scholarly journals Cyanidin Stimulates Insulin Secretion and Pancreatic β-Cell Gene Expression through Activation of l-type Voltage-Dependent Ca2+ Channels

Nutrients ◽  
2017 ◽  
Vol 9 (8) ◽  
pp. 814 ◽  
Author(s):  
Tanyawan Suantawee ◽  
Sara Elazab ◽  
Walter Hsu ◽  
Shaomian Yao ◽  
Henrique Cheng ◽  
...  
2016 ◽  
Vol 40 (5) ◽  
pp. 486-500 ◽  
Author(s):  
Zahra Mehrfarjam ◽  
Fariba Esmaeili ◽  
Leila Shabani ◽  
Esmaeil Ebrahimie

1985 ◽  
Vol 13 (4) ◽  
pp. 680-681 ◽  
Author(s):  
ROSA FERRER ◽  
SALVADOR SALA ◽  
JUAN VICENTE SANCHEZ-ANDRES ◽  
BERNAT SORIA

2018 ◽  
Vol 61 (1) ◽  
pp. 1-12 ◽  
Author(s):  
Weijuan Shao ◽  
Vivian Szeto ◽  
Zhuolun Song ◽  
Lili Tian ◽  
Zhong-Ping Feng ◽  
...  

Pancreatic β-cell Tcf7l2 deletion or its functional knockdown suggested the essential role of this Wnt pathway effector in controlling insulin secretion, glucose homeostasis and β-cell gene expression. As the LIM homeodomain protein ISL1 is a suggested Wnt pathway downstream target, we hypothesize that it mediates metabolic functions of TCF7L2. We aimed to determine the role of ISL1 in mediating the function of TCF7L2 and the incretin hormone GLP-1 in pancreatic β-cells. The effect of dominant negative TCF7L2 (TCF7L2DN) mediated Wnt pathway functional knockdown on Isl1 expression was determined in βTCFDN mouse islets and in the rat insulinoma cell line INS-1 832/13. Luciferase reporter assay and chromatin immunoprecipitation were utilized to determine whether Isl1 is a direct downstream target of Tcf7l2. TCF7L2DN adenovirus infection and siRNA-mediated Isl1 knockdown on β-cell gene expression were compared. Furthermore, Isl1 knockdown on GLP-1 stimulated β-catenin S675 phosphorylation and insulin secretion was determined. We found that TCF7L2DN repressed ISL1 levels in βTCFDN islets and the INS-1 832/13 cell line. Wnt stimulators enhanced Isl1 promoter activity and binding of TCF7L2 on Isl1 promoter. TCF7L2DN adenovirus infection and Isl1 knockdown generated similar repression on expression of β-cell genes, including the ones that encode GLUT2 and GLP-1 receptor. Either TCF7L2DN adenovirus infection or Isl1 knockdown attenuated GLP-1-stimulated β-catenin S675 phosphorylation in INS-1 832/13 cells or mouse islets and GLP-1 stimulated insulin secretion in INS-1 832/13 or MIN6 cells. Our observations support the existence of TCF7L2–ISL1 transcriptional network, and we suggest that this network also mediates β-cell function of GLP-1.


2017 ◽  
Vol 9 (4) ◽  
pp. 1246-1261 ◽  
Author(s):  
Maja Borup Kjær Petersen ◽  
Ajuna Azad ◽  
Camilla Ingvorsen ◽  
Katja Hess ◽  
Mattias Hansson ◽  
...  

PLoS ONE ◽  
2018 ◽  
Vol 13 (10) ◽  
pp. e0204595
Author(s):  
Ahmed I. Mahmoud ◽  
Francisco X. Galdos ◽  
Katherine A. Dinan ◽  
Mark P. Jedrychowski ◽  
Jeffrey C. Davis ◽  
...  

2020 ◽  
Vol 598 (21) ◽  
pp. 4887-4905 ◽  
Author(s):  
Matthew T. Dickerson ◽  
Prasanna K. Dadi ◽  
Regan B. Butterworth ◽  
Arya Y. Nakhe ◽  
Sarah M. Graff ◽  
...  

Endocrinology ◽  
2015 ◽  
Vol 156 (7) ◽  
pp. 2440-2450 ◽  
Author(s):  
Danielle Andrzejewski ◽  
Melissa L. Brown ◽  
Nathan Ungerleider ◽  
Amy Burnside ◽  
Alan L. Schneyer

TGFβ superfamily ligands, receptors, and second messengers, including activins A and B, have been identified in pancreatic islets and proposed to have important roles regulating development, proliferation, and function. We previously demonstrated that Fstl3 (an antagonist of activin activity) null mice have larger islets with β-cell hyperplasia and improved glucose tolerance and insulin sensitivity in the absence of altered β-cell proliferation. This suggested the hypothesis that increased activin signaling influences β-cell expansion by destabilizing the α-cell phenotype and promoting transdifferentiation to β-cells. We tested the first part of this hypothesis by treating α- and β-cell lines and sorted mouse islet cells with activin and related ligands. Treatment of the αTC1-6 α cell line with activins A or B suppressed critical α-cell gene expression, including Arx, glucagon, and MafB while also enhancing β-cell gene expression. In INS-1E β-cells, activin A treatment induced a significant increase in Pax4 (a fate determining β-cell gene) and insulin expression. In sorted primary islet cells, α-cell gene expression was again suppressed by activin treatment in α-cells, whereas Pax4 was enhanced in β-cells. Activin treatment in both cell lines and primary cells resulted in phosphorylated mothers against decapentaplegic-2 phosphorylation. Finally, treatment of αTC1-6 cells with activins A or B significantly inhibited proliferation. These results support the hypothesis that activin signaling destabilized the α-cell phenotype while promoting a β-cell fate. Moreover, these results support a model in which the β-cell expansion observed in Fstl3 null mice may be due, at least in part, to enhanced α- to β-cell transdifferentiation.


Sign in / Sign up

Export Citation Format

Share Document