Skip to main content
Erschienen in: Cardiovascular Diabetology 1/2019

Open Access 01.12.2019 | Original investigation

Temporal relationship between inflammation and insulin resistance and their joint effect on hyperglycemia: the Bogalusa Heart Study

verfasst von: Yinkun Yan, Shengxu Li, Yang Liu, Lydia Bazzano, Jiang He, Jie Mi, Wei Chen

Erschienen in: Cardiovascular Diabetology | Ausgabe 1/2019

Abstract

Background

Inflammation and insulin resistance play crucial roles in the development of type 2 diabetes mellitus (T2DM). We aim to examine the temporal relationship between high-sensitivity C-reactive protein (hsCRP) and insulin resistance in non-diabetic adults and their joint effect on the development of hyperglycemia.

Methods

The longitudinal cohort from the Bogalusa Heart Study consisted of 509 non-diabetic adults (360 whites and 149 blacks, mean age = 42.8 years at follow-up) who had hsCRP, fasting glucose and insulin measured twice at baseline and follow-up over 6.8 years. Cross-lagged panel model was used to examine the temporal relationship between hsCRP and homeostasis model assessment for insulin resistance (HOMA-IR). Information on incident T2DM was collected in a survey in 6.1 years after the follow-up survey.

Results

After adjusting for race, sex, age, body mass index, smoking, alcohol drinking and follow-up years, the path coefficient from baseline hsCRP to follow-up HOMA-IR (β2 = 0.105, p = 0.009) was significant and greater than the path from baseline HOMA-IR to follow-up hsCRP (β1 = 0.005, p = 0.903), with p = 0.011 for the difference between β1 and β2. This one-directional path from baseline hsCRP to follow-up HOMA-IR was significant in the hyperglycemia group but not in the normoglycemia group. In addition, participants with high levels of baseline hsCRP and follow-up HOMA-IR had greater risks of T2DM (odds ratio, OR = 2.38, p = 0.035), pre-T2DM (OR = 2.27, p = 0.006) and hyperglycemia (OR = 2.18, p = 0.003) than those with low–low levels.

Conclusions

These findings suggest that elevated hsCRP is associated with future insulin resistance in non-diabetic adults, and their joint effect is predictive of the development of T2DM.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12933-019-0913-2) contains supplementary material, which is available to authorized users.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
T2DM
type 2 diabetes
hsCRP
high-sensitivity C-reactive protein
HOMA-IR
homeostatic model assessment for insulin resistance
RMR
root mean square residual
CFI
comparative fit index
OR
odds ratio
CI
confidence interval

Background

Chronic inflammation has been shown to play a crucial role in the development of type 2 diabetes (T2DM) [14] and cardiovascular disease [59]. High-sensitivity C-reactive protein (hsCRP) is a sensitive marker of subclinical inflammation and strongly predicts increased risks of T2DM [10, 11]. Cross-sectional studies have shown that multiple inflammation markers including hsCRP are correlated with insulin resistance in diabetic and nondiabetic individuals [12, 13]. It is generally considered that hsCRP, insulin resistance, and T2DM are linked through obesity [14]. To date, there have been no studies focusing on the joint effect of increased hsCRP and insulin resistance on the development of T2DM independent of obesity.
The strong association between hsCRP and insulin resistance suggests that chronic inflammation and insulin resistance can influence each other based on pathophysiological and metabolic mechanisms [3, 1517]. Inflammation may activate the innate immune system and interact with adipose tissue-specific macrophages [15, 17]. Although their relationship is likely bidirectional based on the existing literature in this regard, convincing evidence is lacking from the general population regarding the temporal relationship (causal sequence) between hsCRP and insulin resistance. Several longitudinal studies have demonstrated that higher baseline hsCRP and greater change in hsCRP are associated with the development of insulin resistance [1821]; however, Mendelian randomization studies failed to demonstrate a causal relationship between hsCRP levels and insulin resistance/T2DM [22, 23]. Understanding the potential causal relationship between inflammation and insulin resistance may help provide new insights into the underlying mechanisms and yield novel approaches to prevent T2DM and cardiovascular diseases by focusing on interventions targeting at the causal factor.
Utilizing longitudinal data from the Bogalusa Heart Study, the current study aims to examine the temporal relationship between inflammation measured by hsCRP and homeostasis model assessment for insulin resistance (HOMA-IR) in non-diabetic adults and their joint effect on the development of T2DM.

Methods

Study population

The Bogalusa Heart Study is a series of long-term epidemiologic studies in a semi-rural, biracial (65% white and 35% black) community in Bogalusa, Louisiana founded by Dr. Gerald Berenson in 1973. This study focuses on the early natural history of cardiovascular disease from childhood [24]. Between 2000 and 2016, three cross-sectional surveys (baseline, follow-up and outcome surveys) of adults aged 24 to 58 years were conducted in Bogalusa, Louisiana. By linking the first two surveys (baseline in 2000–2002 and follow-up in 2006–2010), 755 non-diabetic adult participants were identified who had cardiovascular risk factors, including hsCRP, fasting plasma glucose and insulin, measured twice in both baseline and follow-up surveys, with an average follow-up period of 6.8 years. Among these 755 non-diabetic participants, 509 adults (360 Whites and 149 Blacks; 34.6% males, mean age = 49.3 years) were enrolled in the last survey (the outcome survey) in 2013–2016. These 509 participants who had incident T2DM and anti-diabetic medication information in the last survey formed the longitudinal cohort for the current study, with an average follow-up period of 6.1 years (range = 3.5–10.0 years) from the second (follow-up) survey.
All subjects gave informed consent for each survey. Study protocols were approved by the Institutional Review Board of the Tulane University Health Sciences Center.

Measurements

Insulin, hsCRP, and glucose were measured at the same time at respective baseline and follow-up surveys. Standardized protocols were used by trained examiners across all surveys. Subjects were instructed to fast for 12 h before screening. Replicate measurements of height and weight were made, and the mean values were used for analysis. Body mass index (BMI, weight in kilograms divided by height in meters squared) was used as a measure of overall adiposity. Information on medication history, smoking and alcohol drinking was obtained in a questionnaire survey. Current smoking and alcohol drinking were defined as smoking at least one cigarette per day and consuming alcohol every day, respectively, during the prior 12 months.
Plasma hsCRP was measured by latex particle-enhanced immunoturbidimetric assay on Hitachi 902 Automatic Analyzer. Plasma glucose levels were measured as part of a multiple chemistry profile (SMA20; Laboratory Corporation of America, Burlington, NC). A commercial radioimmunoassay kit was used for measuring plasma immunoreactive insulin levels (Phadebas; Pharmacia Diagnostics, Piscataway, New Jersey). The intraclass correlation coefficients between blind duplicate values ranged from 0.86 to 0.98 for insulin, hsCRP, and glucose. HOMA-IR was calculated using the formula as described by Matthews [25]: HOMA-IR = fasting plasma insulin (μU/mL) × fasting plasma glucose (mmol/L)/22.5.

Diagnosis of T2DM and pre-T2DM

T2DM was defined as fasting glucose ≥ 126 mg/dL or taking anti-diabetic medications. Prediabetes (pre-T2DM) was defined as fasting glucose of 100–125 mg/dL. Hyperglycemia was defined as being T2DM and/or pre-T2DM.

Statistical analysis

Values for hsCRP, insulin, and HOMA-IR were log-transformed before subsequent analyses because of their skewed distributions. Analyses of covariance (generalized linear models) were performed to test differences in continuous study variables between race and sex groups.
The study design of longitudinal changes of hsCRP, glucose, and insulin measured at two time points in the baseline and follow-up surveys was typically a cross-lagged panel design [26, 27]. The cross-lagged panel analysis is a form of path analysis that simultaneously examines reciprocal and longitudinal relationships among a set of intercorrelated variables. Kenny et al. first proposed the cross-lagged analysis model by calculating partial correlation coefficients to measure the direct paths [26]. In more recent years, structural equation modeling has been used to estimate the path coefficients [27]. A simplified and conceptual version of the model is presented in Fig. 1. The path with β1 describes the effect of baseline HOMA-IR on subsequent hsCRP, and the path with β2 describes the effect of baseline hsCRP on subsequent HOMA-IR. Prior to cross-lagged path analysis, the baseline and follow-up values of log-transformed hsCRP and log-transformed HOMA-IR were adjusted for age, BMI, smoking, and alcohol drinking by regression residual analyses and then standardized by Z-transformation (mean = 0, SD = 1) in race-sex groups. The four variables, i.e. hsCRP and HOMA-IR at baseline and follow-up, generated six pair-wise observed correlations, and five correlations in Fig. 1 were used to estimate β1 and β2 because the connecting path through hsCRP-HOMA-IR correlation at follow-up is illegal according to the path analysis rules. Pearson correlation coefficients of the covariate-adjusted and Z-transformed variables of hsCRP and HOMA-IR at baseline and follow-up were calculated; correlation coefficients between baseline and follow-up values were calculated with additional adjustment for follow-up years. The cross-lagged path coefficients (β1 and β2) in the path diagram in Fig. 1 were estimated simultaneously based on the correlation matrix using the maximum likelihood method by the program LISREL 8.52. The validity of model fitting was indicated by root mean square residual (RMR) and comparative fit index (CFI) [28]. A significant path coefficient (β1 or β2) suggests the directionality, and a significant difference between β1 and β2 provides stronger evidence for the directional path between the two variables measured over time.
Stratified analyses were performed to examine the difference in cross-lagged path parameters between groups classified by glycemic status, race, sex, and follow-up period. The difference between β1 and β2 derived from the standardized variables (Z-scores) was tested using Fisher’s Z-test. Multivariable logistic regression models were used to examine the joint effect of baseline hsCRP and follow-up HOMA-IR on T2DM, pre-T2DM, and hyperglycemia. Low and high levels of baseline hsCRP and follow-up HOMA-IR were defined by their medians.

Results

Characteristics of participants

The mean levels of continuous variables were compared between race and sex groups, adjusting for age (except age itself). Although log-transformed hsCRP, insulin, and HOMA-IR were used for subsequent analyses, the medians (interquartile ranges) of their original values were presented in Table 1. Race differences in BMI, insulin, and HOMA-IR (blacks > whites) in females and sex differences in glucose (males > females), insulin (males > females), HOMA-IR (males > females) and hsCRP (males < females) in whites were observed in both baseline and follow-up surveys. In the outcome survey, race differences in BMI (blacks > whites) in females and incidence of T2DM (blacks > whites) in males and sex differences in hyperglycemia (males > females) and pre-T2DM (males > females) in blacks and whites were found.
Table 1
Characteristics of study participants in baseline, follow-up and outcome surveys by race and sex
 
White
Black
p for race differencea
Male (n = 138)
Female (n = 222)
Male (n = 38)
Female (n = 111)
Male
Female
Baseline survey
 Age (years)
36.4 (4.5)
36.2 (4.2)
36.6 (4.2)
35.0 (4.5)
0.975
0.041
 BMI (kg/m2)
28.1 (4.9)
27.2 (6.2)
29.4 (6.7)
31.3 (7.8)
0.286
< 0.001
 Smoking, n (%)
35 (25.4)
41 (18.5)
15 (39.5)
33 (29.7)
0.088
0.020
 Alcohol drinking, n (%)
54 (39.1)
98 (44.1)
8 (21.1)
42 (37.8)
0.039
0.272
 Glucose (mg/dL)
83.5 (9.4)**
79.6 (8.1)
83.2 (8.9)
81.2 (9.3)
0.861
0.121
 Insulin (μU/mL)b
10.0 (7.0–14.0)*
8.0 (6.0–13.0)
9.0 (5.0–17.0)
11.0 (7.0–17.0)
0.782
0.001
 HOMA-IRb
2.04 (1.45–2.82)*
1.65 (1.11–2.60)
1.89 (1.06–3.66)
2.27 (1.43–3.46)
0.786
0.001
 hsCRP (mg/L)b
0.93 (0.37–1.93)**
1.65 (0.71–4.15)
1.78 (0.60–3.09)
2.07 (0.58–4.80)
0.131
0.830
 Pre-diabetes, n (%)
5 (3.6)
3 (1.4)
2 (5.3)
2 (1.8)
0.155
0.255
Follow-up survey
 Age (years)
43.2 (4.7)
43.0 (4.5)
43.6 (4.5)
41.9 (4.7)
0.674
0.043
 Follow-up period (years)
6.8 (0.9)
6.8 (0.9)
7.0 (0.8)
6.9 (0.9)
0.222
0.123
 BMI (kg/m2)
29.6 (5.6)
28.6 (6.7)
31.8 (8.2)
33.0 (8.2)
0.131
< 0.001
 Smoking, n (%)
32 (23.2)
50 (22.5)
15 (39.5)
33 (29.7)
0.045
0.152
 Alcohol drinking, n (%)
52 (37.7)
91 (41.0)
7 (18.4)
38 (34.2)
0.026
0.233
 Glucose (mg/dL)
90.6 (10.7)**
86.7 (8.8)
92.5 (12.2)**
88.8 (8.6)
0.345
0.036
 Insulin (μU/mL)b
10.1 (6.3–16.2)*
8.2 (5.1–13.0)
13.6 (6.7–24.0)
12.8 (6.96–18.9)
0.052
< 0.001
 HOMA-IRb
2.26 (1.35–3.63)*
1.74 (1.04–3.00)
3.33 (1.20–5.78)
2.82 (1.43–4.56)
0.065
< 0.001
 hsCRP (mg/L)b
0.93 (0.41–1.97)**
1.35 (0.52–3.14)
1.09 (0.56–2.94)
2.07 (0.58–4.80)
0.186
0.062
 Pre-diabetes, n (%)
22 (15.9)*
19 (8.6)
11 (29.0)**
10 (9.0)
0.069
0.891
Outcome survey
 Age (years)
49.6 (4.5)
49.5 (4.2)
50.1 (4.2)*
48.0 (4.7)
0.547
0.003
 Follow-up period (year)c
6.0 (1.1)
6.1 (1.1)
6.2 (1.1)
5.9 (1.1)
0.386
0.067
 BMI (kg/m2)
30.3 (5.4)
29.5 (6.7)
32.3 (8.6)
34.9 (8.5)
0.085
< 0.001
 Smoking, n (%)
33 (23.9)
45 (20.3)
20 (52.6)**
27 (24.3)
< 0.001
0.397
 Alcohol drinking, n (%)
71 (51.5)
88 (55.4)
18 (47.4)**
18 (16.2)
0.656
< 0.001
 Hyperglycemia, n (%)
76 (55.1)**
78 (35.1)
27 (71.1)*
50 (45.1)
0.077
0.080
 Pre-diabetes, n (%)
61 (44.2)**
51 (22.9)
17 (44.8)*
34 (30.7)
0.288
0.091
 T2DM, n (%)
15 (10.9)
27 (12.2)
10 (26.3)
16 (14.4)
0.016
0.563
Continuous variables are presented as means (SD) or medians (interquartile range)
BMI, body mass index; hsCRP, high-sensitivity C-reactive protein; HOMA-IR, homeostasis model assessment for insulin resistance; T2DM, type 2 diabetes mellitus
Sex difference within race: * p < 0.05; ** p < 0.01
ap values for race difference in continuous variables were adjusted for age
bMedian (interquartile range)
cFollow-up period from the time point of the follow-up survey

Correlations between hsCRP and HOMA-IR at baseline and follow-up

After adjusting for race, sex, age, BMI, smoking and alcohol use, with additional adjustment for follow-up years for baseline-follow-up correlations, most correlation coefficients between baseline and follow-up values of log-hsCRP and log-HOMA-IR were significant in the total sample and subgroups based on the cut-off values for the significance level (p < 0.05) listed below the table. All the coefficients did not differ between subgroups except for the difference in coefficients of follow-up log-hsCRP and log-HOMA-IR between races (0.248 vs 0.043) (Table 2).
Table 2
Pearson correlation coefficients between log-transformed hsCRP and HOMA-IR at baseline and follow-up in the total cohort and subgroups, adjusted for covariates
Group
Variable
Baseline hsCRP
Baseline HOMA-IR
Follow-up hsCRP
Totala
Baseline HOMA-IR
0.102
  
Follow-up hsCRP
0.473
0.053
 
Follow-up HOMA-IR
0.147
0.418
0.187
White/blackb
Baseline HOMA-IR
0.108/0.090
  
Follow-up hsCRP
0.494/0.416
0.082/− 0.014
 
Follow-up HOMA-IR
0.154/0.152
0.437/0.369
0.248/0.043*
Male/femalec
Baseline HOMA-IR
0.036/0.139
  
Follow-up hsCRP
0.444/0.495
0.029/0.068
 
Follow-up HOMA-IR
0.060/0.190
0.460/0.396
0.161/0.201
Normo/hyperd
Baseline HOMA-IR
0.146/0.043
  
Follow-up hsCRP
0.483/0.455
0.034/0.064
 
Follow-up HOMA-IR
0.129/0.175
0.340/0.437
0.121/0.268
Covariates included in the models were race, sex, age, BMI, smoking and alcohol use for the total sample and Normo/Hyper groups, with additional adjustment for follow-up years for baseline-follow-up correlations
hsCRP, high-sensitivity C-reactive protein; HOMA-IR, homeostasis model assessment for insulin resistance; Normo, normoglycemia in the outcome survey; Hyper, hyperglycemia in the outcome survey
aCorrelation coefficients greater than 0.087 are significant (p < 0.05)
bCorrelation coefficients greater than 0.103 for whites and 0.161 for blacks are significant (p < 0.05)
cCorrelation coefficients greater than 0.148 for males, and 0.108 for females are significant (p < 0.05)
dCorrelation coefficients greater than 0.118 for normoglycemia, and 0.129 for hyperglycemia are significant (p < 0.05)
* p < 0.05 for race difference

Cross-lagged path analysis of hsCRP and HOMA-IR

After adjusting for race, sex, age, BMI, smoking, alcohol drinking, and follow-up years, the path coefficient from baseline hsCRP to follow-up HOMA-IR (β2 = 0.105, p = 0.009) was greater than the path coefficient from baseline HOMA-IR to follow-up hsCRP (β1 = 0.005, p = 0.903), with p = 0.011 for the difference between β1 and β2. The tracking correlations of hsCRP and HOMA-IR from baseline to follow-up and the synchronous correlation between hsCRP and HOMA-IR at baseline were significant. The variance (R2) of follow-up hsCRP (0.224) and follow-up HOMA-IR (0.186) explained by baseline predictors was significant. RMR and CFI were 0.037 and 0.960, respectively, indicating a good fit to the observed data according to the criteria of RMR < 0.05 and CFI > 0.90 (Fig. 1).

Cross-lagged path analyses by subgroups

The path coefficients (β1) of baseline HOMA-IR → follow-up hsCRP in all subgroups were nonsignificant (p > 0.05). The path coefficients (β2) of baseline hsCRP → follow-up HOMA-IR in subgroups of hyperglycemia (β2 = 0.156, p = 0.008), pre-T2DM (β2 = 0.177, p = 0.012), whites (β2 = 0.108, p = 0.023), females (β2 = 0.138, p = 0.006) and follow-up years above median (β2 = 0.199, p < 0.001) were significant. Significant differences in the path coefficients (β1 and β2) were noted only between normoglycemia and T2DM groups and between follow-up period groups. The model fitting parameters of RMR ranged from 0.002 to 0.148 and CFI from 0.862 to 1.000, indicating a relatively good fit to the observed data (Table 3). The path coefficient of baseline hsCRP → follow-up insulin (β2 = 0.107, p = 0.008) was greater than the path of baseline insulin → follow-up hsCRP (β1 = − 0.001, p = 0.998), with p = 0.004 for the difference between β1 and β2 (Additional file 1: Fig. S1). In contrast, neither the path coefficient of hsCRP → glucose (β2 = 0.040, p = 0.339) nor the path of glucose → hsCRP (β1 = − 0.025, p = 0.520) was significant. In addition, the path coefficient of hsCRP → insulin (β2 = 0.152, p = 0.009) was significant in the hyperglycemia group, but not significant in the normoglycemia group (β2 = 0.088, p = 0.121) (Additional file 1: Fig. S2).
Table 3
Cross-lagged path coefficients between log-transformed hsCRP and HOMA-IR by glycemia status, race, sex, and follow-up period
 
Path coefficients
Goodness-of-fit
HOMA-IR → hsCRP
hsCRP → HOMA-IR
pa
RMR
CFI
β1
p
β2
p
Glycemia status
 Normoglycemia (n = 278)
− 0.037
0.483
0.081
0.155
0.032
0.022
0.990
 Hyperglycemia (n = 231)
0.045
0.449
0.156
0.008
0.059
0.054
0.914
 T2DM (n = 68)
0.131
0.206
0.143
0.199
0.911
0.031
0.994
 Pre-T2DM (n = 163)
0.006
0.938
0.177
0.012
0.015
0.061
0.884
 pb
0.141
 
0.194
    
 pc
0.011
 
0.380
    
 pd
0.475
 
0.121
    
Race
 White (n = 360)
0.029
0.530
0.108
0.023
0.111
0.097
0.932
 Black (n = 149)
− 0.052
0.490
0.120
0.117
0.022
0.148
1.000
 pe
0.148
 
0.843
    
Sex
 Male (n = 176)
0.013
0.848
0.044
0.518
0.645
0.104
0.928
 Female (n = 333)
− 0.001
0.986
0.138
0.006
0.005
0.111
0.962
 pf
0.801
 
0.095
    
Follow-up period
 Below median (n = 245)
− 0.018
0.756
0.035
0.560
0.352
0.067
0.862
 Above median (n = 264)
0.026
0.627
0.199
< 0.001
< 0.001
0.002
0.981
 pg
0.885
 
0.003
    
β, standardized regression coefficient; RMR, root mean square residual; CFI, comparative fit index; hsCRP, high-sensitivity C-reactive protein; HOMA-IR, homeostasis model assessment for insulin resistance; T2DM, type 2 diabetes mellitus
ap value for difference between β1 and β2
bp value for difference in β1 and β2 between hyperglycemia and normoglycemia groups
cp value for difference in β1 and β2 between T2DM and normoglycemia groups
dp value for difference in β1 and β2 between pre-T2DM and normoglycemia groups
ep value for difference in β1 and β2 between whites and blacks
fp value for difference in β1 and β2 between males and females
gp value for difference in β1 and β2 between follow-up period groups (median = 6.7 years)

Joint effect of inflammation and insulin resistance on hyperglycemia

The percentages of T2DM (p = 0.010), pre-T2DM (p = 0.006) and hyperglycemia (p = 0.001) in the outcome survey were significantly greater among participants with high hsCRP at baseline and high HOMA-IR at follow-up (high–high group) than among participants with low hsCRP at baseline and low HOMA-IR at follow-up (low–low group) (Fig. 2). After adjusting for race, sex, age, BMI, smoking, and alcohol drinking, participants in the high–high group had greater risks of T2DM (OR = 2.38, 95% CI 1.06–5.53), pre-T2DM (OR = 2.27, 95% CI 1.27–4.05) and hyperglycemia in the outcome survey (OR = 2.18, 95% CI 1.30–3.64) than those in the low–low group (Table 4).
Table 4
ORs for T2DM, Pre-T2DM, and hyperglycemia in the outcome survey associated with the high–high group with the low–low group as reference, adjusting for covariates
Independent variable
Outcome
T2DM (37 vs 155)
Pre-T2DM (89 vs 155)
Hyperglycemia (126 vs 155)
OR (95% CI)
p
OR (95% CI)
p
OR (95% CI)
p
Black race
1.14 (0.50–2.63)
0.758
0.98 (0.51–1.90)
0.953
1.07 (0.60–1.91)
0.823
Female sex
0.72 (0.30–1.73)
0.467
0.39 (0.21–0.72)
0.003
0.49 (0.28–0.84)
0.010
Age
1.05 (0.96–1.16)
0.286
1.10 (1.02–1.17)
0.010
1.08 (1.02–1.15)
0.011
BMI
1.07 (1.01–1.12)
0.014
1.09 (1.05–1.14)
< 0.001
1.08 (1.04–1.12)
< 0.001
Smoking
1.74 (0.71–4.25)
0.224
1.17 (0.59–2.32)
0.647
1.28 (0.70–2.34)
0.419
Alcohol drinking
0.30 (0.11–0.86)
0.025
1.06 (0.57–1.96)
0.855
0.77 (0.44–1.36)
0.367
Groupa
2.38 (1.06–5.33)
0.035
2.27 (1.27–4.05)
0.006
2.18 (1.30–3.64)
0.003
High and low levels of baseline hsCRP and follow-up HOMA-IR were defined by their medians
OR, odds ratio; CI, confidence interval; T2DM, type 2 diabetes mellitus; BMI, body mass index; hsCRP, high-sensitivity C-reactive protein; HOMA-IR, homeostasis model assessment for insulin resistance
aCoding: high–high group = 1; low–low group = 0

Discussion

Principal findings

This community-based longitudinal cohort study of black and white adults examined the temporal relationship between inflammation measured by hsCRP and insulin resistance estimated by HOMA-IR in a community-based cohort using a cross-lagged path analysis model, a statistical approach to dissecting a causal relationship between inter-correlated variables [26, 27]. We found that higher hsCRP preceded HOMA-IR rather than vice versa in non-diabetic adults, and this one-directional relationship was found to be significant in pre-T2DM and hyperglycemia groups but not in the normoglycemia group; high levels of baseline hsCRP and follow-up HOMA-IR were significantly associated with increased risks of T2DM, pre-T2DM and hyperglycemia. The findings suggest that hsCRP levels are associated with subsequent HOMA-IR in non-diabetic adults, and their joint effect predicts the development of T2DM.

Temporal relationship between inflammation and insulin resistance

Inflammation and insulin resistance are two well-known mechanisms linking obesity, T2DM and cardiovascular disease [29, 30]. Multiple inflammation markers including hsCRP are correlated with hyperglycemia and insulin resistance in majority of previous studies [13, 21, 3136]. An important question raised by these observations is whether adiposity-induced inflammation reaction precedes insulin resistance or vice versa, or whether this relationship is bidirectional, especially in the evolution of T2DM. Cross-sectional study design limits the inference regarding the temporal relationship between hsCRP and insulin resistance. Several longitudinal studies have attempted to demonstrate the temporal relationship between inflammation, insulin resistance, and T2DM [10, 11, 18, 19]. Multiple inflammatory markers, including hsCRP, interleukin-6, and plasminogen activator inhibitor-1, have been shown to independently predict incident T2DM [10, 11]. The Coronary Artery Risk Development in Young Adults study showed that elevated hsCRP was associated with concurrent and future insulin resistance after accounting for adiposity and oxidative stress markers [18]. To date, only one longitudinal study has examined the bidirectional relationship between inflammation and insulin resistance and showed that baseline levels of hsCRP and interleukin-6 were positively associated with subsequent increases in fasting insulin, HOMA-IR, and beta-cell function. In the reverse analysis, baseline HOMA-IR was not associated with change in inflammation biomarkers [19]. However, these traditional longitudinal analysis models cannot address the causal relationship. The present study confirmed and extended previous findings by showing that higher hsCRP preceded insulin resistance among non-diabetic adults using a cross-lagged path analysis model. This one-directional temporal relationship was observed only in participants who had hyperglycemia but not in normoglycemic adults. In addition to HOMA-IR, we found that hsCRP elevation preceded increased fasting insulin but not for fasting glucose, suggesting that insulin levels contribute more than glucose to the HOMA-IR index to measure insulin resistance, and fasting insulin alone may be a simple and effective surrogate measure of insulin resistance [37]. The causal relationship between hsCRP and insulin resistance may account for the observation that anti-inflammatory treatments can improve glycemia and insulin resistance among diabetic patients [38, 39].
The mechanisms linking inflammation to insulin resistance are incompletely elucidated. Existing data support the concept that the temporal relationship between chronic inflammation and insulin resistance is bidirectional like a “two-way street”, i.e. they are mutually influenced based on the underlying metabolic and physiologic mechanisms [3, 15]. Previous studies suggest that inflammation may promote insulin resistance through the production of proinflammatory cytokines such as interleukin-1β, interacting with adipose tissue-specific macrophages and activation of innate immune system [3, 15, 40]. In addition, inhibition of inflammatory signaling by knockout of key pathways in obese mice can directly prevent the development of insulin resistance [41, 42]. On the other hand, experimental studies in mice have suggested that insulin resistance may cause inflammation through the production of the chemokine monocyte protein 1 [16]. It is well-known that inflammation and insulin resistance are linked through obesity [14]. In the present observational population study, the results from the cross-lagged analyses indicated that elevated hsCRP occurred prior to increased HOMA-IR independent of BMI in non-diabetic participants, but the reverse was not true. The finding of this one-directional time sequence from our study provided a necessary first step in establishing causation between systemic inflammation and insulin resistance. Further large and well-controlled prospective clinical trials targeting inflammatory pathways for its treatment are warranted [43].
Several studies have reported significant sex and black-white differences in the cross-sectional association between hsCRP and HOMA-IR [4446]. In the present study, however, we did not find significant sex and race differences in the cross-sectional correlations (except for the correlation at follow-up) and longitudinal directional path parameters between hsCRP and HOMA-IR. These inconsistencies may be due to limited sample size and different age periods, and thus future studies with larger sample sizes are needed to assess the sex- and race-specific association, particularly in the temporal relationship analysis.

Joint effect of inflammation and insulin resistance on T2DM

Previous longitudinal studies have demonstrated independent effects of hsCRP and HOMA-IR in predicting the risk of T2DM [47, 48]; however, their joint effect on the development of T2DM has not been reported. In the present study, two approaches were applied to test the joint effect of hsCRP and HOMA-IR on T2DM, pre-T2DM and hyperglycemia, i.e. estimating the effect of baseline hsCRP on follow-up HOMA-IR in glycemic groups and assessing the risk of hyperglycemia of the group with high levels of baseline hsCRP and follow-up HOMA-IR with the low–low group as reference. We found that the path coefficients of baseline hsCRP → follow-up HOMA-IR were significant in the pre-T2DM and hyperglycemia groups; individuals with high levels of baseline hsCRP and follow-up HOMA-IR had significantly increased risks of T2DM, pre-T2DM and hyperglycemia compared with those who had low–low levels. The results suggest that inflammation and insulin resistance have a joint effect on the pathogenesis of hyperglycemia.

Strengths and limitations

This community-based longitudinal cohort study provides a unique opportunity to examine the temporal relationship between increased hsCRP and insulin resistance and their joint effect on hyperglycemia. However, several limitations should be noted. First, the relatively small sample size of subgroups, especially the number of T2DM patients, had a limited statistical power to detect a weak-to-moderate association. Second, we assessed chronic inflammation state using hsCRP, which is a downstream inflammatory marker. Further studies are required involving a wider spectrum of inflammatory biomarkers such as fibrinogen, interleukin 6, and tumor necrosis factor-α. Third, T2DM was diagnosed based on fasting blood glucose and anti-diabetic medication history in this study without 2 h blood glucose data, which may lead to missed diagnosis.

Conclusions

This study demonstrates that elevated CRP is associated with future insulin resistance in non-diabetic adults, and their joint effect is predictive of the development of T2DM. The findings will improve our understanding of the pathobiology, mechanisms, and natural history of T2DM. Interventional studies are warranted regarding whether anti-inflammatory therapy is an effective strategy for reducing the risk of insulin resistance and T2DM.

Acknowledgements

We thank all the investigators and staff members who contributed to the Bogalusa Heart Study. We especially thank the children and adults who have participated in this study for many years.
Study protocols were approved by the Institutional Review Board of the Tulane University Health Sciences Center and written informed consent was obtained from all study participants.
All authors have reviewed the final version of the manuscript and approve of its publication.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Duncan BB, Schmidt MI, Pankow JS, Ballantyne CM, Couper D, Vigo A, Hoogeveen R, Folsom AR, Heiss G. Low-grade systemic inflammation and the development of type 2 diabetes: the atherosclerosis risk in communities study. Diabetes. 2003;52(7):1799–805.PubMedCrossRef Duncan BB, Schmidt MI, Pankow JS, Ballantyne CM, Couper D, Vigo A, Hoogeveen R, Folsom AR, Heiss G. Low-grade systemic inflammation and the development of type 2 diabetes: the atherosclerosis risk in communities study. Diabetes. 2003;52(7):1799–805.PubMedCrossRef
2.
Zurück zum Zitat Donath MY. Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat Rev Drug Discov. 2014;13(6):465–76.PubMedCrossRef Donath MY. Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat Rev Drug Discov. 2014;13(6):465–76.PubMedCrossRef
3.
Zurück zum Zitat Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107.PubMedCrossRef Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107.PubMedCrossRef
4.
Zurück zum Zitat Ebtehaj S, Gruppen EG, Parvizi M, Tietge U, Dullaart R. The anti-inflammatory function of HDL is impaired in type 2 diabetes: role of hyperglycemia, paraoxonase-1 and low grade inflammation. Cardiovasc Diabetol. 2017;16(1):132.PubMedPubMedCentralCrossRef Ebtehaj S, Gruppen EG, Parvizi M, Tietge U, Dullaart R. The anti-inflammatory function of HDL is impaired in type 2 diabetes: role of hyperglycemia, paraoxonase-1 and low grade inflammation. Cardiovasc Diabetol. 2017;16(1):132.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Kaptoge S, Di Angelantonio E, Lowe G, Pepys MB, Thompson SG, Collins R, Danesh J. C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet. 2010;375(9709):132–40.PubMedCrossRef Kaptoge S, Di Angelantonio E, Lowe G, Pepys MB, Thompson SG, Collins R, Danesh J. C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet. 2010;375(9709):132–40.PubMedCrossRef
6.
Zurück zum Zitat Bao X, Borne Y, Johnson L, Muhammad IF, Persson M, Niu K, Engstrom G. Comparing the inflammatory profiles for incidence of diabetes mellitus and cardiovascular diseases: a prospective study exploring the ‘common soil’ hypothesis. Cardiovasc Diabetol. 2018;17(1):87.PubMedPubMedCentralCrossRef Bao X, Borne Y, Johnson L, Muhammad IF, Persson M, Niu K, Engstrom G. Comparing the inflammatory profiles for incidence of diabetes mellitus and cardiovascular diseases: a prospective study exploring the ‘common soil’ hypothesis. Cardiovasc Diabetol. 2018;17(1):87.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Ridker PM, MacFadyen JG, Everett BM, Libby P, Thuren T, Glynn RJ. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial. Lancet. 2018;391(10118):319–28.PubMedCrossRef Ridker PM, MacFadyen JG, Everett BM, Libby P, Thuren T, Glynn RJ. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial. Lancet. 2018;391(10118):319–28.PubMedCrossRef
8.
Zurück zum Zitat Yang W, Li Y, Wang JY, Han R, Wang L. Circulating levels of adipose tissue-derived inflammatory factors in elderly diabetes patients with carotid atherosclerosis: a retrospective study. Cardiovasc Diabetol. 2018;17(1):75.PubMedPubMedCentralCrossRef Yang W, Li Y, Wang JY, Han R, Wang L. Circulating levels of adipose tissue-derived inflammatory factors in elderly diabetes patients with carotid atherosclerosis: a retrospective study. Cardiovasc Diabetol. 2018;17(1):75.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Herder C, Kannenberg JM, Carstensen-Kirberg M, Huth C, Meisinger C, Koenig W, Peters A, Rathmann W, Roden M, Thorand B. Serum levels of interleukin-22, cardiometabolic risk factors and incident type 2 diabetes: KORA F4/FF4 study. Cardiovasc Diabetol. 2017;16(1):17.PubMedPubMedCentralCrossRef Herder C, Kannenberg JM, Carstensen-Kirberg M, Huth C, Meisinger C, Koenig W, Peters A, Rathmann W, Roden M, Thorand B. Serum levels of interleukin-22, cardiometabolic risk factors and incident type 2 diabetes: KORA F4/FF4 study. Cardiovasc Diabetol. 2017;16(1):17.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat 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(3):327–34.PubMedCrossRef 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(3):327–34.PubMedCrossRef
11.
Zurück zum Zitat Wang X, Bao W, Liu J, Ouyang YY, Wang D, Rong S, Xiao X, Shan ZL, Zhang Y, Yao P, et al. Inflammatory markers and risk of type 2 diabetes: a systematic review and meta-analysis. Diabetes Care. 2013;36(1):166–75.PubMedCrossRef Wang X, Bao W, Liu J, Ouyang YY, Wang D, Rong S, Xiao X, Shan ZL, Zhang Y, Yao P, et al. Inflammatory markers and risk of type 2 diabetes: a systematic review and meta-analysis. Diabetes Care. 2013;36(1):166–75.PubMedCrossRef
12.
Zurück zum Zitat Festa A, D’Agostino R, Howard G, Mykkänen L, Tracy RP, Haffner SM. Chronic subclinical inflammation as part of the insulin resistance syndrome: the Insulin Resistance Atherosclerosis Study (IRAS). Circulation. 2000;102(1):42–7.PubMedCrossRef Festa A, D’Agostino R, Howard G, Mykkänen L, Tracy RP, Haffner SM. Chronic subclinical inflammation as part of the insulin resistance syndrome: the Insulin Resistance Atherosclerosis Study (IRAS). Circulation. 2000;102(1):42–7.PubMedCrossRef
13.
Zurück zum Zitat Wu T, Dorn JP, Donahue RP, Sempos CT, Trevisan M. Associations of serum C-reactive protein with fasting insulin, glucose, and glycosylated hemoglobin: the Third National Health and Nutrition Examination Survey, 1988–1994. Am J Epidemiol. 2002;155(1):65–71.PubMedCrossRef Wu T, Dorn JP, Donahue RP, Sempos CT, Trevisan M. Associations of serum C-reactive protein with fasting insulin, glucose, and glycosylated hemoglobin: the Third National Health and Nutrition Examination Survey, 1988–1994. Am J Epidemiol. 2002;155(1):65–71.PubMedCrossRef
14.
Zurück zum Zitat Kahn SE, Zinman B, Haffner SM, O’Neill MC, Kravitz BG, Yu D, Freed MI, Herman WH, Holman RR, Jones NP, et al. Obesity is a major determinant of the association of C-reactive protein levels and the metabolic syndrome in type 2 diabetes. Diabetes. 2006;55(8):2357–64.PubMedCrossRef Kahn SE, Zinman B, Haffner SM, O’Neill MC, Kravitz BG, Yu D, Freed MI, Herman WH, Holman RR, Jones NP, et al. Obesity is a major determinant of the association of C-reactive protein levels and the metabolic syndrome in type 2 diabetes. Diabetes. 2006;55(8):2357–64.PubMedCrossRef
15.
Zurück zum Zitat Chen L, Chen R, Wang H, Liang F. Mechanisms linking inflammation to insulin resistance. Int J Endocrinol. 2015;2015:508409.PubMedPubMedCentral Chen L, Chen R, Wang H, Liang F. Mechanisms linking inflammation to insulin resistance. Int J Endocrinol. 2015;2015:508409.PubMedPubMedCentral
16.
Zurück zum Zitat Shimobayashi M, Albert V, Woelnerhanssen B, Frei IC, Weissenberger D, Meyer-Gerspach AC, Clement N, Moes S, Colombi M, Meier JA, et al. Insulin resistance causes inflammation in adipose tissue. J Clin Invest. 2018;128(4):1538–50.PubMedPubMedCentralCrossRef Shimobayashi M, Albert V, Woelnerhanssen B, Frei IC, Weissenberger D, Meyer-Gerspach AC, Clement N, Moes S, Colombi M, Meier JA, et al. Insulin resistance causes inflammation in adipose tissue. J Clin Invest. 2018;128(4):1538–50.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Savage DB, Petersen KF, Shulman GI. Mechanisms of insulin resistance in humans and possible links with inflammation. Hypertension. 2005;45(5):828–33.PubMedCrossRef Savage DB, Petersen KF, Shulman GI. Mechanisms of insulin resistance in humans and possible links with inflammation. Hypertension. 2005;45(5):828–33.PubMedCrossRef
18.
Zurück zum Zitat Park K, Steffes M, Lee DH, Himes JH, Jacobs DR. Association of inflammation with worsening HOMA-insulin resistance. Diabetologia. 2009;52(11):2337–44.PubMedPubMedCentralCrossRef Park K, Steffes M, Lee DH, Himes JH, Jacobs DR. Association of inflammation with worsening HOMA-insulin resistance. Diabetologia. 2009;52(11):2337–44.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Herder C, Færch K, Carstensen-Kirberg M, Lowe GD, Haapakoski R, Witte DR, Brunner EJ, Roden M, Tabák AG, Kivimäki M, et al. Biomarkers of subclinical inflammation and increases in glycaemia, insulin resistance and beta-cell function in non-diabetic individuals: the Whitehall II study. Eur J Endocrinol. 2016;175(5):367–77.PubMedCrossRef Herder C, Færch K, Carstensen-Kirberg M, Lowe GD, Haapakoski R, Witte DR, Brunner EJ, Roden M, Tabák AG, Kivimäki M, et al. Biomarkers of subclinical inflammation and increases in glycaemia, insulin resistance and beta-cell function in non-diabetic individuals: the Whitehall II study. Eur J Endocrinol. 2016;175(5):367–77.PubMedCrossRef
20.
Zurück zum Zitat Uemura H, Katsuura-Kamano S, Yamaguchi M, Bahari T, Ishizu M, Fujioka M, Arisawa K. Relationships of serum high-sensitivity C-reactive protein and body size with insulin resistance in a Japanese cohort. PLoS ONE. 2017;12(6):e0178672.PubMedPubMedCentralCrossRef Uemura H, Katsuura-Kamano S, Yamaguchi M, Bahari T, Ishizu M, Fujioka M, Arisawa K. Relationships of serum high-sensitivity C-reactive protein and body size with insulin resistance in a Japanese cohort. PLoS ONE. 2017;12(6):e0178672.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Gala T, Herder C, Rutters F, Carstensen-Kirberg M, Huth C, Stehouwer C, Nijpels G, Schalkwijk C, Flyvbjerg A, Franks PW, et al. Association of changes in inflammation with variation in glycaemia, insulin resistance and secretion based on the KORA study. Diabetes Metab Res Rev. 2018;34(8):e3063.CrossRef Gala T, Herder C, Rutters F, Carstensen-Kirberg M, Huth C, Stehouwer C, Nijpels G, Schalkwijk C, Flyvbjerg A, Franks PW, et al. Association of changes in inflammation with variation in glycaemia, insulin resistance and secretion based on the KORA study. Diabetes Metab Res Rev. 2018;34(8):e3063.CrossRef
22.
Zurück zum Zitat Brunner EJ, Kivimäki M, Witte DR, Lawlor DA, Davey Smith G, Cooper JA, Miller M, Lowe GD, Rumley A, Casas JP, et al. Inflammation, insulin resistance, and diabetes–Mendelian randomization using CRP haplotypes points upstream. PLoS Med. 2008;5(8):e155.PubMedPubMedCentralCrossRef Brunner EJ, Kivimäki M, Witte DR, Lawlor DA, Davey Smith G, Cooper JA, Miller M, Lowe GD, Rumley A, Casas JP, et al. Inflammation, insulin resistance, and diabetes–Mendelian randomization using CRP haplotypes points upstream. PLoS Med. 2008;5(8):e155.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Noordam R, Oudt CH, Bos MM, Smit R, van Heemst D. High-sensitivity C-reactive protein, low-grade systemic inflammation and type 2 diabetes mellitus: a two-sample Mendelian randomization study. Nutr Metab Cardiovasc Dis. 2018;28(8):795–802.PubMedCrossRef Noordam R, Oudt CH, Bos MM, Smit R, van Heemst D. High-sensitivity C-reactive protein, low-grade systemic inflammation and type 2 diabetes mellitus: a two-sample Mendelian randomization study. Nutr Metab Cardiovasc Dis. 2018;28(8):795–802.PubMedCrossRef
24.
Zurück zum Zitat Berenson GS. Bogalusa Heart Study: a long-term community study of a rural biracial (black/white) population. Am J Med Sci. 2001;322(5):267–74.PubMedCrossRef Berenson GS. Bogalusa Heart Study: a long-term community study of a rural biracial (black/white) population. Am J Med Sci. 2001;322(5):267–74.PubMedCrossRef
25.
Zurück zum Zitat Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985;28(7):412–9.PubMedCrossRef Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985;28(7):412–9.PubMedCrossRef
26.
Zurück zum Zitat Kenny DA. Cross-lagged panel correlation: a test for spuriousness. Psychol Bull. 1975;82:887–903.CrossRef Kenny DA. Cross-lagged panel correlation: a test for spuriousness. Psychol Bull. 1975;82:887–903.CrossRef
27.
Zurück zum Zitat Kivimäki M, Feldt T, Vahtera J, Nurmi JE. Sense of coherence and health: evidence from two cross-lagged longitudinal samples. Soc Sci Med. 2000;50(4):583–97.PubMedCrossRef Kivimäki M, Feldt T, Vahtera J, Nurmi JE. Sense of coherence and health: evidence from two cross-lagged longitudinal samples. Soc Sci Med. 2000;50(4):583–97.PubMedCrossRef
28.
Zurück zum Zitat Joreskog K, Sorbom D. LISREL 8.52: User’s Reference Guide. 2001. Chicago. Scientifc Software International. Joreskog K, Sorbom D. LISREL 8.52: User’s Reference Guide. 2001. Chicago. Scientifc Software International.
29.
Zurück zum Zitat Ormazabal V, Nair S, Elfeky O, Aguayo C, Salomon C, Zuniga FA. Association between insulin resistance and the development of cardiovascular disease. Cardiovasc Diabetol. 2018;17(1):122.PubMedPubMedCentralCrossRef Ormazabal V, Nair S, Elfeky O, Aguayo C, Salomon C, Zuniga FA. Association between insulin resistance and the development of cardiovascular disease. Cardiovasc Diabetol. 2018;17(1):122.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Van Gaal LF, Mertens IL, De Block CE. Mechanisms linking obesity with cardiovascular disease. Nature. 2006;444(7121):875–80.PubMedCrossRef Van Gaal LF, Mertens IL, De Block CE. Mechanisms linking obesity with cardiovascular disease. Nature. 2006;444(7121):875–80.PubMedCrossRef
31.
Zurück zum Zitat Natali A, Toschi E, Baldeweg S, Ciociaro D, Favilla S, Saccà L, Ferrannini E. Clustering of insulin resistance with vascular dysfunction and low-grade inflammation in type 2 diabetes. Diabetes. 2006;55(4):1133–40.PubMedCrossRef Natali A, Toschi E, Baldeweg S, Ciociaro D, Favilla S, Saccà L, Ferrannini E. Clustering of insulin resistance with vascular dysfunction and low-grade inflammation in type 2 diabetes. Diabetes. 2006;55(4):1133–40.PubMedCrossRef
32.
Zurück zum Zitat Meng YX, Ford ES, Li C, Quarshie A, Al-Mahmoud AM, Giles W, Gibbons GH, Strayhorn G. Association of C-reactive protein with surrogate measures of insulin resistance among nondiabetic US from National Health and Nutrition Examination Survey 1999–2002. Clin Chem. 2007;53(12):2152–9.PubMedCrossRef Meng YX, Ford ES, Li C, Quarshie A, Al-Mahmoud AM, Giles W, Gibbons GH, Strayhorn G. Association of C-reactive protein with surrogate measures of insulin resistance among nondiabetic US from National Health and Nutrition Examination Survey 1999–2002. Clin Chem. 2007;53(12):2152–9.PubMedCrossRef
33.
Zurück zum Zitat Deepa R, Velmurugan K, Arvind K, Sivaram P, Sientay C, Uday S, Mohan V. Serum levels of interleukin 6, C-reactive protein, vascular cell adhesion molecule 1, and monocyte chemotactic protein 1 in relation to insulin resistance and glucose intolerance–the Chennai Urban Rural Epidemiology Study (CURES). Metabolism. 2006;55(9):1232–8.PubMedCrossRef Deepa R, Velmurugan K, Arvind K, Sivaram P, Sientay C, Uday S, Mohan V. Serum levels of interleukin 6, C-reactive protein, vascular cell adhesion molecule 1, and monocyte chemotactic protein 1 in relation to insulin resistance and glucose intolerance–the Chennai Urban Rural Epidemiology Study (CURES). Metabolism. 2006;55(9):1232–8.PubMedCrossRef
34.
Zurück zum Zitat Bahceci M, Tuzcu A, Ogun C, Canoruc N, Iltimur K, Aslan C. Is serum C-reactive protein concentration correlated with HbA1c and insulin resistance in Type 2 diabetic men with or without coronary heart disease. J Endocrinol Invest. 2005;28(2):145–50.PubMedCrossRef Bahceci M, Tuzcu A, Ogun C, Canoruc N, Iltimur K, Aslan C. Is serum C-reactive protein concentration correlated with HbA1c and insulin resistance in Type 2 diabetic men with or without coronary heart disease. J Endocrinol Invest. 2005;28(2):145–50.PubMedCrossRef
35.
Zurück zum Zitat Hwu CM, Lin YC, Lin MW. High levels of C-reactive protein are positively associated with isolated postchallenge hyperglycemia in postmenopausal women. Metab Syndr Relat Disord. 2016;14(7):334–9.PubMedCrossRef Hwu CM, Lin YC, Lin MW. High levels of C-reactive protein are positively associated with isolated postchallenge hyperglycemia in postmenopausal women. Metab Syndr Relat Disord. 2016;14(7):334–9.PubMedCrossRef
36.
Zurück zum Zitat Carstensen-Kirberg M, Kannenberg JM, Huth C, Meisinger C, Koenig W, Heier M, Peters A, Rathmann W, Roden M, Herder C, et al. Inverse associations between serum levels of secreted frizzled-related protein-5 (SFRP5) and multiple cardiometabolic risk factors: KORA F4 study. Cardiovasc Diabetol. 2017;16(1):109.PubMedPubMedCentralCrossRef Carstensen-Kirberg M, Kannenberg JM, Huth C, Meisinger C, Koenig W, Heier M, Peters A, Rathmann W, Roden M, Herder C, et al. Inverse associations between serum levels of secreted frizzled-related protein-5 (SFRP5) and multiple cardiometabolic risk factors: KORA F4 study. Cardiovasc Diabetol. 2017;16(1):109.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Laakso M. How good a marker is insulin level for insulin resistance. Am J Epidemiol. 1993;137(9):959–65.PubMedCrossRef Laakso M. How good a marker is insulin level for insulin resistance. Am J Epidemiol. 1993;137(9):959–65.PubMedCrossRef
38.
Zurück zum Zitat Larsen CM, Faulenbach M, Vaag A, Vølund A, Ehses JA, Seifert B, Mandrup-Poulsen T, Donath MY. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356(15):1517–26.PubMedCrossRef Larsen CM, Faulenbach M, Vaag A, Vølund A, Ehses JA, Seifert B, Mandrup-Poulsen T, Donath MY. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356(15):1517–26.PubMedCrossRef
39.
Zurück zum Zitat Goldfine AB, Silver R, Aldhahi W, Cai D, Tatro E, Lee J, Shoelson SE. Use of salsalate to target inflammation in the treatment of insulin resistance and type 2 diabetes. Clin Transl Sci. 2008;1(1):36–43.PubMedPubMedCentralCrossRef Goldfine AB, Silver R, Aldhahi W, Cai D, Tatro E, Lee J, Shoelson SE. Use of salsalate to target inflammation in the treatment of insulin resistance and type 2 diabetes. Clin Transl Sci. 2008;1(1):36–43.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Robertson RP, Harmon J, Tran PO, Poitout V. Beta-cell glucose toxicity, lipotoxicity, and chronic oxidative stress in type 2 diabetes. Diabetes. 2004;53(Suppl 1):S119–24.PubMedCrossRef Robertson RP, Harmon J, Tran PO, Poitout V. Beta-cell glucose toxicity, lipotoxicity, and chronic oxidative stress in type 2 diabetes. Diabetes. 2004;53(Suppl 1):S119–24.PubMedCrossRef
41.
Zurück zum Zitat Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest. 2006;116(11):3015–25.PubMedPubMedCentralCrossRef Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest. 2006;116(11):3015–25.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Hirosumi J, Tuncman G, Chang L, Görgün CZ, Uysal KT, Maeda K, Karin M, Hotamisligil GS. A central role for JNK in obesity and insulin resistance. Nature. 2002;420(6913):333–6.PubMedCrossRef Hirosumi J, Tuncman G, Chang L, Görgün CZ, Uysal KT, Maeda K, Karin M, Hotamisligil GS. A central role for JNK in obesity and insulin resistance. Nature. 2002;420(6913):333–6.PubMedCrossRef
43.
Zurück zum Zitat Tenenbaum A, Fisman EZ. Mirroring the CANTOS revolution: is anti-inflammatory therapy for diabetes just around the corner. Cardiovasc Diabetol. 2017;16(1):91.PubMedPubMedCentralCrossRef Tenenbaum A, Fisman EZ. Mirroring the CANTOS revolution: is anti-inflammatory therapy for diabetes just around the corner. Cardiovasc Diabetol. 2017;16(1):91.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Lai MM, Li CI, Kardia SL, Liu CS, Lin WY, Lee YD, Chang PC, Lin CC, Li TC. Sex difference in the association of metabolic syndrome with high sensitivity C-reactive protein in a Taiwanese population. BMC Public Health. 2010;10:429.PubMedPubMedCentralCrossRef Lai MM, Li CI, Kardia SL, Liu CS, Lin WY, Lee YD, Chang PC, Lin CC, Li TC. Sex difference in the association of metabolic syndrome with high sensitivity C-reactive protein in a Taiwanese population. BMC Public Health. 2010;10:429.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Hyatt TC, Phadke RP, Hunter GR, Bush NC, Muñoz AJ, Gower BA. Insulin sensitivity in African-American and white women: association with inflammation. Obesity (Silver Spring). 2009;17(2):276–82.CrossRef Hyatt TC, Phadke RP, Hunter GR, Bush NC, Muñoz AJ, Gower BA. Insulin sensitivity in African-American and white women: association with inflammation. Obesity (Silver Spring). 2009;17(2):276–82.CrossRef
46.
Zurück zum Zitat 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(9):3251–8.PubMedPubMedCentralCrossRef 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(9):3251–8.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Martin BC, Warram JH, Krolewski AS, Bergman RN, Soeldner JS, Kahn CR. Role of glucose and insulin resistance in development of type 2 diabetes mellitus: results of a 25-year follow-up study. Lancet. 1992;340(8825):925–9.PubMedCrossRef Martin BC, Warram JH, Krolewski AS, Bergman RN, Soeldner JS, Kahn CR. Role of glucose and insulin resistance in development of type 2 diabetes mellitus: results of a 25-year follow-up study. Lancet. 1992;340(8825):925–9.PubMedCrossRef
48.
Zurück zum Zitat Effoe VS, Correa A, Chen H, Lacy ME, Bertoni AG. High-sensitivity C-Reactive protein is associated with incident type 2 diabetes among African Americans: the Jackson Heart Study. Diabetes Care. 2015;38(9):1694–700.PubMedPubMedCentralCrossRef Effoe VS, Correa A, Chen H, Lacy ME, Bertoni AG. High-sensitivity C-Reactive protein is associated with incident type 2 diabetes among African Americans: the Jackson Heart Study. Diabetes Care. 2015;38(9):1694–700.PubMedPubMedCentralCrossRef
Metadaten
Titel
Temporal relationship between inflammation and insulin resistance and their joint effect on hyperglycemia: the Bogalusa Heart Study
verfasst von
Yinkun Yan
Shengxu Li
Yang Liu
Lydia Bazzano
Jiang He
Jie Mi
Wei Chen
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Cardiovascular Diabetology / Ausgabe 1/2019
Elektronische ISSN: 1475-2840
DOI
https://doi.org/10.1186/s12933-019-0913-2

Weitere Artikel der Ausgabe 1/2019

Cardiovascular Diabetology 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.