Skip to main content
  • Research article
  • Open access
  • Published:

The plasma level of mCRP is linked to cardiovascular disease in antineutrophil cytoplasmic antibody-associated vasculitis

Abstract

Background

C-reactive protein (CRP) has two natural isomers: C-reactive protein pentamer (pCRP) and C-reactive protein monomer (mCRP). The levels of CRP are significantly elevated in patients with anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). mCRP not only activates the endothelial cells, platelets, leukocytes, and complements, but also has a proinflammatory structural subtype that can localize and deposit in inflammatory tissues. Thus, it regulates a variety of clinical diseases, such as ischemia/reperfusion (I/R) injury, Alzheimer’s disease, age-related macular degeneration, and cardiovascular disease. We hypothesized that plasma mCRP levels are related to cardiovascular disease in AAV.

Methods

In this cross-sectional study, 37 patients with AAV were assessed. Brain natriuretic peptide (BNP) and mCRP in plasma were assessed by enzyme-linked immunosorbent assay (ELISA). The acute ST-segment elevation myocardial infarction (STEMI) was diagnosed by coronary angiography, and the Gensini score calculated. Echocardiography evaluated the ejection fraction (EF%), left ventricular end-diastolic volume (LVEDV), left ventricular end-systolic volume (LVESV), and left ventricular mass index (LVMI). Estimated glomerular filtration rate (eGFR) was calculated based on serum creatinine, age, and gender.

Results

The plasma level of mCRP in AAV was significantly higher than that in healthy volunteers (P < 0.001). Then, mCRP and CRP levels were compared with and without STEMI complications in AAV. The plasma level of mCRP was higher, but that of CRP was lower in STEMI. The plasma level of mCRP was correlated with Birmingham vasculitis activity score (BVAS), eGFR, BNP, EF%, LVEDV, LVESV, LVMI, and STEMI complications’ Gensini score in AAV; however, CRP did not correlate with BNP, EF%, LVEDV, LVESV, LVMI, and Gensini score.

Conclusions

The plasma level of mCRP was related to cardiovascular diseases in AAV patients.

Introduction

Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is commonly characterized by a small amount of immunoglobulin deposition and segmental vascular wall necrotic inflammation [1]. AAV patients are classified into granulomas with polyangiitis (GPA), eosinophilic granulomas with polyangiitis (EGPA), and microvascular polyangiitis (MPA), according to pathological and clinical characteristics [2]. Target antigens for AAV diseases include protease 3 (PR3) and myeloperoxidase (MPO) [3, 4]. The majority of the Chinese patients with AAV are positive for MPO-ANCA [5,6,7], which activates and damages glomerular endothelial cells (GEnC) in patients with AAV [7,8,9].

C-reactive protein (CRP) is an acute-phase protein that serves as an inflammatory response biomarker [10, 11]. During activity and relapse of AAV disease, serum CRP levels increase rapidly, followed by a decrease during remission of the disease [12, 13]. Human CRP in blood circulation consists of C-reactive protein pentamer (pCRP) and tissue-related C-reactive protein monomer (mCRP) [14]. In atherosclerosis-related experiments in mice, CRP bound to the Fc receptor CD32, while mCRP bound to another human Fc receptor subtype CD16 in neutrophils and performed function that was opposite to that of CRP [15]. Thus, FC receptor may be the key that links CRP and mCRP to AAV.

The most common pathogenesis of ST-segment elevation myocardial infarction (STEMI) is atherosclerotic fragile plaque rupture and thrombus-induced cardiac muscle cell necrosis. Also, inflammatory reactions are crucial responses [16, 17]. CRP is involved in acute myocardial infarction. mCRP is obtained from the dissociation process of necrobiosis. Moreover, mCRP explains the function of CRP, while pCRP does not promote inflammation [18]. In vitro tests have shown that mCRP induces the synthesis of neutrophil-derived chemokines, stimulates the expression of monocyte integrin [19], and promotes neutrophil adhesion to vascular endothelial cells [20].

CRP is the classical acute-phase protein named for its ability to precipitate and bind to the pneumococcal C-polysaccharide. Although the blood circulation concentration is low in healthy individuals, the level of CRP increases sharply when tissue damage and inflammation occur [21, 22]. Currently, CRP is used not only as a marker of inflammation but also as a crucial and independent predictor of atherosclerotic thrombosis (including cardiovascular events) [22].

Active AAV patients have high levels of CRP [13, 23]. Also, renal involvement is common in patients with AAV, including renal tubulointerstitial (TI) lesions [24, 25]. mCRP is an acute-phase protein in tissues or cells, wherein it might act as an antigen in acute tubular interstitium (ATIN) [25]. Serum CRP is decomposed into mCRP and induces the generation of free oxygen radicals on the surface of apoptotic and necrotic cells, resulting in inflammatory reactions [26]. It was also found that mCRP is not only related to thrombosis [27] and atherosclerotic plaque rupture but can also be concentrated in localized areas after transient myocardial ischemia and related to acute myocardial infarction (AMI) [28]. Therefore, we hypothesized that plasma mCRP levels are related to cardiovascular disease in AAV.

Methods

Collecting plasma samples

This cross-sectional study utilized plasma samples collected from 37 patients with AAV and 20 healthy volunteers. The AAV patients were diagnosed in the Department of Nephrology, Affiliated Hospital of Inner Mongolia Medical University, from October 2018 to November 2019. The patients met the criteria set at the Chapel Hill Consensus Meeting [29]. All the participants provided informed consent. Subjects with other autoimmune diseases, such as IgA nephropathy, rheumatoid arthritis, Graves’ disease, and lupus nephritis, as well as secondary and various acute infectious vasculitis, were excluded. A volume of 5 mL venous blood from each patient was withdrawn and collected in EDTA anticoagulation tubes within 24 h of admission. The supernatant was separated by centrifugation at 3000 rpm, 10 min, and stored at − 80 °C for subsequent use. Repeated freeze-thaw cycles are prohibited for this sample. The disease activity was based on the Birmingham vasculitis activity score (BVAS) in AAV patients [30]. We collected baseline patient data including status, age, gender, tobacco, alcohol, hypertension, diabetes, obesity, and biochemical indexes. These cardiovascular risk factors included triglyceride (TG), total cholesterol (Tch), low-density lipoprotein (LDL-C), high-density lipoprotein (HDL-C), CRP, and fibrinogen (Fib). The study was approved by the Ethics Committee of the Helsinki Declaration of Inner Mongolia Medical University.

Detection of mCRP by enzyme-linked immunosorbent assay (ELISA)

Plasma mCRP levels were tested by ELISA using a commercial kit (Kamaishu Biotechnology Co, Shanghai, China). The 96-well plate was coated with mCRP and blocked to reduce non-specific binding. Antibodies compete for binding to a plate or sample. This kit uses the double-antibody sandwich method to determine the level of human mCRP in the specimen. The microtiter plates were coated with purified human mCRP antibodies to make solid-phase antibodies. Then, mCRP was added to the microwells, followed by the addition of HRP-labeled mCRP antibodies to form an antibody-antigen complex, as assessed by ELISA. Subsequently, the substrate tetramethylbenzidine (TMB) was used to develop the color. The absorbance (OD value) of the colored complex was measured at 450 nm, and the human mCRP concentration in the sample was calculated by a standard curve [31].

Detection of brain natriuretic peptide (BNP) by ELISA

BNP was detected by a double-antibody sandwich immunoenzyme method, using Unicel-TM-DXI800 (Beckman, CA, USA) full-automated immunoassay instrument. The ELISA was performed, as described previously [32].

Detection of circulating blood serum creatinine (Scr), estimated glomerular filtration rate (eGFR), and CRP

Scr was determined by a rating method using a commercial kit (Beckman, CA, USA) [33]. The level of CRP was determined by immunoturbidimetry using a commercially available kit (Goldsite, Shenzhen, China) [34]. According to Scr, age, and gender, the eGFR of the patient was calculated. The modification of diet in renal disease (MDRD) formula was adapted by Chinese experts. The female eGFR calculation formula was as follows: eGFR (mL/min/1.73 m2) = 186 × Scr − 1.154 × (age) − 0.203 × 0.742 × 1.227 and male eGFR calculation formula eGFR (mL/min/1.73 m2) = 186 × Scr − 1.154 × (age) − 0.203 × 1.227 [35].

Detection of ejection fraction (EF%), left ventricular end-diastolic volume (LVEDV), left ventricular end-systolic volume (LVESV), and left ventricular mass index (LVMI)

The values of EF%, LVEDV, LVESV, left ventricular septal end-diastolic thickness (IVSTD), and left ventricular posterior wall diastolic thickness (LVPWTD) were obtained by echocardiography. Then, the value of left ventricular myocardial mass (LVM) was calculated based on LVIDD, IVSTD, and LVPWTD. Body surface area (BSA) was calculated through weight (W) and height (H).

LVM (g) = 1.04 {[(LVIDD + PWTD + IVSTD)3 − (LVIDD)3] − 13.6}.

BSA (m2) = (W0.425 × H0.725) × 0.007184 [36].

Statistical analysis

All data were statistically analyzed using SPSS19.0. All grouped data were tested for normality before further analysis. The two groups of enumeration data that conformed to the normal distribution were compared using an independent t test. If any of the groups did not match, a non-parametric rank-sum test was used. If the data fulfilled the normal distribution, it would be expressed as means (x̅) ± standard deviation (SD). The Shapiro-Wilk test, median, and interquartile range (IQR) were used to check the degree of dispersion of the data. The correlation between the two groups of data was analyzed; if both groups met the normal distribution, Pearson’s rank correlation was used, and if any group did not meet the normal distribution, Spearman’s rank correlation was utilized. Cross-tabulation χ2 test was used for gender comparison. P < 0.05 indicated statistical significance.

Results

General data from AAV patients and healthy volunteers (normal control)

All the 37 AAV patients, including 17 (45.9%) females and 20 (54.1%) males, harbored MPO-ANCA. Among the 4 AAV patients with STEMI complications, 2 (50.0%) were males and 2 (50.0%) were females, with average age 59.5 ± 14.3 years, while that of the 37 AAV patients was 58.5 ± 10.6 years. The healthy volunteers consisted of 10 (50.0%) males and 10 (50.0%) females, aged 55.3 ± 10.7 years. The comparison of age and sex between 37 AAV patient groups and healthy volunteers (n = 20, P = 0.266; n = 20, P = 0.788) did not show significant differences. Next, we used the same method to compare the age and gender of 4 patients with STEMI complications and 33 patients without STEMI complications in the AAV group. Therefore, both the AAV patient group and the normal control, as well as patients with and without STEMI complications in the AAV group, showed no significant difference in the age and sex (P > 0.05). Also, the relevant biochemical indicators were analyzed in AAV patients (Table 1).

Table 1 General data (biochemical indexes) in AAV

Correlation between mCRP and cardiovascular disease risk factors

The risk factors of cardiovascular diseases include status, age, gender, tobacco, alcohol, hypertension, diabetes, and obesity. Moreover, the correlation between clinical indicators of cardiovascular risk factors (lipid- and inflammation-related factors) and mCRP were compared. We found that mCRP was negatively correlated with total cholesterol (TG) and CRP (r = − 0.592, P < 0.001; r = − 0.340, P = 0.040, respectively), (Table 2).

Table 2 Comparison of the relationship between clinical indicators of cardiovascular risk factors in AAV

Plasma levels of mCRP were higher in AAV patients than in normal controls

The plasma level of mCRP in AAV patients was 244.12 (226.12, 331.725) μg/mL and that in the normal control was 170.0 (135.7, 199.3) μg/mL. Next, we compared the plasma levels of mCRP in AAV patients and healthy volunteers (P < 0.001) and found significant differences (Fig. 1).

Fig. 1
figure 1

Comparison of the plasma levels of mCRP in AAV patients (n = 37) and healthy volunteers (n = 20, P < 0.001)

Plasma levels of mCRP were highest in patients with than in those without STEMI complications

The AAV patients were divided into groups with (n = 4) and without STEMI complications (n = 33) groups (Fig. 2a). The plasma concentration of mCRP in patients with and without STEMI complications in AAV group was 581.4 (508.7, 647.3) and 240.8 (219.2, 292.1) μg/mL, respectively, while that in normal controls was 170.0 (135.7, 199.3) μg/mL (Fig. 2b). We also compared the BVAS of patients with and without STEMI complications (Fig. 2c).

Fig. 2
figure 2

Plasma levels of mCRP were highest in STEMI complications in AAV than without STEMI complications in AAV and normal controls. a Thirty-seven AAV patients were divided into two groups: with STEMI complications (n = 4) and without STEMI complications (n = 33). b Plasma levels of mCRP in STEMI complications were highest in the three groups (with STEMI complications in AAV, without STEMI complications, normal). c Comparison of BVAS in groups with and without STEMI complications

Plasma levels of mCRP were positively correlated with BVAS and Scr and negatively correlated with eGFR

BVAS in 37 AAV patients and normal controls was 29.9 ± 9.1 and 0, while Scr was 291.0 (161.5, 434.5), respectively. The eGFR was calculated based on gender, age, and Scr levels. The eGFR was 17.7 (10.1, 41.0) mL/min/1.73 m2 in AAV patients and 170.0 (135.7, 199.3) mL/min/1.73 m2 in normal controls. Thus, we analyzed the correlation between mCRP and BVAS (r = 0.404, P = 0.013, Fig. 3a) and mCRP and eGFR (r = − 0.341, P = 0.039, Fig. 3b).

Fig. 3
figure 3

Plasma levels of mCRP were compared with BVAS and eGFR in AAV. a Correlation between mCRP and BVAS. The plasma levels of mCRP were positively correlated with BVAS (r = 0.404, P = 0.013). BVAS, Birmingham vasculitis activity score. b Correlation between mCRP and eGFR. The plasma levels of mCRP were negatively correlated with eGFR (r = − 0.341, P = 0.039). eGFR, estimated glomerular filtration rate

Plasma levels of mCRP were correlated with BNP, EF%, LVEDV, LVESV, LVMI, and STEMI complications’ Gensini score, but not CRP

The Gensini score of patients with STEMI complications in AAV was 72.0 ± 15.1. The BNP level in circulating blood was 155.7 (104.4, 204.3) pg/mL, while the EF (%) levels were 63.0 ± 8.8%, LVEDV was 124.2 ± 25.3 mL, LVESV was 44.2 ± 10.7 mL, LVMI was 86.5 (50.6, 112.6), and the Gensini score of four STEMI complications was 72.0 ± 15.1 in AAV patients.

Furthermore, we speculated that the plasma levels of mCRP might be related to the patient’s heart disease, and hence, the correlation between mCRP and BNP, EF%, LVEDV, LVESV, LVMI, and Gensini score of STEMI complications was analyzed (r = 0.349, P = 0.034, Fig. 4a; r = − 0.362, P = 0.027, Fig. 4b; r = 0.375, P = 0.022, Fig. 4c, r = 0.334, P = 0.043, Fig. 4d; r = 0.736, P < 0.001, Fig. 4e; r = 0.997, P = 0.023, Fig. 4f, respectively), while CRP was not correlated (Table 3).

Fig. 4
figure 4

Plasma levels of mCRP were correlated with BNP, EF%, LVEDV, LVESV, LVMI, and Gensini score of STEMI complications in the AAV group. a Correlation between mCRP and BNP. The plasma levels of mCRP were positively correlated with BNP (r = 0.349, P = 0.034). BNP, brain natriuretic peptide. b Correlation of mCRP and EF%. The plasma levels of mCRP were negatively correlated with EF% (r = − 0.362, P = 0.027). EF%, ejection fraction. c Correlation between mCRP and LVEDV. The plasma levels of mCRP were positively correlated with LVEVD (r = 0.375, P = 0.022). LVEDV, left ventricular end-diastolic volume. d Correlation between mCRP and LVESV. The plasma levels of mCRP were positively correlated with LVESV (r = 0.334, P = 0.043). LVESV, left ventricular end-systolic volume. e Correlation between mCRP and LVMI. The plasma levels of mCRP were positively correlated with LVMI (r = 0.736, P < 0.001). LVMI, left ventricular mass index. f Correlation between mCRP and Gensini score. The plasma levels of mCRP were positively correlated with Gensini score of STEMI complications in the AAV group (r = 0.977, P = 0.02). STEMI, ST-segment elevation myocardial infarction

Table 3 Comparison of mCRP and CRP with BNP, EF%, LVEDV, LVESV, LVMI, and Gensini score with STEMI complications

Discussion

Accumulating evidence has shown that CRP is pathogenic in atherosclerosis, acute myocardial infarction, cerebral infarction, and AAV disease [37]. CRP is one of the biomarkers for assessing AAV disease activity. The serum CRP concentration increased during the AAV active phase and decreased rapidly with the remission of the disease [13, 38]. mCRP can bind with complement factor H (CFH) and is more effective than pCRP in inhibiting alternative complement [39] and plays a critical role in the development of AAV [40, 41]. Xu et al. showed that MPO inhibits the binding between CFH and mCRP, thereby inhibiting the regulatory activation of alternative complements. PR3 does not bind to either pCRP or mCRP, while MPO can bind to mCRP, which blocks the binding between mCRP and CFH [41]. Therefore, the mechanism of mCRP and AAV disease is indistinguishable.

CRP can be decomposed into mCRP at high temperature, urea, or acidic microenvironment [42, 43]. mCRP activates platelets [20], monocytes [44], and endothelial cells [45]. It is a key substance that promotes inflammatory response [21, 46]. The mechanism underlying the inflammatory response of endothelial cells is caused by the interaction of mCRP with neutrophils, macrophages, and platelets [46, 47]. Furthermore, mCRP binds to CFH and directs it to the damaged part of the cell [48, 49], inactivates C3b, and limits the progression of inflammation [39]. Therefore, we speculated that the role of mCRP in the pathogenesis of AAV disease might be related to CFH.

In patients with lupus, the level of anti-mCRP autoantibodies is related to the degree of renal interstitial lesions [50], and mCRP has been reported as an autoantigen in interstitial nephritis-associated uveitis (TINU) syndrome [51]. In AAV patients, anti-mCRP antibodies may be the cause of severe TI lesions [25].

Serum CRP levels increase due to acute infection, trauma, and inflammation [52]. Thus, CRP is commonly used for the risk stratification of cardiovascular disease [53, 54], as well as a marker of generalized atherosclerosis [54, 55]. CRP levels are significantly increased in cardiovascular patients without any symptoms, obvious cardiovascular disease, unstable angina pectoris, myocardial infarction, and other diseases [22, 56]. Also, it is directly involved in the response process of acute myocardial infarction [18]. Thielle et al. demonstrated that mCRP stimulates inflammation, while pCRP has no proinflammatory effect [45]. In addition, Diehl et al. detected mCRP deposition in human atherosclerotic plaque vessels [57]. Similarly, studies on cardiovascular disease have found that mCRP is present in endothelial cells cultured in vitro and can promote inflammatory responses [20]. In the current study, 37 patients with AAV were collected and 4 patients with STEMI complications were found.

mCRP has lectin-like properties and can bind galactose-containing residues [12, 58, 59]. ANCA-induced NETs activate the platelets and then promote mCRP formation on these platelets. Subsequently, the newly generated mCRP can further enhance thrombosis and inflammatory response during platelet activation. mCRP may be a potential link between thrombosis and inflammation in AAV. Therefore, we suspected that the occurrence of STEMI complications in patients was related to mCRP [12]. AAV exhibits various echocardiographic characteristics, such as decreased systolic function, increased diastolic dysfunction, and pulmonary hypertension at the time of diagnosis [60]. The current study focused on cardiovascular disease in patients with AAV, which reflected the clinical examination of cardiovascular function by echocardiography. EF%, LVEDV, and LVESV are indicators of heart function. LVMI is associated with the mortality of patients with chronic kidney disease (CKD) and is a high-risk factor of heart failure [61]. Therefore, we analyzed the correlation between EF%, LVEDV, LVESV, LVMI, and mCRP. In this study, we proved that circulating blood levels of CRP were significantly higher in AAV patients than in healthy volunteers. Also, mCRP levels, but not CRP levels, were significantly increased in patients with STEMI complications. Next, we analyzed the correlation between mCRP levels and the Gensini score of STEMI complications and that between mCRP levels, BNP, and EF% in patients with AAV and found that the level of mCRP was positively correlated with the Gensini score and BNP and negatively correlated with EF%.

Nevertheless, the present study had some limitations. First, it was not a longitudinal study because while collecting cardiovascular clinical indicators, a large amount of experimental data were collected first, followed by comparison analysis. Finally, it was found that the mCRP level was related to the different parameters mentioned in this study. However, AAV patients have low morbidity but high mortality [12]. Most AAV patients visit our department for treatment due to abnormal renal function. The patients we collected in this study were MPO-ANCA because it is a predominant condition in Chinese AAV patients [14, 17]. Therefore, follow-up studies are imperative.

Conclusions

The level of mCRP is related not only to AAV disease but also to the occurrence of cardiovascular disease in patients with AAV. Therefore, mCRP can be used as a plasma biomarker of cardiovascular diseases in AAV.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

CRP:

C-reactive protein

pCRP:

C-reactive protein pentamer

mCRP:

C-reactive protein monomer

ANCA:

Anti-neutrophil cytoplasmic antibody

AAV:

Anti-neutrophil cytoplasmic antibody-associated vasculitis

PR3:

Protease 3

MPO:

Myeloperoxidase

Scr:

Serum creatinine

Hb:

Hemoglobin

PTH:

Parathyroid hormone

CTnT:

Cardiac troponin T

CK:

Creatine kinase

CK-MB:

Creatine kinase isoenzyme

HDL-C:

High-density lipoprotein

Tch:

Total serum cholesterol

TG:

Triglyceride

LDL-C:

Low-density lipoprotein

Fib:

Fibrinogen

ELISA:

Enzyme-linked immunosorbent assay

TMB:

The substrate tetramethylbenzidine

BNP:

Brain natriuretic peptide

STEMI:

ST-segment elevation myocardial infarction

EF%:

Ejection fraction

LVEDV:

Left ventricular end-diastolic volume

LVESV:

Left ventricular end-systolic volume

LVMI:

Left ventricular mass index

AMI:

Acute myocardial infarction

LVIDD:

Left ventricular end-diastolic diameter

IVSTD:

Left ventricular septal end-diastolic thickness

LVPWTD:

Left ventricular posterior wall diastolic thickness

LVM:

Left ventricular myocardial mass

GEnC:

Glomerular endothelial cells

TI:

Tubulointerstitial

ATIN:

Acute tubulointerstitial

eGFR:

Glomerular filtration rate

BVAS:

Birmingham Vasculitis Activity Score

References

  1. Jennette JC, Falk RJ. Small-vessel vasculitis. N Engl J Med. 1997;337(21):1512–23.

    Article  CAS  PubMed  Google Scholar 

  2. Jennette JC, Falk R, Bacon P, et al. 2012 revised international Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheumatism. 2013;65(1):1–11.

    Article  CAS  PubMed  Google Scholar 

  3. Segelmark M, Wieslander J. IgG subclasses of antineutrophil cytoplasm autoantibodies (ANCA). Nephrol Dialysis Transplantation. 1993;8(8):696–702.

    Article  CAS  Google Scholar 

  4. Hao J, Huang Y-M, Zhao M-H, Chen M. The interaction between C5a and sphingosine-1-phosphate in neutrophils for antineutrophil cytoplasmic antibody mediated activation. Arthritis Res Therapy. 2014;16(4):R142.

    Article  CAS  Google Scholar 

  5. Chen M, Yu F, Zhang Y, Zou W-Z, Zhao M-H, Wang H-Y. Characteristics of Chinese patients with Wegener’s granulomatosis with anti-myeloperoxidase autoantibodies. Kidney Int. 2005;68(5):2225–9.

    Article  CAS  PubMed  Google Scholar 

  6. Li ZY, Chang DY, Zhao MH, Chen M. Predictors of treatment resistance and relapse in antineutrophil cytoplasmic antibody–associated vasculitis: a study of 439 cases in a single Chinese center. Arthritis Rheumatol. 2014;66(7):1920–6.

    Article  CAS  PubMed  Google Scholar 

  7. Sun X-J, Chen M, Zhao M-H. Thrombin contributes to anti-myeloperoxidase antibody positive IgG-mediated glomerular endothelial cells activation through SphK1-S1P-S1PR3 signaling. Front Immunol. 2019;10:237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Nagao T, Matsumura M, Mabuchi A, et al. Up-regulation of adhesion molecule expression in glomerular endothelial cells by anti-myeloperoxidase antibody. Nephrol Dialysis Transplantation. 2007;22(1):77–87.

    Article  CAS  Google Scholar 

  9. Nagao T, Suzuki K, Utsunomiya K, et al. Direct activation of glomerular endothelial cells by anti-moesin activity of anti-myeloperoxidase antibody. Nephrol Dialysis Transplantation. 2011;26(9):2752–60.

    Article  CAS  Google Scholar 

  10. Ten Oever J, Netea MG, Kullberg B-J. Utility of immune response-derived biomarkers in the differential diagnosis of inflammatory disorders. J Infect. 2016;72(1):1–18.

    Article  PubMed  Google Scholar 

  11. Coster D, Wasserman A, Fisher E, et al. Using the kinetics of C-reactive protein response to improve the differential diagnosis between acutebacterial and viral infections. Infection. 2020;48:241–8.

  12. Xu P-C, Lin S, Yang X-W, et al. C-reactive protein enhances activation of coagulation system and inflammatory response through dissociating into monomeric form in antineutrophil cytoplasmic antibody-associated vasculitis. BMC Immunology. 2015;16(1):10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Kälsch A-I, Csernok E, Münch D, et al. Use of highly sensitive C-reactive protein for followup of Wegener’s granulomatosis. J Rheumatol. 2010;37(11):2319–25.

    Article  PubMed  CAS  Google Scholar 

  14. Shrive AK, Cheetham GM, Holden D, et al. Three dimensional structure of human C-reactive protein. Nat Struct Biol. 1996;3(4):346–54.

    Article  CAS  PubMed  Google Scholar 

  15. Yeh ETH. A new perspective on the biology of C-reactive protein. Circ Res. 2005;97:609–11.

  16. Nakajima T, Yamazaki K. Periodontal disease and risk of atherosclerotic coronary heart disease. Odontology. 2009;97(2):84–91.

    Article  PubMed  Google Scholar 

  17. Papathanasiou AI, Pappas KD, Korantzopoulos P, et al. An epidemiologic study of acute coronary syndromes in northwestern Greece. Angiology. 2004;55(2):187–94.

    Article  CAS  PubMed  Google Scholar 

  18. Ridker PM. C-reactive protein and the prediction of cardiovascular events among those at intermediate risk: moving an inflammatory hypothesis toward consensus. J Am Coll Cardiol. 2007;49(21):2129–38.

    Article  CAS  PubMed  Google Scholar 

  19. Khreiss T, József L, Potempa LA, Filep JG. Loss of pentameric symmetry in C-reactive protein induces interleukin-8 secretion through peroxynitrite signaling in human neutrophils. Circ Res. 2005;97(7):690–7.

    Article  CAS  PubMed  Google Scholar 

  20. Eisenhardt SU, Habersberger J, Murphy A, et al. Dissociation of pentameric to monomeric C-reactive protein on activated platelets localizes inflammation to atherosclerotic plaques. Circ Res. 2009;105(2):128–37.

    Article  CAS  PubMed  Google Scholar 

  21. Wu Y, Potempa LA, El Kebir D, Filep JG. C-reactive protein and inflammation: conformational changes affect function. Biol Chem. 2015;396(11):1181–97.

    Article  CAS  PubMed  Google Scholar 

  22. Boncler M, Wu Y, Watala C. The multiple faces of C-reactive protein—physiological and pathophysiological implications in cardiovascular disease. Molecules. 2019;24(11):2062.

    Article  CAS  PubMed Central  Google Scholar 

  23. Chen S-F, Wang F-M, Li Z-Y, Yu F, Chen M, Zhao M-H. The functional activities of complement factor H are impaired in patients with ANCA-positive vasculitis. Clin Immunol. 2017;175:41–50.

    Article  CAS  PubMed  Google Scholar 

  24. Zonozi R, Niles JL, Cortazar FB. Renal involvement in antineutrophil cytoplasmic antibody–associated vasculitis. Rheum Dis Clin. 2018;44(4):525–43.

    Article  Google Scholar 

  25. Lin Z-S, Liu X-L, Cui Z, et al. Acute tubulointerstitial nephritis with germinal centers in antineutrophil cytoplasmic antibody-associated vasculitis: a case report and literature review. Medicine. 2019;98(48):e18178.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Wang H-W, Sui S-F. Dissociation and subunit rearrangement of membrane-bound human C-reactive proteins. Biochem Biophys Res Commun. 2001;288(1):75–9.

    Article  CAS  PubMed  Google Scholar 

  27. Molins B, Peña E, de la Torre R, Badimon L. Monomeric C-reactive protein is prothrombotic and dissociates from circulating pentameric C-reactive protein on adhered activated platelets under flow. Cardiovasc Res. 2011;92(2):328–37.

    Article  CAS  PubMed  Google Scholar 

  28. Wang J, Tang B, Liu X, et al. Increased monomeric CRP levels in acute myocardial infarction: a possible new and specific biomarker for diagnosis and severity assessment of disease. Atherosclerosis. 2015;239(2):343–9.

    Article  CAS  PubMed  Google Scholar 

  29. Schreiber A, Xiao H, Jennette JC, Schneider W, Luft FC, Kettritz R. C5a receptor mediates neutrophil activation and ANCA-induced glomerulonephritis. J Am Soc Nephrol. 2009;20(2):289–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Luqmani R, Bacon P, Moots R, et al. Birmingham vasculitis activity score (BVAS) Dim system necrotizinig vasculitis. QJM Int J Med. 1994;87(11):671–8.

    CAS  Google Scholar 

  31. Tan Y, Yu F, Qu Z, et al. Modified C-reactive protein might be a target autoantigen of TINU syndrome. Clin J Am Soc Nephrol. 2011;6(1):93–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Mayo D, Colletti JE, Kuo D. Brain natriuretic peptide (BNP) testing in the emergency department. J Emerg Med. 2006;31(2):201–10.

    Article  PubMed  Google Scholar 

  33. World Health Organization. Use of anticoagulants in diagnostic laboratory investigations. World Health Organization. 2002. p. 1–62.

  34. Rosen M. C-reactive protein: a marker of infection, inflammation, tissue damage and malignancy. Diag Clin Testing. 1990;28:18–22.

    Google Scholar 

  35. Ma Y-C, Zuo L, Chen J-H, et al. Modified glomerular filtration rate estimating equation for Chinese patients with chronic kidney disease. J Am Soc Nephrol. 2006;17(10):2937–44.

    Article  PubMed  Google Scholar 

  36. Mostfa SA. Impact of obesity and surgical weight reduction on cardiac remodeling. Indian Heart J. 2018;70(Suppl3):S224–8.

  37. Gill R, Kemp JA, Sabin C, Pepys MB. Human C-reactive protein increases cerebral infarct size after middle cerebral artery occlusion in adult rats. J Cereb Blood Flow Metab. 2004;24(11):1214–8.

    Article  CAS  PubMed  Google Scholar 

  38. Xu P-C, Hao J, Yang X-W, D-y C, Chen M, M-h Z. C-reactive protein enhances the respiratory burst of neutrophils-induced by antineutrophil cytoplasmic antibody. Mol Immunol. 2012;52(3–4):148–54.

    Article  CAS  PubMed  Google Scholar 

  39. Mihlan M, Blom AM, Kupreishvili K, et al. Monomeric C-reactive protein modulates classic complement activation on necrotic cells. FASEB J. 2011;25(12):4198–210.

    Article  CAS  PubMed  Google Scholar 

  40. Hao J, Meng L-Q, Xu P-C, et al. p38MAPK, ERK and PI3K signaling pathways are involved in C5a-primed neutrophils for ANCA-mediated activation. PLoS ONE. 2012;7:e38317.

  41. Xu P-c, Z-y L, Yang X-w, M-h Z, Chen M. Myeloperoxidase influences the complement regulatory function of modified C-reactive protein. Innate Immunity. 2014;20(4):440–8.

    Article  PubMed  CAS  Google Scholar 

  42. 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(11):1165–75.

    Article  CAS  PubMed  Google Scholar 

  43. Kresl JJ, Potempa LA, Anderson BE. Conversion of native oligomeric to a modified monomeric form of human C-reactive protein. Int J Biochem Cell Biol. 1998;30(12):1415–26.

    Article  CAS  PubMed  Google Scholar 

  44. Braig D, Nero TL, Koch H-G, et al. Transitional changes in the CRP structure lead to the exposure of proinflammatory binding sites. Nat Commun. 2017;8(1):1–19.

    Article  CAS  Google Scholar 

  45. Thiele JR, Habersberger J, Braig D, et al. Dissociation of pentameric to monomeric C-reactive protein localizes and aggravates inflammation: in vivo proof of a powerful proinflammatory mechanism and a new anti-inflammatory strategy. Circulation. 2014;130(1):35–50.

    Article  CAS  PubMed  Google Scholar 

  46. Caprio V, Badimon L, Di Napoli M, et al. pCRP-mCRP dissociation mechanisms as potential targets for the development of small-molecule anti-inflammatory chemotherapeutics. Front Immunol. 2018;9:1089.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Eisenhardt SU, Thiele JR, Bannasch H, Stark GB, Peter K. C-reactive protein: how conformational changes influence inflammatory properties. Cell Cycle. 2009;8(23):3885–92.

    Article  CAS  PubMed  Google Scholar 

  48. Mihlan M, Stippa S, Józsi M, Zipfel P. Monomeric CRP contributes to complement control in fluid phase and on cellular surfaces and increases phagocytosis by recruiting factor H. Cell Death Differentiation. 2009;16(12):1630–40.

    Article  CAS  PubMed  Google Scholar 

  49. O'Flynn J, van der Pol P, Dixon KO, Prohászka Z, Daha MR, van Kooten C. Monomeric C-reactive protein inhibits renal cell-directed complement activation mediated by properdin. Am J Physiol-Renal Physiol. 2016;310(11):F1308–16.

    Article  CAS  PubMed  Google Scholar 

  50. Tan Y, Yu F, Yang H, Chen M, Fang Q, Zhao M-H. Autoantibodies against monomeric C-reactive protein in sera from patients with lupus nephritis are associated with disease activity and renal tubulointerstitial lesions. Hum Immunol 2008;69(12):0–844.

  51. Tan Y, Yu F, Qu Z, Su T, Zhao MH. Modified C-reactive protein might be a target autoantigen of TINU syndrome. Clin J Am Soc Nephrol Cjasn. 2011;6(1):93–100.

    Article  CAS  PubMed  Google Scholar 

  52. Pepys MB, Hirschfield GM. C-reactive protein: a critical update. J Clin Invest. 2003;111(12):1805–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ridker PM. A test in context: high-sensitivity C-reactive protein. J Am Coll Cardiol. 2016;67(6):712–23.

    Article  PubMed  Google Scholar 

  54. Li Y, Zhong X, Cheng G, et al. Hs-CRP and all-cause, cardiovascular, and cancer mortality risk: a meta-analysis. Atherosclerosis. 2017;259:75–82.

    Article  CAS  PubMed  Google Scholar 

  55. Elias-Smale SE, Kardys I, Oudkerk M, Hofman A, Witteman JC. C-reactive protein is related to extent and progression of coronary and extra-coronary atherosclerosis; results from the Rotterdam study. Atherosclerosis. 2007;195(2):e195–202.

    Article  CAS  PubMed  Google Scholar 

  56. Ridker PM. Clinical application of C-reactive protein for cardiovascular disease detection and prevention. Circulation. 2003;107(3):363–9.

    Article  PubMed  Google Scholar 

  57. Diehl EE, Haines GK, Radosevich JA, Potempa LA. Immunohistochemical localization of modified C-reactive protein antigen in normal vascular tissue. Am J Med Sci. 2000;319(2):79–83.

    Article  CAS  PubMed  Google Scholar 

  58. Lee RT, Lee YC. Carbohydrate ligands of human C-reactive protein: binding of neoglycoproteins containing galactose-6-phosphate and galactose-terminated disaccharide. Glycoconj J. 2006;23(5–6):317–27.

    Article  CAS  PubMed  Google Scholar 

  59. Köttgen E, Hell B, Kage A, Tauber R. Lectin specificity and binding characteristics of human C-reactive protein. J Immunol. 1992;149(2):445–53.

    PubMed  Google Scholar 

  60. Ahn SSPE, Jung SM, Song JJ, Park YB, Lee SW. Echocardiographic features in patients with ANCA-associated vasculitis within 3 months before and after diagnosis. Clin Rheumatol. 2017;36(12):2751–9.

    Article  PubMed  Google Scholar 

  61. Tripepi G, D'Arrigo G, Mallamaci F, et al. Prognostic values of left ventricular mass index in chronic kidney disease patients. Nephrol Dial Transplant. 2019;1–8.

Download references

Acknowledgements

The Affiliated Hospital of Inner Mongolia Medical University. Funding details are included above.

Funding

The two funds are the China Natural Science Foundation (NO81460145) and Inner Mongolia Natural Science Foundation Project Number (2018MS08105).

Author information

Authors and Affiliations

Authors

Contributions

Kai-Li Wu and Qing-Hui Liang conducted experiments, analyzed the data, and drafted the manuscript. Na Ding and Bo-Wei Li conducted experiments. Bin-Tao Huang participated in revising the manuscript. Jian Hao conceived the study, participated in revising the manuscript, and provided final approval to submit the version of the document. All authors read, and the report was approved.

Corresponding author

Correspondence to Jian Hao.

Ethics declarations

Ethics approval and consent to participate

All patients provided their informed written consent. The study was approved by the Ethics Committee of the Helsinki Declaration of Inner Mongolia Medical University.

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, KL., Liang, QH., Huang, BT. et al. The plasma level of mCRP is linked to cardiovascular disease in antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Res Ther 22, 228 (2020). https://doi.org/10.1186/s13075-020-02321-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13075-020-02321-w

Keywords