Topic Highlight Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Diabetes. Dec 15, 2013; 4(6): 263-269
Published online Dec 15, 2013. doi: 10.4239/wjd.v4.i6.263
Down-regulation of pancreatic transcription factors and incretin receptors in type 2 diabetes
Hideaki Kaneto, Taka-aki Matsuoka, Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
Author contributions: Kaneto H and Matsuoka TA wrote the paper.
Correspondence to: Hideaki Kaneto, MD, PhD, Department of Metabolic Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan. kaneto@endmet.med.osaka-u.ac.jp
Telephone: +81-6-68793743 Fax: +81-6-68793739
Received: August 26, 2013
Revised: October 26, 2013
Accepted: November 15, 2013
Published online: December 15, 2013

Abstract

Type 2 diabetes is one of the most prevalent and serious metabolic diseases. Under diabetic conditions, chronic hyperglycemia and subsequent induction of oxidative stress deteriorate pancreatic β-cell function, which leads to the aggravation of type 2 diabetes. Although such phenomena are well known as glucose toxicity, its molecular mechanism remains unclear. In this review article, we describe the possible molecular mechanism for β-cell dysfunction found in type 2 diabetes, focusing on (1) oxidative stress, (2) pancreatic transcription factors (PDX-1 and MafA) and (3) incretin receptors (GLP-1 and GIP receptors). Under such conditions, nuclear expression levels of PDX-1 and MafA are decreased, which leads to suppression of insulin biosynthesis and secretion. In addition, expression levels of GLP-1 and GIP receptors are decreased, which likely contributes to the impaired incretin effects found in diabetes. Taken together, it is likely that down-regulation of pancreatic transcription factors (PDX-1 and MafA) and down-regulation of incretin receptors (GLP-1 and GIP receptors) explain, at least in part, the molecular mechanism for β-cell dysfunction found in type 2 diabetes.

Key Words: Pancreatic β-cells, Oxidative stress, Pancreatic duodenal homeobox-1, MafA, Incretin receptor

Core tip: Expression of pancreatic transcription factors and incretin receptors is decreased in diabetes.



INVOLVEMENT OF OXIDATIVE STRESS IN THE DETERIORATION OF β-CELL FUNCTION FOUND IN TYPE 2 DIABETES

The development of type 2 diabetes is associated with pancreatic β-cell dysfunction and insulin resistance. First, overeating and/or obesity lead to the development of insulin resistance and normal β-cells secrete a larger amount of insulin to compensate for the increased insulin resistance. Next, large adipocytes secrete a larger amount of free fatty acids (FFAs) and/or various inflammatory cytokines which gradually deteriorate β-cell function and finally lead to the onset of diabetes. This process is known as “β-cell lipotoxicity”. Indeed, it has been reported that when islets or β-cell-derived cell line are exposed to FFAs, oxidative stress is induced, which leads to the reduction of insulin secretion[1-5]. It has also been reported that FFA-mediated induction of inducible nitric oxide synthase (iNOS) and excess nitric oxide (NO) generation are involved in the progression of β-cell dysfunction[6]. Once hyperglycemia becomes apparent, β-cell function such as insulin biosynthesis and secretion progressively deteriorates. This process is known as “β-cell glucose toxicity” which is often observed under diabetic conditions. In the diabetic state, hyperglycemia per se and subsequent induction of oxidative stress decrease insulin biosynthesis and secretion and finally bring about apoptosis[7-28] (Figure 1).

Figure 1
Figure 1 Typical progress of type 2 diabetes. The development of type 2 diabetes is associated with pancreatic β-cell dysfunction and insulin resistance. First, overeating and/or obesity lead to the development of insulin resistance and normal β-cells secrete a larger amount of insulin to compensate for the increased insulin resistance. Next, large adipocytes secrete a larger amount of free fatty acids and/or various inflammatory cytokines which gradually deteriorate β-cell function and finally lead to the onset of diabetes. This process is known as “β-cell lipotoxicity”. Once hyperglycemia becomes apparent, β-cell function progressively deteriorates; insulin biosynthesis and secretion are reduced. This process is known as “β-cell glucose toxicity” which is often observed in type 2 diabetes.

Under diabetic conditions, oxidative stress is induced and involved in the β-cell glucose toxicity[22-36]. β-cells express GLUT2, a high-Km glucose transporter, and thereby display highly efficient glucose uptake when exposed to a high glucose concentration. Indeed, it was shown that expression levels of oxidative stress markers such as 8-hydroxy-2'-deoxyguanosine (8-OHdG) and 4-hydroxy-2, 3-nonenal (4-HNE) were increased in islets under diabetic conditions[14,16]. In addition, β-cells are rather vulnerable to oxidative stress due to the relatively low expression of antioxidant enzymes such as catalase and glutathione peroxidase. Therefore, it is likely that oxidative stress is involved in the deterioration of β-cell function found in diabetes. It was shown that when β-cell-derived cell lines or isolated islets were exposed to oxidative stress, insulin gene promoter activity and mRNA expression were suppressed[19-26]. In addition, when they were exposed to oxidative stress, bindings of pancreatic transcription factors PDX-1 and/or MafA to the insulin gene promoter were reduced. It is noted here that PDX-1 plays a crucial role in pancreas development, β-cell differentiation, induction of surrogate β-cells, and maintenance of mature β-cell function[29-41] and that MafA is a β-cell-specific transcription factor and functions as a potent activator of insulin gene transcription[42-47]. Furthermore, it was shown that the decrease of insulin gene expression after chronic exposure to a high glucose concentration was prevented by treatment with antioxidants[16,19,25,26]. Reduction of expression and/or DNA binding activities of PDX-1 and/or MafA by chronic exposure to high glucose was also prevented by an antioxidant treatment. These results suggest that chronic hyperglycemia suppresses insulin biosynthesis and secretion by increasing oxidative stress, accompanied by reduction of expression and/or DNA binding activities of two important pancreatic transcription factors, PDX-1 and MafA. Therefore, it is likely that the alteration of such transcription factors explains, at least in part, the suppression of insulin biosynthesis and secretion, and thereby is involved in β-cell glucose toxicity (Figure 2).

Figure 2
Figure 2 Possible molecular mechanism for suppression of insulin biosynthesis in type 2 diabetes. Under diabetic conditions, hyperglycemia induces oxidative stress and thereby leads to suppression of insulin biosynthesis and secretion which is accompanied by reduction of nuclear pancreatic duodenal homeobox 1 (PDX-1) and MafA expression. Oxidative stress and subsequent activation of the JNK pathway translocate Foxo1 from cytoplasm to nuclei, leading to translocation of PDX-1 from nuclei to cytoplasm in β-cells. In addition, oxidative stress and subsequent induction of c-Jun expression suppress nuclear expression of MafA in β-cells. Therefore, it is likely that induction of oxidative stress and suppression of nuclear PDX-1 and MafA expression are involved in β-cell glucose toxicity found in type 2 diabetes.
MOLECULAR MECHANISM FOR DOWN-REGULATION OF NUCLEAR PDX-1 EXPRESSION UNDER DIABETIC CONDITIONS: POSSIBLE INVOLVEMENT OF OXIDATIVE STRESS AND SUBSEQUENT ACTIVATION OF THE JNK PATHWAY

It has been suggested that activation of the c-Jun N-terminal kinase (JNK) pathway is involved in pancreatic β-cell dysfunction found in type 2 diabetes. It was reported that activation of the JNK pathway is involved in reduction of insulin gene expression by oxidative stress and that suppression of the JNK pathway can protect β-cells from oxidative stress[48]. When isolated islets were exposed to oxidative stress, the JNK pathway was activated, preceding the decrease of insulin gene expression. Adenoviral overexpression of dominant-negative type JNK1 (DN-JNK) protected insulin gene expression and secretion from oxidative stress. These results were correlated with change in the binding of PDX-1 to insulin gene promoter. Adenoviral overexpression of DN-JNK preserved PDX-1 DNA binding activity in the face of oxidative stress, while wild type JNK (WT-JNK) overexpression decreased PDX-1 DNA binding activity[48]. Taken together, it is likely that activation of the JNK pathway leads to decreased PDX-1 activity and consequent suppression of insulin gene transcription found in the diabetic state. Also, it was shown that PDX-1 was transported from the nuclei to the cytoplasm in response to oxidative stress. When β-cell-derived HIT cells were exposed to oxidative stress, both intrinsically expressed PDX-1 and exogenously introduced PDX-1 moved from the nuclei to the cytoplasm[49]. DN-JNK overexpression inhibited the oxidative stress-induced PDX-1 translocation, suggesting that activation of the JNK pathway is involved in PDX-1 translocation by oxidative stress. Furthermore, leptomycin B, a specific inhibitor of the classical, leucine-rich nuclear export signal (NES), inhibited nucleo-cytoplasmic translocation of PDX-1 induced by oxidative stress[49]. Taken together, it is likely that oxidative stress induces nucleo-cytoplasmic translocation of PDX-1 through activation of the JNK pathway, which leads to reduction of its DNA binding activity and suppression of insulin biosynthesis (Figure 2).

The forkhead transcription factor Foxo1 is known as one of the important fundamental transcription factors playing a key role in the process of apoptosis, cellular proliferation and differentiation, and glucose metabolism through regulating the transcription of various target genes[50,51]. Foxo1 regulates hepatic gluconeogenesis and thus contributes to insulin resistance[52]. Insulin inhibits the function of Foxo1 through Akt/PKB-mediated phosphorylation and nuclear exclusion[53], and thereby suppresses hepatic gluconeogenesis. In addition, Foxo1 exhibits a counter localization to PDX-1 in β-cells[54], suggesting that it is involved in the deterioration of β-cell function. Moreover, Foxo1 plays a role as a mediator between the JNK pathway and PDX-1[55]. In β-cell-derived cell line HIT cells, Foxo1 changed its intracellular localization from the cytoplasm to the nucleus after exposure to oxidative stress. In contrast to Foxo1, the nuclear expression of PDX-1 was decreased and its cytoplasmic distribution was increased by oxidative stress. Activation of the JNK pathway also induced the nuclear localization of Foxo1, whereas suppression of the JNK pathway reduced the oxidative stress-induced nuclear localization of Foxo1, suggesting an involvement of the JNK pathway in Foxo1 translocation[55]. In addition, oxidative stress or activation of the JNK pathway decreased Akt phosphorylation in HIT cells, leading to the decreased phosphorylation of Foxo1 following nuclear localization. Furthermore, adenoviral Foxo1 overexpression reduced the nuclear expression of PDX-1, whereas suppression of Foxo1 by Foxo1-specific small interfering RNA retained the nuclear expression of PDX-1[55]. Taken together, oxidative stress and subsequent activation of the JNK pathway induce nuclear translocation of Foxo1 through the modification of the insulin signaling in β-cells, which leads to the nucleo-cytoplasmic translocation of PDX-1 and reduction of its DNA binding activity (Figure 2).

MOLECULAR MECHANISM FOR DOWN-REGULATION OF NUCLEAR MAFA EXPRESSION UNDER DIABETIC CONDITIONS: POSSIBLE INVOLVEMENT OF OXIDATIVE STRESS AND SUBSEQUENT INDUCTION OF C-JUN EXPRESSION

It is known that c-Jun protein level and activity are increased in response to oxidative stress in various cells[56,57]. We recently reported that c-Jun expression was not clearly detected in islets of control m/m mice and young diabetic db/db mice, but that the number of c-Jun-positive cells gradually increased with age in the islets of diabetic db/db mice[58]. This expression pattern of c-Jun paralleled the loss of insulin gene transcription factor MafA expression. Quantitative real-time PCR analysis using freshly isolated islets from db/db mice clearly showed that c-Jun mRNA level was significantly increased but that both MafA and insulin mRNA levels were markedly decreased with age[58]. These results imply that the increased level of c-Jun caused a decrease in MafA and insulin gene expression in old diabetic mice. Furthermore, in immunostaining, in db/db mice nuclear MafA expression in pancreatic islets was markedly decreased with age and was not clearly detected in old mice, whereas in control m/m mice MafA expression retained up to old age[58]. In db/db mice insulin expression was also decreased in some β-cells in which MafA was undetectable or weakly expressed. Furthermore, MafA and insulin expression was suppressed in most c-Jun-positive β-cells. Similarly, in islets of diabetic KKAy mice, the number of c-Jun-positive β-cells was increased with marked hyperglycemia, and both MafA and insulin protein levels were decreased in those cells[58]. These findings suggest that c-Jun is involved in the suppression of MafA and insulin expression under diabetic conditions. In addition, c-Jun overexpression markedly decreased insulin promoter activity, which was consistent with previous reports[59,60] (Figure 2).

Although c-Jun protein expression was almost undetectable in MIN6 cells, adenoviral c-Jun overexpression markedly suppressed MafA protein level and its DNA-binding activity in MIN6 cells[58]. Northern blotting and real-time PCR analysis also showed that c-Jun overexpression significantly suppressed MafA mRNA level. Adenoviral overexpression of c-Jun in isolated mouse islets also markedly suppressed MafA mRNA and protein levels. Consistent with these results, mRNA levels of insulin 1 and 2 and insulin content were suppressed by c-Jun overexpression in both MIN6 cells and islets[58]. These findings directly demonstrate that c-Jun suppresses the expression of both MafA and insulin. In addition, since MafA appears to not only regulate insulin expression but also to be involved in insulin secretion[61,62], it is likely that the suppression of MafA protein levels by c-Jun leads to insulin secretory defects that are often observed under diabetic conditions. In conclusion, the augmented expression of c-Jun in diabetic islets decreases MafA activity followed by reduced insulin biosynthesis and secretion, and thereby explains, at least in part, the molecular mechanism for β-cell glucose toxicity that is often observed in type 2 diabetes (Figure 2).

MOLECULAR MECHANISM FOR DOWN-REGULATION OF INCRETIN RECEPTOR EXPRESSION IN β-CELLS UNDER DIABETIC CONDITIONS: POSSIBLE INVOLVEMENT OF HYPERGLYCEMIA AND SUBSEQUENT REDUCTION OF TCF7L2 EXPRESSION

The incretin effect causes more insulin to be secreted when glucose is orally taken compared to when given intravenously, even when blood glucose levels have the same profile. This effect is thought to be very important for maximizing insulin response during meals, thereby limiting postprandial glucose excursions. Two incretins have been identified: glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP). It is thought that such incretins play an important role in glucose homeostasis by promoting insulin secretion immediately on meal ingestion. It is well known that incretins (GLP-1 and GIP) bind their incretin receptors (GLP-1 and GIP receptors) in β-cells and increase intracellular cAMP levels, leading to stimulation of insulin secretion, suppression of β-cell apoptosis and increase of β-cell growth (Figure 3).

Figure 3
Figure 3 A role of incretin signaling in pancreatic β-cells. Incretins (GLP-1 and GIP) bind their incretin receptors (GLP-1 and GIP receptors) in β-cells and increase intracellular cAMP levels, leading to stimulation of insulin secretion, suppression of β-cell apoptosis and increase of β-cell growth.

Although plasma GLP-1 and GIP levels after meals are almost normal in type 2 diabetes, striking abnormalities are observed in the action of incretin hormones in type 2 diabetes[63]. It was reported that GLP-1 and GIP receptor expression was decreased in a glucose-dependent manner in islets isolated from 90% pancreatectomized diabetic (Px) rats[64]. Such decrease was not observed after normalization of blood glucose levels with phlorizin which is known to lower blood glucose levels by preventing glucose reabsorption from the glomerular filtrate in the kidney. These results suggest that hyperglycemia per se leads to down-regulation of GLP-1 and GIP receptor expression. Furthermore, insulin response to GLP-1 or GIP was markedly reduced in islets isolated from diabetic rats compared to those from control rats[65]. These results indicate that down-regulation of GLP-1 and GIP receptor expression leads to the deterioration of β-cell function. Similar results were reported in obese type 2 diabetic db/db mice; incretin receptor expression in islets was markedly decreased at 16 wk of age in db/db mice but was preserved by normalization of blood glucose levels with insulin therapy[66]. These results strengthen the hypothesis that hyperglycemia per se leads to down-regulation of GLP-1 and GIP receptor expression (Figure 4).

Figure 4
Figure 4 Down-regulation of incretin receptors in pancreatic β-cells under diabetic conditions. Under diabetic conditions, expression of incretin receptors (GLP-1 and GIP receptors) in β-cells are down-regulated, leading to decrease of insulin secretion, increase of β-cell apoptosis and decrease of β-cell growth.

Furthermore, down-regulation of GLP-1 and GIP receptor expression was observed in type 2 diabetic subjects, as observed in diabetic rodents[66]. In addition, it was shown that transcription factor TCF7L2 is involved in down-regulation of GLP-1 and GIP receptor expression found under diabetic conditions[66]. Recent human genetics studies have revealed that common variants of the TCF7L2 gene are strongly associated with type 2 diabetes mellitus. It was shown that expression levels of GLP-1 and GIP receptors were lower in islets of type 2 diabetic subjects as well as in isolated human islets treated with siRNA to TCF7L2 (siTCF7L2). Insulin secretion stimulated by glucose, GLP-1 or GIP was also impaired in siTCF7L2-treated isolated human islets. In conclusion, we think that the down-regulation of incretin receptors by hyperglycemia is largely responsible for the impaired incretin effects and thus, at least in part, explains the molecular mechanism for β-cell dysfunction found in diabetes (Figure 4).

Footnotes

P- Reviewers: Bilotta F, Hsieh PS, Hsu YH, Salles GF S- Editor: Cui XM L- Editor: Roemmele A E- Editor: Liu XM

References
1.  Carlsson C, Borg LA, Welsh N. Sodium palmitate induces partial mitochondrial uncoupling and reactive oxygen species in rat pancreatic islets in vitro. Endocrinology. 1999;140:3422-3428.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Joseph JW, Koshkin V, Saleh MC, Sivitz WI, Zhang CY, Lowell BB, Chan CB, Wheeler MB. Free fatty acid-induced beta-cell defects are dependent on uncoupling protein 2 expression. J Biol Chem. 2004;279:51049-51056.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Wang X, Li H, De Leo D, Guo W, Koshkin V, Fantus IG, Giacca A, Chan CB, Der S, Wheeler MB. Gene and protein kinase expression profiling of reactive oxygen species-associated lipotoxicity in the pancreatic beta-cell line MIN6. Diabetes. 2004;53:129-140.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Oprescu AI, Bikopoulos G, Naassan A, Allister EM, Tang C, Park E, Uchino H, Lewis GF, Fantus IG, Rozakis-Adcock M. Free fatty acid-induced reduction in glucose-stimulated insulin secretion: evidence for a role of oxidative stress in vitro and in vivo. Diabetes. 2007;56:2927-2937.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Bikopoulos G, da Silva Pimenta A, Lee SC, Lakey JR, Der SD, Chan CB, Ceddia RB, Wheeler MB, Rozakis-Adcock M. Ex vivo transcriptional profiling of human pancreatic islets following chronic exposure to monounsaturated fatty acids. J Endocrinol. 2008;196:455-464.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Shimabukuro M, Ohneda M, Lee Y, Unger RH. Role of nitric oxide in obesity-induced beta cell disease. J Clin Invest. 1997;100:290-295.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Weir GC, Laybutt DR, Kaneto H, Bonner-Weir S, Sharma A. Beta-cell adaptation and decompensation during the progression of diabetes. Diabetes. 2001;50 Suppl 1:S154-S159.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Prentki M, Nolan CJ. Islet beta cell failure in type 2 diabetes. J Clin Invest. 2006;116:1802-1812.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Poitout V, Olson LK, Robertson RP. Chronic exposure of betaTC-6 cells to supraphysiologic concentrations of glucose decreases binding of the RIPE3b1 insulin gene transcription activator. J Clin Invest. 1996;97:1041-1046.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Poitout V, Robertson RP. Minireview: Secondary beta-cell failure in type 2 diabetes--a convergence of glucotoxicity and lipotoxicity. Endocrinology. 2002;143:339-342.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Sharma A, Fusco-DeMane D, Henderson E, Efrat S, Stein R. The role of the insulin control element and RIPE3b1 activators in glucose-stimulated transcription of the insulin gene. Mol Endocrinol. 1995;9:1468-1476.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Moran A, Zhang HJ, Olson LK, Harmon JS, Poitout V, Robertson RP. Differentiation of glucose toxicity from beta cell exhaustion during the evolution of defective insulin gene expression in the pancreatic islet cell line, HIT-T15. J Clin Invest. 1997;99:534-539.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Are oxidative stress-activated signaling pathways mediators of insulin resistance and beta-cell dysfunction? Diabetes. 2003;52:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Gorogawa Si, Kajimoto Y, Umayahara Y, Kaneto H, Watada H, Kuroda A, Kawamori D, Yasuda T, Matsuhisa M, Yamasaki Y. Probucol preserves pancreatic beta-cell function through reduction of oxidative stress in type 2 diabetes. Diabetes Res Clin Pract. 2002;57:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Harmon JS, Stein R, Robertson RP. Oxidative stress-mediated, post-translational loss of MafA protein as a contributing mechanism to loss of insulin gene expression in glucotoxic beta cells. J Biol Chem. 2005;280:11107-11113.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Ihara Y, Toyokuni S, Uchida K, Odaka H, Tanaka T, Ikeda H, Hiai H, Seino Y, Yamada Y. Hyperglycemia causes oxidative stress in pancreatic beta-cells of GK rats, a model of type 2 diabetes. Diabetes. 1999;48:927-932.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Kajimoto Y, Matsuoka T, Kaneto H, Watada H, Fujitani Y, Kishimoto M, Sakamoto K, Matsuhisa M, Kawamori R, Yamasaki Y. Induction of glycation suppresses glucokinase gene expression in HIT-T15 cells. Diabetologia. 1999;42:1417-1424.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Maechler P, Jornot L, Wollheim CB. Hydrogen peroxide alters mitochondrial activation and insulin secretion in pancreatic beta cells. J Biol Chem. 1999;274:27905-27913.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Kaneto H, Kajimoto Y, Miyagawa J, Matsuoka T, Fujitani Y, Umayahara Y, Hanafusa T, Matsuzawa Y, Yamasaki Y, Hori M. Beneficial effects of antioxidants in diabetes: possible protection of pancreatic beta-cells against glucose toxicity. Diabetes. 1999;48:2398-2406.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Kaneto H, Xu G, Song KH, Suzuma K, Bonner-Weir S, Sharma A, Weir GC. Activation of the hexosamine pathway leads to deterioration of pancreatic beta-cell function through the induction of oxidative stress. J Biol Chem. 2001;276:31099-31104.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Kaneto H, Matsuoka TA, Nakatani Y, Kawamori D, Miyatsuka T, Matsuhisa M, Yamasaki Y. Oxidative stress, ER stress, and the JNK pathway in type 2 diabetes. J Mol Med (Berl). 2005;83:429-439.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Matsuoka T, Kajimoto Y, Watada H, Kaneto H, Kishimoto M, Umayahara Y, Fujitani Y, Kamada T, Kawamori R, Yamasaki Y. Glycation-dependent, reactive oxygen species-mediated suppression of the insulin gene promoter activity in HIT cells. J Clin Invest. 1997;99:144-150.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Robertson RP, Harmon J, Tran PO, Tanaka Y, Takahashi H. Glucose toxicity in beta-cells: type 2 diabetes, good radicals gone bad, and the glutathione connection. Diabetes. 2003;52:581-587.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Robertson RP. Chronic oxidative stress as a central mechanism for glucose toxicity in pancreatic islet beta cells in diabetes. J Biol Chem. 2004;279:42351-42354.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Tanaka Y, Gleason CE, Tran PO, Harmon JS, Robertson RP. Prevention of glucose toxicity in HIT-T15 cells and Zucker diabetic fatty rats by antioxidants. Proc Natl Acad Sci USA. 1999;96:10857-10862.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Tanaka Y, Tran PO, Harmon J, Robertson RP. A role for glutathione peroxidase in protecting pancreatic beta cells against oxidative stress in a model of glucose toxicity. Proc Natl Acad Sci USA. 2002;99:12363-12368.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Sakai K, Matsumoto K, Nishikawa T, Suefuji M, Nakamaru K, Hirashima Y, Kawashima J, Shirotani T, Ichinose K, Brownlee M. Mitochondrial reactive oxygen species reduce insulin secretion by pancreatic beta-cells. Biochem Biophys Res Commun. 2003;300:216-222.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Kaneto H, Fujii J, Myint T, Miyazawa N, Islam KN, Kawasaki Y, Suzuki K, Nakamura M, Tatsumi H, Yamasaki Y. Reducing sugars trigger oxidative modification and apoptosis in pancreatic beta-cells by provoking oxidative stress through the glycation reaction. Biochem J. 1996;320:855-863.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Ohlsson H, Karlsson K, Edlund T. IPF1, a homeodomain-containing transactivator of the insulin gene. EMBO J. 1993;12:4251-4259.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Leonard J, Peers B, Johnson T, Ferreri K, Lee S, Montminy MR. Characterization of somatostatin transactivating factor-1, a novel homeobox factor that stimulates somatostatin expression in pancreatic islet cells. Mol Endocrinol. 1993;7:1275-1283.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Miller CP, McGehee RE, Habener JF. IDX-1: a new homeodomain transcription factor expressed in rat pancreatic islets and duodenum that transactivates the somatostatin gene. EMBO J. 1994;13:1145-1156.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Jonsson J, Carlsson L, Edlund T, Edlund H. Insulin-promoter-factor 1 is required for pancreas development in mice. Nature. 1994;371:606-609.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Stoffers DA, Zinkin NT, Stanojevic V, Clarke WL, Habener JF. Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence. Nat Genet. 1997;15:106-110.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Ahlgren U, Jonsson J, Jonsson L, Simu K, Edlund H. beta-cell-specific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the beta-cell phenotype and maturity onset diabetes. Genes Dev. 1998;12:1763-1768.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Ferber S, Halkin A, Cohen H, Ber I, Einav Y, Goldberg I, Barshack I, Seijffers R, Kopolovic J, Kaiser N. Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nat Med. 2000;6:568-572.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Holland AM, Hale MA, Kagami H, Hammer RE, MacDonald RJ. Experimental control of pancreatic development and maintenance. Proc Natl Acad Sci USA. 2002;99:12236-12241.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Noguchi H, Kaneto H, Weir GC, Bonner-Weir S. PDX-1 protein containing its own antennapedia-like protein transduction domain can transduce pancreatic duct and islet cells. Diabetes. 2003;52:1732-1737.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Miyazaki S, Yamato E, Miyazaki J. Regulated expression of pdx-1 promotes in vitro differentiation of insulin-producing cells from embryonic stem cells. Diabetes. 2004;53:1030-1037.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Kaneto H, Nakatani Y, Miyatsuka T, Matsuoka TA, Matsuhisa M, Hori M, Yamasaki Y. PDX-1/VP16 fusion protein, together with NeuroD or Ngn3, markedly induces insulin gene transcription and ameliorates glucose tolerance. Diabetes. 2005;54:1009-1022.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Kaneto H, Miyatsuka T, Shiraiwa T, Yamamoto K, Kato K, Fujitani Y, Matsuoka TA. Crucial role of PDX-1 in pancreas development, beta-cell differentiation, and induction of surrogate beta-cells. Curr Med Chem. 2007;14:1745-1752.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Kaneto H, Miyatsuka T, Kawamori D, Yamamoto K, Kato K, Shiraiwa T, Katakami N, Yamasaki Y, Matsuhisa M, Matsuoka TA. PDX-1 and MafA play a crucial role in pancreatic beta-cell differentiation and maintenance of mature beta-cell function. Endocr J. 2008;55:235-252.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Olbrot M, Rud J, Moss LG, Sharma A. Identification of beta-cell-specific insulin gene transcription factor RIPE3b1 as mammalian MafA. Proc Natl Acad Sci USA. 2002;99:6737-6742.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Kataoka K, Han SI, Shioda S, Hirai M, Nishizawa M, Handa H. MafA is a glucose-regulated and pancreatic beta-cell-specific transcriptional activator for the insulin gene. J Biol Chem. 2002;277:49903-49910.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Matsuoka TA, Zhao L, Artner I, Jarrett HW, Friedman D, Means A, Stein R. Members of the large Maf transcription family regulate insulin gene transcription in islet beta cells. Mol Cell Biol. 2003;23:6049-6062.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Matsuoka TA, Artner I, Henderson E, Means A, Sander M, Stein R. The MafA transcription factor appears to be responsible for tissue-specific expression of insulin. Proc Natl Acad Sci USA. 2004;101:2930-2933.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Kaneto H, Matsuoka TA, Nakatani Y, Miyatsuka T, Matsuhisa M, Hori M, Yamasaki Y. A crucial role of MafA as a novel therapeutic target for diabetes. J Biol Chem. 2005;280:15047-15052.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Matsuoka TA, Kaneto H, Stein R, Miyatsuka T, Kawamori D, Henderson E, Kojima I, Matsuhisa M, Hori M, Yamasaki Y. MafA regulates expression of genes important to islet beta-cell function. Mol Endocrinol. 2007;21:2764-2774.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Kaneto H, Xu G, Fujii N, Kim S, Bonner-Weir S, Weir GC. Involvement of c-Jun N-terminal kinase in oxidative stress-mediated suppression of insulin gene expression. J Biol Chem. 2002;277:30010-30018.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Kawamori D, Kajimoto Y, Kaneto H, Umayahara Y, Fujitani Y, Miyatsuka T, Watada H, Leibiger IB, Yamasaki Y, Hori M. Oxidative stress induces nucleo-cytoplasmic translocation of pancreatic transcription factor PDX-1 through activation of c-Jun NH(2)-terminal kinase. Diabetes. 2003;52:2896-2904.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Ogg S, Paradis S, Gottlieb S, Patterson GI, Lee L, Tissenbaum HA, Ruvkun G. The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature. 1997;389:994-999.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Accili D, Arden KC. FoxOs at the crossroads of cellular metabolism, differentiation, and transformation. Cell. 2004;117:421-426.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Nakae J, Biggs WH, Kitamura T, Cavenee WK, Wright CV, Arden KC, Accili D. Regulation of insulin action and pancreatic beta-cell function by mutated alleles of the gene encoding forkhead transcription factor Foxo1. Nat Genet. 2002;32:245-253.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Biggs WH, Meisenhelder J, Hunter T, Cavenee WK, Arden KC. Protein kinase B/Akt-mediated phosphorylation promotes nuclear exclusion of the winged helix transcription factor FKHR1. Proc Natl Acad Sci USA. 1999;96:7421-7426.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Kitamura T, Nakae J, Kitamura Y, Kido Y, Biggs WH, Wright CV, White MF, Arden KC, Accili D. The forkhead transcription factor Foxo1 links insulin signaling to Pdx1 regulation of pancreatic beta cell growth. J Clin Invest. 2002;110:1839-1847.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Kawamori D, Kaneto H, Nakatani Y, Matsuoka TA, Matsuhisa M, Hori M, Yamasaki Y. The forkhead transcription factor Foxo1 bridges the JNK pathway and the transcription factor PDX-1 through its intracellular translocation. J Biol Chem. 2006;281:1091-1098.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Devary Y, Gottlieb RA, Lau LF, Karin M. Rapid and preferential activation of the c-jun gene during the mammalian UV response. Mol Cell Biol. 1991;11:2804-2811.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Nose K, Shibanuma M, Kikuchi K, Kageyama H, Sakiyama S, Kuroki T. Transcriptional activation of early-response genes by hydrogen peroxide in a mouse osteoblastic cell line. Eur J Biochem. 1991;201:99-106.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Matsuoka TA, Kaneto H, Miyatsuka T, Yamamoto T, Yamamoto K, Kato K, Shimomura I, Stein R, Matsuhisa M. Regulation of MafA expression in pancreatic beta-cells in db/db mice with diabetes. Diabetes. 2010;59:1709-1720.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Inagaki N, Maekawa T, Sudo T, Ishii S, Seino Y, Imura H. c-Jun represses the human insulin promoter activity that depends on multiple cAMP response elements. Proc Natl Acad Sci USA. 1992;89:1045-1049.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Henderson E, Stein R. c-jun inhibits transcriptional activation by the insulin enhancer, and the insulin control element is the target of control. Mol Cell Biol. 1994;14:655-662.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Zhang C, Moriguchi T, Kajihara M, Esaki R, Harada A, Shimohata H, Oishi H, Hamada M, Morito N, Hasegawa K. MafA is a key regulator of glucose-stimulated insulin secretion. Mol Cell Biol. 2005;25:4969-4976.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Wang H, Brun T, Kataoka K, Sharma AJ, Wollheim CB. MAFA controls genes implicated in insulin biosynthesis and secretion. Diabetologia. 2007;50:348-358.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W. Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest. 1993;91:301-307.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Xu G, Kaneto H, Laybutt DR, Duvivier-Kali VF, Trivedi N, Suzuma K, King GL, Weir GC, Bonner-Weir S. Downregulation of GLP-1 and GIP receptor expression by hyperglycemia: possible contribution to impaired incretin effects in diabetes. Diabetes. 2007;56:1551-1558.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Kawashima S, Matsuoka TA, Kaneto H, Tochino Y, Kato K, Yamamoto K, Yamamoto T, Matsuhisa M, Shimomura I. Effect of alogliptin, pioglitazone and glargine on pancreatic β-cells in diabetic db/db mice. Biochem Biophys Res Commun. 2011;404:534-540.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Shu L, Matveyenko AV, Kerr-Conte J, Cho JH, McIntosh CH, Maedler K. Decreased TCF7L2 protein levels in type 2 diabetes mellitus correlate with downregulation of GIP- and GLP-1 receptors and impaired beta-cell function. Hum Mol Genet. 2009;18:2388-2399.  [PubMed]  [DOI]  [Cited in This Article: ]