Skip to main content
Erschienen in: Diabetology & Metabolic Syndrome 1/2019

Open Access 01.12.2019 | Research

Serum fibroblast growth factor 19 and endogenous islet beta cell function in type 2 diabetic patients

verfasst von: Meng-jie Tang, Jian‑bin Su, Tian-li Xu, Xue‑qin Wang, Dong-mei Zhang, Xiao-hua Wang

Erschienen in: Diabetology & Metabolic Syndrome | Ausgabe 1/2019

Abstract

Background

Fibroblast growth factor 19 (FGF19) takes part in maintaining the balance of glycolipids and may be involved in regulating the secretory activity of islet beta cells in patients with type 2 diabetes. This study aimed to evaluate the relationship between the levels of serum FGF19 and endogenous islet beta cell function in type 2 diabetic patients.

Methods

Samples were obtained from 271 subjects: 85 drug-naïve type 2 diabetes participants exclusively on lifestyle intervention (N-DM group), 122 type 2 diabetes subjects previously used medications (DM group) and 64 normal controls (NC group). Serum FGF19 concentrations were measured by ELISA. The insulin sensitivity (MI), insulin secretion (AUCins/AUCglu) and insulin secretion-sensitivity index-2 (ISSI-2) were also measured in the N-DM and DM.

Results

Serum FGF19 levels decreased, in order, from the NC group [median (interquartile range), 245.03 (126.23–317.43) pg/mL] to the N-DM group [170.05 (89.01–244.70) pg/mL] and, finally, to the DM group [142.25 (55.55–187.58) pg/mL] (p for trend < 0.05). Among subjects in the DM group, there was a positive trend in the serum FGF19 concentration; plasma insulin levels at 60 min, 120 min (INS60, INS120, respectively); and area under the insulin curve (AUCins) at two points (r = 0.214, p = 0.025; r = 0.189, p = 0.048; r = 0.188, p = 0.049). However, the differences were no longer observed among the N-DM subjects. Simultaneously, the ISSI-2 was closely related to the serum FGF19 levels (r = 0.297, p = 0.002) among DM subjects. Furthermore, after adjusting for age, sex, duration, therapy and other clinical factors via multiple logistic regression analysis, ISSI-2 was a key independent factor in the levels of FGF19 (β = 0.281, t = 2.557, p = 0.013).

Conclusions

The serum FGF19 level has a close relation with endogenous beta cell function among DM subjects, as assessed by the ISSI-2. As ISSI-2 is higher in N-DM group, FGF19 may be a main protector in dysfunction of beta cell.
Hinweise
Meng-jie Tang, Jian‑bin Su and Tian-li Xu contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BMI
body mass index
W
waist
SBP
systolic blood pressure
DBP
diastolic blood pressure
FPG
fasting plasma glucose
FIN
fasting plasma insulin
2hPG
2-hour postprandial blood glucose
HbA1c
glycosylated haemoglobin A1c
HOMA-IR
homeostatic model assessment for insulin resistance
TC
total cholesterol
TG
triglyceride
HDL-c
high-density lipoprotein cholesterol
LDL-c
low-density lipoprotein cholesterol
FGF19
fibroblast growth factor 19
MI
Matsuda Index
AUCins/glu
the ratio of the total area under the insulin curve to the total area under the glucose curve
ISSI-2
insulin secretion-sensitivity index-2
OGTT
oral glucose tolerance test
GE
glucose effectiveness
HGP
hepatic glucose production
GSK3
glycogen synthase kinase 3
CREB-PGC1α
cyclic adenosine monophosphate regulatory element binding protein/peroxisome proliferator-activated receptor γ coactivator-1α-dependent
GLP-1
glucagon-like peptide-1
ER
endoplasmic reticulum

Background

Type 2 diabetes (T2D) affects up to 10% of the global adult population, causing a decline in human health. Insulin resistance and an islet beta cell defect are the main pathogenic mechanisms of diabetes and have severe complications, including microangiopathy and cardiovascular disease.
Fibroblast growth factor 19 (FGF19), a hormone expressed by ileal enterocytes, is a member of the FGF family involved in regulating lipid and nutrient metabolism [15]. The main physiological function of FGF19 is bile acid (BA) metabolism [6]. FGF19 (the mouse orthologue of which is called FGF15) binds to FGF receptor 4 (FGFR4) and generates physiological effects via the FGFR4-β-Klotho complex [7].
FGF19 has been widely discussed and has potential as a therapeutic target for metabolic disorders, including diabetes. In rodent model studies, FGF19 is able to decrease weight and improve metabolic disorders and insulin resistance [811]. In addition, FGF19 restores metabolic function in hormonal remodelling following bariatric surgery [1214]. Clinical studies have observed that the insulin-independent beneficial effects of FGF19 in glucose metabolism can be explained by glucose effectiveness (GE) [15]. However, the aetiology of endogenous islet beta cell function is not fully understood, and not only insulin-independent but also insulin-dependent mechanisms may contribute to islet beta cell dysfunction. Glucose homeostasis is regulated through the BA-G-protein-coupled receptor 5 (TGR5)/FXR axis via incretin hormone: glucagon-like peptide 1 (GLP-1), which eventually stimulates the expression of FXR [16]. GLP-1 can regulate insulin secretion and glucose homeostasis by mimicking insulin action [17, 18]. However, the regulatory role of FGF19 in endogenous beta cell function among type 2 diabetic patients of different therapies remains insufficiently understood.
Our previous studies have shown that insulin sensitivity and insulin secretion were measured by the indices from oral glucose tolerance test (OGTT) results: insulin sensitivity, assessed by Matsuda Index (MI), insulin secretion, assessed by area under the insulin curve/area under the glucose curve (AUCins/glu) [19]. Furthermore, the insulin secretion-sensitivity index-2 (ISSI-2), which evaluates integrated islet beta cell function, can be determined by the MI multiplied by the AUCins/glu [1921].
Therefore, the current study was designed to identify the correlation between serum FGF19 and endogenous beta cell function in type 2 diabetic subjects with treatment before, naïve type 2 diabetic patients and normal controls. We also measured and compared the concentrations of FGF19 among these three groups.

Methods

Study design and subjects

The study was conducted during visits and follow-ups with patients from the Endocrinology Department at Affiliated Hospital 2 of Nantong University and First People’s Hospital of Nantong City from Apr 2017 to Oct 2018. The non-T2DM normal controls (NC) were required to be 20–60 years old and to have fasting plasma glucose < 6.1 mmol/L and 2-h plasma glucose (2hPG) < 7.8 mmol/L [1]. The inclusion criteria for newly diagnosed diabetic patients (N-DM) were being between 20 and 75 years old, having newly diagnosed type 2 diabetes as previously described [1] and being drug naïve exclusively on lifestyle intervention. The inclusion criteria for previously diagnosed diabetes patients (DM) were age between 20 and 75 years old, diagnosis of type 2 diabetes and with medications used. The exclusion criteria for all groups were type 1 diabetes, hyperthyroidism or hypothyroidism, severe hepatic disease, chronic renal insufficiency, cancer, acute diabetic complications, and current treatment with systemic corticosteroids. Finally, we recruited 64 NC, 122 DM and 85 N-DM participants. All participants gave written informed consent. The study was approved by the institution review board of Second People’s Hospital of Nantong and complied with the Declaration of Helsinki.

Basic data collection

Upon enrolment, all participants were questioned and examined by experienced investigators to complete a questionnaire including parameters on their age, sex, weight, height, waist circumference, blood pressure, illness and medical therapy history. Body mass index (BMI) was calculated as the weight/the height squared. Blood pressure (BP) was tested in triplicate after at least 30 min of rest, and the average of three recordings was used for the analysis.

OGTT procedures and calculation

After an overnight fast, all patients received OGTT (75 g of glucose) and plasma glucose at fasting (0) and 30, 60, 120 and 180 min (FPG, PG30, PG60, PG120, PG180, respectively) were determined. Measurements of serum insulin and C-peptide were also performed. The insulin sensitivity index was determined with the following equations: MI = 10,000/(FPG × fasting insulin × mean glucose × mean insulin)1/2 [2]. AUCins/glu was measured by area under the insulin curve/area under the glucose curve [2]. The ISSI-2 was calculated as the product of MI * AUCins/glu [24]. Insulin resistance was evaluated by HOMA-IR = FPG [mmol/L] × fasting insulin [mU/L]/22.5.

Laboratory examination

Fasting blood samples were also collected to measure laboratory parameters. Measurements of serum insulin level, plasma glucose level, HbA1c concentration, low-density lipoprotein cholesterol (LDL-c), high-density lipoprotein cholesterol (HDL-c), total cholesterol (TC), and triglycerides (TG) were determined by standard laboratory procedures [5, 6]. Circulating FGF19 levels were detected by sandwich ELISA (FGF19 Quantikine® DF1900; R&D Systems, Minneapolis, MN, USA). The range of the standard curve was 15.6–1000 pg/mL.

Statistical analysis

Statistical analyses were conducted using SPSS software version 25.0 (Inc., Chicago, IL). Continuous variables with normal distribution are presented as the mean ± SD, skewed data as medians with interquartile ranges, and categorical variables as frequencies. All variables with skewed distributions were log transformed for further analysis. The skewness and kurtosis tests were used to assess the normality of the distribution. ANOVA/general linear model were conducted to compare inter-group differences between continuous variables, and the Pearson Chi-squared test was conducted to compare categorical variables. Moreover, a bivariate correlation analysis was used to select risk factors associated with FGF19. Then, multivariable linear regression analysis was undertaken to explore and identify independent contributors to FGF19. A p value < 0.05 was considered significant.

Results

Clinical characteristics of study participants

A total of 271 participants (mean age of 47.98 ± 13.188, 50.2% male) were recruited, and 195 subjects underwent an OGTT. We divided the population into NC group (n = 64), N-DM group (n = 85), and DM group (n = 122). Table 1 shows the clinical paraments of the three groups. Fasting FGF19 levels decreased in order from the NC group [245.03 (126.23–317.43) pg/mL] to the N-DM group [170.05 (89.01–244.70) pg/mL] and finally to the DM group [142.25 (55.55–187.58) pg/mL] (p < 0.05; Table 1, Fig. 1). After adjustment for age, sex, BMI and SBP, FGF19 levels still significantly decreased in order from NC, N-DM to DM (p < 0.001, Table 2). After adjustment for medications, the difference of serum FGF19 was significantly between the N-DM and DM groups (p < 0.05, Table 2). Compared with participants in the NC group, participants in the N-DM and DM groups were older and had higher BMI, SBP and FPG, TG (all p < 0.01) and LDL-c levels (p < 0.05) but lower HDL-c levels (p < 0.01). Additionally, individuals in the DM group exhibited higher FINS and HOMA-IR levels than subjects among N-DM group, but a lower level of ISSI-2 (p < 0.05; Table 1). Meanwhile, HbA1c did not differ between the N-DM and DM groups. Moreover, HbA1c, a reflection of 3-months glucose control was negatively correlated with serum FGF19 levels in DM group (r = − 0.247, p = 0.012; Fig. 2).
Table 1
Characteristics of the study participants
Variable
NC
N-DM
DM
n
64
85
122
Female n (%)
82.8
34.1**
43.4**
Age (years)
33.61 ± 10.64
51.38 ± 10.87**
53.15 ± 10.14**
BMI (kg/m2)
22.20 ± 2.59
25.41 ± 4.25**
25.60 ± 3.16**
W (cm)
N
92.62 ± 10.16
92.95 ± 9.82
SBP (mmHg)
110.3 (108.0–112.0)
137.2 (124.0–150.5)**
135.4 (127.0–145.0)**
DBP (mmHg)
90.1 (70.0–82.0)
83.8 (76.0–91.0)*
80.63 (74.0–89.0)*
FPG (mmol/L)
4.41 ± 0.30
5.97 ± 1.62**
5.81 ± 1.80**
FINS (mU/L)
N
112.0 (56.72–121.32)
160.19 (84.22–179.49)##
2hPG (mmol/L)
N
16.09 ± 3.98
16.81 ± 4.08
HbA1c (%)
N
9.48 ± 2.56
9.48 ± 2.44
HOMA-IR
N
28.92 (14.38–33.00)
41.55 (18.67–50.06)#
TC (mmol/L)
4.31 ± 0.79
4.55 ± 0.75
4.46 ± 1.04
TG (mmol/L)
1.21 ± 0.89
2.25 ± 2.05*
2.36 ± 2.23**
HDL-c (mmol/L)
1.34 ± 0.17
1.10 ± 0.28**
1.17 ± 0.32**
LDL-c (mmol/L)
2.52 ± 0.56
2.90 ± 0.68*
2.79 ± 0.93
FGF19 (pg/mL)
245.03 (126.23–317.43)
170.05 (89.01–244.70)*
142.25 (55.55–187.58)**,#
MI
N
9.39 ± 2.95
8.42 ± 3.58
AUCins/glu
N
16.23 (6.92–19.43)
16.45 (7.09–20.93)
ISSI-2
N
146.44 (62.03–187.35)
122.94 (55.48–146.59)#
Categorical variables are frequency (percentage), normally distributed values in the table are mean ± SD and non-normally distributed values are median (25 and 75% interquartiles)
BMI body mass index, W waist, SBP/DBP systolic/diastolic blood pressure, FPG fasting plasma glucose, FIN fasting plasma insulin, 2hPG 2-hour postprandial blood glucose, HbA1c glycosylated hemoglobin A1c, HOMA-IR homeostatic model assessment for insulin resistance, TC total cholesterol, TG triglyceride, HDL-c high density lipoprotein cholesterol, LDL-c low density lipoprotein cholesterol, FGF19 fibroblast growth factor 19, MI Matsuda index, AUCins/glu the ratio of total area-under-the- insulin-curve to glucose-curve, ISSI-2 Insulin Secretion-Sensitivity Index-2
p-values were determined using Student’s t-test, the Mann–Whitney U-test or the Chi-square test as appropriate
* p < 0.05, ** p < 0.01, the comparison of N-DM and DM with CN
#p < 0.05, ##p < 0.01, the comparison of N-DM and DM
Table 2
Univariate analysis of serum FGF19 among NC, N-DM and DM group
Group
Adjusted for age
Adjusted for s ex
Mean
95% CI
p
Mean
95% CI
p
NC
5.255
5.001–5.510
 
5.230
5.007–5.452
 
N-DM
4.886
4.700–5.073
< 0.001
4.909
4.723–5.095
< 0.001
DM
4.589
4.428–4.751
 
4.587
4.434–4.740
 
Group
Adjusted for BMI
Adjusted for SBP
Mean
95% CI
p
Mean
95% CI
p
NC
5.258
5.033–5.484
 
5.250
4.995–5.504
 
N-DM
4.887
4.702–5.072
< 0.001
4.894
4.703–5.085
< 0.001
DM
4.590
4.434–4.747
 
4.587
4.429–4.745
 
Group
Adjusted for medication
Mean
95% CI
p
N-DM
4.873
4.635–5.111
0.018
DM
4.491
4.299–4.682
FGF19 did log transformation
p value < 0.05 was considered significant

Serum FGF19 levels and beta cell function

To detect the relationship between FGF19 and glucose, insulin, and beta cell function from the OGTT, Spearman analysis was performed. Serum FGF19 levels were positively correlated with insulin levels at 60 and 120 min after a glucose challenge in the DM group (r = 0.214, p = 0.025; r = 0.189, p = 0.048). Simultaneously, FGF19 level was positively correlated with the AUCins at two time points in the DM group (r = 0.188, p = 0.049; Fig. 3). Moreover, the MI, AUCins/glu, and ISSI-2 among participants in DM group were analysed. Interestingly, only the AUCins/glu and ISSI-2 increased with increasing plasma FGF19 levels in the DM group (r = 0.227, p = 0.018; r = 0.297, p = 0.002; Fig. 4). However, no significant differences in AUCins/glu and ISSI-2 were observed in the N-DM group (r = 0.040, p = 0.720; r = 0.027, p = 0.812; Fig. 4).

The determinants of FGF19 in DM group

To analyse whether serum FGF19 is independently correlated with insulin resistance, secretion, sensitivity and diabetes therapy, we performed multiple stepwise regression analysis. As is shown in Table 3, Model 1 considers gender, age, BMI, W, duration, hypertension, HbA1c, 2hPG, HOMA-IR, LDL-c, HDL-c, TG and TD. The results showed that ISSI-2 (β = 0.321, t = 3.233, p = 0.002, Table 3) was independently correlated with serum FGF19 levels. Based on Model 1, Model 2 further condensed the use of therapy (lifestyle intervention alone, insulin injection, insulin secretagogues and insulin sensitizers), and showed that W was independently and negatively connected with FGF19 levels. (β =− 0.220, t =− 2.002, p = 0.049, Table 3).
Table 3
Multivariate linear regression analyses on serum FGF19 levels
Model
Variable
β
t
p
Model 1
ISSI-2
0.321
3.233
0.002
Model 2
ISSI-2
0.281
2.557
0.013
W
− 0.220
− 2.002
0.049
Original independent variables included
Model 1: gender, age, BMI, W, duration, hypertension, HbA1c, 2hPG, HOMA-IR, LDL-c, HDL-c, TG, TD
Model 2: Model 1+ antidiabetic treatments (lifestyle intervention alone, insulin injection, insulin secretagogues and insulin sensitizers)

Discussion

In the current study, we compared FGF19 levels and investigated the relationship between FGF19 levels and islet beta cell function among a middle cohort of participants. The main findings of the present study are as follows. First, the serum concentrations of FGF19 were lower in N-DM group and even lower in DM group than in NC group regardless of the degree of insulin resistance. Second, serum FGF19 concentration was positively associated with INS60, INS120 level and the AUCins at two points. The AUCins/glu increased with increasing plasma FGF19 levels, but the correlation between ISSI-2 and FGF19 level was closer than the correlation between AUCins/glu and FGF19 level. However, these relationships were not significant in N-DM group. Third, after adjusting for sex, age, the use of therapy and other clinical factors via multiple logistic regression analysis, the ISSI-2 was still related to plasma FGF19 level. Additionally, as ISSI-2 is lower in DM group, FGF19 may be a crucial protector in dysfunction of islet beta cell.
Although the pancreas is an FGF19 target organ, data regarding the function of FGF19 in pancreatic islets remain scarce. Previous studies have detected that FGF19 plays an important role in keeping glucose homeostasis [811]. A study of the Chinese population demonstrated that the levels of fasting FGF19 were decreased in participants with decreased fasting glucose levels and naive diabetic patients and were inversely correlation with fasting glucose [5]. These data are agreement with our study. In addition, FGF19 could mediate its function via activating the FGFR4-β-Klotho complex [7]. Izaguirre et al. found that FGF19 combined with the FGFR4-β-Klotho complex to activate an insulin-independent endocrine pathway and mediate different metabolic effects [22]. Kir et al. reveal that FGF19 may play an insulin-like role in mouse liver and regulates hepatic glycogen via an insulin-independent mechanism [23]. Zhang et al. showed that FGF19 levels are positively associated with glucose effectiveness (GE) and negatively associated with hepatic glucose production (HGP) The increase in HGP in humans is partially due to the insulin-independent decrease in FGF19, which disagrees with our data [15]. ISSI-2 represents insulin sensitivity and insulin secretion during the OGTT. ISSI-2 and serum FGF19 levels were positively related in our research, which means ISSI-2 participates in insulin-dependent glucose regulation. Different durations of diabetes and treatments may cause these differences.
Previous studies have shown that when FGF19 was administered to diabetic mice in animal studies, plasma glucose levels were reduced, and glucose homeostasis was maintained [11]. These effects may be linked with glycogen synthase kinase 3 (GSK3), which is increased in hepatic glycogen storage [23] and inhibits the cyclic adenosine monophosphate regulatory element binding protein/peroxisome proliferator-activated receptor γ coactivator-1α-dependent (CREB/PGC1α) pathway [1]. Simultaneously, FGF19 contributed to weight loss and decreased glucose and insulin levels by β-Klotho in neurons [24].
Type 2 diabetes is associated with islet beta cell dysfunction. Fatty acid, hyperglycaemia, endoplasmic reticulum (ER) stress, or beta cell oxidative stress are factors that result in impaired islet beta cell function [25]. The present results suggest that serum FGF19 is independently correlated with insulin secretion and sensitivity and that increased serum FGF19 may improve the function of islet beta cells. However, in our study, the DM subjects were treated with insulin, insulin secretagogues and/or insulin sensitizers whose mechanisms of action are different. Wang et al. revealed that FGF19 serum decreased in DM rats and was continuously reduced after metformin treatment via AMPK-FXR crosstalk [26]. However, Sonne et al. showed no differences in FGF19 level in metformin-treated patients vs. patients treated with sulfonylureas and diet only [27]. We conducted univariate analysis and showed that serum FGF19 was significantly different between the N-DM and DM groups after adjustment for diabetic medications. Furthermore, multiple stepwise regression analysis was performed and showed that serum FGF19 was independently correlated with ISSI-2 in diabetic patients receiving treatment. However, further studies and larger sample sizes are needed to analyse the relationship between FGF19 and islet beta cell function in a sub-group of diabetic medications.
The mechanism of the relationship between FGF19 and islet beta cells remains unclear, but a possible explanation for this link is as follows. The circulating FGF19 level follows a diurnal rhythm depending on transintestinal BA flux after insulin secretion [28]. Isolated impaired fasting glucose (IFG), which depends on glucose metabolism, is due to impaired basal insulin secretion and increased HGP [29]. Therefore, individuals with isolated IFG have lower serum FGF19 levels than those without isolated IFG, suggesting that FGF19 plays a role in the secretion of impaired basal insulin [5]. The effect of the incretin GLP-1, a hormone that releases insulin by a G-protein coupled receptor, is dependent upon glucose [30]. The present study showed that GLP-1 secretion is inhibited by FXR through decreased glycolysis [16]. In addition, FXR/ mice are insulin resistant; their insulin levels were lower [31], and their islet size was smaller than those in control mice [32]. FGF19 clearly regulates BA homeostasis via the FXR-FGF19 axis [33]. Glucose homeostasis is also regulated by activation of FXR-TGR5 and GLP-1 secretion [16]. Our study reveals that serum FGF19 and ISSI-2 are lower in DM participants than in N-DM participants, which means that FGF19 may regulate islet β-cell secretion and insulin levels via the BA-FXR-FGF19 axis and GLP-1 secretion. The reason for this discrepancy in the serum levels of FGF19 in subjects with diabetes is unclear and requires exploration through further studies.
Several limitations in our study must be addressed. First, this was a cross-sectional design with medium sample sizes. This study could not explain the cause-effect connection between decreased serum FGF19 and islet beta cell secretion in diabetic patients. In other words, the secretion of islet beta cells may cause a decrease in the levels of serum FGF19. As this study was prospective, this weakness must be compensated for. Second, our research was conducted in Chinese participants, and the generalizability of our results should be evaluated. Third, our study consisted of the NC, N-DM, and DM groups; an increased number of different glucose tolerance states are warranted to assess the correlation between FGF19 levels and islet beta cell function. Fourth, ISSI-2 is closely related to FGF19 levels in the DM group. As the treatments of patients are different, the results may be influenced by therapies. However, serum FGF19 was still significantly different between the N-DM and DM groups after adjustment for medications and was independently associated with ISSI-2 after multiple stepwise regression analysis in DM group. Fifth, considering that previous studies showed that FGF19 was not significantly related with insulin sensitivity and secretion among NC subjects, only N-DM and DM groups underwent OGTT in our study; nonetheless, the correlation of FGF19 and islet beta cell function should also be observed in NC group. Therefore, further studies are needed to validate the results of the current study and address these limitations.
In summary, the serum FGF19 level is lower in DM subjects than in N-DM and NC subjects, but ISSI-2 is higher among the N-DM group. FGF19 may regulate endogenous islet beta cell function in diabetic patients with medical therapies.

Conclusions

The serum FGF19 level has a close relationship with endogenous beta cell function among DM subjects, as assessed by the ISSI-2, but no significant association between serum FGF19 and integrated beta cell function was detected in the N-DM group. As ISSI-2 is higher in the N-DM group, FGF19 may be a main protector in beta cell dysfunction

Acknowledgements

Not applicable.
The study was approved by the institutional review board of Affiliated Hospital 2 of Nantong University and First People’s Hospital of Nantong City, and written informed consent was obtained from all participants.
Not applicable.

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 Alberti KG, Zimmet PZ. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med. 1998;15:539–53.CrossRef Alberti KG, Zimmet PZ. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med. 1998;15:539–53.CrossRef
2.
Zurück zum Zitat Somm E, Jornayvaz FR. Fibroblast growth factor 15/19: from basic functions to therapeutic perspectives. Endocr Rev. 2018;39(6):960–89.CrossRef Somm E, Jornayvaz FR. Fibroblast growth factor 15/19: from basic functions to therapeutic perspectives. Endocr Rev. 2018;39(6):960–89.CrossRef
3.
Zurück zum Zitat Hui Q, Jin Z, Li X, Liu C, Wang X. FGF family: from drug development to clinical application. Int J Mol Sci. 2018;19(7):1875.CrossRef Hui Q, Jin Z, Li X, Liu C, Wang X. FGF family: from drug development to clinical application. Int J Mol Sci. 2018;19(7):1875.CrossRef
5.
Zurück zum Zitat Fang Q, Li H, Song Q, Yang W, Hou X, Ma X, Lu J, Xu A, Jia W. Serum fibroblast growth factor 19 levels are decreased in Chinese subjects with impaired fasting glucose and inversely associated with fasting plasma glucose levels. Diabetes Care. 2013;36(9):2810–4.CrossRef Fang Q, Li H, Song Q, Yang W, Hou X, Ma X, Lu J, Xu A, Jia W. Serum fibroblast growth factor 19 levels are decreased in Chinese subjects with impaired fasting glucose and inversely associated with fasting plasma glucose levels. Diabetes Care. 2013;36(9):2810–4.CrossRef
7.
Zurück zum Zitat Shi SY, Lu YW, Richardson J, Min X, Weiszmann J, Richards WG, Wang Z, Zhang Z, Zhang J, Li Y. A systematic dissection of sequence elements determining beta-Klotho and FGF interaction and signaling. Sci Rep. 2018;8(1):11045.CrossRef Shi SY, Lu YW, Richardson J, Min X, Weiszmann J, Richards WG, Wang Z, Zhang Z, Zhang J, Li Y. A systematic dissection of sequence elements determining beta-Klotho and FGF interaction and signaling. Sci Rep. 2018;8(1):11045.CrossRef
8.
Zurück zum Zitat Zhou M, Learned RM, Rossi SJ, DePaoli AM, Tian H, Ling L. Engineered FGF19 eliminates bile acid toxicity and lipotoxicity leading to resolution of steatohepatitis and fibrosis in mice. Hepatol Commun. 2017;1(10):1024–42.CrossRef Zhou M, Learned RM, Rossi SJ, DePaoli AM, Tian H, Ling L. Engineered FGF19 eliminates bile acid toxicity and lipotoxicity leading to resolution of steatohepatitis and fibrosis in mice. Hepatol Commun. 2017;1(10):1024–42.CrossRef
9.
Zurück zum Zitat Tomlinson E, Fu L, John L, Hultgren B, Huang X, Renz M, Stephan JP, Tsai SP, Powell-Braxton L, French D, et al. Transgenic mice expressing human fibroblast growth factor-19 display increased metabolic rate and decreased adiposity. Endocrinology. 2002;143(5):1741–7.CrossRef Tomlinson E, Fu L, John L, Hultgren B, Huang X, Renz M, Stephan JP, Tsai SP, Powell-Braxton L, French D, et al. Transgenic mice expressing human fibroblast growth factor-19 display increased metabolic rate and decreased adiposity. Endocrinology. 2002;143(5):1741–7.CrossRef
10.
Zurück zum Zitat Massafra V, Milona A, Vos HR, Burgering BM, van Mil SW. Quantitative liver proteomics identifies FGF19 targets that couple metabolism and proliferation. PLoS ONE. 2017;12(2):e0171185.CrossRef Massafra V, Milona A, Vos HR, Burgering BM, van Mil SW. Quantitative liver proteomics identifies FGF19 targets that couple metabolism and proliferation. PLoS ONE. 2017;12(2):e0171185.CrossRef
11.
Zurück zum Zitat Hansen AMK, Vienberg SG, Lykkegaard K, Zhao X, Tingqing G, Han D, Zhang X, Thogersen H, Sass-Orum K, Tagmose T. Differential receptor selectivity of the FGF15/FGF19 orthologues determines distinct metabolic activities in db/db mice. Biochem J. 2018;475(18):2985–96.CrossRef Hansen AMK, Vienberg SG, Lykkegaard K, Zhao X, Tingqing G, Han D, Zhang X, Thogersen H, Sass-Orum K, Tagmose T. Differential receptor selectivity of the FGF15/FGF19 orthologues determines distinct metabolic activities in db/db mice. Biochem J. 2018;475(18):2985–96.CrossRef
13.
Zurück zum Zitat Nemati R, Lu J, Dokpuang D, Booth M, Plank LD, Murphy R. Increased bile acids and FGF19 after sleeve gastrectomy and Roux-en-Y gastric bypass correlate with improvement in type 2 diabetes in a randomized trial. Obes Surg. 2018;28(9):2672–86.CrossRef Nemati R, Lu J, Dokpuang D, Booth M, Plank LD, Murphy R. Increased bile acids and FGF19 after sleeve gastrectomy and Roux-en-Y gastric bypass correlate with improvement in type 2 diabetes in a randomized trial. Obes Surg. 2018;28(9):2672–86.CrossRef
14.
Zurück zum Zitat Sachdev S, Wang Q, Billington C, Connett J, Ahmed L, Inabnet W, Chua S, Ikramuddin S, Korner J. FGF 19 and bile acids increase following Roux-en-Y gastric bypass but not after medical management in patients with type 2 diabetes. Obes Surg. 2016;26(5):957–65.CrossRef Sachdev S, Wang Q, Billington C, Connett J, Ahmed L, Inabnet W, Chua S, Ikramuddin S, Korner J. FGF 19 and bile acids increase following Roux-en-Y gastric bypass but not after medical management in patients with type 2 diabetes. Obes Surg. 2016;26(5):957–65.CrossRef
15.
Zurück zum Zitat Zhang J, Li H, Bai N, Xu Y, Song Q, Zhang L, Wu G, Chen S, Hou X, Wang C, Wei L, Xu A, Fang Q, Jia W. Decrease of FGF19 contributes to the increase of fasting glucose in human in an insulin-independent manner. J Endocrinol Invest. 2019;42:1019–27.CrossRef Zhang J, Li H, Bai N, Xu Y, Song Q, Zhang L, Wu G, Chen S, Hou X, Wang C, Wei L, Xu A, Fang Q, Jia W. Decrease of FGF19 contributes to the increase of fasting glucose in human in an insulin-independent manner. J Endocrinol Invest. 2019;42:1019–27.CrossRef
16.
Zurück zum Zitat Yan X, Li P, Tang Z, Feng B. The relationship between bile acid concentration, glucagon-like-peptide 1, fibroblast growth factor 15 and bile acid receptors in rats during progression of glucose intolerance. BMC Endocr Disord. 2017;17(1):60.CrossRef Yan X, Li P, Tang Z, Feng B. The relationship between bile acid concentration, glucagon-like-peptide 1, fibroblast growth factor 15 and bile acid receptors in rats during progression of glucose intolerance. BMC Endocr Disord. 2017;17(1):60.CrossRef
17.
Zurück zum Zitat Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev. 2007;87(4):1409–39.CrossRef Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev. 2007;87(4):1409–39.CrossRef
18.
Zurück zum Zitat Orskov C, Holst JJ, Nielsen OV. Effect of truncated glucagon-like peptide-1 [proglucagon-(78-107) amide] on endocrine secretion from pig pancreas, antrum, and nonantral stomach. Endocrinology. 1988;123(4):2009–13.CrossRef Orskov C, Holst JJ, Nielsen OV. Effect of truncated glucagon-like peptide-1 [proglucagon-(78-107) amide] on endocrine secretion from pig pancreas, antrum, and nonantral stomach. Endocrinology. 1988;123(4):2009–13.CrossRef
19.
Zurück zum Zitat Zhuang L, Su JB, Zhang XL, Huang HY, Zhao LH, Xu F, Chen T, Wang XQ, Wu G, Wang XH. Serum amylase levels in relation to islet beta cell function in patients with early type 2 diabetes. PLoS ONE. 2016;11(9):e0162204.CrossRef Zhuang L, Su JB, Zhang XL, Huang HY, Zhao LH, Xu F, Chen T, Wang XQ, Wu G, Wang XH. Serum amylase levels in relation to islet beta cell function in patients with early type 2 diabetes. PLoS ONE. 2016;11(9):e0162204.CrossRef
20.
Zurück zum Zitat Retnakaran R, Shen S, Hanley AJ, Vuksan V, Hamilton JK, Zinman B. Hyperbolic relationship between insulin secretion and sensitivity on oral glucose tolerance test. Obesity (Silver Spring). 2008;16(8):1901–7.CrossRef Retnakaran R, Shen S, Hanley AJ, Vuksan V, Hamilton JK, Zinman B. Hyperbolic relationship between insulin secretion and sensitivity on oral glucose tolerance test. Obesity (Silver Spring). 2008;16(8):1901–7.CrossRef
21.
Zurück zum Zitat Retnakaran R, Qi Y, Goran MI, Hamilton JK. Evaluation of proposed oral disposition index measures in relation to the actual disposition index. Diabet Med. 2009;26(12):1198–203.CrossRef Retnakaran R, Qi Y, Goran MI, Hamilton JK. Evaluation of proposed oral disposition index measures in relation to the actual disposition index. Diabet Med. 2009;26(12):1198–203.CrossRef
22.
Zurück zum Zitat Izaguirre M, Gil MJ, Monreal I, Montecucco F, Fruhbeck G, Catalan V. The role and potential therapeutic implications of the fibroblast growth factors in energy balance and type 2 diabetes. Curr Diab Rep. 2017;17(6):43.CrossRef Izaguirre M, Gil MJ, Monreal I, Montecucco F, Fruhbeck G, Catalan V. The role and potential therapeutic implications of the fibroblast growth factors in energy balance and type 2 diabetes. Curr Diab Rep. 2017;17(6):43.CrossRef
23.
Zurück zum Zitat Kir S, Beddow SA, Samuel VT, Miller P, Previs SF, Suino-Powell K, Xu HE, Shulman GI, Kliewer SA, Mangelsdorf DJ. FGF19 as a postprandial, insulin-independent activator of hepatic protein and glycogen synthesis. Science. 2011;331(6024):1621–4.CrossRef Kir S, Beddow SA, Samuel VT, Miller P, Previs SF, Suino-Powell K, Xu HE, Shulman GI, Kliewer SA, Mangelsdorf DJ. FGF19 as a postprandial, insulin-independent activator of hepatic protein and glycogen synthesis. Science. 2011;331(6024):1621–4.CrossRef
24.
Zurück zum Zitat Lan T, Morgan DA, Rahmouni K, Sonoda J, Fu X, Burgess SC, Holland WL, Kliewer SA, Mangelsdorf DJ. FGF19, FGF21, and an FGFR1/beta-Klotho-activating antibody act on the nervous system to regulate body weight and glycemia. Cell Metab. 2017;26(5):709.e703–718.e703.CrossRef Lan T, Morgan DA, Rahmouni K, Sonoda J, Fu X, Burgess SC, Holland WL, Kliewer SA, Mangelsdorf DJ. FGF19, FGF21, and an FGFR1/beta-Klotho-activating antibody act on the nervous system to regulate body weight and glycemia. Cell Metab. 2017;26(5):709.e703–718.e703.CrossRef
26.
Zurück zum Zitat Wang Y, Dang N, Sun P, Xia J, Zhang C, Pang S. The effects of metformin on fibroblast growth factor 19, 21 and fibroblast growth factor receptor 1 in high-fat diet and streptozotocin induced diabetic rats. Endocr J. 2017;64:543–52.CrossRef Wang Y, Dang N, Sun P, Xia J, Zhang C, Pang S. The effects of metformin on fibroblast growth factor 19, 21 and fibroblast growth factor receptor 1 in high-fat diet and streptozotocin induced diabetic rats. Endocr J. 2017;64:543–52.CrossRef
27.
Zurück zum Zitat Sonne DP, van Nierop FS, Kulik W, Soeters MR, Vilsbøll T, Knop FK. Postprandial plasma concentrations of individual bile acids and FGF-19 in patients with type 2 diabetes. J Clin Endocrinol Metab. 2016;101:3002–9.CrossRef Sonne DP, van Nierop FS, Kulik W, Soeters MR, Vilsbøll T, Knop FK. Postprandial plasma concentrations of individual bile acids and FGF-19 in patients with type 2 diabetes. J Clin Endocrinol Metab. 2016;101:3002–9.CrossRef
28.
Zurück zum Zitat Lundasen T, Galman C, Angelin B, Rudling M. Circulating intestinal fibroblast growth factor 19 has a pronounced diurnal variation and modulates hepatic bile acid synthesis in man. J Intern Med. 2006;260(6):530–6.CrossRef Lundasen T, Galman C, Angelin B, Rudling M. Circulating intestinal fibroblast growth factor 19 has a pronounced diurnal variation and modulates hepatic bile acid synthesis in man. J Intern Med. 2006;260(6):530–6.CrossRef
29.
Zurück zum Zitat Meyer C, Pimenta W, Woerle HJ, Van Haeften T, Szoke E, Mitrakou A, Gerich J. Different mechanisms for impaired fasting glucose and impaired postprandial glucose tolerance in humans. Diabetes Care. 2006;29(8):1909–14.CrossRef Meyer C, Pimenta W, Woerle HJ, Van Haeften T, Szoke E, Mitrakou A, Gerich J. Different mechanisms for impaired fasting glucose and impaired postprandial glucose tolerance in humans. Diabetes Care. 2006;29(8):1909–14.CrossRef
31.
Zurück zum Zitat Prawitt J, Abdelkarim M, Stroeve JH, Popescu I, Duez H, Velagapudi VR, Dumont J, Bouchaert E, van Dijk TH, Lucas A, et al. Farnesoid X receptor deficiency improves glucose homeostasis in mouse models of obesity. Diabetes. 2011;60(7):1861–71.CrossRef Prawitt J, Abdelkarim M, Stroeve JH, Popescu I, Duez H, Velagapudi VR, Dumont J, Bouchaert E, van Dijk TH, Lucas A, et al. Farnesoid X receptor deficiency improves glucose homeostasis in mouse models of obesity. Diabetes. 2011;60(7):1861–71.CrossRef
32.
Zurück zum Zitat Jiang C, Xie C, Lv Y, Li J, Krausz KW, Shi J, Brocker CN, Desai D, Amin SG, Bisson WH, et al. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat Commun. 2015;6:10166.CrossRef Jiang C, Xie C, Lv Y, Li J, Krausz KW, Shi J, Brocker CN, Desai D, Amin SG, Bisson WH, et al. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat Commun. 2015;6:10166.CrossRef
33.
Zurück zum Zitat Struik D, Dommerholt MB, Jonker JW. Fibroblast growth factors in control of lipid metabolism: from biological function to clinical application. Curr Opin Lipidol. 2019;30(3):235–43.CrossRef Struik D, Dommerholt MB, Jonker JW. Fibroblast growth factors in control of lipid metabolism: from biological function to clinical application. Curr Opin Lipidol. 2019;30(3):235–43.CrossRef
Metadaten
Titel
Serum fibroblast growth factor 19 and endogenous islet beta cell function in type 2 diabetic patients
verfasst von
Meng-jie Tang
Jian‑bin Su
Tian-li Xu
Xue‑qin Wang
Dong-mei Zhang
Xiao-hua Wang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Diabetology & Metabolic Syndrome / Ausgabe 1/2019
Elektronische ISSN: 1758-5996
DOI
https://doi.org/10.1186/s13098-019-0475-1

Weitere Artikel der Ausgabe 1/2019

Diabetology & Metabolic Syndrome 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

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

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.

Update Innere Medizin

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