Skip to main content
Erschienen in: BMC Endocrine Disorders 1/2019

Open Access 01.12.2019 | Research article

Poor short-term glycemic control in patients with type 2 diabetes impairs the intestinal mucosal barrier: a prospective, single-center, observational study

verfasst von: Lijuan Shen, Li Ao, Haoben Xu, Junfeng Shi, Dali You, Xiuwen Yu, Weixin Xu, Jie Sun, Fei Wang

Erschienen in: BMC Endocrine Disorders | Ausgabe 1/2019

Abstract

Background

To determine the relation between daily glycemic fluturation and the intestinal mucosal barrier dysfunction in type 2 diabetes mellitus (T2DM).

Methods

Totally 66 patients with T2DM were enrolled, 33 healthy volunteers were also recruited according to the enrolled patients’ gender and age in a ratio of 2: 1. Patients were bisected by the median of endotoxins level into low(< 12.31 μ/l, n = 33) and high(≥12.31 μ/l, n = 33) blood endotoxin groups. Clinical data and blood glucose fluctuations were compared between groups. Multivariate regression analysis was used to determine the independent factors affecting the intestinal mucosal barrier.

Results

Serum endotoxin [12.1 (4.2~22.0) vs 3.2 (1.3~6.0), P < 0.001] and fasting blood glucose levels [9.8 ± 3.6 vs 5.4 ± 0.7, P < 0.001] were significantly higher in patients with T2DM than the control group. The standard deviation of blood glucose (SDBG) within 1 day [2.9 (2.0~3.3) vs. 2.1 (1.6~2.5), P = 0.012] and the largest amplitude of glycemic excursions (LAGE) [7.5 (5.4~8.9) vs. 5.9 (4.3~7.4), P = 0.034] were higher in the high endotoxin group than in the low endotoxin group. A multiple linear stepwise regression revealed a positive correlation between SDBG with endotoxin (standard partial regression coefficient = 0.255, P = 0.039).

Conclusions

T2DM patients who incapable of maintaining stable blood glucose level are at a higher risk to associated with intestinal mucosal barrier injury.
Hinweise
Lijuan Shen and Li Ao contributed equally to this work.
Abkürzungen
BMI
Body Mass Index
DAO
Diamine oxidase
DGA
Daily glucose average
FPG
Fasting Plasma Glucose
GA
glycated albumin
HbA1c
Glycosylated hemoglobin
hs-CRP
High-sensitivity C-reactive protein
LAGE
Largest amplitude of glycemic excursions
PBG
Postprandial Blood Glucose
PPGE
Postprandial glucose excursion
SDBG
Standard Deviation Of Blood Glucose
T2DM
Type 2 diabetes mellitus

Background

Type 2 diabetes mellitus (T2DM) is a long-term metabolic disorder caused by both genetic and environmental factors. The defects in insulin secretion or function (or both) can cause disorder in carbohydrates, proteins, fats, electrolytes, and water metabolism [13]. It is clinically characterized by chronic persistent hyperglycemia and high volatility. T2DM patients also tend to be associated with a series of chronic complications, such as nerve, blood vessel and gastrointestinal tract defects including intestinal mucosal barrier damage, seriously affecting the life quality [4, 5]. According to the latest statistics from the National Vital Statistics System, diabetes is the seventh of the top ten causes of death in the United States in 2016 [6].
The intestine is an important organ that should not be omitted during the treatment of DM [79]. The intestinal mucosal barrier prevents the translocation of bacteria and endotoxins into blood and lymph circulatory systems under normal physiological conditions. Dysregulation of intestinal mucosal barrier function would increase the permeability to the intestinal pathogens and endotoxins, which cause infection or inflammation [10, 11]. Previous studies suggested that the low-grade inflammatory state of patients with T2DM was possibily related to intestinal endotoxin [12, 13]. On the other hand, disorder of intestinal microflora composition is associated with the intestinal mucosal barrier damages [14, 15], which leads to nonspecific inflammatory, and in turn aggravates insulin resistance and T2DM metabolism disorders through the NF-κB pathway and JNK signal transduction pathway [1618]. Therefore, the intestinal mucosal barrier function of patients with T2DM should be carefully monitored.
Currently, levels of the serum D-lactic acid, diamine oxidase (DAO), and endotoxin, which reflect the permeability of intestinal mucosa and bacterial translocation, are used to determine the intestinal mucosal barrier function in clinical practice [19, 20]. For patients with T2DM, glycemic monitoring (including short- and long-term blood glucose level fluctuations, as well as average blood glucose levels) is substantial. Studies have shown that persistent hyperglycemia can lead to intestinal mucosal barrier damage [5], however, the relationship between fluctuation of blood glucose level and intestinal mucosal barrier damage remains unclear. Therefore, this study aimed to investigate the relationship between glycemic control and intestinal mucosal barrier dysfunction in patients with T2DM.

Methods

Subjects

We recruited 66 patients diagnosed with T2DM from September 2017 to June 2018. All patients included in the study met the diagnostic criteria of the 2018 AACE/ACE Consensus Statement: Comprehensive Management of T2DM [21]. And their eating pattern were controlled according to the suggestion of ‘Standards of Medical Care in Diabetes’ in 2014 [22]. The control group was composed of 33 healthy volunteers who were recruited in a 2:1 ratio according to the patients’ sex and age. All subjects with digestive diseases, chronic malnutrition, malignant tumors, and intestinal infections which can inhibit the intestinal mucosal barrier function within 2 weeks were excluded.
The study protocol was approved by the Ethics Review Board of Jiading District Central Hospital (2017-ZD-03). All subjects were anonymized. All subjects signed informed consent form.

Study methods

For all subjects included, the fasting venous blood sample was collected, the fasting blood glucose was examined, and the functionality of intestinal mucosal barrier of subjects were determined by the serum D-lactic acid, DAO, and endotoxin. Patients with T2DM were further divided into the low-value group (< 12.31u/l, n = 33) and high-value group (≥12.31u/l, n = 33) based on the median endotoxin levels. For the patients with T2DM, clinical characteristic were collected [DM duration, body mass index (BMI), history of hypertension, use of drugs (such as insulin and metformin etc.), smoking history, and family history of DM]. The abnormal fasting blood glucose, 2 h blood glucose level after breakfast, glycated hemoglobin (HbA1c), and glycated albumin (GA) of patients were also examined at the first day of enrollment. The daily blood glucose level (before breakfast (A), 2 h after breakfast (B), before lunch (C), 2 h after lunch (D), before dinner (E), 2 h after dinner (F), and before sleep (G)) was monitered by active blood glucose meter (Accu-Chek®, Germany) using the finger capillary blood samples. Short-term glycemic excursions, including the magnitude of postprandial glucose excursion (PPGE), the largest amplitude of glycemic excursions (LAGE), and the standard deviation of blood glucose (SDBG) within 1 day were calculated by:
PPGE = \( \frac{\left(B-A\right)+\left(D-C\right)+\left(F-E\right)}{3} \);
LAGE = D (maximum glycemic value) - A (minimum glycemic value);
SDBG =\( \sqrt{\frac{\ {\left(A-X\right)}^2+{\left(B-X\right)}^2+{\left(C-X\right)}^2+{\left(D-X\right)}^2+{\left(E-X\right)}^2+{\left(F-X\right)}^2+{\left(G-X\right)}^2}{7-1}} \);
X (glycemic average within 1 day) = \( \frac{\left(A+B+C+D+E+F+G\right)}{7} \).

Functionality of the intestinal mucosal barrier

The functionality of the intestinal mucosal barrier was determined using the DAO/lactic acid/bacterial endotoxin combined test kit (enzymatic method; Beijing Zhongsheng Jinyu Diagnostic Technology Co., Ltd.), supporting by the JY-DLT intestinal barrier function biochemical indicator analysis system. The experiments were undergone according to the protocols suggested by the manufacturer and conducted within 4 h after serum extraction.

Statistical analysis

Statistical analysis was done using the SPSS 19.0 software (SPSS Inc., Chicago, IL, USA). Normally distributed data were expressed in mean ± standard deviations (SD) and compared using Student’s t test; otherwise was indicated as Median (Q1~Q3) and compared using the non-parametric Mann-Whitney test. Numerical data were expressed in frequency and compared using χ2 test. Multiple logistic regression was conducted to identify the factors that influenced the functionality of intestinal mucosal barrier. P < 0.05 was considered statistically significant.

Results

Baseline characteristics

The serum endotoxin [12.1(4.2~22.0) vs 3.2(1.3~6.0), P < 0.001] and FPG [(9.8 ± 3.6) vs (5.4 ± 0.7), P < 0.001] in patients with T2DM were significantly higher than those in the control group. No significant difference was observed in DAO and D-lactic acid (Table 1).
Table 1
Background characteristics of particippants
 
Control groups (n = 33)
T2DM (n = 66)
Statistics
P
Male [n(%)]
19 (57.6)
38 (57.6)
1.000
Age [(Mean ± SD), years]
61.2 ± 9.6
60.9 ± 11.8
t = 0.121
0.904
BMI [(Mean ± SD), Kg/M2]
26.5 ± 5.8
25.1 ± 3.9
t = 1.417
0.160
Smoking [n(%)]
13 (39.4)
16 (24.2)
x2 = 2.438
0.118
Hypertension [n(%)]
16 (48.5)
40 (60.6)
x2 = 1.316
0.251
Diabetes history [(Mean ± SD), years]
NA
6.0 (1–14)
Diabetic complication [n(%)]
NA
31 (46.9)
Medications [n(%)]
 Insulin
NA
21 (31.8)
 Metformin
NA
32 (48.5)
 α-glucosidase inhibitor
NA
29 (43.9)
 sulfonylureas
NA
9 (13.6)
 glinide
NA
5 (7.6)
 DPP-4 inhibitor
NA
1 (1.5)
Laboratory data
 HbAlc [M(Q1~Q3), %]
5.2 (4.9~5.6)
9.3 (7.4~11.0)
z = −7.340
< 0.001
 GA [M(Q1~Q3), %]
14.1 (13.3~14.8)
23.5 (19.4~28.5)
z = −6.751
< 0.001
 Creatinine [M(Q1~Q3), μmol/L]
76 (64.5~83)
65 (54.5~80)
z = −1.620
0.105
 GFR [M(Q1~Q3), ml/min]
90.3 (83.7~98.8)
94.9 (78.1~104.1)
z = − 0.672
0.502
 hs-CRP [M(Q1~Q3), ng/L]
3 (1.5~6.7)
3.5 (1.2~18.9)
z = −0.801
0.423
Index of the intestinal mucosal barrier
 DAO [M(Q1~Q3), u/l]
6.7 (5.4~9.3)
5.9 (4.1~9.6)
z = −1.413
0.158
 D-lactic acid [M(Q1~Q3), mg/l]
49.0 (43.1~53.3)
47.0 (37.2~54.8)
z = −0.572
0.567
 Endotoxins [M(Q1~Q3), μ/l]
3.2 (1.3~6.0)
12.1 (4.2~22.0)
z = −4.315
< 0.001
FPG[(Mean ± SD), mmol/l]
5.4 ± 0.7
9.8 ± 3.6
t = −9.070
< 0.001
HbAlc Glycosylated hemoglobin, GA glycated albumin, GFR Glomerular filtration rate, DAO diamine oxidase, hs-CRP high-sensitivity C-reactive protein, FPG:Fasting Plasma Glucose

Clinical charateristics comparation between high- and low-endotoxin groups

The SDBG [2.9(2.0~3.3) vs 2.1(1.6~2.5), P = 0.012] and LAGE [7.5(5.4~8.9) vs 5.9(4.3~7.4), P = 0.034] in the high endotoxin group were higher than those in the low endotoxin group. No significant difference was observed in the other indicators (all P > 0.05) (Table 2).
Table 2
Between the two groups of clinical data and glucose control comparison
 
Low groups (< 12.31u/l, n = 33)
High groups (12.31 ≥ u/l, n = 33)
Statistics
P
Male [n(%)]
16 (48.5)
22 (66.7)
x2 = 2.233
0.135
Age [(Mean ± SD), years]
62.3 ± 11.1
59.5 ± 12.5
t = 0.990
0.326
Hypertension [n(%)]
21 (63.6)
19 (57.6)
x2 = 0.254
0.614
Smoking [n(%)]
5 (15.2)
11 (33.3)
x2 = 2.970
0.085
Positive family history [n(%)]
7 (21.2)
7 (21.2)
1.000
BMI [(Mean ± SD), Kg/M2]
25.0 ± 4.4
25.2 ± 3.4
t = − 0.176
0.861
Diabetes history [(Mean ± SD), mouth]
9.0 (2~15.5)
5.0 (1.0~10.0)
z = −1.649
0.099
hs-CRP [M(Q1~Q3), mmol/l]
2.1 (0.5~6.6)
1.8 (0.5~6.7)
z = −0.250
0.802
Medications [n(%)]
 Insulin
11.0 (33.3)
10.0 (30.3)
x2 = 0.070
0.792
 Metformin
17.0 (51.5)
15.0 (45.5)
x2 = 0.243
0.622
 α-glucosidase inhibitor
12.0 (36.4)
17.0 (51.5)
x2 = 1.538
0.215
 Sulfonylureas
2.0 (6.1)
7.0 (21.2)
0.149
 Glinide
1.0 (3.0)
4.0 (12.1)
0.355
 DPP-4 inhibitor
0 (0.0)
1 (3.0)
1.000
Glucose control data
 FPG [M(Q1~Q3), mmol/l]
8.6 (6.8~12.9)
8.8 (7.5~11.1)
z = − 0.023
0.982
 2h PBG [M(Q1~Q3), mmol/l]
12.8 (10.6~18.1)
17.0 (15.2~20.8)
z = − 1.858
0.063
 DGA [M(Q1~Q3), mmol/l]
11.0 (9.0~15.0)
11.9 (10.1~13.7)
z = − 0.301
0.763
 HbAlc [M(Q1~Q3), %]
9.2 (8.2~10.5)
8.2 (8.2~9.4)
z = − 1.320
0.187
 GA [M(Q1~Q3), %]
23.3 (19.3~27.2)
25.8 (20.0~36.3)
z = − 1.247
0.212
Glucose fluctuations of Short-term
 SDBG [M(Q1~Q3), mmol/l]
2.1 (1.6~2.5)
2.9 (2.0~3.3)
z = − 2.520
0.012
 PPGE [M(Q1~Q3), mmol/l]
2.8 (1.8~3.1)
2.2 (1.9~3.8)
z = − 0.917
0.359
 LAGE [M(Q1~Q3), mmol/l]
5.9 (4.3~7.4)
7.5 (5.4~8.9)
z = − 2.123
0.034
 SDBG < 2.0 mmol/l [n(%)]
15.0 (45.5)
9.0 (27.3)
x2 = 2.357
0.125
 PPGE < 2.2 mmol/l [n(%)]
15.0 (45.5)
11.0 (33.3)
x2 = 1.015
0.314
 LAGE< 4.4 mmol/l [n(%)]
9.0 (27.3)
4.0 (12.1)
x2 = 2.395
0.122
DGA Daily glucose average,FPG:Fasting Plasma Glucose, PBG:Postprandial Blood Glucose, SDBG Standard Deviation Of Blood Glucose, PPGE Postprandial Glucose Excursion, LAGE Largest amplitude of glycemic excursions, BMI Body Mass Index, HbAlc Glycosylated hemoglobin, GA glycated albumin, hs-CRP high-sensitivity C-reactive protein

Regression analysis

Multivariable linear regression analysis showed that after adjusting for gender, age, and LAGE, SDBG was positively correlated with endotoxin independently (standard partial regression coefficient = 0.255, P = 0.039). The scatter-plot is shown in Fig. 1.

Discussion

Previous reports exhibited that patients with T2DM are more susceptible to intestinal mucosal barrier dysfunction [23, 24], however factors remains to be investigated. In this study, T2DM patients with higher daily blood glucose fluctuation tended to have a higher serum endotoxin level, which indicates the function of the intestinal mucosal barrier was possibly compromised.
In this study patients with T2DM and high SDBG were associated with higher serum endotoxin level; meanwhile there was no significant change in serum DAO and D-lactic acid levels. Diamine oxidase (DAO) is an enzyme mainly produced in the small intestine involved in the histamine metabolism [25, 26]. Recent studies showed that a established first-line treatment for patients in T2DM, metformin, inhibits DAO activity [27]. Such inhabitation possibly compensate the increased DAO due to the dysfunction of intestinal mucosal barrier. D-lactate is a bydroxycarboxylic acid produced by bacterial fermentation [28, 29]. Studies revealed that the gut microbiome composition is altered for T2DM patients treated with metformin [30], in which possibly explain the inconsistent result in serum D-lactate and endotoxin level.
Endotoxins, also known as Lipopolysaccharides (LPS), are large molecules found in the outer membrane of Gram-negative bacteria. The increase in LPS is associated with bacterial translocation due to the impairment of intestinal epithelial cell [31]. The increase in endotoxins of T2DM patients are probably result from the change in gut microbial composition, epithelial cell impairment, responses to inflammatory mediators or secondary action of endotoxins [3134]. A resent study on mice suggested that fluctuant hyperglycemia had more potential to cause oxidative stress and inflammation, and eventually endothelial dysfunction [35]. In this study, although the amplitude of blood glucose fluctuation of patients are much less than that of mouse model (2.1/2.9 mmol/l vs ~ 15 mmol/l), the SDBG is still an independent factor positively correlated to the serum LPS level. In addition, blood glucose fluctuations are associated with ketoacidosis in patients with DM, and also an independent risk factors for the progression of atherosclerosis [4, 36]. These pevious reports, combined with this study, further indicate the importance of blood glucose stabilization to health.
Previous studies revealed that T2DM patients are more probably associated with higher LPS, in which long-term hyperglycemia in these patients is one of the causes of intestinal mucosal barrier damage [37]. The compromised mucosal barrier could be a potential cause of the large blood glucose fluctuartion. In this study, however, no sign of inflammatory damage of intestinal mucosa was observed as the CRP level of patients are at normal level.
Multivariable linear regression analysis revealed that the amplitude of blood glucose fluctuations was an independent risk factor of increased intestinal permeability. The blood glucose fluctuation compromise the intestinal permeability in several possible ways: (1) Increased blood glucose fluctuations affect the integrity of the intestinal epithelial cells and the contact structure between cells [38]. (2) Abnormal metabolic pathways caused by fluctuations in blood glucose lead to increased inflammatory factors and stimulate intestinal mucosa [38]. (3) Function of liver is compromised due to the flucturation, thereby preventing timely and effective removal of endotoxins.
The positively correlation between intestinal mucosal barrier damage and SDBG indicates that clinicans/ T2DM patients should carefully maintain stable blood glucose level. Subsequent studies should be done to confirm our findings by increasing the sample size. Although DAO, D-lactic acid, and endotoxin can be used as indicators of the intestinal mucosal barrier status, the impairment mechanism of intestinal mucosal barrier remains to be clearly elucidated. Factors such as intestinal microenvironment affected by medication and immunity of patients should be further investigated.

Conclusions

In summary, impairment of the intestinal mucosal barrier function, characterized by impaired intestinal permeability, may occur in patients with T2DM with large SDBG. Clinical attention should be focused on monitoring and controlling blood glucose fluctuations in patients with T2DM.

Funding

The work was supported by grants from Shanghai Municipal Jiading District Natural Science Foundation (2015–023), Foundation of the Public Health Bureau of Jiading (2017-KY-06) and New Key Subjects of Jiading District (2017-ZD-03).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
The study was approved by the Ethics Committee of Shanghai Jiading District Central Hospital, and the acquisition of specimens from all cases was performed with the consent of the patient.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
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.
Literatur
1.
Zurück zum Zitat Yang W, Lu J, Weng J, et al. Prevalence of diabetes among men and women in China. N Engl J Med. 2010;362:1090–101.CrossRef Yang W, Lu J, Weng J, et al. Prevalence of diabetes among men and women in China. N Engl J Med. 2010;362:1090–101.CrossRef
2.
Zurück zum Zitat Malo MS. A high level of intestinal alkaline phosphatase is protective against type 2 diabetes mellitus irrespective of obesity. EBioMedicine. 2015;2:2016–23.CrossRef Malo MS. A high level of intestinal alkaline phosphatase is protective against type 2 diabetes mellitus irrespective of obesity. EBioMedicine. 2015;2:2016–23.CrossRef
3.
Zurück zum Zitat Sato J, Kanazawa A, Ikeda F, et al. Gut dysbiosis and detection of "live gut bacteria" in blood of Japanese patients with type 2 diabetes. Diabetes Care. 2014;37(8):2343–50.CrossRef Sato J, Kanazawa A, Ikeda F, et al. Gut dysbiosis and detection of "live gut bacteria" in blood of Japanese patients with type 2 diabetes. Diabetes Care. 2014;37(8):2343–50.CrossRef
4.
Zurück zum Zitat Zhao H, Li Z, Tian G, et al. Effects of traditional Chinese medicine on rats with type II diabetes induced by high-fat diet and streptozotocin: a urine metabonomic study. Afr Health Sci. 2013;13:673–81.PubMedPubMedCentral Zhao H, Li Z, Tian G, et al. Effects of traditional Chinese medicine on rats with type II diabetes induced by high-fat diet and streptozotocin: a urine metabonomic study. Afr Health Sci. 2013;13:673–81.PubMedPubMedCentral
5.
Zurück zum Zitat Thaiss CA, Levy M, Grosheva I, et al. Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science. 2018;359(6382):1376–83. Thaiss CA, Levy M, Grosheva I, et al. Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science. 2018;359(6382):1376–83.
6.
Zurück zum Zitat Heron M. Deaths: Leading Causes for 2016. Natl Vital Stat Rep. 2018;67(6):1–77.PubMed Heron M. Deaths: Leading Causes for 2016. Natl Vital Stat Rep. 2018;67(6):1–77.PubMed
7.
Zurück zum Zitat Hayes MT, Foo J, Besic V, et al. Is intestinal gluconeogenesis a key factor in the early changes in glucose homeostasis following gastric bypass? Obes Surg. 2011;21:759–62.CrossRef Hayes MT, Foo J, Besic V, et al. Is intestinal gluconeogenesis a key factor in the early changes in glucose homeostasis following gastric bypass? Obes Surg. 2011;21:759–62.CrossRef
8.
Zurück zum Zitat Li B1, Zhou X, Wu J, et al. From gut changes to type 2 diabetes remission after gastric bypass surgeries. Front Med. 2013;7:191–200.CrossRef Li B1, Zhou X, Wu J, et al. From gut changes to type 2 diabetes remission after gastric bypass surgeries. Front Med. 2013;7:191–200.CrossRef
9.
Zurück zum Zitat Koliaki C, Liatis S, le Roux CW, et al. The role of bariatric surgery to treat diabetes: current challenges and perspectives. BMC Endocr Disord. 2017;17(1):50.CrossRef Koliaki C, Liatis S, le Roux CW, et al. The role of bariatric surgery to treat diabetes: current challenges and perspectives. BMC Endocr Disord. 2017;17(1):50.CrossRef
10.
Zurück zum Zitat Scaldaferri F, Pizzoferrato M, Gerardi V, et al. The gut barrier: new acquisitions and therapeutic approaches. J Clin Gastroenterol. 2012;46(Suppl):S12–7.CrossRef Scaldaferri F, Pizzoferrato M, Gerardi V, et al. The gut barrier: new acquisitions and therapeutic approaches. J Clin Gastroenterol. 2012;46(Suppl):S12–7.CrossRef
11.
Zurück zum Zitat Yang X, Gao XC, Liu J, et al. Effect of EPEC endotoxin and bifidobacteria on intestinal barrier function through modulation of toll-like receptor 2 and toll-like receptor 4 expression in intestinal epithelial cell-18. World J Gastroenterol. 2017;23(26):4744–51.CrossRef Yang X, Gao XC, Liu J, et al. Effect of EPEC endotoxin and bifidobacteria on intestinal barrier function through modulation of toll-like receptor 2 and toll-like receptor 4 expression in intestinal epithelial cell-18. World J Gastroenterol. 2017;23(26):4744–51.CrossRef
12.
Zurück zum Zitat Al-Attas OS, Al-Daghri NM, Al-Rubeaan K, et al. Changes in endotoxin levels in T2DM subjects on anti-diabetic therapies. Cardiovasc Diabetol. 2009;15:8–20. Al-Attas OS, Al-Daghri NM, Al-Rubeaan K, et al. Changes in endotoxin levels in T2DM subjects on anti-diabetic therapies. Cardiovasc Diabetol. 2009;15:8–20.
13.
Zurück zum Zitat Creely SJ, McTernan PG, Kusminski CM, et al. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol Metab. 2007;292(3):E740–7.CrossRef Creely SJ, McTernan PG, Kusminski CM, et al. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol Metab. 2007;292(3):E740–7.CrossRef
14.
Zurück zum Zitat Lozupone CA, Stombaugh JI, Gordon JI, et al. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489:220–30.CrossRef Lozupone CA, Stombaugh JI, Gordon JI, et al. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489:220–30.CrossRef
15.
Zurück zum Zitat Genton L, Cani PD, Schrenzel J. Alterations of gut barrier and gut microbiota in food restriction, food deprivation and protein-energy wasting. Clinl Nutr. 2015;34:341–9.CrossRef Genton L, Cani PD, Schrenzel J. Alterations of gut barrier and gut microbiota in food restriction, food deprivation and protein-energy wasting. Clinl Nutr. 2015;34:341–9.CrossRef
16.
Zurück zum Zitat Cyphert TJ, Morris RT, House LM, et al. NF-κB-dependent airway inflammation triggers systemic insulin resistance. Am J Physiol Regul Integr Comp Physiol. 2015;309:R1144–52.CrossRef Cyphert TJ, Morris RT, House LM, et al. NF-κB-dependent airway inflammation triggers systemic insulin resistance. Am J Physiol Regul Integr Comp Physiol. 2015;309:R1144–52.CrossRef
17.
Zurück zum Zitat Kaĭdashev IP. NF-kB activation as a molecular basis of pathological process by metabolic syndrome. Fiziol Zh. 2012;58:93–101.PubMed Kaĭdashev IP. NF-kB activation as a molecular basis of pathological process by metabolic syndrome. Fiziol Zh. 2012;58:93–101.PubMed
18.
Zurück zum Zitat Ferdaoussi M, Abdelli S, Yang JY, et al. Exendin-4 protects beta-cells from interleukin-1 beta-induced apoptosis by interfering with the c-Jun NH2-terminal kinase pathway. Diabetes. 2008;57:1205–15.CrossRef Ferdaoussi M, Abdelli S, Yang JY, et al. Exendin-4 protects beta-cells from interleukin-1 beta-induced apoptosis by interfering with the c-Jun NH2-terminal kinase pathway. Diabetes. 2008;57:1205–15.CrossRef
19.
Zurück zum Zitat Xu J, Liu Z, Zhan W, et al. Recombinant TsP53 modulates intestinal epithelial barrier integrity via upregulation of ZO-1 in LPS-induced septic mice. Mol Med Rep. 2018;17(1):1212–8.PubMed Xu J, Liu Z, Zhan W, et al. Recombinant TsP53 modulates intestinal epithelial barrier integrity via upregulation of ZO-1 in LPS-induced septic mice. Mol Med Rep. 2018;17(1):1212–8.PubMed
20.
Zurück zum Zitat Gao X, Miao R, Tao Y, et al. Effect of montmorillonite powder on intestinal mucosal barrier in children with abdominal Henoch-Schonlein purpura: a randomized controlled study. Medicine (Baltimore). 2018;97(39):e12577.CrossRef Gao X, Miao R, Tao Y, et al. Effect of montmorillonite powder on intestinal mucosal barrier in children with abdominal Henoch-Schonlein purpura: a randomized controlled study. Medicine (Baltimore). 2018;97(39):e12577.CrossRef
21.
Zurück zum Zitat Garber AJ, Abrahamson MJ, Barzilay JI, et al. Consensus statement by the American Association of Clinical Endocrinologists and American College of endocrinology on the comprehensive type 2 diabetes management algorithm-2018 executive summary. Endocr Pract. 2018;24:91–120.CrossRef Garber AJ, Abrahamson MJ, Barzilay JI, et al. Consensus statement by the American Association of Clinical Endocrinologists and American College of endocrinology on the comprehensive type 2 diabetes management algorithm-2018 executive summary. Endocr Pract. 2018;24:91–120.CrossRef
22.
Zurück zum Zitat American Diabetes Association. Standards of medical care in diabetes--2014. Diabetes Care. 2014;37(Suppl 1):S14–80. American Diabetes Association. Standards of medical care in diabetes--2014. Diabetes Care. 2014;37(Suppl 1):S14–80.
23.
Zurück zum Zitat Zhong H, Yuan Y, Xie W, et al. Type 2 Diabetes Mellitus Is Associated with More Serious Small Intestinal Mucosal Injuries. PLoS ONE. 11(9):e0162354. Zhong H, Yuan Y, Xie W, et al. Type 2 Diabetes Mellitus Is Associated with More Serious Small Intestinal Mucosal Injuries. PLoS ONE. 11(9):e0162354.
24.
Zurück zum Zitat Horton F, Wright J, Smith L, et al. Short Report: Pathophysiology Increased intestinal permeability to oral chromium (51Cr)-EDTA in human Type 2 diabetes. Diabet Med. 2014;31:559–63.CrossRef Horton F, Wright J, Smith L, et al. Short Report: Pathophysiology Increased intestinal permeability to oral chromium (51Cr)-EDTA in human Type 2 diabetes. Diabet Med. 2014;31:559–63.CrossRef
25.
Zurück zum Zitat Zhao L, Luo L, Jia W, et al. Serum diamine oxidase as a hemorrhagic shock biomarker in a rabbit model. PLoS One. 2014;9(8):e102285.CrossRef Zhao L, Luo L, Jia W, et al. Serum diamine oxidase as a hemorrhagic shock biomarker in a rabbit model. PLoS One. 2014;9(8):e102285.CrossRef
26.
Zurück zum Zitat Bragg LE, Thompson JS, West WW. Intestinal diamine oxidase levels reflect ischemic injury. J Surg Res. 1991;50(3):228–33.CrossRef Bragg LE, Thompson JS, West WW. Intestinal diamine oxidase levels reflect ischemic injury. J Surg Res. 1991;50(3):228–33.CrossRef
27.
Zurück zum Zitat Yee SW, Lin L, Merski M, et al. Prediction and validation of enzyme and transporter off-targets for metformin. J Pharmacokinet Pharmacodyn. 2015;42(5):463–75.CrossRef Yee SW, Lin L, Merski M, et al. Prediction and validation of enzyme and transporter off-targets for metformin. J Pharmacokinet Pharmacodyn. 2015;42(5):463–75.CrossRef
28.
Zurück zum Zitat Demircan M, Cetin S, Uguralp S, et al. Plasma D-lactic acid level: a useful marker to distinguish perforated from acute simple appendicitis. Asian J Surg. 2004;27(4):303–5.CrossRef Demircan M, Cetin S, Uguralp S, et al. Plasma D-lactic acid level: a useful marker to distinguish perforated from acute simple appendicitis. Asian J Surg. 2004;27(4):303–5.CrossRef
29.
Zurück zum Zitat Sun XQ, Fu XB, Zhang R, et al. Relationship between plasma D(−)-lactate and intestinal damage after severe injuries in rats. World J Gastroenterol. 2001;7(4):555–8.CrossRef Sun XQ, Fu XB, Zhang R, et al. Relationship between plasma D(−)-lactate and intestinal damage after severe injuries in rats. World J Gastroenterol. 2001;7(4):555–8.CrossRef
30.
Zurück zum Zitat McCreight LJ, Bailey CJ, Pearson ER. Metformin and the gastrointestinal tract. Diabetologia. 2016;59(3):426–35.CrossRef McCreight LJ, Bailey CJ, Pearson ER. Metformin and the gastrointestinal tract. Diabetologia. 2016;59(3):426–35.CrossRef
31.
Zurück zum Zitat Cavaillon JM. Exotoxins and endotoxins: inducers of inflammatory cytokines. Toxicon. 2018;149:45–53.CrossRef Cavaillon JM. Exotoxins and endotoxins: inducers of inflammatory cytokines. Toxicon. 2018;149:45–53.CrossRef
32.
Zurück zum Zitat Wu X, Ma C, Han L, et al. Molecular characterisation of the faecal microbiota in patients with type II diabetes. Curr Microbiol. 2010;61:69–78.CrossRef Wu X, Ma C, Han L, et al. Molecular characterisation of the faecal microbiota in patients with type II diabetes. Curr Microbiol. 2010;61:69–78.CrossRef
33.
Zurück zum Zitat Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761–72.CrossRef Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761–72.CrossRef
34.
Zurück zum Zitat Al-Obaide MAI, Singh R, Datta P, et al. Gut Microbiota-Dependent Trimethylamine-N-oxide and Serum Biomarkers in Patients with T2DM and Advanced CKD. J Clin Med. 2017;6(9). Al-Obaide MAI, Singh R, Datta P, et al. Gut Microbiota-Dependent Trimethylamine-N-oxide and Serum Biomarkers in Patients with T2DM and Advanced CKD. J Clin Med. 2017;6(9).
35.
Zurück zum Zitat Wu N, Shen H, Liu H, et al. Acute blood glucose fluctuation enhances rat aorta endothelial cell apoptosis, oxidative stress and pro-inflammatory cytokine expression in vivo. Cardiovasc Diabetol. 2016;15:109.CrossRef Wu N, Shen H, Liu H, et al. Acute blood glucose fluctuation enhances rat aorta endothelial cell apoptosis, oxidative stress and pro-inflammatory cytokine expression in vivo. Cardiovasc Diabetol. 2016;15:109.CrossRef
36.
Zurück zum Zitat Martín-Timón I, Sevillano-Collantes C, Segura-Galindo A, et al. Type 2 diabetes and cardiovascular disease: have all risk factors the same strength? World J Diabetes. 2014;5:444–70.CrossRef Martín-Timón I, Sevillano-Collantes C, Segura-Galindo A, et al. Type 2 diabetes and cardiovascular disease: have all risk factors the same strength? World J Diabetes. 2014;5:444–70.CrossRef
37.
Zurück zum Zitat Jayashree B, Bibin YS, Prabhu D, et al. Increased circulatory levels of lipopolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation in patients with type 2 diabetes. Mol Cell Biochem. 2014;388(1–2):203–10.CrossRef Jayashree B, Bibin YS, Prabhu D, et al. Increased circulatory levels of lipopolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation in patients with type 2 diabetes. Mol Cell Biochem. 2014;388(1–2):203–10.CrossRef
38.
Zurück zum Zitat Oliveira RB, Canuto LP, Collares-Buzato CB. Intestinal luminal content from high-fat-fed prediabetic mice changes epithelial barrier function in vitro. Life Sci. 2019;216:10–21.CrossRef Oliveira RB, Canuto LP, Collares-Buzato CB. Intestinal luminal content from high-fat-fed prediabetic mice changes epithelial barrier function in vitro. Life Sci. 2019;216:10–21.CrossRef
Metadaten
Titel
Poor short-term glycemic control in patients with type 2 diabetes impairs the intestinal mucosal barrier: a prospective, single-center, observational study
verfasst von
Lijuan Shen
Li Ao
Haoben Xu
Junfeng Shi
Dali You
Xiuwen Yu
Weixin Xu
Jie Sun
Fei Wang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Endocrine Disorders / Ausgabe 1/2019
Elektronische ISSN: 1472-6823
DOI
https://doi.org/10.1186/s12902-019-0354-7

Weitere Artikel der Ausgabe 1/2019

BMC Endocrine Disorders 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

Update Innere Medizin

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