Preventing β-cell loss and diabetes with calcium channel blockers

Guanlan Xu, Junqin Chen, Gu Jing, Anath Shalev, Guanlan Xu, Junqin Chen, Gu Jing, Anath Shalev

Abstract

Although loss of functional β-cell mass is a hallmark of diabetes, no treatment approaches that halt this process are currently available. We recently identified thioredoxin-interacting protein (TXNIP) as an attractive target in this regard. Glucose and diabetes upregulate β-cell TXNIP expression, and TXNIP overexpression induces β-cell apoptosis. In contrast, genetic ablation of TXNIP promotes endogenous β-cell survival and prevents streptozotocin (STZ)- and obesity-induced diabetes. Finding an oral medication that could inhibit β-cell TXNIP expression would therefore represent a major breakthrough. We were surprised to discover that calcium channel blockers inhibited TXNIP expression in INS-1 cells and human islets and that orally administered verapamil reduced TXNIP expression and β-cell apoptosis, enhanced endogenous insulin levels, and rescued mice from STZ-induced diabetes. Verapamil also promoted β-cell survival and improved glucose homeostasis and insulin sensitivity in BTBR ob/ob mice. Our data further suggest that this verapamil-mediated TXNIP repression is conferred by reduction of intracellular calcium, inhibition of calcineurin signaling, and nuclear exclusion and decreased binding of carbohydrate response element-binding protein to the E-box repeat in the TXNIP promoter. Thus, for the first time, we have identified an oral medication that can inhibit proapoptotic β-cell TXNIP expression, enhance β-cell survival and function, and prevent and even improve overt diabetes.

Figures

FIG. 1.
FIG. 1.
Effects of calcium channel blockers on β-cell TXNIP expression. A: INS-1 cells were incubated for 24 h with diltiazem (50 μmol/L), verapamil (50 μmol/L), NiCl2 (500 μmol/L), or the calcium chelator EGTA (500 μmol/L), and TXNIP expression was assessed by quantitative RT-PCR. B: INS-1 cells were incubated for 48 h in the presence (V) or absence (C) of verapamil (50 μmol/L), and effects on TXNIP protein levels were assessed by Western blotting and corrected for β-actin. C: Isolated human islets were incubated for 24 h at the designated concentrations of verapamil, and TXNIP expression was assessed by quantitative RT-PCR. D: C57BL/6 mice received verapamil (100 mg/kg/day) in their drinking water for 3 weeks, and their islet TXNIP expression was assessed by quantitative RT-PCR. Bars represent means ± SEM of at least three independent experiments.
FIG. 2.
FIG. 2.
Verapamil effects on TXNIP promoter activity. INS-1 cells were grown in 12-well plates and transfected with different TXNIP promoter reporter plasmids (0.4 μg/well) together with pRL-TK control plasmid (20 ng/well) using DharmaFECT Duo transfection reagent (1 μL/well). Verapamil was added to the medium with a final concentration of 100 μmol/L. At 24 h after transfection, cells were harvested and luciferase activity was determined by Dual Luciferase assay kit. Bars represent mean luciferase activity ± SEM of at least three independent experiments as compared with no verapamil control.
FIG. 3.
FIG. 3.
ChIP analysis of ChREBP binding to the TXNIP promoter in response to verapamil. INS-1 cells were treated with verapamil for the designated times, and ChIP assays were performed as described previously (11) using 4 μg ChREBP antibody (A) or normal rabbit IgG (B), and real-time PCR was performed using primers flanking the E-boxes. Effects of ChREBP overexpression on verapamil-mediated inhibition of TXNIP expression. INS-1 cells were grown in 6-well plates and transfected with control LacZ (C) or ChREBP plasmids (2 μg/well) (D). At 24 h after transfection, verapamil was added to the medium with a final concentration of 50 μmol/L; 48 h after transfection, cells were harvested for RNA extraction and analyzed by quantitative RT-PCR. Bars represent means ± SEM of at least three independent experiments.
FIG. 4.
FIG. 4.
Changes in nuclear ChREBP in response to verapamil and CyA. INS-1 cells were incubated in the presence or absence of verapamil (50 μmol/L) for 24 h, and ChREBP levels were analyzed by Western blotting in whole cell extracts (A) or nuclear fractions (B) as detailed in Research Design and Methods. C: INS-1 cells were transfected with the FL-TXNIP promoter reporter plasmid (0.4 μg/well) together with pRL-TK control plasmid (20 ng/well) using DharmaFECT Duo transfection reagent (1 μL/well) and incubated in the presence or absence of the calcineurin inhibitor CyA (5 μmol/L) for 24 h. Bars represent luciferase activity (mean ± SEM) of three independent experiments. D: INS-1 cells were incubated in the presence or absence of CyA (5 μmol/L) for 24 h, and ChREBP levels were assessed in the nuclear fractions (n = 3).
FIG. 5.
FIG. 5.
Verapamil effects on STZ-induced diabetes and β-cell apoptosis. Male C57BL/6 mice received multiple low-dose STZ injections (40 mg/kg/day) for 5 days and then half the mice received verapamil (V) in their drinking water (100 mg/kg/day). A: Blood glucose (mean ± SEM) in STZ- and STZ + verapamil–treated mice (n = 3 mice per group). B: Verapamil effects on islet TXNIP protein levels in STZ-induced diabetic mice. S, STZ. C: Comparison of pancreas cross-section of STZ- and STZ + verapamil–treated mice (blue = insulin staining). D and E: β-Cell apoptosis as assessed by TUNEL in response to verapamil treatment in STZ-induced diabetic mice. pos, positive. White arrows point at apoptotic nuclei. In total, >2,000 β-cell nuclei were analyzed per group; bars represent means ± SEM. (A high-quality digital representation of this figure is available in the online issue.)
FIG. 6.
FIG. 6.
Verapamil (V) effects on glucose homeostasis and β-cell function and survival in overtly diabetic mice. Male C57BL/6 mice received multiple low-dose STZ injections (40 mg/kg/day) for 5 days. At day 15, overtly diabetic mice (blood glucose >250 mg/dL) were selected to receive verapamil in their drinking water at a regular dose of 1 mg/mL or at a 50% lower dose of 0.5 mg/mL or to serve as controls. A: Blood glucose (mean ± SEM) in the three experimental groups. P values designate level of significance compared with the STZ control group (n = 4–8 mice per group). B: Serum insulin levels (mean ± SEM) in STZ-induced diabetic mice and STZ + verapamil–treated (0.5 mg/mL) mice (n = 6–7). C: Comparison of pancreas cross-section of STZ- and STZ + verapamil–treated mice (blue = insulin staining). (A high-quality digital representation of this figure is available in the online issue.)
FIG. 7.
FIG. 7.
Verapamil (V) effects on obesity-induced diabetes and β-cell survival. BTBRlepob/ob mice, as a model of severe obesity, insulin resistance, and diabetes, received verapamil (1 mg/mL) in their drinking water starting at age 4 weeks. A: Blood glucose (mean ± SEM) in verapamil-treated and control ob/ob mice (n = 8–10 mice per group). W, weeks. B: ITTs were performed as described in research design and methods. Blood glucose (mean ± SEM) in verapamil-treated and control ob/ob mice is shown (n = 4). ■, verapamil-treated mice; ○, control ob/ob mice. C: Serum insulin levels (mean ± SEM) in 8-week-old ob/ob (white bar) and verapamil-treated ob/ob (black bar) mice (n = 4–5). D and E: β-Cell apoptosis as assessed by TUNEL in response to verapamil treatment in obesity-induced BTBR ob/ob mice. pos, positive. White arrows point at apoptotic nuclei. In total, >3,000 β-cell nuclei were analyzed per group; bars represent means ± SEM. (A high-quality digital representation of this figure is available in the online issue.)

References

    1. Shalev A, Pise-Masison CA, Radonovich M, et al. Oligonucleotide microarray analysis of intact human pancreatic islets: identification of glucose-responsive genes and a highly regulated TGFbeta signaling pathway. Endocrinology 2002;143:3695–3698
    1. Chen J, Hui ST, Couto FM, et al. Thioredoxin-interacting protein deficiency induces Akt/Bcl-xL signaling and pancreatic beta-cell mass and protects against diabetes. FASEB J 2008;22:3581–3594
    1. Chen J, Saxena G, Mungrue IN, Lusis AJ, Shalev A. Thioredoxin-interacting protein: a critical link between glucose toxicity and beta-cell apoptosis. Diabetes 2008;57:938–944
    1. Minn AH, Hafele C, Shalev A. Thioredoxin-interacting protein is stimulated by glucose through a carbohydrate response element and induces beta-cell apoptosis. Endocrinology 2005;146:2397–2405
    1. Minn AH, Pise-Masison CA, Radonovich M, et al. Gene expression profiling in INS-1 cells overexpressing thioredoxin-interacting protein. Biochem Biophys Res Commun 2005;336:770–778
    1. Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol 2010;11:136–140
    1. Chen J, Cha-Molstad H, Szabo A, Shalev A. Diabetes induces and calcium channel blockers prevent cardiac expression of proapoptotic thioredoxin-interacting protein. Am J Physiol Endocrinol Metab 2009;296:E1133–E1139
    1. Minn AH, Lan H, Rabaglia ME, et al. Increased insulin translation from an insulin splice-variant overexpressed in diabetes, obesity, and insulin resistance. Mol Endocrinol 2005;19:794–803
    1. Lan H, Rabaglia ME, Stoehr JP, et al. Gene expression profiles of nondiabetic and diabetic obese mice suggest a role of hepatic lipogenic capacity in diabetes susceptibility. Diabetes 2003;52:688–700
    1. Cohn RD, Durbeej M, Moore SA, Coral-Vazquez R, Prouty S, Campbell KP. Prevention of cardiomyopathy in mouse models lacking the smooth muscle sarcoglycan-sarcospan complex. J Clin Invest 2001;107:R1–R7
    1. Cha-Molstad H, Saxena G, Chen J, Shalev A. Glucose-stimulated expression of Txnip is mediated by carbohydrate response element-binding protein, p300, and histone H4 acetylation in pancreatic beta cells. J Biol Chem 2009;284:16898–16905
    1. Saxena G, Chen J, Shalev A. Intracellular shuttling and mitochondrial function of thioredoxin-interacting protein. J Biol Chem 2010;285:3997–4005
    1. Denechaud PD, Bossard P, Lobaccaro JM, et al. ChREBP, but not LXRs, is required for the induction of glucose-regulated genes in mouse liver. J Clin Invest 2008;118:956–964
    1. Parikh H, Carlsson E, Chutkow WA, et al. TXNIP regulates peripheral glucose metabolism in humans. PLoS Med 2007;4:e158.
    1. Chutkow WA, Patwari P, Yoshioka J, Lee RT. Thioredoxin-interacting protein (Txnip) is a critical regulator of hepatic glucose production. J Biol Chem 2008;283:2397–2406
    1. Davies MN, O’Callaghan BL, Towle HC. Glucose activates ChREBP by increasing its rate of nuclear entry and relieving repression of its transcriptional activity. J Biol Chem 2008;283:24029–24038
    1. Yamashita H, Takenoshita M, Sakurai M, et al. A glucose-responsive transcription factor that regulates carbohydrate metabolism in the liver. Proc Natl Acad Sci U S A 2001;98:9116–9121
    1. Soleimanpour SA, Crutchlow MF, Ferrari AM, et al. Calcineurin signaling regulates human islet beta-cell survival. J Biol Chem 2010;285:40050–40059
    1. Afzal N, Ganguly PK, Dhalla KS, Pierce GN, Singal PK, Dhalla NS. Beneficial effects of verapamil in diabetic cardiomyopathy. Diabetes 1988;37:936–942
    1. Cooper-Dehoff R, Cohen JD, Bakris GL, et al. ; INVEST Investigators Predictors of development of diabetes mellitus in patients with coronary artery disease taking antihypertensive medications (findings from the INternational VErapamil SR-Trandolapril STudy [INVEST]). Am J Cardiol 2006;98:890–894
    1. Cooper-DeHoff RM, Aranda JM, Jr, Gaxiola E, et al. ; INVEST Investigators Blood pressure control and cardiovascular outcomes in high-risk Hispanic patients—findings from the International Verapamil SR/Trandolapril Study (INVEST). Am Heart J 2006;151:1072–1079

Source: PubMed

3
Abonner