This work is licensed under http://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
1. Introduction
Cardiovascular disease (CVD) remains the leading cause of death and disability in developed countries, and it accounted for the death of approximately 17 million people worldwide in 2013 [1]. Cardiometabolic disorders, including obesity, type 2 diabetes (T2D), insulin resistance (IR), hypertension, and dyslipidemia, are common risk factors for CVD [2]. Moreover, the steady increase in the occurrence of cardiometabolic disorders compels the search for the involved pathophysiology and more effective preventive strategies.
Cytokeratin 18 (CK18) is involved in cell death pathway, and it is an important cell death marker. Cell death (apoptosis, necrosis, autophagy, etc.) can help to get rid of damaged cells and protect cell integrity when a cell responds to the aberrant cellular stresses like endoplasmic reticulum (ER) stress and oxidative stress, which have been the hallmarks of cardiometabolic disorders [3–5]. Massive beta cell death [6, 7] and cell death-induced decrease in the delta cell volume and number cause T2D [8]. Endothelial apoptosis can lead to endothelial dysfunction and development of hypertension [9]. Apoptosis in response to glucose excess was observed in cardiometabolic diseases [10], and decreased myocardial levels of apoptosis were found to significantly reduce obesity-associated cardiomyopathy [11]. The cellular content of the soluble fraction of CK18 has been shown to be released into the extracellular space during cell death both in vitro and in vivo [12]. A recent study indicated that CK18 played a role in apoptosis, which may be mediated via a feedback regulation loop and may involve regulation of transcription and alternative splicing of a number of genes in NF-κB pathway and certain apoptosis pathways [13]. In addition to caspase-induced apoptosis, other forms of cell death can lead to the release of CK18. For example, CK18 is released during pyroptosis, nonlethal processes of apoptosis independent of caspase activation, or secondary lysis following programmed cell death [14, 15].
However, limited studies have focused on the role of serum CK18 in nonalcoholic fatty liver disease (NAFLD) and cardiometabolic disorders. Serum CK18 was found to be closely associated with hepatic inflammation in patients with NAFLD [16–18]. Chang et al. found that elevated levels of CK18 (assessed by M30 and M65 assays) in patients with T2D had a significant positive association with fasting plasma glucose levels, and it was independent of NAFLD status [19]. Tan et al. demonstrated the correlation between elevated levels of CK18 M30 and IR in patients with polycystic ovary syndrome [20]. Civera et al. showed that increased insulin resistance induces an increase in the levels of CK18 M30 in patients with severe obesity [21]. M30 was highly correlated to diastolic dysfunction indices in adolescents with obesity [22]. However, the role of CK18 in hypertension or dyslipidemia in adults has not been studied yet. Furthermore, the association between serum CK18 M65ED levels and cardiometabolic disorders has also not been investigated. In the present study, we therefore investigated this association with a focus on cardiometabolic disorders including overweight or central obesity, diabetes, dyslipidemia, and hypertension.
2. Methods
2.1. Study Participants
Subjects aged 18 years or older were enrolled from the local communities in Shanghai. All subjects underwent comprehensive physical examinations, routine biochemical analyses of blood, 75 g oral glucose tolerance test, hepatitis B surface antigen test, hepatitis C virus antibody test, and B-ultrasonography. The histories of present and past illnesses, medical therapy, and alcohol consumption were obtained through standard questionnaires. Subjects with the following conditions were excluded from this study: biliary obstructive disease, acute or chronic viral hepatitis, drug-induced liver disease, autoimmune hepatitis, Wilson’s disease, known hyperthyroidism or hypothyroidism, presence of cancer, ongoing treatment with systemic corticosteroids, pregnancy, and current drinkers and exdrinkers. A total of 588 subjects were enrolled in the study. Of them, 310 had cardiometabolic disorders, and the control group consisted of 278 age-matched participants without conditions of obesity, central obesity, diabetes, dyslipidemia, or hypertension. The study was approved by the Ethics Committee of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital and was conducted in accordance the principles of the declaration of Helsinki. All data were handled without compromising the privacy of the study participants. All participants provided written informed consent prior to enrollment.
2.2. Anthropometric and Laboratory Measurements
Blood pressure, weight, height, waist circumference (WC), and biomedical indices were measured according to our previous standardized protocols [23].
Cytokeratin 18 is a major intermediate filament protein of the cytoskeleton that is expressed in simple epithelial cells, hepatocytes, cholangiocytes, pancreas, and colon [16, 24, 25]. During apoptosis, full-length CK18 can be cleaved at two sites into three fragments [26]. The M30 assay can detect the caspase-cleaved fragments of CK18 by using the monoclonal antibody, M30, showing the extent of apoptosis [24]. The CK18 M65 assay uses the capture antibody M6 and the detection antibody M5 that are directed against two different epitopes of CK18 and can recognize both full-length CK18 and cleaved fragments of CK18, regardless of whether they are cleaved by caspases or not [18]. The M65ED assay uses the M6 antibody for detection and the M5 as a capture antibody, which can achieve improved binding specificity and lower signals in healthy controls [24, 27].
2.3. Diagnostic Criteria
Cardiometabolic disorders were identified by the presence of at least one of the components including overweight or central obesity, diabetes, dyslipidemia, and hypertension [28]. An overweight condition was defined as a BMI between 25.0 and 29.9 kg/m2, and obesity was defined as a
2.4. Statistical Analysis
All analyses were performed using SPSS 25.0 (Chicago, IL, USA). Normally distributed data were expressed as
3. Results
3.1. Clinical Characteristics of the Study Subjects
A total of 588 subjects were enrolled in this study (median age: 40.00 (33.00–49.75) years) and included 340 men and 248 women. Table 1 lists the clinical and laboratory characteristics of the subjects. Subjects with cardiometabolic disorders (
Table 1
Characteristics of subjects with and without cardiometabolic disorders.
Variables | Total | Without cardiometabolic disorders | With cardiometabolic disorders | |
588 (340/248) | 278 (109/169) | 310 (231/79) | <0.001 | |
Age (years) | 40.0 (33.0–49.8) | 40.0 (32.0–47.0) | 40.0 (34.0–52.0) | 0.101 |
Body mass index (kg/m2)§ | 24.8 (22.1–27.9) | 22.2 (20.4–23.6) | 27.7 (25.4–30.4) | <0.001 |
Waist circumference (cm)§ | 86.5 (72.0–97.5) | 71.0 (66.0–77.0) | 96.7 (90.6–103.0) | <0.001 |
Systolic blood pressure (mmHg)§ | 118 (110–124) | 114 (109–120) | 120 (112–128) | <0.001 |
Diastolic blood pressure (mmHg)§ | 80 (72–82) | 76 (70–80) | 80 (74–85) | <0.001 |
Fasting plasma glucose (mmol/L)§ | 5.08 (4.72–5.58) | 4.96 (4.63–5.27) | 5.28 (4.84–5.98) | 0.251 |
2-hour plasma glucose (mmol/L)§ | 6.69 (5.61–8.12) | 5.69 (4.93–6.58) | 7.71 (6.70–9.82) | <0.001 |
HbA1c (%)§ | 5.6 (5.4–5.9) | 5.5 (5.3–5.7) | 5.7 (5.4–6.3) | 0.005 |
Fasting insulin (μU/mL)§ | 10.10 (5.28–13.10) | 5.51 (3.93–8.51) | 12.65 (10.99–16.73) | <0.001 |
HOMA-IR§ | 2.26 (1.18–3.23) | 1.21 (0.85–1.89) | 3.10 (2.49–4.39) | <0.001 |
HOMA-B§ | 30.57 (16.92–49.4) | 21.81 (13.88–34.93) | 41.04 (28.37–64.9) | 0.001 |
Alanine aminotransferase (IU/L)§ | 21 (13–33) | 14 (10.75–19.00) | 29 (20–43) | <0.001 |
Aspartate transaminase (IU/L)§ | 20 (17–25) | 18 (16–21) | 22 (19–28) | <0.001 |
Gamma-glutamyl-transferase (IU/L)§ | 24 (15–40) | 16 (13–24) | 37 (24–49) | <0.001 |
Triglycerides (mmol/L)§ | 1.31 (0.89–1.86) | 1.01 (0.72–1.30) | 1.74 (1.28–2.38) | <0.001 |
Total cholesterol (mmol/L) | <0.001 | |||
HDL cholesterol (mmol/L)§ | 1.21 (1.05–1.4) | 1.37 (1.23–1.57) | 1.08 (0.95–1.21) | <0.001 |
LDL cholesterol (mmol/L) | <0.001 | |||
Cytokeratin 18 M65ED (U/L)§ | 137.54 (76.51–254.39) | 83.85 (52.80–153.75) | 197.36 (121.13–354.50) | <0.001 |
Data are
3.2. Serum CK18 M65ED Levels in Different Components of Cardiometabolic Disorders
As shown in Table 1, the median serum CK18 M65ED level for all subjects was 137.54 (76.51–254.39) U/L. The subjects with cardiometabolic disorders showed higher serum CK18 M65ED levels (197.36 (121.13–354.5)) than those without cardiometabolic disorders (83.85 (52.8–153.75) U/L) (
[figures omitted; refer to PDF]
3.3. Clinical Characteristics and Prevalence of Cardiometabolic Disorders Stratified by Quartiles of Serum CK18 M65ED Levels
The CK18 M65ED quartiles were defined according to the median and interquartile values for the serum CK18 M65ED levels for the entire study population as follows: quartile 1, ≤76.51 U/L (
Table 2
Characteristics of subjects and prevalence of cardiometabolic disorders and individual components according to CK18 M65ED quartiles.
Variables | Serum CK18 M65ED quartiles | ||||
Quartile 1 | Quartile 2 | Quartile 3 | Quartile 4 | ||
71/76 | 74/73 | 90/57 | 105/42 | — | |
Age (years)§ | 39 (33–45) | 42 (34–52) | 40 (34–48) | 38 (32–52) | 0.159 |
Body mass index (kg/m2)§ | 22.8 (20.8–24.5) | 24.1 (21.8–26.6) | 25.9 (22.9–29.3) | 27.4 (24.5–30.5) | <0.001 |
Waist circumference (cm)§ | 76.0 (67.0–86.0) | 81.0 (71.0–95.0) | 90.1 (73.0–100.0) | 95.0 (87.6–102.9) | <0.001 |
Systolic blood pressure (mmHg)§ | 112.0 (108.5–120.0) | 116.0 (109.0–124.0) | 119.0 (110.0–125.0) | 120.0 (114.0–128.5) | <0.001 |
Diastolic blood pressure (mmHg)§ | 75 (70–80) | 78 (71–80.5) | 80 (73.8–84) | 80 (76–85) | <0.001 |
Fasting plasma glucose (mmol/L)§ | 4.94 (4.61–5.3) | 5.06 (4.72–5.49) | 5.07 (4.79–5.68) | 5.33 (4.90–6.14) | <0.001 |
2-hour plasma glucose (mmol/L)§ | 5.98 (5.16–7.21) | 6.56 (5.44–8.05) | 6.800 (5.67–8.18) | 7.72 (6.49–10.42) | <0.001 |
HbA1c (%)§ | 5.5 (5.3–5.7) | 5.55 (5.3–5.8) | 5.6 (5.4–5.8) | 5.7 (5.4–6.8) | <0.001 |
Fasting insulin (μU/mL)§ | 6.50 (4.60–11.58) | 8.40 (5.09–12.30) | 10.69 (5.24–13.79) | 11.90 (10.16–16.71) | <0.001 |
HOMA-IR§ | 1.42 (1.02–2.48) | 1.80 (1.11–2.86) | 2.44 (1.16–3.44) | 2.94 (2.38–4.78) | <0.001 |
HOMA-B§ | 26.21 (16.27–48.58) | 29.41 (15.47–43.94) | 30.97 (15.91–57.90) | 36.34 (22.78–55.74) | 0.078 |
Alanine aminotransferase (IU/L)§ | 14 (11–20) | 16 (11.75–23) | 22 (14–35) | 41 (24–62) | <0.001 |
Aspartate transaminase (IU/L)§ | 18 (16–21) | 18 (16–22) | 21 (17–25) | 27 (21–32) | <0.001 |
Gamma-glutamyl-transferase (IU/L)§ | 17 (13–26) | 19.5 (14–28) | 31 (17–41) | 40 (24–56) | <0.001 |
Triglycerides (mmol/L)§ | 1.08 (0.77–1.45) | 1.17 (0.79–1.69) | 1.49 (1.03–2.15) | 1.74 (1.14–2.38) | <0.001 |
Total cholesterol (mmol/L) | <0.001 | ||||
HDL cholesterol (mmol/L)§ | 1.31 (1.20–1.48) | 1.28 (1.11–1.47) | 1.13 (0.97–1.35) | 1.10 (0.95–1.21) | <0.001 |
LDL cholesterol (mmol/L) | <0.001 | ||||
Cytokeratin 18 M65 (U/L)§ | 53.00 (43.08–62.25) | 104.69 (87.88–120.78) | 175.22 (155.98–210.06) | 412.52 (321.94–594.20) | <0.001 |
Cardiometabolic disorders ( |
29 (19.73%) | 70 (47.62%) | 90 (61.22%) | 121 (82.31%) | <0.001 |
Obesity ( |
4 (2.72%) | 11 (7.48%) | 28 (19.05%) | 45 (30.61%) | <0.001 |
Central obesity ( |
25 (17.01%) | 59 (40.14%) | 81 (55.1%) | 107 (72.79%) | <0.001 |
Diabetes ( |
1 (0.68%) | 14 (9.52%) | 19 (12.93%) | 42 (28.57%) | <0.001 |
Dyslipidemia ( |
2 (1.36%) | 29 (19.73%) | 65 (44.22%) | 78 (53.06%) | <0.001 |
Hypertension ( |
1 (0.68%) | 12 (8.16%) | 20 (13.61%) | 38 (25.85%) | <0.001 |
No. of CD components | |||||
One or more ( |
29 (19.73%) | 70 (47.62%) | 90 (61.22%) | 122 (82.99%) | <0.001 |
Two or more ( |
0 (0%) | 29 (19.73%) | 67 (45.58%) | 90 (61.22%) | <0.001 |
Three or more ( |
0 (0%) | 11 (7.48%) | 22 (14.97%) | 38 (25.85%) | <0.001 |
Four ( |
0 (0%) | 4 (2.72%) | 6 (4.08%) | 15 (10.2%) | <0.001 |
§Log transformed before analysis. CK18: cytokeratin 18; CD: cardiometabolic disorder.
[figure omitted; refer to PDF]3.4. Association of Clinical Parameters with Serum CK18 M65ED Levels
Pearson’s correlation analysis revealed positive correlations between BMI, WC, SBP, DBP, FPG, 2hPG, HbA1c, HOMA-IR, ALT, AST, GGT, TG, TC, and LDL-C, and serum CK18 M65ED levels (all
Table 3
Pearson correlation analysis and multivariate stepwise regression analysis for serum CK18 M65ED levels.
Variables | Pearson correlation analysis | Multiple stepwise regression analysis | |||
Standardized |
|||||
Gender | -0.16 | <0.001 | — | — | — |
Age (years) | 0.01 | 0.806 | — | — | — |
Body mass index (kg/m2)§ | 0.404 | <0.001 | — | — | — |
Waist circumference (cm)§ | 0.401 | <0.001 | — | — | — |
Systolic blood pressure (mmHg)§ | 0.303 | <0.001 | — | — | — |
Diastolic blood pressure (mmHg)§ | 0.315 | <0.001 | 0.207 | 5.279 | <0.001 |
Fasting plasma glucose (mmol/L)§ | 0.26 | <0.001 | — | — | — |
2-hour plasma glucose (mmol/L)§ | 0.323 | <0.001 | — | — | — |
HbA1c (%)§ | 0.234 | <0.001 | 0.171 | 4.526 | <0.001 |
Fasting insulin (μU/mL)§ | 0.249 | <0.001 | — | — | — |
HOMA-IR§ | 0.297 | <0.001 | — | — | — |
HOMA-B§ | 0.081 | 0.06 | — | — | — |
Alanine aminotransferase (IU/L)§ | 0.522 | <0.001 | 0.474 | 10.464 | <0.001 |
Aspartate transaminase (IU/L)§ | 0.431 | <0.001 | — | — | — |
Gamma-glutamyl-transferase (IU/L)§ | 0.388 | <0.001 | — | — | — |
Triglycerides (mmol/L)§ | 0.329 | <0.001 | — | — | — |
Total cholesterol (mmol/L) | 0.199 | <0.001 | — | — | — |
HDL cholesterol (mmol/L)§ | -0.339 | <0.001 | -0.135 | -3.035 | 0.003 |
LDL cholesterol (mmol/L) | 0.153 | <0.001 | — | — | — |
§Log transformed before analysis. aOriginal variables included all the variables with
3.5. Serum CK18 M65ED Is an Independent Risk Factor for Cardiometabolic Disorders
We performed a binary logistic regression analysis in which the presence of cardiometabolic disorders was designated as the dependent variable in three different models, and adjustments were made for age, gender, BMI, and other related clinical parameters. The multivariable-adjusted (age, gender, and BMI, model 1) OR of cardiometabolic disorders across the quartiles of CK18 M65ED were 1.00 (95% confidence interval (CI), 1.00–1.00), 2.77 (95% CI, 1.31–5.85), 3.14 (95% CI, 1.43–6.89), and 9.08 (95% CI, 3.91–21.08) (all
Table 4
Odds ratios of cardiometabolic disorders based on serum cytokeratin 18 M65ED as a continuous or categorical variable.
Odds ratios (95% CI) | ||||||||
Model 1a | Model 2b | Model 3c | Model 4d | |||||
M65ED as categories | ||||||||
Quartile 1 | 1.00 (1.00–1.00) | — | 1.00 (1.00–1.00) | — | 1.00 (1.00–1.00) | — | 1.00 (1.00–1.00) | — |
Quartile 2 | 2.77 (1.31–5.85) | 0.008 | 3.41 (1.13–10.29) | 0.03 | 3.24 (1.00–10.44) | 0.049 | 4.02 (2.31–7.03) | <0.001 |
Quartile 3 | 3.14 (1.43–6.89) | 0.004 | 3.74 (1.00–13.95) | 0.049 | 4.96 (1.20–20.47) | 0.027 | 6.64 (3.79–11.65) | <0.001 |
Quartile 4 | 9.08 (3.91–21.08) | <0.001 | 14.28 (2.79–73.21) | 0.001 | 16.83 (2.75–103.19) | 0.002 | 18.39 (9.89–34.21) | <0.001 |
<0.001 | 0.012 | 0.018 | <0.001 | |||||
M65ED (per 1 SD) | 1.92 (1.39–2.65) | <0.001 | 1.90 (1.13–3.19) | 0.015 | 1.97 (1.11–3.48) | 0.020 | 2.75 (2.07–3.66) | <0.001 |
aModel 1 adjusted for age, gender, and BMI. bModel 2 adjusted for variables in model 1 and also for SBP, DBP, ALT, AST, GGT, TG, TC, HDL-C, LDL-C, HOMA-IR, HOMA-B, and HbA1c. cModel 3 adjusted for variables in model 1 and also for SBP, DBP, ALT, AST, GGT, TG, TC, HDL-C, LDL-C, FPG, 2hPG, HbA1c, and FINS. dModel 4 adjusted for age, gender, and nonalcoholic fatty liver disease (yes, no). Serum cytokeratin 18 M65ED levels: quartile 1: ≤76.51 U/L; quartile 2: 76.51–137.54 U/L; quartile 3: 137.54–254.39 U/L; quartile 4: ≥254.39 U/L. CI: confidence interval; SD: standard deviation.
4. Discussion
In the present study, we have demonstrated that serum CK18 M65ED levels are significantly increased in Chinese subjects with multiple cardiometabolic disorders and these levels positively correlate with DBP, HbA1c, ALT, and HDL-C levels. Our analyses showed that increased serum CK18 M65ED levels is an independent risk factor for cardiometabolic disorders, after accounting for general cardiometabolic risk parameters or the presence of NAFLD. Additionally, we found that the prevalence of cardiometabolic disorders and its components increased with increasing quartiles of CK18 M65ED.
Several studies have been conducted to assess serum CK18 levels in NAFLD, as it is one of the most extensively evaluated biomarkers of steatohepatitis [35]. However, to our knowledge, limited studies have examined the association between serum CK18 M65ED levels and the risk for cardiometabolic disorders. In accordance with the results of Tan et al. and Civera et al., we showed that CK18 M65ED is positively correlated with the insulin resistance index of HOMA-IR [20, 21]; however, the correlation was not observed in the multiple stepwise regression analysis. The CK18 M65ED levels positively correlated with fasting glucose levels, and it was associated with cardiometabolic disorders independent of NAFLD status, which is in agreement with the results of Chang et al. [19]. In addition to adult hepatocytes [16], CK18 can be expressed in simple epithelial cells, cholangiocytes, pancreas, and colon [16, 24, 25]. CK18 M65ED can act as a cell death marker in cardiometabolic disorders because of its ability to detect both native and intact CK18 [18]. Previous studies have demonstrated that apoptosis can cause a decrease in the number of circulating mucosal-associated invariant T cells in cardiometabolic diseases [7]. Moreover, myocardial cells undergo death in obesity-associated cardiomyopathy in high-fat diet-fed mice [8]. In the present study, we extend these investigations by providing further evidences of the possibility of cell death in cardiometabolic disorders.
The strong correlation between serum CK18 M65ED levels and the risk for cardiometabolic disorders might be explained by the interactions among various metabolic stress inducers that are activated by nutritional surplus, inflammation, innate immune system, and gut microbiota during the induction of cell death. Under conditions of glucotoxicity, lipotoxicity, and exposure to proinflammatory cytokines or biologically active sphingolipids, cell death mediated by ER stress, 4-hydroxynonenal, and endogenous reactive oxygen species may contribute to the pathogenesis of cardiometabolic disorders [9, 10, 36]. Long-lasting and low-grade chronic inflammation mediated by cytokines and chemokines, which are primarily released by innate immune cells under cardiometabolic stress, is a distinguishing feature of cardiometabolic disorders, and their overactive responses can directly contribute to the pathogenesis of such diseases [37–39]. For example, pancreatic β-cell apoptosis due to hyperglycemia and ER stress can contribute to the decreased β-cell mass that characterizes T2D, and the activation of inflammasomes by these harmful factors can lead to β-cell death [9]. Thus, β-cell death and cell death-associated inflammation interact with each other through innate immune receptors [9]. Another explanation might be that altered microbial compositions in patients with cardiometabolic disorders may foster inflammation by producing more proinflammatory molecules such as lipopolysaccharide and peptidoglycans that can interact with host pattern recognition receptors of the innate immune system [40–42]. Winer et al. have demonstrated that obesity-altered gut microbiome can increase the recruitment of macrophages and reduce the proportion of regulatory T cells within the gut [43]. Touch et al. have suggested that apoptosis of mucosal-associated invariant T cells in patients with cardiometabolic disorders occurs due to gut microbiome dysbiosis and enhanced proapoptotic signaling, which is caused by long-term and low-grade systemic inflammation [7]. The pathway in which CK18 is involved in cell death at a molecular level is, however, not very clear. CK18 could regulate the transcription of apoptotic genes FAS and FADD, as well as immune genes CXCL2 and CD79B, in addition to alternative splicing of FAS and CTNNB1 [13]. More precisely designed studies should be conducted to isolate peripheral blood mononuclear cells from the whole blood of the subjects and measure the expression of cell death-related genes and proteins to explore why CK18 M65ED is positively associated with cardiometabolic disorders.
Recent studies demonstrated that some serum metabolites, ratios of several traditional metabolic parameters, and adipokines could have closed relationship with the cardiometabolic risk. Serum glycans could improve type 2 diabetes and CVD prediction beyond established risk markers [44]. TG : HDL-C ratio and triglyceride-glucose index are all reported to be associated with the cardiometabolic disorders [45, 46]. Leptin, adiponectin, and their ratio were associated with insulin resistance and other cardiometabolic comorbidities [47]. In the latest researches, branched chain amino acids were found to be passive biomarkers or the key to the pathogenesis of cardiometabolic diseases [48]. Based on our data, instead of ratios and in an easier testing method, CK18 M65ED is an independent risk factor for cardiometabolic disorders, which remained statistically significant after adjustment with age, gender, NAFLD, and traditional metabolic variables.
The limitation of this study is its cross-sectional study design and relatively small sample size, which made it difficult to address the cause-effect relationship between serum CK18 M65ED levels and cardiometabolic disorders. Secondly, the serum levels of CK18 M65ED provided limited information about the cell death condition in the body. Hence, further prospective studies are warranted to confirm the present findings in a larger population.
5. Conclusions
In this study, the serum levels of CK18 M65ED, as a cell death marker, were significantly increased among subjects with cardiometabolic disorders, and these increased levels were associated with the risk for cardiometabolic disorders, independent of other cardiometabolic risk parameters, and NAFLD. Our study suggested that elevated serum CK18 M65ED levels could be used to predict the risk for cardiometabolic disorders.
Ethical Approval
The study was approved and supervised by the Ethics Committee of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, following the principles of the declaration of Helsinki. All subjects provided written informed consent.
Acknowledgments
This work was supported by the National Natural Science Foundation major international (regional) joint research project of China (grant number 81220108006) and National Natural Science Foundation Council-National Health and Medical Research Council joint research grant of China (grant number 81561128016) to Weiping Jia, General Program of Natural Science Foundation of China (grant number 81870598), Shanghai Pujiang Program (grant number 17PJ1407500), and Municipal Human Resources Development Program for Outstanding Young Talents in Medical and Health Sciences in Shanghai of China (grant number 2017YQ009) to Huating Li. We are very grateful to all of the involved clinicians, nurses, and technicians for dedicating their time and skill for helping with the present study.
[1] GBD 2013 Mortality and Causes of Death Collaborators, I. I. Abubakar, T. Tillmann, A. Banerjee, "Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013," Lancet, vol. 385 no. 9963, pp. 117-171, DOI: 10.1016/S0140-6736(14)61682-2, 2015.
[2] D. Mozaffarian, E. J. Benjamin, A. S. Go, D. K. Arnett, M. J. Blaha, M. Cushman, S. R. Das, S. de Ferranti, J.-P. Després, H. J. Fullerton, V. J. Howard, M. D. Huffman, C. R. Isasi, M. C. Jiménez, S. E. Judd, B. M. Kissela, J. H. Lichtman, L. D. Lisabeth, S. Liu, R. H. Mackey, D. J. Magid, D. K. McGuire, E. R. Mohler, C. S. Moy, P. Muntner, M. E. Mussolino, K. Nasir, R. W. Neumar, G. Nichol, L. Palaniappan, D. K. Pandey, M. J. Reeves, C. J. Rodriguez, W. Rosamond, P. D. Sorlie, J. Stein, A. Towfighi, T. N. Turan, S. S. Virani, D. Woo, R. W. Yeh, M. B. Turner, "Heart disease and stroke statistics-2016 update: a report from the American Heart Association," Circulation, vol. 133 no. 4, pp. e38-360, DOI: 10.1161/CIR.0000000000000350, 2016.
[3] L. Ozcan, I. Tabas, "Role of endoplasmic reticulum stress in metabolic disease and other disorders," Annual Review of Medicine, vol. 63 no. 1, pp. 317-328, DOI: 10.1146/annurev-med-043010-144749, 2012.
[4] S. Dalleau, M. Baradat, F. Guéraud, L. Huc, "Cell death and diseases related to oxidative stress:4-hydroxynonenal (HNE) in the balance," Cell Death and Differentiation, vol. 20 no. 12, pp. 1615-1630, DOI: 10.1038/cdd.2013.138, 2013.
[5] K. S. Twayana, P. Ravanan, "Eukaryotic cell survival mechanisms: disease relevance and therapeutic intervention," Life Sciences, vol. 205, pp. 73-90, DOI: 10.1016/j.lfs.2018.05.002, 2018.
[6] A. Swisa, B. Glaser, Y. Dor, "Metabolic stress and compromised identity of pancreatic beta cells," Frontiers in Genetics, vol. 8,DOI: 10.3389/fgene.2017.00021, 2017.
[7] S. Gezginci-Oktayoglu, E. Onay-Ucar, S. Sancar-Bas, A. Karatug-Kacar, E. S. N. Arda, S. Bolkent, "Involvement of dying beta cell originated messenger molecules in differentiation of pancreatic mesenchymal stem cells under glucotoxic and glucolipotoxic conditions," Journal of Cellular Physiology, vol. 233 no. 5, pp. 4235-4244, DOI: 10.1002/jcp.26242, 2018.
[8] R. Guardado Mendoza, C. Perego, G. Finzi, S. la Rosa, C. Capella, L. M. Jimenez-Ceja, L. A. Velloso, M. J. Saad, F. Sessa, F. Bertuzzi, S. Moretti, Dick EJ Jr, A. M. Davalli, F. Folli, "Delta cell death in the islet of Langerhans and the progression from normal glucose tolerance to type 2 diabetes in non-human primates (baboon, Papio hamadryas )," Diabetologia, vol. 58 no. 8, pp. 1814-1826, DOI: 10.1007/s00125-015-3625-5, 2015.
[9] J. Guo, Z. Wang, J. Wu, M. Liu, M. Li, Y. Sun, W. Huang, Y. Li, Y. Zhang, W. Tang, X. Li, C. Zhang, F. Hong, N. Li, J. Nie, F. Yi, "Endothelial SIRT6 is vital to prevent hypertension and associated cardiorenal injury through targeting Nkx3.2-GATA5 signaling," Circulation Research, vol. 124 no. 10, pp. 1448-1461, DOI: 10.1161/CIRCRESAHA.118.314032, 2019.
[10] S. Touch, K. E. Assmann, J. Aron-Wisnewsky, F. Marquet, C. Rouault, M. Fradet, H. Mosbah, M. C. Consortium, R. Isnard, G. Helft, A. Lehuen, C. Poitou, K. Clément, S. André, "Mucosal-associated invariant T (MAIT) cells are depleted and prone to apoptosis in cardiometabolic disorders," FASEB Journal, vol. 32 no. 9, pp. 5078-5089, DOI: 10.1096/fj.201800052RR, 2018.
[11] H. Tronchere, M. Cinato, A. Timotin, L. Guitou, C. Villedieu, H. Thibault, D. Baetz, B. Payrastre, P. Valet, A. Parini, O. Kunduzova, F. Boal, "Inhibition of PIKfyve prevents myocardial apoptosis and hypertrophy through activation of SIRT3 in obese mice," EMBO Molecular Medicine, vol. 9 no. 6, pp. 770-785, DOI: 10.15252/emmm.201607096, 2017.
[12] M. Hesse, T. M. Magin, K. Weber, "Genes for intermediate filament proteins and the draft sequence of the human genome: novel keratin genes and a surprisingly high number of pseudogenes related to keratin genes 8 and 18," Journal of Cell Science, vol. 114 no. 14, pp. 2569-2575, 2001.
[13] Y. Cheng, K. Qin, N. Huang, Z. Zhou, H. Xiong, J. Zhao, Y. Zhang, S. Yu, "Cytokeratin 18 regulates the transcription and alternative splicing of apoptotic-related genes and pathways in HeLa cells," Oncology Reports, vol. 42 no. 1, pp. 301-312, DOI: 10.3892/or.2019.7166, 2019.
[14] M. C. Abraham, S. Shaham, "Death without caspases, caspases without death," Trends in Cell Biology, vol. 14 no. 4, pp. 184-193, DOI: 10.1016/j.tcb.2004.03.002, 2004.
[15] A. Eguchi, A. Wree, A. E. Feldstein, "Biomarkers of liver cell death," Journal of Hepatology, vol. 60 no. 5, pp. 1063-1074, DOI: 10.1016/j.jhep.2013.12.026, 2014.
[16] M. B. Omary, N. O. Ku, P. Strnad, S. Hanada, "Toward unraveling the complexity of simple epithelial keratins in human disease," Journal of Clinical Investigation, vol. 119 no. 7, pp. 1794-1805, DOI: 10.1172/JCI37762, 2009.
[17] A. E. Feldstein, A. Wieckowska, A. R. Lopez, Y. C. Liu, N. N. Zein, A. McCullough, "Cytokeratin-18 fragment levels as noninvasive biomarkers for nonalcoholic steatohepatitis: a multicenter validation study," Hepatology, vol. 50 no. 4, pp. 1072-1078, DOI: 10.1002/hep.23050, 2009.
[18] D. Joka, K. Wahl, S. Moeller, J. Schlue, B. Vaske, M. J. Bahr, M. P. Manns, K. Schulze-Osthoff, H. Bantel, "Prospective biopsy-controlled evaluation of cell death biomarkers for prediction of liver fibrosis and nonalcoholic steatohepatitis," Hepatology, vol. 55 no. 2, pp. 455-464, DOI: 10.1002/hep.24734, 2012.
[19] Y. H. Chang, H. C. Lin, D. W. Hwu, D. M. Chang, K. C. Lin, Y. J. Lee, "Elevated serum cytokeratin-18 concentration in patients with type 2 diabetes mellitus and non-alcoholic fatty liver disease," Annals of Clinical Biochemistry, vol. 56 no. 1, pp. 141-147, DOI: 10.1177/0004563218796259, 2019.
[20] S. Tan, L. P. Bechmann, S. Benson, T. Dietz, S. Eichner, S. Hahn, O. E. Janssen, H. Lahner, G. Gerken, K. Mann, A. Canbay, "Apoptotic markers indicate nonalcoholic steatohepatitis in polycystic ovary syndrome," Journal of Clinical Endocrinology & Metabolism, vol. 95 no. 1, pp. 343-348, DOI: 10.1210/jc.2009-1834, 2010.
[21] M. Civera, A. Urios, M. L. Garcia-Torres, J. Ortega, J. Martinez-Valls, N. Cassinello, J. del Olmo, A. Ferrandez, J. M. Rodrigo, C. Montoliu, "Relationship between insulin resistance, inflammation and liver cell apoptosis in patients with severe obesity," Diabetes Metabolism Research and Reviews, vol. 26 no. 3, pp. 187-192, DOI: 10.1002/dmrr.1070, 2010.
[22] M. C. Yang, H. K. Liu, Y. T. Su, C. C. Tsai, J. R. Wu, "Serum apoptotic marker M30 is positively correlated with early diastolic dysfunction in adolescent obesity," PloS One, vol. 14 no. 5,DOI: 10.1371/journal.pone.0217429, 2019.
[23] H. Li, Q. Fang, F. Gao, J. Fan, J. Zhou, X. Wang, H. Zhang, X. Pan, Y. Bao, K. Xiang, A. Xu, W. Jia, "Fibroblast growth factor 21 levels are increased in nonalcoholic fatty liver disease patients and are correlated with hepatic triglyceride," Journal of Hepatology, vol. 53 no. 5, pp. 934-940, DOI: 10.1016/j.jhep.2010.05.018, 2010.
[24] T. Ueno, M. Toi, S. Linder, "Detection of epithelial cell death in the body by cytokeratin 18 measurement," Biomedicine & Pharmacotherapy, vol. 59, pp. S359-S362, DOI: 10.1016/S0753-3322(05)80078-2, 2005.
[25] S. Aho, J. Uitto, "180-kD bullous pemphigoid antigen/type XVII collagen: tissue-specific expression and molecular interactions with keratin 18," Journal of Cellular Biochemistry, vol. 72 no. 3, pp. 356-367, DOI: 10.1002/(sici)1097-4644(19990301)72:3<356::aid-jcb5>3.0.co;2-m, 1999.
[26] B. Schutte, M. Henfling, W. Kölgen, M. Bouman, S. Meex, M. P. G. Leers, M. Nap, V. Björklund, P. Björklund, B. Björklund, E. B. Lane, M. B. Omary, H. Jörnvall, F. C. S. Ramaekers, "Keratin 8/18 breakdown and reorganization during apoptosis," Experimental Cell Research, vol. 297 no. 1, pp. 11-26, DOI: 10.1016/j.yexcr.2004.02.019, 2004.
[27] L. De Petris, E. Brandén, R. Herrmann, B. C. Sanchez, H. Koyi, B. Linderholm, R. Lewensohn, S. Linder, J. Lehtiö, "Diagnostic and prognostic role of plasma levels of two forms of cytokeratin 18 in patients with non-small-cell lung cancer," European Journal of Cancer, vol. 47 no. 1, pp. 131-137, DOI: 10.1016/j.ejca.2010.08.006, 2011.
[28] T. Wang, Z. Zhao, Y. Xu, L. Qi, M. Xu, J. Lu, M. Li, Y. Chen, M. Dai, W. Zhao, G. Ning, W. Wang, Y. Bi, "Insulin resistance and beta-cell dysfunction in relation to cardiometabolic risk patterns," Journal of Clinical Endocrinology & Metabolism, vol. 103 no. 6, pp. 2207-2215, DOI: 10.1210/jc.2017-02584, 2018.
[29] World Health Organization, Obesity: preventing and managing the global epidemic, . May 2019 http://www.who.int/nutrition/publications/obesity_executive_summary.pdf
[30] The World Health Organization Western Pacific Region, The Asia-Pacific perspective: redefining obesity and its treatment, . May 2019 http://www.wpro.who.int/nutrition/documents/docs/Redefiningobesity.pdf
[31] American Diabetes Association, "2. Classification and diagnosis of Diabetes:Standards of medical care in diabetes-2018," Diabetes Care, vol. 41, pp. S13-s27, DOI: 10.2337/dc18-S002, 2018.
[32] Expert Panel on Detection, Evaluation, Treatment of High Blood Cholesterol in Adults, "Executive summary of the third report of the National Cholesterol Education Program (NCEP) expert panel on detection, evaluation, and treatment of high blood cholesterol in adults (Adult Treatment Panel III)," Journal of the American Medical Association, vol. 285 no. 19, pp. 2486-2497, DOI: 10.1001/jama.285.19.2486, 2001.
[33] A. Chobanian, G. Bakris, H. Black, W. C. Cushman, L. A. Green, Izzo JL Jr, D. W. Jones, B. J. Materson, S. Oparil, Wright JT Jr, E. J. Roccella, National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure, National High Blood Pressure Education Program Coordinating Committee, "The seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure: the JNC 7 report," Journal of the American Medical Association, vol. 289 no. 19, pp. 2560-2572, DOI: 10.1001/jama.289.19.2560, 2003.
[34] S. Chitturi, G. C. Farrell, E. Hashimoto, T. Saibara, G. K. Lau, J. D. Sollano, Asia-Pacific Working Party on NAFLD, "Non-alcoholic fatty liver disease in the Asia-Pacific region: definitions and overview of proposed guidelines," Journal of Gastroenterology and Hepatology, vol. 22 no. 6, pp. 778-787, DOI: 10.1111/j.1440-1746.2007.05001.x, 2007.
[35] V. W. S. Wong, L. A. Adams, V. de Lédinghen, G. L. H. Wong, S. Sookoian, "Noninvasive biomarkers in NAFLD and NASH -- current progress and future promise," Nature Reviews Gastroenterology Hepatology, vol. 15 no. 8, pp. 461-478, DOI: 10.1038/s41575-018-0014-9, 2018.
[36] I. Tabas, D. Ron, "Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress," Nature Cell Biology, vol. 13 no. 3, pp. 184-190, DOI: 10.1038/ncb0311-184, 2011.
[37] M. Xu, P. P. Liu, H. Li, "Innate immune signaling and its role in metabolic and cardiovascular diseases," Physiological Reviews, vol. 99 no. 1, pp. 893-948, DOI: 10.1152/physrev.00065.2017, 2019.
[38] Y. Tang, S. Purkayastha, D. Cai, "Hypothalamic microinflammation: a common basis of metabolic syndrome and aging," Trends in Neurosciences, vol. 38 no. 1, pp. 36-44, DOI: 10.1016/j.tins.2014.10.002, 2015.
[39] G. P. Shi, I. Bot, P. T. Kovanen, "Mast cells in human and experimental cardiometabolic diseases," Nature Reviews. Cardiology, vol. 12 no. 11, pp. 643-658, DOI: 10.1038/nrcardio.2015.117, 2015.
[40] A. S. Neish, "Microbes in gastrointestinal health and disease," Gastroenterology, vol. 136 no. 1, pp. 65-80, DOI: 10.1053/j.gastro.2008.10.080, 2009.
[41] W. H. Tang, T. Kitai, S. L. Hazen, "Gut microbiota in cardiovascular health and disease," Circulation Research, vol. 120 no. 7, pp. 1183-1196, DOI: 10.1161/CIRCRESAHA.117.309715, 2017.
[42] E. Larsson, V. Tremaroli, Y. S. Lee, O. Koren, I. Nookaew, A. Fricker, J. Nielsen, R. E. Ley, F. Bäckhed, "Analysis of gut microbial regulation of host gene expression along the length of the gut and regulation of gut microbial ecology through MyD88," Gut, vol. 61 no. 8, pp. 1124-1131, DOI: 10.1136/gutjnl-2011-301104, 2012.
[43] D. A. Winer, H. Luck, S. Tsai, S. Winer, "The intestinal immune system in obesity and insulin resistance," Cell Metabolism, vol. 23 no. 3, pp. 413-426, DOI: 10.1016/j.cmet.2016.01.003, 2016.
[44] C. Wittenbecher, T. Štambuk, O. Kuxhaus, N. Rudman, F. Vučković, J. Štambuk, C. Schiborn, D. Rahelić, S. Dietrich, O. Gornik, M. Perola, H. Boeing, M. B. Schulze, G. Lauc, "PlasmaN-Glycans as emerging biomarkers of cardiometabolic risk: a prospective investigation in the EPIC-Potsdam cohort study," Diabetes Care, vol. 43 no. 3, pp. 661-668, DOI: 10.2337/dc19-1507, 2020.
[45] A. K. N. Z. Iwani, M. Y. Jalaludin, R. M. W. M. Zin, M. Z. Fuziah, J. Y. H. Hong, Y. Abqariyah, A. H. Mokhtar, W. N. W. Mohamud, "TG : HDL-C Ratio Is a Good Marker to Identify Children Affected by Obesity with Increased Cardiometabolic Risk and Insulin Resistance," International Journal of Endocrinology, vol. 2019,DOI: 10.1155/2019/8586167, 2019.
[46] A. da Silva, A. P. S. Caldas, H. H. M. Hermsdorff, Â. C. Bersch-Ferreira, C. R. Torreglosa, B. Weber, J. Bressan, "Triglyceride-glucose index is associated with symptomatic coronary artery disease in patients in secondary care," Cardiovascular Diabetology, vol. 18 no. 1,DOI: 10.1186/s12933-019-0893-2, 2019.
[47] C. Frithioff‐Bøjsøe, M. A. V. Lund, U. Lausten‐Thomsen, P. L. Hedley, O. Pedersen, M. Christiansen, J. L. Baker, T. Hansen, J.‐. C. Holm, "Leptin, adiponectin, and their ratio as markers of insulin resistance and cardiometabolic risk in childhood obesity," Pediatric Diabetes, vol. 21 no. 2, pp. 194-202, DOI: 10.1111/pedi.12964, 2020.
[48] S. Marta, D. Jacek, "Branched chain amino acids: passive biomarkers or the key to the pathogenesis of cardiometabolic diseases?," Advances in Clinical and Experimental Medicine, vol. 28 no. 9, pp. 1263-1269, 2019.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2020 Lingling Qian et al. This work is licensed under http://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Background. Recent studies have suggested that cell death might be involved in the pathophysiology of metabolic disorders. The cytokeratin 18 (CK18) fragment, as a cell death marker, plays an important role in nonalcoholic fatty liver disease (NAFLD). However, only a limited number of studies have found elevated serum levels of CK18 in patients with type 2 diabetes. Moreover, no studies have been conducted yet to investigate the role of CK18 in hypertension or dyslipidemia. In particular, CK18 M65ED is a more sensitive marker of cell death, and its role in cardiometabolic disorders has not been revealed yet. Methods. A total of 588 subjects were enrolled from the local communities of Shanghai. Serum CK18 M65ED were determined using the enzyme-linked immunosorbent assay. A cardiometabolic disorder was identified by the presence of at least one of the components including overweight or central obesity, diabetes, dyslipidemia, and hypertension. Results. Subjects with cardiometabolic disorders exhibited significantly higher serum levels of CK18 M65ED than those without cardiometabolic disorders (197.36 (121.13–354.50) U/L versus 83.85 (52.80–153.75) U/L, respectively,
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details



1 Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai 200233, China; Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
2 Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai 200233, China