scholarly journals C-terminal site-specific PEGylated Exendin-4 analog: A long-acting glucagon like Peptide-1 receptor agonist, on glycemic control and beta cell function in diabetic db/db mice

2018 ◽  
Vol 138 (1) ◽  
pp. 23-30 ◽  
Author(s):  
Daoqi Tang ◽  
Hong Tian ◽  
Jicheng Wu ◽  
Jiaxiao Cheng ◽  
Cheng Luo ◽  
...  
2019 ◽  
Vol 22 (4) ◽  
pp. 640-647 ◽  
Author(s):  
Roberto Visentin ◽  
Michele Schiavon ◽  
Britta Göbel ◽  
Michela Riz ◽  
Claudio Cobelli ◽  
...  

2019 ◽  
Vol 13 (3) ◽  
pp. 2142-2147 ◽  
Author(s):  
Kalyani Sridharan ◽  
Raja Kalayarasan ◽  
Sadishkumar Kamalanathan ◽  
Jayaprakash Sahoo ◽  
Sitanshu Sekhar Kar ◽  
...  

2006 ◽  
Vol 74 (2) ◽  
pp. 189-193 ◽  
Author(s):  
Michelle Fung ◽  
David Thompson ◽  
R. Jean Shapiro ◽  
Garth L. Warnock ◽  
Dana K. Andersen ◽  
...  

2015 ◽  
Vol 2015 ◽  
pp. 1-17 ◽  
Author(s):  
Shaocong Hou ◽  
Caina Li ◽  
Yi Huan ◽  
Shuainan Liu ◽  
Quan Liu ◽  
...  

Glucagon like peptide-1 (GLP-1) receptor agonists such as exendin-4 have been widely used but their short half-life limits their therapeutic value. The recombinant protein, E2HSA, is a novel, long-acting GLP-1 receptor agonist generated by the fusion of exendin-4 with human serum albumin. In mouse pancreatic NIT-1 cells, E2HSA activated GLP-1 receptor with similar efficacy as exendin-4. After single-dose administration in ICR mice, E2HSA showed prolonged glucose lowering effects which lasted up to four days and extended inhibition on gastric emptying for at least 72 hours. Chronic E2HSA treatment in db/db mice significantly improved glucose tolerance, reduced elevated nonfasting and fasting plasma glucose levels, and also decreased HbA1c levels. E2HSA also increased insulin secretion and decreased body weight and appetite. Furthermore, immunofluorescence analysis showed that E2HSA increasedβ-cell area, improved islet morphology, and reducedβ-cell apoptosis. In accordance with the promotion ofβ-cell function and survival, E2HSA upregulated genes such as Irs2, Pdx-1, Nkx6.1, and MafA and downregulated the expression levels of FoxO1 and proapoptotic Bcl-2 family proteins. In conclusion, with prolonged glucose lowering effects and promotingβ-cell function and survival, the fusion protein, E2HSA, is a promising new therapeutic for once weekly treatment of type 2 diabetes.


Diabetes ◽  
2020 ◽  
Vol 69 (Supplement 1) ◽  
pp. 2106-P
Author(s):  
YANNA SU ◽  
WEN XU ◽  
BEISI LIN ◽  
ZIYU LIU ◽  
YALAN CHEN ◽  
...  

2012 ◽  
Vol 2012 ◽  
pp. 1-5 ◽  
Author(s):  
Haoyong Yu ◽  
Ruixia Li ◽  
Lei Zhang ◽  
Haibing Chen ◽  
Yuqian Bao ◽  
...  

CA19-9 is a tumor-associated antigen. It is also a marker of pancreatic tissue damage that might be caused by diabetes. Long-term poor glycemic control may lead to pancreatic beta cell dysfunction which is reflected by elevated serum CA19-9 level. Intracellular cholesterol accumulation leads to islet dysfunction and impaired insulin secretion which provide a new lipotoxic model. This study firstly found total cholesterol was one of the independent contributors to CA19-9. Elevated serum CA19-9 level in diabetic patients may indicate further investigations of glycemic control, pancreatic beta cell function, and total cholesterol level.


2018 ◽  
Vol 315 (4) ◽  
pp. R595-R608 ◽  
Author(s):  
Jacob D. Brown ◽  
Danielle McAnally ◽  
Jennifer E. Ayala ◽  
Melissa A. Burmeister ◽  
Camilo Morfa ◽  
...  

Long-acting glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists (GLP-1RA), such as exendin-4 (Ex4), promote weight loss. On the basis of a newly discovered interaction between GLP-1 and oleoylethanolamide (OEA), we tested whether OEA enhances GLP-1RA-mediated anorectic signaling and weight loss. We analyzed the effect of GLP-1+OEA and Ex4+OEA on canonical GLP-1R signaling and other proteins/pathways that contribute to the hypophagic action of GLP-1RA (AMPK, Akt, mTOR, and glycolysis). We demonstrate that OEA enhances canonical GLP-1R signaling when combined with GLP-1 but not with Ex4. GLP-1 and Ex4 promote phosphorylation of mTOR pathway components, but OEA does not enhance this effect. OEA synergistically enhanced GLP-1- and Ex4-stimulated glycolysis but did not augment the hypophagic action of GLP-1 or Ex4 in lean or diet-induced obese (DIO) mice. However, the combination of Ex4+OEA promoted greater weight loss in DIO mice than Ex4 or OEA alone during a 7-day treatment. This was due in part to transient hypophagia and increased energy expenditure, phenotypes also observed in Ex4-treated DIO mice. Thus, OEA augments specific GLP-1RA-stimulated signaling but appears to work in parallel with Ex4 to promote weight loss in DIO mice. Elucidating cooperative mechanisms underlying Ex4+OEA-mediated weight loss could, therefore, be leveraged toward more effective obesity therapies.


Sign in / Sign up

Export Citation Format

Share Document