosis whereas FcγRIIB is atheroprotective, and activating FcγR additionally enhance thrombotic and non-thrombotic vascular occlusion (Fig. 1B).

Due to its upregulation under inflammatory conditions and perhaps also its stability in stored samples, the FcγR ligand CRP has been intensely studied. In mice and rats CRP causes hypertension and insulin resistance, and it exaggerates myocardial infarct size and related cardiac dysfunction (Fig. 1B). In contrast, CRP has been found to not impact atherosclerosis in animal models. A large volume of work has demonstrated an association between modest, chronic elevations in circulating CRP and coronary heart disease in humans. However, Mendelian randomization studies, which employ genetic variants influencing CRP abundance as unconfounded proxies for CRP levels, have shown that CRP is likely a marker of inflammation and not a mediator of coronary vascular disease. Studies evaluating CRP genetics and hypertension risk are currently insufficient to provide meaningful insight into a possible causal relationship.

Paralleling the genetic strategies assessing how CRP impacts cardiovascular health, inherited variants of activating FcγR have been studied. The information available to date does not yet add clarity to our understanding of how the receptors may influence vascular health and disease in humans. However, the first investigation of a loss-of-function variant of FcγRIIB in endothelium suggests that the receptor may contribute to the hypertension that often develops in individuals with chronically elevated circulating CRP (Fig. 3, Table 4).

At the same time that these new insights are recognized, multiple remaining knowledge gaps become apparent. Since the vast majority of mouse studies evaluating FcγR participation in vascular disease pathogenesis entailed global FcγR gene deletion, how individual FcγR in specific cell types impact cardiovascular health is unknown. This will require cell-specific gain- or loss-of-function studies in vivo. It is also poorly understood how individual FcγR ligands impact cardiovascular health in vivo, with different IgG subclasses having varying affinity for particular FcγR, and with FcγR additionally varying in their affinity for a given subclass of IgG. The identity of FcγR ligands that may be relevant to cardiovascular disease remains even more open-ended considering the potential diversity in circulating immune complexes, whose actions via FcγR may explain the greater predisposition for disease in individuals with autoimmune disorders. Finally, having observed that there is a common SNP in FcγRIIB that dramatically alters the endothelial actions of CRP (Fig. 3), it is critical to recognize that Mendelian randomization studies of FcγR ligands such as CRP or FcγR themselves may have limited utility because there is functionally-relevant genetic variation in multiple components in the FcγR ligand-receptor signaling pathway. Recognizing the various complexities that characterize FcγR biology, further attempts to understand how the receptors and their ligands impact vascular health will likely require the simplification that can only be afforded by discrete genetic manipulation or highly specific targeted pharmacologic intervention. Such efforts are warranted, however, because FcγR biology may underlie a considerable component of vascular disease predisposition that is not explained by traditional risk factors.

Acknowledgments

The authors would like to thank their numerous colleagues and collaborators who have contributed to the effort to better understand Fcγ receptors, their ligands and cardiovascular health and disease.

Sources of Funding

This work was supported by National Institutes of Health grant HL115122 (PWS), American Diabetes Association grant ADA 1–10-BS-124 (CM), American Heart Association grant 13GRNT16080003 (CM), and the Associates First Capital Distinguished Chair in Pediatrics (PWS).

Non-standard Abbreviations and Acronyms

AT1

angiotensin II receptor subtype 1

AT2

angiotensin II receptor subtype 2

BCR

B cell receptor

BAEC

bovine aortic endothelial cells

BTK

Bruton’s tyrosine kinase

CRP

C-reactive protein

DC

dendritic cell

DHS

Dallas Heart Study

eNOS

endothelial NO synthase

FcγR

Fcγ receptors

GADD153

growth arrest- and DNA damage-inducible gene 153

ITAM

immunoreceptor tyrosine-based activation motif

ITIM

immunoreceptor tyrosine-based inhibitory motif

IVIG

intravenous immunoglobulin

LDL-IC

LDL cholesterol-containing immune complex

MMP-2

matrix metalloproteinase-2

NOX4

NADPH oxidase 4 isoform

PLCγ

phospholipase Cγ

SAP

serum amyloid P component

SHIP

SH2 domain-containing inositol polyphosphate 5′ phosphatase

SHP1

SH2-domain -containing protein tyrosine phosphatase 1

TF

tissue factor

TG-CRP

CRP-transgenic mice

Footnotes

Conflict-of-Interest Disclosure

None.

References

  • 1.Ravetch JV, Bolland S. IgG Fc receptors. Annu Rev Immunol. 2001;19:275–290. doi: 10.1146/annurev.immunol.19.1.275. [DOI] [PubMed] [Google Scholar]
  • 2.Nimmerjahn F, Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat Rev Immunol. 2008;8:34–47. doi: 10.1038/nri2206. [DOI] [PubMed] [Google Scholar]
  • 3.Nimmerjahn F, Ravetch JV. Fcgamma receptors: old friends and new family members. Immunity. 2006;24:19–28. doi: 10.1016/j.immuni.2005.11.010. [DOI] [PubMed] [Google Scholar]
  • 4.Ravetch JV, Kinet JP. Fc receptors. Annu Rev Immunol. 1991;9:457–492. doi: 10.1146/annurev.iy.09.040191.002325. [DOI] [PubMed] [Google Scholar]
  • 5.van de Winkel JG, Anderson CL. Biology of human immunoglobulin G Fc receptors. J Leukoc Biol. 1991;49:511–524. doi: 10.1002/jlb.49.5.511. [DOI] [PubMed] [Google Scholar]
  • 6.Rothe G, Gabriel H, Kovacs E, Klucken J, Stohr J, Kindermann W, Schmitz G. Peripheral blood mononuclear phagocyte subpopulations as cellular markers in hypercholesterolemia. Arterioscler Thromb Vasc Biol. 1996;16:1437–1447. doi: 10.1161/01.atv.16.12.1437. [DOI] [PubMed] [Google Scholar]
  • 7.Dhodapkar KM, Banerjee D, Connolly J, Kukreja A, Matayeva E, Veri MC, Ravetch JV, Steinman RM, Dhodapkar MV. Selective blockade of the inhibitory Fcgamma receptor (FcgammaRIIB) in human dendritic cells and monocytes induces a type I interferon response program. J Exp Med. 2007;204(6):1359–1360. doi: 10.1084/jem.20062545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Molfetta R, Quatrini L, Gasparrini F, Zitti B, Santoni A, Paolini R. Regulation of fc receptor endocytic trafficking by ubiquitination. Front Immunol. 2014;5:449. doi: 10.3389/fimmu.2014.00449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Devaraj S, Du Clos TW, Jialal I. Binding and internalization of C-reactive protein by Fcgamma receptors on human aortic endothelial cells mediates biological effects. Arterioscler Thromb Vasc Biol. 2005;25:1359–1363. doi: 10.1161/01.ATV.0000168573.10844.ae. [DOI] [PubMed] [Google Scholar]
  • 10.Bruhns P. Properties of mouse and human IgG receptors and their contribution to disease models. Blood. 2012;119:5640–5649. doi: 10.1182/blood-2012-01-380121. [DOI] [PubMed] [Google Scholar]
  • 11.Du Clos TW. Function of C-reactive protein. Ann Med. 2000;32:274–278. doi: 10.3109/07853890009011772. [DOI] [PubMed] [Google Scholar]
  • 12.Du Clos TW. Pentraxins: structure, function, and role in inflammation. ISRN Inflamm. 2013;2013:379040. doi: 10.1155/2013/379040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Du Clos TW, Mold C. The role of C-reactive protein in the resolution of bacterial infection. Curr Opin Infect Dis. 2001;14:289–293. doi: 10.1097/00001432-200106000-00007. [DOI] [PubMed] [Google Scholar]
  • 14.Yeh ET, Anderson HV, Pasceri V, Willerson JT. C-reactive protein: linking inflammation to cardiovascular complications. Circulation. 2001;104:974–975. doi: 10.1161/01.cir.104.9.974. [DOI] [PubMed] [Google Scholar]
  • 15.Yasojima K, Schwab C, McGeer EG, McGeer PL. Generation of C-reactive protein and complement components in atherosclerotic plaques. Am J Pathol. 2001;158:1039–1051. doi: 10.1016/S0002-9440(10)64051-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kuji T, Masaki T, Li L, Cheung AK. Expression of C-reactive protein in myointimal hyperplasia in a porcine arteriovenous graft model. Nephrol Dial Transplant. 2007;22:2469–2475. doi: 10.1093/ndt/gfm240. [DOI] [PubMed] [Google Scholar]
  • 17.Venugopal SK, 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;166:1265–1271. doi: 10.1016/S0002-9440(10)62345-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Chu CS, Wang YC, Lu LS, Walton B, Yilmaz HR, Huang RY, Sawamura T, Dixon RA, Lai WT, Chen CH, Lu J. Electronegative low-density lipoprotein increases C-reactive protein expression in vascular endothelial cells through the LOX-1 receptor. PLoS One. 2013;8:e70533. doi: 10.1371/journal.pone.0070533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Pang X, Liu J, Zhao J, Mao J, Zhang X, Feng L, Han C, Li M, Wang S, Wu D. Homocysteine induces the expression of C-reactive protein via NMDAr-ROS-MAPK-NF-kappaB signal pathway in rat vascular smooth muscle cells. Atherosclerosis. 2014;236:73–81. doi: 10.1016/j.atherosclerosis.2014.06.021. [DOI] [PubMed] [Google Scholar]
  • 20.Peng N, Liu JT, Gao DF, Lin R, Li R. Angiotensin II-induced C-reactive protein generation: inflammatory role of vascular smooth muscle cells in atherosclerosis. Atherosclerosis. 2007;193:292–298. doi: 10.1016/j.atherosclerosis.2006.09.007. [DOI] [PubMed] [Google Scholar]
  • 21.Wang C, Liu J, Guo F, Ji Y, Liu N. Endothelin-1 induces the expression of C-reactive protein in rat vascular smooth muscle cells. Biochem Biophys Res Commun. 2009;389:537–542. doi: 10.1016/j.bbrc.2009.09.023. [DOI] [PubMed] [Google Scholar]
  • 22.Memoli B, Procino A, Calabro P, Esposito P, Grandaliano G, Pertosa G, Prete MD, Andreucci M, Lillo SD, Ferulano G, Cillo C, Savastano S, Colao A, Guida B. Inflammation may modulate IL-6 and C-reactive protein gene expression in the adipose tissue: the role of IL-6 cell membrane receptor. Am J Physiol Endocrinol Metab. 2007;293:E1030–E1035. doi: 10.1152/ajpendo.00697.2006. [DOI] [PubMed] [Google Scholar]
  • 23.Ouchi N, Kihara S, Funahashi T, Nakamura T, Nishida M, Kumada M, Okamoto Y, Ohashi K, Nagaretani H, Kishida K, Nishizawa H, Maeda N, Kobayashi H, Hiraoka H, Matsuzawa Y. Reciprocal association of C-reactive protein with adiponectin in blood stream and adipose tissue. Circulation. 2003;107:671–674. doi: 10.1161/01.cir.0000055188.83694.b3. [DOI] [PubMed] [Google Scholar]
  • 24.Anty R, Bekri S, Luciani N, Saint-Paul MC, Dahman M, Iannelli A, Amor IB, Staccini-Myx A, Huet PM, Gugenheim J, Sadoul JL, Le Marchand-Brustel Y, Tran A, Gual P. The inflammatory C-reactive protein is increased in both liver and adipose tissue in severely obese patients independently from metabolic syndrome, Type 2 diabetes, and NASH. Am J Gastroenterol. 2006;101:1824–1833. doi: 10.1111/j.1572-0241.2006.00724.x. [DOI] [PubMed] [Google Scholar]
  • 25.Diehl EE, Haines GK, III, Radosevich JA, Potempa LA. Immunohistochemical localization of modified C-reactive protein antigen in normal vascular tissue. Am J Med Sci. 2000;319:79–83. doi: 10.1097/00000441-200002000-00002. [DOI] [PubMed] [Google Scholar]
  • 26.Potempa LA, Maldonado BA, Laurent P, Zemel ES, Gewurz H. Antigenic, electrophoretic and binding alterations of human C-reactive protein modified selectively in the absence of calcium. Mol Immunol. 1983;20:1165–1175. doi: 10.1016/0161-5890(83)90140-2. [DOI] [PubMed] [Google Scholar]
  • 27.Rees RF, Gewurz H, Siegel JN, Coon J, Potempa LA. Expression of a C-reactive protein neoantigen (neo-CRP) in inflamed rabbit liver and muscle. Clin Immunol Immunopathol. 1988;48:95–107. doi: 10.1016/0090-1229(88)90160-2. [DOI] [PubMed] [Google Scholar]
  • 28.Eisenhardt SU, Habersberger J, Murphy A, Chen YC, Woollard KJ, Bassler N, Qian H, von Zur MC, Hagemeyer CE, Ahrens I, Chin-Dusting J, Bobik A, Peter K. Dissociation of pentameric to monomeric C-reactive protein on activated platelets localizes inflammation to atherosclerotic plaques. Circ Res. 2009;105:128–137. doi: 10.1161/CIRCRESAHA.108.190611. [DOI] [PubMed] [Google Scholar]
  • 29.Khreiss T, Jozsef L, Potempa LA, Filep JG. Conformational rearrangement in C-reactive protein is required for proinflammatory actions on human endothelial cells. Circulation. 2004;109:2016–2022. doi: 10.1161/01.CIR.0000125527.41598.68. [DOI] [PubMed] [Google Scholar]
  • 30.Sundgren NC, Zhu W, Yuhanna IS, Chambliss KL, Ahmed M, Tanigaki K, Umetani M, Mineo C, Shaul PW. Coupling of fcgamma receptor I to fcgamma receptor IIb by SRC kinase mediates C-reactive protein impairment of endothelial function. Circ Res. 2011;109:1132–1140. doi: 10.1161/CIRCRESAHA.111.254573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ji SR, Ma L, Bai CJ, Shi JM, Li HY, Potempa LA, Filep JG, Zhao J, Wu Y. Monomeric C-reactive protein activates endothelial cells via interaction with lipid raft microdomains. FASEB J. 2009;23:1806–1816. doi: 10.1096/fj.08-116962. [DOI] [PubMed] [Google Scholar]
  • 32.Schwedler SB, Kuhlencordt PJ, Ponnuswamy PP, Hatiboglu G, Quaschning T, Widder J, Wanner C, Potempa LA, Galle J. Native C-reactive protein induces endothelial dysfunction in ApoE−/− mice: implications for iNOS and reactive oxygen species. Atherosclerosis. 2007;195:e76–e84. doi: 10.1016/j.atherosclerosis.2007.06.013. [DOI] [PubMed] [Google Scholar]
  • 33.Crowell RE, Du Clos TW, Montoya G, Heaphy E, Mold C. C-reactive protein receptors on the human monocytic cell line U-937 Evidence for additional binding to Fc gamma RI. J Immunol. 1991;147:3445–3451. [PubMed] [Google Scholar]
  • 34.Marnell LL, Mold C, Volzer MA, Burlingame RW, Du Clos TW. C-reactive protein binds to Fc gamma RI in transfected COS cells. J Immunol. 1995;155:2185–2193. [PubMed] [Google Scholar]
  • 35.Bharadwaj D, Stein MP, Volzer M, Mold C, Du Clos TW. The major receptor for C-reactive protein on leukocytes is fcgamma receptor II. J Exp Med. 1999;190:585–590. doi: 10.1084/jem.190.4.585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Stein MP, Edberg JC, Kimberly RP, Mangan EK, Bharadwaj D, Mold C, Du Clos TW. C-reactive protein binding to FcgammaRIIa on human monocytes and neutrophils is allele-specific. J Clin Invest. 2000;105:369–376. doi: 10.1172/JCI7817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Stein MP, Mold C, Du Clos TW. C-reactive protein binding to murine leukocytes requires Fc gamma receptors. J Immunol. 2000;164:1514–1520. doi: 10.4049/jimmunol.164.3.1514. [DOI] [PubMed] [Google Scholar]
  • 38.Mold C, Baca R, Du Clos TW. Serum Amyloid P Component and C-Reactive Protein Opsonize Apoptotic Cells for Phagocytosis through Fcgamma Receptors. J Autoimmun. 2002;19:147–154. doi: 10.1006/jaut.2002.0615. [DOI] [PubMed] [Google Scholar]
  • 39.Lu J, Marnell LL, Marjon KD, Mold C, Du Clos TW, Sun PD. Structural recognition and functional activation of FcgammaR by innate pentraxins. Nature. 2008;456:989–992. doi: 10.1038/nature07468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Venugopal SK, Devaraj S, Yuhanna I, Shaul P, Jialal I. Demonstration that C-reactive protein decreases eNOS expression and bioactivity in human aortic endothelial cells. Circulation. 2002;106:1439–1441. doi: 10.1161/01.cir.0000033116.22237.f9. [DOI] [PubMed] [Google Scholar]
  • 41.Devaraj S, Xu DY, Jialal I. C-reactive protein increases plasminogen activator inhibitor-1 expression and activity in human aortic endothelial cells: implications for the metabolic syndrome and atherothrombosis. Circulation. 2003;107:398–404. doi: 10.1161/01.cir.0000052617.91920.fd. [DOI] [PubMed] [Google Scholar]
  • 42.Devaraj S, Kumaresan PR, Jialal I. Effect of C-reactive protein on chemokine expression in human aortic endothelial cells. J Mol Cell Cardiol. 2004;36:405–410. doi: 10.1016/j.yjmcc.2003.12.005. [DOI] [PubMed] [Google Scholar]
  • 43.Devaraj S, Davis B, Simon SI, Jialal I. CRP promotes monocyte-endothelial cell adhesion via Fcgamma receptors in human aortic endothelial cells under static and shear flow conditions. Am J Physiol Heart Circ Physiol. 2006;291:H1170–H1176. doi: 10.1152/ajpheart.00150.2006. [DOI] [PubMed] [Google Scholar]
  • 44.Schwartz R, Osborne-Lawrence S, Hahner L, Gibson LL, Gormley AK, Vongpatanasin W, Zhu W, Word RA, Seetharam D, Black S, Samols D, Mineo C, Shaul PW. C-reactive protein downregulates endothelial NO synthase and attenuates reendothelialization in vivo in mice. Circ Res. 2007;100:1452–1459. doi: 10.1161/01.RES.0000267745.03488.47. [DOI] [PubMed] [Google Scholar]
  • 45.Mineo C, Gormley AK, Yuhanna IS, Osborne-Lawrence S, Gibson LL, Hahner L, Shohet RV, Black S, Salmon JE, Samols D, Karp DR, Thomas GD, Shaul PW. FcgammaRIIB mediates C-reactive protein inhibition of endothelial NO synthase. Circ Res. 2005;97:1124–1131. doi: 10.1161/01.RES.0000194323.77203.fe. [DOI] [PubMed] [Google Scholar]
  • 46.Hein TW, Singh U, Vasquez-Vivar J, Devaraj S, Kuo L, Jialal I. Human C-reactive protein induces endothelial dysfunction and uncoupling of eNOS in vivo. Atherosclerosis. 2009;206:61–68. doi: 10.1016/j.atherosclerosis.2009.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Black S, Kushner I, Samols D. C-reactive Protein. J Biol Chem. 2004;279:48487–48490. doi: 10.1074/jbc.R400025200. [DOI] [PubMed] [Google Scholar]
  • 48.Teoh H, Quan A, Lovren F, Wang G, Tirgari S, Szmitko PE, Szalai AJ, Ward ME, Verma S. Impaired endothelial function in C-reactive protein overexpressing mice. Atherosclerosis. 2008;201:318–325. doi: 10.1016/j.atherosclerosis.2008.02.034. [DOI] [PubMed] [Google Scholar]
  • 49.Tanigaki K, Mineo C, Yuhanna IS, Chambliss KL, Quon MJ, Bonvini E, Shaul PW. C-reactive protein inhibits insulin activation of endothelial nitric oxide synthase via the immunoreceptor tyrosine-based inhibition motif of FcgammaRIIB and SHIP-1. Circ Res. 2009;104:1275–1282. doi: 10.1161/CIRCRESAHA.108.192906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Muniyappa R, Montagnani M, Koh KK, Quon MJ. Cardiovascular actions of insulin. Endocr Rev. 2007;28:463–491. doi: 10.1210/er.2007-0006. [DOI] [PubMed] [Google Scholar]
  • 51.Veri MC, Gorlatov S, Li H, Burke S, Johnson S, Stavenhagen J, Stein KE, Bonvini E, Koenig S. Monoclonal antibodies capable of discriminating the human inhibitory Fcgamma-receptor IIB (CD32B) from the activating Fcgamma-receptor IIA (CD32A): biochemical, biological and functional characterization. Immunology. 2007;121:392–404. doi: 10.1111/j.1365-2567.2007.02588.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Blaschke F, Bruemmer D, Yin F, Takata Y, Wang W, Fishbein MC, Okura T, Higaki J, Graf K, Fleck E, Hsueh WA, Law RE. C-reactive protein induces apoptosis in human coronary vascular smooth muscle cells. Circulation. 2004;110:579–587. doi: 10.1161/01.CIR.0000136999.77584.A2. [DOI] [PubMed] [Google Scholar]
  • 53.Ryu J, Lee CW, Shin JA, Park CS, Kim JJ, Park SJ, Han KH. FcgammaRIIa mediates C-reactive protein-induced inflammatory responses of human vascular smooth muscle cells by activating NADPH oxidase 4. Cardiovasc Res. 2007;75:555–565. doi: 10.1016/j.cardiores.2007.04.027. [DOI] [PubMed] [Google Scholar]
  • 54.Hattori Y, Matsumura M, Kasai K. Vascular smooth muscle cell activation by C-reactive protein. Cardiovasc Res. 2003;58:186–195. doi: 10.1016/s0008-6363(02)00855-6. [DOI] [PubMed] [Google Scholar]
  • 55.Doronzo G, Russo I, Mattiello L, Trovati M, Anfossi G. C-reactive protein increases matrix metalloproteinase-2 expression and activity in cultured human vascular smooth muscle cells. J Lab Clin Med. 2005;146:287–298. doi: 10.1016/j.lab.2005.07.010. [DOI] [PubMed] [Google Scholar]
  • 56.Wu J, Stevenson MJ, Brown JM, Grunz EA, Strawn TL, Fay WP. C-reactive protein enhances tissue factor expression by vascular smooth muscle cells: mechanisms and in vivo significance. Arterioscler Thromb Vasc Biol. 2008;28:698–704. doi: 10.1161/ATVBAHA.107.160903. [DOI] [PubMed] [Google Scholar]
  • 57.Wang CH, Li SH, Weisel RD, Fedak PW, Dumont AS, Szmitko P, Li RK, Mickle DA, Verma S. C-reactive protein upregulates angiotensin type 1 receptors in vascular smooth muscle. Circulation. 2003;107:1783–1790. doi: 10.1161/01.CIR.0000061916.95736.E5. [DOI] [PubMed] [Google Scholar]
  • 58.Lopes-Virella MF, Binzafar N, Rackley S, Takei A, La VM, Virella G. The uptake of LDL-IC by human macrophages: predominant involvement of the Fc gamma RI receptor. Atherosclerosis. 1997;135:161–170. doi: 10.1016/s0021-9150(97)00157-3. [DOI] [PubMed] [Google Scholar]
  • 59.Huang Y, Fleming AJ, Wu S, Virella G, Lopes-Virella MF. Fc-gamma receptor cross-linking by immune complexes induces matrix metalloproteinase-1 in U937 cells via mitogen-activated protein kinase. Arterioscler Thromb Vasc Biol. 2000;20:2533–2538. doi: 10.1161/01.atv.20.12.2533. [DOI] [PubMed] [Google Scholar]
  • 60.Luo Y, Pollard JW, Casadevall A. Fcgamma receptor cross-linking stimulates cell proliferation of macrophages via the ERK pathway. J Biol Chem. 2010;285:4232–4242. doi: 10.1074/jbc.M109.037168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Oksjoki R, Kovanen PT, Lindstedt KA, Jansson B, Pentikainen MO. OxLDL-IgG immune complexes induce survival of human monocytes. Arterioscler Thromb Vasc Biol. 2006;26:576–583. doi: 10.1161/01.ATV.0000201041.14438.8d. [DOI] [PubMed] [Google Scholar]
  • 62.Hansson GK, Bondjers G, Bylock A, Hjalmarsson L. Ultrastructural studies on the localization of IgG in the aortic endothelium and subendothelial intima of atherosclerotic and nonatherosclerotic rabbits. Exp Mol Pathol. 1980;33:302–315. doi: 10.1016/0014-4800(80)90028-3. [DOI] [PubMed] [Google Scholar]
  • 63.Parums D, Mitchinson MJ. Demonstration of immunoglobulin in the neighbourhood of advanced atherosclerotic plaques. Atherosclerosis. 1981;38:211–216. doi: 10.1016/0021-9150(81)90118-0. [DOI] [PubMed] [Google Scholar]
  • 64.Yla-Herttuala S, Palinski W, Butler SW, Picard S, Steinberg D, Witztum JL. Rabbit and human atherosclerotic lesions contain IgG that recognizes epitopes of oxidized LDL. Arterioscler Thromb. 1994;14:32–40. doi: 10.1161/01.atv.14.1.32. [DOI] [PubMed] [Google Scholar]
  • 65.Boulaftali Y, Hess PR, Kahn ML, Bergmeier W. Platelet immunoreceptor tyrosine-based activation motif (ITAM) signaling and vascular integrity. Circ Res. 2014;114:1174–1184. doi: 10.1161/CIRCRESAHA.114.301611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cines DB, Schreiber AD. Immune thrombocytopenia Use of a Coombs antiglobulin test to detect IgG and C3 on platelets. N Engl J Med. 1979;300:106–111. doi: 10.1056/NEJM197901183000302. [DOI] [PubMed] [Google Scholar]
  • 67.Clawson CC, Rao GH, White JG. Platelet interaction with bacteria. IV. Stimulation of the release reaction. Am J Pathol. 1975;81:411–420. [PMC free article] [PubMed] [Google Scholar]
  • 68.Davoren A, Aster RH. Heparin-induced thrombocytopenia and thrombosis. Am J Hematol. 2006;81:36–44. doi: 10.1002/ajh.20490. [DOI] [PubMed] [Google Scholar]
  • 69.Reilly MP, McKenzie SE. Insights from mouse models of heparin-induced thrombocytopenia and thrombosis. Curr Opin Hematol. 2002;9:395–400. doi: 10.1097/00062752-200209000-00002. [DOI] [PubMed] [Google Scholar]
  • 70.Warkentin TE. Heparin-induced thrombocytopenia: pathogenesis and management. Br J Haematol. 2003;121:535–555. doi: 10.1046/j.1365-2141.2003.04334.x. [DOI] [PubMed] [Google Scholar]
  • 71.Cox D, Kerrigan SW, Watson SP. Platelets and the innate immune system: mechanisms of bacterial-induced platelet activation. J Thromb Haemost. 2011;9:1097–1107. doi: 10.1111/j.1538-7836.2011.04264.x. [DOI] [PubMed] [Google Scholar]
  • 72.Fitzgerald JR, Foster TJ, Cox D. The interaction of bacterial pathogens with platelets. Nat Rev Microbiol. 2006;4:445–457. doi: 10.1038/nrmicro1425. [DOI] [PubMed] [Google Scholar]
  • 73.McKenzie SE, Taylor SM, Malladi P, Yuhan H, Cassel DL, Chien P, Schwartz E, Schreiber AD, Surrey S, Reilly MP. The role of the human Fc receptor Fc gamma RIIA in the immune clearance of platelets: a transgenic mouse model. J Immunol. 1999;162:4311–4318. [PubMed] [Google Scholar]
  • 74.Hansson GK, Libby P, Schonbeck U, Yan ZQ. Innate and adaptive immunity in the pathogenesis of atherosclerosis. Circ Res. 2002;91:281–291. doi: 10.1161/01.res.0000029784.15893.10. [DOI] [PubMed] [Google Scholar]
  • 75.Hernandez-Vargas P, Ortiz-Munoz G, Lopez-Franco O, Suzuki Y, Gallego-Delgado J, Sanjuan G, Lazaro A, Lopez-Parra V, Ortega L, Egido J, Gomez-Guerrero C. Fcgamma receptor deficiency confers protection against atherosclerosis in apolipoprotein E knockout mice. Circ Res. 2006;99:1188–1196. doi: 10.1161/01.RES.0000250556.07796.6c. [DOI] [PubMed] [Google Scholar]
  • 76.Sumiyoshi K, Mokuno H, Iesaki T, Shimada K, Miyazaki T, Kume A, Kiyanagi T, Kuremoto K, Watanabe Y, Tada N, Daida H. Deletion of the Fc receptors gamma chain preserves endothelial function affected by hypercholesterolaemia in mice fed on a high-fat diet. Cardiovasc Res. 2008;80:463–470. doi: 10.1093/cvr/cvn206. [DOI] [PubMed] [Google Scholar]
  • 77.Mallavia B, Oguiza A, Lopez-Franco O, Recio C, Ortiz-Munoz G, Lazaro I, Lopez-Parra V, Egido J, Gomez-Guerrero C. Gene Deficiency in Activating Fcgamma Receptors Influences the Macrophage Phenotypic Balance and Reduces Atherosclerosis in Mice. PLoS One. 2013;8:e66754. doi: 10.1371/journal.pone.0066754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ng HP, Burris RL, Nagarajan S. Attenuated atherosclerotic lesions in apoE-Fcgamma-chain-deficient hyperlipidemic mouse model is associated with inhibition of Th17 cells and promotion of regulatory T cells. J Immunol. 2011;187:6082–6093. doi: 10.4049/jimmunol.1004133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kelly JA, Griffin ME, Fava RA, Wood SG, Bessette KA, Miller ER, Huber SA, Binder CJ, Witztum JL, Morganelli PM. Inhibition of arterial lesion progression in CD16-deficient mice: evidence for altered immunity and the role of IL-10. Cardiovasc Res. 2010;85:224–231. doi: 10.1093/cvr/cvp300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Mendez-Fernandez YV, Stevenson BG, Diehl CJ, Braun NA, Wade NS, Covarrubias R, van LS, Witztum JL, Major AS. The inhibitory FcgammaRIIb modulates the inflammatory response and influences atherosclerosis in male apoE(−/−) mice. Atherosclerosis. 2011;214:73–80. doi: 10.1016/j.atherosclerosis.2010.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zhao M, Wigren M, Duner P, Kolbus D, Olofsson KE, Bjorkbacka H, Nilsson J, Fredrikson GN. FcgammaRIIB inhibits the development of atherosclerosis in low-density lipoprotein receptor-deficient mice. J Immunol. 2010;184:2253–2260. doi: 10.4049/jimmunol.0902654. [DOI] [PubMed] [Google Scholar]
  • 82.Nicoletti A, Kaveri S, Caligiuri G, Bariety J, Hansson GK. Immunoglobulin treatment reduces atherosclerosis in apo E knockout mice. J Clin Invest. 1998;102:910–918. doi: 10.1172/JCI119892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Paul A, Ko KW, Li L, Yechoor V, McCrory MA, Szalai AJ, Chan L. C-reactive protein accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2004;109:647–655. doi: 10.1161/01.CIR.0000114526.50618.24. [DOI] [PubMed] [Google Scholar]
  • 84.Hirschfield GM, Gallimore JR, Kahan MC, Hutchinson WL, Sabin CA, Benson GM, Dhillon AP, Tennent GA, Pepys MB. Transgenic human C-reactive protein is not proatherogenic in apolipoprotein E-deficient mice. Proc Natl Acad Sci U S A. 2005;102:8309–8314. doi: 10.1073/pnas.0503202102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tennent GA, Hutchinson WL, Kahan MC, Hirschfield GM, Gallimore JR, Lewin J, Sabin CA, Dhillon AP, Pepys MB. Transgenic human CRP is not pro-atherogenic, pro-atherothrombotic or pro-inflammatory in apoE−/− mice. Atherosclerosis. 2008;196:248–255. doi: 10.1016/j.atherosclerosis.2007.05.010. [DOI] [PubMed] [Google Scholar]
  • 86.Trion A, de Maat MP, Jukema JW, van der Laarse A, Maas MC, Offerman EH, Havekes LM, Szalai AJ, Princen HM, Emeis JJ. 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;25:1635–1640. doi: 10.1161/01.ATV.0000171992.36710.1e. [DOI] [PubMed] [Google Scholar]
  • 87.Kovacs A, Tornvall P, Nilsson R, Tegner J, Hamsten A, Bjorkegren J. Human C-reactive protein slows atherosclerosis development in a mouse model with human-like hypercholesterolemia. Proc Natl Acad Sci U S A. 2007;104:13768–13773. doi: 10.1073/pnas.0706027104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ortiz MA, Campana GL, Woods JR, Boguslawski G, Sosa MJ, Walker CL, Labarrere CA. Continuously-infused human C-reactive protein is neither proatherosclerotic nor proinflammatory in apolipoprotein E-deficient mice. Exp Biol Med (Maywood) 2009;234:624–631. doi: 10.3181/0812-RM-347. [DOI] [PubMed] [Google Scholar]
  • 89.Koike T, Kitajima S, Yu Y, Nishijima K, Zhang J, Ozaki Y, Morimoto M, Watanabe T, Bhakdi S, Asada Y, Chen YE, Fan J. Human C-reactive protein does not promote atherosclerosis in transgenic rabbits. Circulation. 2009;120:2088–2094. doi: 10.1161/CIRCULATIONAHA.109.872796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Teupser D, Weber O, Rao TN, Sass K, Thiery J, Fehling HJ. No reduction of atherosclerosis in C-reactive protein (CRP)-deficient mice. J Biol Chem. 2011;286:6272–6279. doi: 10.1074/jbc.M110.161414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yu Q, Liu Z, Waqar AB, Ning B, Yang X, Shiomi M, Graham MJ, Crooke RM, Liu E, Dong S, Fan J. Effects of antisense oligonucleotides against C-reactive protein on the development of atherosclerosis in WHHL rabbits. Mediators Inflamm. 2014;2014:979132. doi: 10.1155/2014/979132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wang D, Oparil S, Chen YF, McCrory MA, Skibinski GA, Feng W, Szalai AJ. Estrogen treatment abrogates neointima formation in human C-reactive protein transgenic mice. Arterioscler Thromb Vasc Biol. 2005;25:2094–2099. doi: 10.1161/01.ATV.0000179602.85797.3f. [DOI] [PubMed] [Google Scholar]
  • 93.Xing D, Hage FG, Chen YF, McCrory MA, Feng W, Skibinski GA, Majid-Hassan E, Oparil S, Szalai AJ. Exaggerated neointima formation in human C-reactive protein transgenic mice is IgG Fc receptor type I (Fc gamma RI)-dependent. Am J Pathol. 2008;172:22–30. doi: 10.2353/ajpath.2008.070154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Danenberg HD, Szalai AJ, Swaminathan RV, Peng L, Chen Z, Seifert P, Fay WP, Simon DI, Edelman ER. Increased thrombosis after arterial injury in human C-reactive protein-transgenic mice. Circulation. 2003;108:512–515. doi: 10.1161/01.CIR.0000085568.13915.1E. [DOI] [PubMed] [Google Scholar]
  • 95.Danenberg HD, Grad E, Swaminathan RV, Chen Z, Seifert P, Szalai AJ, Lotan C, Simon DI, Edelman ER. Neointimal formation is reduced after arterial injury in human crp transgenic mice. Atherosclerosis. 2008;201:85–91. doi: 10.1016/j.atherosclerosis.2008.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Konishi H, Katoh Y, Takaya N, Kashiwakura Y, Itoh S, Ra C, Daida H. Platelets activated by collagen through immunoreceptor tyrosine-based activation motif play pivotal role in initiation and generation of neointimal hyperplasia after vascular injury. Circulation. 2002;105:912–916. doi: 10.1161/hc0802.105256. [DOI] [PubMed] [Google Scholar]
  • 97.Hage FG. C-reactive protein and hypertension. J Hum Hypertens. 2014;28:410–415. doi: 10.1038/jhh.2013.111. [DOI] [PubMed] [Google Scholar]
  • 98.Vongpatanasin W, Thomas GD, Schwartz R, Cassis LA, Osborne-Lawrence S, Hahner L, Gibson LL, Black S, Samols D, Shaul PW. C-reactive protein causes downregulation of vascular angiotensin subtype 2 receptors and systolic hypertension in mice. Circulation. 2007;115:1020–1028. doi: 10.1161/CIRCULATIONAHA.106.664854. [DOI] [PubMed] [Google Scholar]
  • 99.Guan H, Wang P, Hui R, Edin ML, Zeldin DC, Wang DW. Adeno-associated virus-mediated human C-reactive protein gene delivery causes endothelial dysfunction and hypertension in rats. Clin Chem. 2009;55:274–284. doi: 10.1373/clinchem.2008.115857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pravenec M, Kajiya T, Zidek V, Landa V, Mlejnek P, Simakova M, Silhavy J, Malinska H, Oliyarnyk O, Kazdova L, Fan J, Wang J, Kurtz TW. Effects of human C-reactive protein on pathogenesis of features of the metabolic syndrome. Hypertension. 2011;57:731–737. doi: 10.1161/HYPERTENSIONAHA.110.164350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Pietila KO, Harmoinen AP, Jokiniitty J, Pasternack AI. Serum C-reactive protein concentration in acute myocardial infarction and its relationship to mortality during 24 months of follow-up in patients under thrombolytic treatment. Eur Heart J. 1996;17:1345–1349. doi: 10.1093/oxfordjournals.eurheartj.a015068. [DOI] [PubMed] [Google Scholar]
  • 102.Suleiman M, Aronson D, Reisner SA, Kapeliovich MR, Markiewicz W, Levy Y, Hammerman H. Admission C-reactive protein levels and 30-day mortality in patients with acute myocardial infarction. Am J Med. 2003;115:695–701. doi: 10.1016/j.amjmed.2003.06.008. [DOI] [PubMed] [Google Scholar]
  • 103.Suleiman M, Khatib R, Agmon Y, Mahamid R, Boulos M, Kapeliovich M, Levy Y, Beyar R, Markiewicz W, Hammerman H, Aronson D. Early inflammation and risk of long-term development of heart failure and mortality in survivors of acute myocardial infarction predictive role of C-reactive protein. J Am Coll Cardiol. 2006;47:962–968. doi: 10.1016/j.jacc.2005.10.055. [DOI] [PubMed] [Google Scholar]
  • 104.Griselli M, Herbert J, Hutchinson WL, Taylor KM, Sohail M, Krausz T, Pepys MB. C-reactive protein and complement are important mediators of tissue damage in acute myocardial infarction. J Exp Med. 1999;190:1733–1740. doi: 10.1084/jem.190.12.1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Pepys MB, Hirschfield GM, Tennent GA, Gallimore JR, Kahan MC, Bellotti V, Hawkins PN, Myers RM, Smith MD, Polara A, Cobb AJ, Ley SV, Aquilina JA, Robinson CV, Sharif I, Gray GA, Sabin CA, Jenvey MC, Kolstoe SE, Thompson D, Wood SP. Targeting C-reactive protein for the treatment of cardiovascular disease. Nature. 2006;440:1217–1221. doi: 10.1038/nature04672. [DOI] [PubMed] [Google Scholar]
  • 106.Szalai AJ, McCrory MA, Xing D, Hage FG, Miller A, Oparil S, Chen YF, Mazzone M, Early R, Henry SP, Zanardi TA, Graham MJ, Crooke RM. Inhibiting C-reactive protein for the treatment of cardiovascular disease: promising evidence from rodent models. Mediators Inflamm. 2014;2014:353614. doi: 10.1155/2014/353614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Casas JP, Shah T, Hingorani AD, Danesh J, Pepys MB. C-reactive protein and coronary heart disease: a critical review. J Intern Med. 2008;264:295–314. doi: 10.1111/j.1365-2796.2008.02015.x. [DOI] [PubMed] [Google Scholar]
  • 108.Festa A, D'Agostino R, Jr, Howard G, Mykkanen L, Tracy RP, Haffner SM. Chronic subclinical inflammation as part of the insulin resistance syndrome: the Insulin Resistance Atherosclerosis Study (IRAS) Circulation. 2000;102:42–47. doi: 10.1161/01.cir.102.1.42. [DOI] [PubMed] [Google Scholar]
  • 109.Haffner SM. Insulin resistance, inflammation, and the prediabetic state. Am J Cardiol. 2003;92:18J–26J. doi: 10.1016/s0002-9149(03)00612-x. [DOI] [PubMed] [Google Scholar]
  • 110.Pradhan AD, Cook NR, Buring JE, Manson JE, Ridker PM. C-reactive protein is independently associated with fasting insulin in nondiabetic women. Arterioscler Thromb Vasc Biol. 2003;23:650–655. doi: 10.1161/01.ATV.0000065636.15310.9C. [DOI] [PubMed] [Google Scholar]
  • 111.Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA. 2001;286:327–334. doi: 10.1001/jama.286.3.327. [DOI] [PubMed] [Google Scholar]
  • 112.Tanigaki K, Vongpatanasin W, Barrera JA, Atochin DN, Huang PL, Bonvini E, Shaul PW, Mineo C. C-reactive Protein Causes Insulin Resistance in Mice Through Fcgamma Receptor IIB-Mediated Inhibition of Skeletal Muscle Glucose Delivery. Diabetes. 2013;62:721–731. doi: 10.2337/db12-0133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Yudkin JS, Stehouwer CD, Emeis JJ, Coppack SW. C-reactive protein in healthy subjects: associations with obesity, insulin resistance, and endothelial dysfunction: a potential role for cytokines originating from adipose tissue? Arterioscler Thromb Vasc Biol. 1999;19:972–978. doi: 10.1161/01.atv.19.4.972. [DOI] [PubMed] [Google Scholar]
  • 114.Vlaicu R, Rus HG, Niculescu F, Cristea A. Immunoglobulins and complement components in human atherosclerotic intima. Atherosclerosis. 1985;55:35–50. doi: 10.1016/0021-9150(85)90164-9. [DOI] [PubMed] [Google Scholar]
  • 115.Doring Y, Manthey HD, Drechsler M, Lievens D, Megens RT, Soehnlein O, Busch M, Manca M, Koenen RR, Pelisek J, Daemen MJ, Lutgens E, Zenke M, Binder CJ, Weber C, Zernecke A. Auto- antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation. 2012;125:1673–1693. doi: 10.1161/CIRCULATIONAHA.111.046755. [DOI] [PubMed] [Google Scholar]
  • 116.Tsimikas S, Brilakis ES, Lennon RJ, Miller ER, Witztum JL, McConnell JP, Kornman KS, Berger PB. Relationship of IgG and IgM autoantibodies to oxidized low density lipoprotein with coronary artery disease and cardiovascular events. J Lipid Res. 2007;48:425–433. doi: 10.1194/jlr.M600361-JLR200. [DOI] [PubMed] [Google Scholar]
  • 117.Zhang X, He M, Cheng L, Chen Y, Zhou L, Zeng H, Pockley AG, Hu FB, Wu T. Elevated heat shock protein 60 levels are associated with higher risk of coronary heart disease in Chinese. Circulation. 2008;118:2687–2693. doi: 10.1161/CIRCULATIONAHA.108.781856. [DOI] [PubMed] [Google Scholar]
  • 118.Meraviglia MV, Maggi E, Bellomo G, Cursi M, Fanelli G, Minicucci F. Autoantibodies against oxidatively modified lipoproteins and progression of carotid restenosis after carotid endarterectomy. Stroke. 2002;33:1139–1141. doi: 10.1161/01.str.0000014420.15948.2e. [DOI] [PubMed] [Google Scholar]
  • 119.Moohebati M, Kabirirad V, Ghayour-Mobarhan M, Esmaily H, Tavallaie S, Akhavan RA, Pourghadamyari H, Sahebkar A. Investigation of serum oxidized low-density lipoprotein IgG levels in patients with angiographically defined coronary artery disease. Int J Vasc Med. 2014;2014:845960. doi: 10.1155/2014/845960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Tsiantoulas D, Diehl CJ, Witztum JL, Binder CJ. B cells and humoral immunity in atherosclerosis. Circ Res. 2014;114:1743–1756. doi: 10.1161/CIRCRESAHA.113.301145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Ravandi A, Boekholdt SM, Mallat Z, Talmud PJ, Kastelein JJ, Wareham NJ, Miller ER, Benessiano J, Tedgui A, Witztum JL, Khaw KT, Tsimikas S. Relationship of IgG and IgM autoantibodies and immune complexes to oxidized LDL with markers of oxidation and inflammation and cardiovascular events: results from the EPIC-Norfolk Study. J Lipid Res. 2011;52:1829–1836. doi: 10.1194/jlr.M015776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ebringer A, Doyle AE. Raised serum IgG levels in hypertension. Br Med J. 1970;2:146–148. doi: 10.1136/bmj.2.5702.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Gudbrandsson T, Hansson L, Herlitz H, Lindholm L, Nilsson LA. Immunological changes in patients with previous malignant essential hypertension. Lancet. 1981;1:406–408. doi: 10.1016/s0140-6736(81)91790-6. [DOI] [PubMed] [Google Scholar]
  • 124.Hilme E, Herlitz H, Soderstrom T, Hansson L. Increased secretion of immunoglobulins in malignant hypertension. J Hypertens. 1989;7:91–95. [PubMed] [Google Scholar]
  • 125.Suryaprabha P, Padma T, Rao UB. Increased serum IgG levels in essential hypertension. Immunol Lett. 1984;8:143–145. doi: 10.1016/0165-2478(84)90067-1. [DOI] [PubMed] [Google Scholar]
  • 126.Chan CT, Lieu M, Toh BH, Kyaw TS, Bobik A, Sobey CG, Drummond GR. Antibodies in the Pathogenesis of Hypertension. Biomed Res Int. 2014;2014:504045. doi: 10.1155/2014/504045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.McMahon M, Hahn BH. Atherosclerosis and systemic lupus erythematosus: mechanistic basis of the association. Curr Opin Immunol. 2007;19:633–639. doi: 10.1016/j.coi.2007.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Danesh J, Wheeler JG, Hirschfield GM, Eda S, Eiriksdottir G, Rumley A, Lowe GD, Pepys MB, Gudnason V. C-reactive protein and other circulating markers of inflammation in the prediction of coronary heart disease. N Engl J Med. 2004;350:1387–1397. doi: 10.1056/NEJMoa032804. [DOI] [PubMed] [Google Scholar]
  • 129.Buckley DI, Fu R, Freeman M, Rogers K, Helfand M. C-reactive protein as a risk factor for coronary heart disease: a systematic review meta-analyses for the U.S Preventive Services Task Force. Ann Intern Med. 2009;151:483–495. doi: 10.7326/0003-4819-151-7-200910060-00009. [DOI] [PubMed] [Google Scholar]
  • 130.Albert MA, Danielson E, Rifai N, Ridker PM. Effect of statin therapy on C-reactive protein levels: the pravastatin inflammation/CRP evaluation (PRINCE): a randomized trial and cohort study. JAMA. 2001;286:64–70. doi: 10.1001/jama.286.1.64. [DOI] [PubMed] [Google Scholar]
  • 131.Nissen SE, Tuzcu EM, Schoenhagen P, Crowe T, Sasiela WJ, Tsai J, Orazem J, Magorien RD, O'Shaughnessy C, Ganz P. Statin therapy, LDL cholesterol, C-reactive protein, and coronary artery disease. N Engl J Med. 2005;352:29–38. doi: 10.1056/NEJMoa042000. [DOI] [PubMed] [Google Scholar]
  • 132.Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Jr, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJ. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359:2195–2207. doi: 10.1056/NEJMoa0807646. [DOI] [PubMed] [Google Scholar]
  • 133.Hage FG, Szalai AJ. The role of C-reactive protein polymorphisms in inflammation and cardiovascular risk. Curr Atheroscler Rep. 2009;11:124–130. doi: 10.1007/s11883-009-0020-z. [DOI] [PubMed] [Google Scholar]
  • 134.Dehghan A, Dupuis J, Barbalic M, et al. Meta-analysis of genome-wide association studies in >80 000 subjects identifies multiple loci for C-reactive protein levels. Circulation. 2011;123:731–738. doi: 10.1161/CIRCULATIONAHA.110.948570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Nordestgaard BG. Does elevated C-reactive protein cause human atherothrombosis? Novel insights from genetics, intervention trials, and elsewhere. Curr Opin Lipidol. 2009;20:393–401. doi: 10.1097/MOL.0b013e3283307bfe. [DOI] [PubMed] [Google Scholar]
  • 136.Wensley F, Gao P, Burgess S, Kaptoge S, et al. Association between C reactive protein and coronary heart disease: mendelian randomisation analysis based on individual participant data. BMJ. 2011;342:d548. doi: 10.1136/bmj.d548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Sesso HD, Buring JE, Rifai N, Blake GJ, Gaziano JM, Ridker PM. C-reactive protein and the risk of developing hypertension. JAMA. 2003;290:2945–2951. doi: 10.1001/jama.290.22.2945. [DOI] [PubMed] [Google Scholar]
  • 138.Wang TJ, Gona P, Larson MG, Levy D, Benjamin EJ, Tofler GH, Jacques PF, Meigs JB, Rifai N, Selhub J, Robins SJ, Newton-Cheh C, Vasan RS. Multiple biomarkers and the risk of incident hypertension. Hypertension. 2007;49:432–438. doi: 10.1161/01.HYP.0000256956.61872.aa. [DOI] [PubMed] [Google Scholar]
  • 139.Davey SG, Lawlor DA, Harbord R, Timpson N, Rumley A, Lowe GD, Day IN, Ebrahim S. Association of C-reactive protein with blood pressure and hypertension: life course confounding and mendelian randomization tests of causality. Arterioscler Thromb Vasc Biol. 2005;25:1051–1056. doi: 10.1161/01.ATV.0000160351.95181.d0. [DOI] [PubMed] [Google Scholar]
  • 140.Komurcu-Bayrak E, Erginel-Unaltuna N, Onat A, Ozsait B, Eklund C, Hurme M, Mononen N, Laaksonen R, Hergenc G, Lehtimaki T. Association of C-reactive protein (CRP) gene allelic variants with serum CRP levels and hypertension in Turkish adults. Atherosclerosis. 2009;206:474–479. doi: 10.1016/j.atherosclerosis.2009.03.030. [DOI] [PubMed] [Google Scholar]
  • 141.Zhao Y, Wang H, Liu S, Zhao X, Chen Y, Yang Y, Wang W, Wu Y, Chen A, Tang J, Yao Y, Li Y, Chen J, Shen C, Yang S. Association study of CRP gene polymorphism and hypertension in Han Chinese population. Gene. 2013;512:41–46. doi: 10.1016/j.gene.2012.09.107. [DOI] [PubMed] [Google Scholar]
  • 142.Kong H, Qian YS, Tang XF, Zhang J, Gao PJ, Zhang Y, Zhu DL. C-reactive protein (CRP) gene polymorphisms, CRP levels and risk of incident essential hypertension: findings from an observational cohort of Han Chinese. Hypertens Res. 2012;35:1019–1023. doi: 10.1038/hr.2012.89. [DOI] [PubMed] [Google Scholar]
  • 143.Hong EP, Kim DH, Suh JG, Park JW. Genetic risk assessment for cardiovascular disease with seven genes associated with plasma C-reactive protein concentrations in Asian populations. Hypertens Res. 2014;37:692–698. doi: 10.1038/hr.2014.56. [DOI] [PubMed] [Google Scholar]
  • 144.Ratcliffe NR, Kennedy SM, Morganelli PM. Immunocytochemical detection of Fcgamma receptors in human atherosclerotic lesions. Immunol Lett. 2001;77:169–174. doi: 10.1016/s0165-2478(01)00217-6. [DOI] [PubMed] [Google Scholar]
  • 145.Gavasso S, Nygard O, Pedersen ER, Aarseth JH, Bleie O, Myhr KM, Vedeler CA. Fcgamma receptor IIIA polymorphism as a risk-factor for coronary artery disease. Atherosclerosis. 2005;180:277–282. doi: 10.1016/j.atherosclerosis.2004.12.011. [DOI] [PubMed] [Google Scholar]
  • 146.Raaz D, Herrmann M, Ekici AB, Klinghammer L, Lausen B, Voll RE, Leusen JH, van de Winkel JG, Daniel WG, Reis A, Garlichs CD. FcgammaRIIa genotype is associated with acute coronary syndromes as first manifestation of coronary artery disease. Atherosclerosis. 2009;205:512–516. doi: 10.1016/j.atherosclerosis.2009.01.013. [DOI] [PubMed] [Google Scholar]
  • 147.Raaz-Schrauder D, Ekici AB, Munoz LE, Klinghammer L, Voll RE, Leusen JH, van de Winkel JG, Reis A, Schett G, Garlichs CD, Herrmann M. Patients with unstable angina pectoris show an increased frequency of the Fc gamma RIIa R131 allele. Autoimmunity. 2012;45:556–564. doi: 10.3109/08916934.2012.682665. [DOI] [PubMed] [Google Scholar]
  • 148.van der Meer IM, Witteman JC, Hofman A, Kluft C, de Maat MP. Genetic variation in Fcgamma receptor IIa protects against advanced peripheral atherosclerosis The Rotterdam Study. Thromb Haemost. 2004;92:1273–1276. doi: 10.1160/TH04-05-0268. [DOI] [PubMed] [Google Scholar]
  • 149.Schneider MP, Leusen JH, Herrmann M, Garlichs CD, Amann K, John S, Schmieder RE. The Fcgamma receptor IIA R131H gene polymorphism is associated with endothelial function in patients with hypercholesterolaemia. Atherosclerosis. 2011;218:411–415. doi: 10.1016/j.atherosclerosis.2011.07.009. [DOI] [PubMed] [Google Scholar]
  • 150.Sampi M, Ukkola O, Paivansalo M, Kesaniemi YA, Horkko S. Early atherosclerosis and IgG2 to bacteria are associated with FcgammaRIIa genotype in non-smokers. Eur J Clin Invest. 2009;39:517–526. doi: 10.1111/j.1365-2362.2009.02138.x. [DOI] [PubMed] [Google Scholar]
  • 151.Gardemann A, Horstmann A, Santoso S. Fc gamma RIIa-R131H polymorphism: its impact on coronary heart disease in a German cohort. Thromb Haemost. 2003;90:1218–1220. [PubMed] [Google Scholar]
  • 152.Karakas M, Hoffmann MM, Vollmert C, Rothenbacher D, Meisinger C, Winkelmann B, Khuseyinova N, Bohm BO, Illig T, Marz W, Koenig W. Genetic variation in Fc gamma receptor IIa and risk of coronary heart disease: negative results from two large independent populations. BMC Med Genet. 2009;10:46. doi: 10.1186/1471-2350-10-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Mollaki V, Steeds RP, Samani NJ, Channer KS, Daly ME. The FcgammaRIIa His131Arg polymorphism and its association with myocardial infarction. J Thromb Haemost. 2004;2:1014–1015. doi: 10.1111/j.1538-7836.2004.00750.x. [DOI] [PubMed] [Google Scholar]
  • 154.Murphy A, White A, Nair R, Kingwell B, Duffy S, Chin-Dusting J, Woollard K. C-reactive protein and Fc gamma RIIa functional polymorphisms are not associated with clinical presentation of stable and unstable angina. Thromb Haemost. 2007l;97:681–682. [PubMed] [Google Scholar]
  • 155.Zuniga R, Markowitz GS, Arkachaisri T, Imperatore EA, D'Agati VD, Salmon JE. Identification of IgG subclasses and C-reactive protein in lupus nephritis: the relationship between the composition of immune deposits and FCgamma receptor type IIA alleles. Arthritis Rheum. 2003;48:460–470. doi: 10.1002/art.10930. [DOI] [PubMed] [Google Scholar]
  • 156.Sanders LA, Feldman RG, Voorhorst-Ogink MM, de HM, Rijkers GT, Capel PJ, Zegers BJ, van de Winkel JG. Human immunoglobulin G (IgG) Fc receptor IIA (CD32) polymorphism and IgG2-mediated bacterial phagocytosis by neutrophils. Infect Immun. 1995;63:73–81. doi: 10.1128/iai.63.1.73-81.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Pfeiffer JR, Howes PS, Waters MA, Hynes ML, Schnurr PP, Demidenko E, Bech FR, Morganelli PM. Levels of expression of Fcgamma receptor IIA (CD32) are decreased on peripheral blood monocytes in patients with severe atherosclerosis. Atherosclerosis. 2001;155:211–218. doi: 10.1016/s0021-9150(00)00541-4. [DOI] [PubMed] [Google Scholar]
  • 158.Calverley DC, Brass E, Hacker MR, Tsao-Wei DD, Espina BM, Pullarkat VA, Hodis HN, Groshen S. Potential role of platelet FcgammaRIIA in collagen-mediated platelet activation associated with atherothrombosis. Atherosclerosis. 2002;164:261–267. doi: 10.1016/s0021-9150(02)00179-x. [DOI] [PubMed] [Google Scholar]
  • 159.Smith KG, Clatworthy MR. FcgammaRIIB in autoimmunity and infection: evolutionary and therapeutic implications. Nat Rev Immunol. 2010;10:328–343. doi: 10.1038/nri2762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Kyogoku C, Dijstelbloem HM, Tsuchiya N, Hatta Y, Kato H, Yamaguchi A, Fukazawa T, Jansen MD, Hashimoto H, van de Winkel JG, Kallenberg CG, Tokunaga K. Fcgamma receptor gene polymorphisms in Japanese patients with systemic lupus erythematosus: contribution of FCGR2B to genetic susceptibility. Arthritis Rheum. 2002;46:1242–1254. doi: 10.1002/art.10257. [DOI] [PubMed] [Google Scholar]
  • 161.Li X, Wu J, Carter RH, Edberg JC, Su K, Cooper GS, Kimberly RP. A novel polymorphism in the Fcgamma receptor IIB (CD32B) transmembrane region alters receptor signaling. Arthritis Rheum. 2003;48:3242–3252. doi: 10.1002/art.11313. [DOI] [PubMed] [Google Scholar]
  • 162.Floto RA, Clatworthy MR, Heilbronn KR, Rosner DR, MacAry PA, Rankin A, Lehner PJ, Ouwehand WH, Allen JM, Watkins NA, Smith KG. Loss of function of a lupus-associated FcgammaRIIb polymorphism through exclusion from lipid rafts. Nat Med. 2005;11:1056–1058. doi: 10.1038/nm1288. [DOI] [PubMed] [Google Scholar]
  • 163.Dries DL, Victor RG, Rame JE, Cooper RS, Wu X, Zhu X, Leonard D, Ho SI, Wu Q, Post W, Drazner MH. Corin gene minor allele defined by 2 missense mutations is common in blacks and associated with high blood pressure and hypertension. Circulation. 2005;112:2403–2410. doi: 10.1161/CIRCULATIONAHA.105.568881. [DOI] [PubMed] [Google Scholar]
  • 164.Khera A, McGuire DK, Murphy SA, Stanek HG, Das SR, Vongpatanasin W, Wians FH, Jr, Grundy SM, de Lemos JA. Race and gender differences in C-reactive protein levels. J Am Coll Cardiol. 2005;46:464–469. doi: 10.1016/j.jacc.2005.04.051. [DOI] [PubMed] [Google Scholar]
  • 165.Khera A, Vega GL, Das SR, Ayers C, McGuire DK, Grundy SM, de Lemos JA. Sex differences in the relationship between C-reactive protein and body fat. J Clin Endocrinol Metab. 2009;94:3251–3258. doi: 10.1210/jc.2008-2406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Khera A, de Lemos JA, Peshock RM, Lo HS, Stanek HG, Murphy SA, Wians FH, Jr, Grundy SM, McGuire DK. Relationship between C-reactive protein and subclinical atherosclerosis: the Dallas Heart Study. Circulation. 2006;113:38–43. doi: 10.1161/CIRCULATIONAHA.105.575241. [DOI] [PubMed] [Google Scholar]
  • 167.Grundy SM, Adams-Huet B, Vega GL. Variable contributions of fat content and distribution to metabolic syndrome risk factors. Metab Syndr Relat Disord. 2008;6:281–288. doi: 10.1089/met.2008.0026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Kozlitina J, Boerwinkle E, Cohen JC, Hobbs HH. Dissociation between APOC3 variants, hepatic triglyceride content and insulin resistance. Hepatology. 2011;53:467–474. doi: 10.1002/hep.24072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Victor RG, Haley RW, Willett DL, Peshock RM, Vaeth PC, Leonard D, Basit M, Cooper RS, Iannacchione VG, Visscher WA, Staab JM, Hobbs HH. The Dallas Heart Study: a population-based probability sample for the multidisciplinary study of ethnic differences in cardiovascular health. Am J Cardiol. 2004;93:1473–1480. doi: 10.1016/j.amjcard.2004.02.058. [DOI] [PubMed] [Google Scholar]
  • 170.Vaeth PA, Willett DL. Level of acculturation and hypertension among Dallas County Hispanics: findings from the Dallas Heart Study. Ann Epidemiol. 2005;15:373–380. doi: 10.1016/j.annepidem.2004.11.003. [DOI] [PubMed] [Google Scholar]
  • 171.Flack JM, Sica DA, Bakris G, Brown AL, Ferdinand KC, Grimm RH, Jr, Hall WD, Jones WE, Kountz DS, Lea JP, Nasser S, Nesbitt SD, Saunders E, Scisney-Matlock M, Jamerson KA. Management of high blood pressure in Blacks: an update of the International Society on Hypertension in Blacks consensus statement. Hypertension. 2010;56:780–800. doi: 10.1161/HYPERTENSIONAHA.110.152892. [DOI] [PubMed] [Google Scholar]
  • 172.Redmond N, Baer HJ, Hicks LS. Health behaviors and racial disparity in blood pressure control in the national health and nutrition examination survey. Hypertension. 2011;57:383–389. doi: 10.1161/HYPERTENSIONAHA.110.161950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Faruque MU, Chen G, Doumatey A, Huang H, Zhou J, Dunston GM, Rotimi CN, Adeyemo AA. Association of ATP1B1, RGS5 and SELE polymorphisms with hypertension and blood pressure in African-Americans. J Hypertens. 2011;29:1906–1912. doi: 10.1097/HJH.0b013e32834b000d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Chang YP, Liu X, Kim JD, Ikeda MA, Layton MR, Weder AB, Cooper RS, Kardia SL, Rao DC, Hunt SC, Luke A, Boerwinkle E, Chakravarti A. Multiple genes for essential-hypertension susceptibility on chromosome 1q. Am J Hum Genet. 2007;80:253–264. doi: 10.1086/510918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Tobin MD, Sheehan NA, Scurrah KJ, Burton PR. Adjusting for treatment effects in studies of quantitative traits: antihypertensive therapy and systolic blood pressure. Stat Med. 2005;24:2911–2935. doi: 10.1002/sim.2165. [DOI] [PubMed] [Google Scholar]

RESOURCES