Glucagon-like peptide-1 prevents methylglyoxal-induced apoptosis of beta cells through improving mitochondrial function and suppressing prolonged AMPK activation

Tien-Jyun Chang, Hsing-Chi Tseng, Meng-Wei Liu, Yi-Cheng Chang, Meng-Lun Hsieh, Lee-Ming Chuang, Tien-Jyun Chang, Hsing-Chi Tseng, Meng-Wei Liu, Yi-Cheng Chang, Meng-Lun Hsieh, Lee-Ming Chuang

Abstract

Accumulation of methylglyoxal (MG) contributes to glucotoxicity and mediates beta cell apoptosis. The molecular mechanism by which GLP-1 protects MG-induced beta cell apoptosis remains unclear. Metformin is a first-line drug for treating type 2 diabetes associated with AMPK activation. However, whether metformin prevents MG-induced beta cell apoptosis is controversial. Here, we explored the signaling pathway involved in the anti-apoptotic effect of GLP-1, and investigated whether metformin had an anti-apoptotic effect on beta cells. MG treatment induced apoptosis of beta cells, impaired mitochondrial function, and prolonged activation of AMP-dependent protein kinase (AMPK). The MG-induced pro-apoptotic effects were abolished by an AMPK inhibitor. Pretreatment of GLP-1 reversed MG-induced apoptosis, and mitochondrial dysfunction, and suppressed prolonged AMPK activation. Pretreatment of GLP-1 reversed AMPK activator 5-aminoimidazole-4-carboxamide riboside (AICAR)-induced apoptosis, and suppressed prolonged AMPK activation. However, metformin neither leads to beta cell apoptosis nor ameliorates MG-induced beta cell apoptosis. In parallel, GLP-1 also prevents MG-induced beta cell apoptosis through PKA and PI3K-dependent pathway. In conclusion, these data indicates GLP-1 but not metformin protects MG-induced beta cell apoptosis through improving mitochondrial function, and alleviating the prolonged AMPK activation. Whether adding GLP-1 to metformin provides better beta cell survival and delays disease progression remains to be validated.

Figures

Figure 1. GLP-1 protects rat insulinoma cells…
Figure 1. GLP-1 protects rat insulinoma cells RINm5F from MG-induced apoptosis.
RINm5F Cells were treated in the presence or absence of 1 mM MG with or without GLP-1 (100 nM or 300 nM). (A) Cell viability was measured by MTT assay. Data are shown as relative cell viability (mean % ± S.E. bar) as compared with that in control (n = 4). *p < 0.05. (B) Apoptosis was demonstrated by Annexin V/ Hoechst 33342 staining after incubated with indicated treatment for 17 hr. Annexin V positive cells showed green fluorescence (upper row). Condense nuclei were shown in apoptotic cells by Hoechst 33342 staining (middle row). The pictures on bright field were shown in the lower row. (C) The percentage of apoptotic cells was calculated by measuring the percentage of cells in the sub-G1 population in the indicated time by using flow cytometry with propidium iodide (PI) staining (n = 3). *p < 0.05. (D) The cell counts and percentage of apoptotic cells in the sub-G1 population after incubation with indicated treatment for 17 hr were measured by using flow cytometry with propidium iodide (PI) staining. (E) Western blot of poly(ADP-ribose) polymerase (PARP) and cleaved caspase-3. The positions of the 113 kDa and 89 kDa in Western blot represent intact PARP protein and its cleavage products, respectively. The positions of the 19 kDa and 17 kDa in Western blot represent cleaved caspase-3. GAPDH was used as an internal control.
Figure 2. GLP-1 suppressed MG-induced beta cell…
Figure 2. GLP-1 suppressed MG-induced beta cell apoptosis through protein kinase A (PKA) and PI3K dependent pathway, respectively.
RINm5F cells were treated in the absence of MG, 1 mM MG, 1 mM MG + 300 nM GLP-1, 1 mM MG + 300 nM GLP-1 + 100 μM Rp-cAMP, 1 mM MG + 300 nM GLP-1 + 10 μM H-89, 1 mM MG + 300 nM GLP-1 + 30 μM LY294002, 1 mM MG + 300 nM GLP-1 + 50 nM wortmannin, respectively. (A) Cell viability was measured by MTT assay. Data are shown as relative cell viability (mean % ± S.E. bar) as compared with that in control (n = 3). *p < 0.05. (B) Annexin-V/PI flow cytometry. LL: left lower quadrant indicated viable cells, UL: upper left quadrant indicated early apoptotic cells, UR: upper right quadrant indicated late apoptotic cells, LR: right lower quadrant indicated necrotic cells. (C) Western blot of pCREB/CREB, PDX1, cleaved caspase-3, and GAPDH was used as an internal control. (D) Western blot of pAkt/Akt, cleaved caspase-3, and GAPDH was used as an internal control.
Figure 3. GLP-1 rescued MG-induced mitochondria dysfunction…
Figure 3. GLP-1 rescued MG-induced mitochondria dysfunction and inhibited prolonged AMPK activation in RINm5F cells.
Cells were treated in the absence or presence of 1 mM MG with or without 300 nM GLP-1 for indicated time. (A) Relative intracellular ATP concentration (%) compared with control in indicated time (n = 6). *p < 0.05 (Control vs. 1 mM MG at 1 hr, 2 hr, 4 hr, 6 hr and 17 hr), #p < 0.05 (1 mM MG vs. 1 mM MG + 300 nM GLP-1 at 17 hr) (B) Relative oxygen consumption rate (%) compared with control in indicated time (n = 3). *p < 0.05 (Control vs. 1 mM MG at 2 hr, 10 hr, and 17 hr), #p < 0.05 (1 mM MG vs. 1 mM MG + 300 nM GLP-1 at 2 hr, 10 hr, and 17 hr). (C) Western blot of p-AMPK/AMPK. GAPDH was used as internal control. The bar graph showed the ratio of pAMPK/GAPDH in Western blot in the absence or presence of 1 mM MG with or without 300 nM GLP-1 in the indicated time (n = 3). *p < 0.05.
Figure 4. AMPK inhibitor partially rescues MG-induced…
Figure 4. AMPK inhibitor partially rescues MG-induced cell death and apoptosis in RINm5F cells.
Cells were treated in the presence or absence of 1 mM MG with or without compound C (C.C. 10 μM) in indicated time. (A) Cell viability was measured by MTT assay. Data are shown as relative cell viability (mean % ± S.E. bar) as compared with that in control (n = 5). *p < 0.05. (B) The percentage of apoptotic cells was calculated by measuring the percentage of cells in the sub-G1 population by using flow cytometry with propidium iodide (PI) staining (n = 3). *p < 0.05. (C) Western blot of PARP, p-AMPK/AMPK, cleaved caspase-3, and GAPDH as internal control.
Figure 5. GLP-1 rescues AMPK activator AICAR-induced…
Figure 5. GLP-1 rescues AMPK activator AICAR-induced cell death, apoptosis and AMPK activation in RINm5F cells.
(A) Cell viability was measured by MTT assay in different concentration of AICAR at indicated time. Data are shown as relative cell viability (mean % ± S.E. bar) as compared with that in control (n = 3). *p < 0.05. (B) Cell viability was measured by MTT assay in cells treated by 1 mM MG or 1.5 mM AICAR for 17 hr with or without pretreatment of 300 nM GLP-1 (n = 3). *p < 0.05. (C) Western blot of p-ACC/ACC and p-AMPK/AMPK in cells treated by 1 mM MG or 1.5 mM AICAR for 17 hr with or without pretreatment of 300 nM GLP-1. GAPDH was used as internal control. (D) Western blot of PARP in cells treated by 1 mM MG or 1.5 mM AICAR for 17 hr with or without pretreatment of 300 nM GLP-1. GAPDH was used as internal control. (E) Cell viability was measured by MTT assay in different concentration of metformin at indicated time. Data are shown as relative cell viability (mean % ± S.E. bar) as compared with that in control (n = 3). (F) Cell viability was measured by MTT assay in cells treated by 1 mM MG for 17 hr with pretreatment of different concentration of metformin. Data are shown as relative cell viability (mean % ± S.E. bar) as compared with that in control (n = 3).
Figure 6. GLP-1 prevents MG-induced apoptosis of…
Figure 6. GLP-1 prevents MG-induced apoptosis of INS-1 and MIN6 cells in part through PKA and PI3 kinase pathway, and partially via improving mitochondrial function and suppressing prolonged AMPK activation.
INS-1 and MIN6 cells were seeded overnight, and then were incubated with indicated treatment for indicated time. (A) INS-1 and MIN6 cells were treated in the absence of MG, 1 mM MG, 1 mM MG + 300 nM GLP-1, 1 mM MG + 300 nM GLP-1 + 100 μM Rp-cAMP, 1 mM MG + 300 nM GLP-1 + 10 μM H-89, 1 mM MG + 300 nM GLP-1 + 30 μM LY294002, 1 mM MG + 300 nM GLP-1 + 50 nM wortmannin, respectively. Cell viability was measured by MTT assay. Data are shown as relative cell viability (mean % ± S.E. bar) as compared with that in control (n = 3). *p 

Figure 7. Schematic representation of the effect…

Figure 7. Schematic representation of the effect of GLP-1 against methylglyoxal (MG) toxicity in the…

Figure 7. Schematic representation of the effect of GLP-1 against methylglyoxal (MG) toxicity in the beta cell.
MG suppresses mitochondria function and leads to decrease ATP/AMP ratio, which in turn steadily activates AMPK, and the activation of AMPK subsequently leads to apoptosis. GLP-1 improved mitochondria function and in turn increases ATP/AMP ratio, which leads to inhibit sustained AMPK activation induced by MG, and subsequently suppress MG-induced apoptosis. In parallel, GLP-1 also exerts the anti-apoptotic effect through activation of PDX-1 in a PKA-dependent pathway. GLP-1 also activates PI3K/Akt pathway and inhibits caspase-3 activity and leads to suppress apoptosis.
All figures (7)
Figure 7. Schematic representation of the effect…
Figure 7. Schematic representation of the effect of GLP-1 against methylglyoxal (MG) toxicity in the beta cell.
MG suppresses mitochondria function and leads to decrease ATP/AMP ratio, which in turn steadily activates AMPK, and the activation of AMPK subsequently leads to apoptosis. GLP-1 improved mitochondria function and in turn increases ATP/AMP ratio, which leads to inhibit sustained AMPK activation induced by MG, and subsequently suppress MG-induced apoptosis. In parallel, GLP-1 also exerts the anti-apoptotic effect through activation of PDX-1 in a PKA-dependent pathway. GLP-1 also activates PI3K/Akt pathway and inhibits caspase-3 activity and leads to suppress apoptosis.

References

    1. Mandrup-Poulsen T. Beta-cell apoptosis: stimuli and signaling. Diabetes 50 Suppl 1, S58–S63 (2001).
    1. Schalkwijk C. G., Brouwers O. & Stehouwer C. D. Modulation of insulin action by advanced glycation end products: a new player in the field. Horm Metab Res 40, 614– 619 (2008).
    1. Thornalley P. J. Endogenous alpha-oxoaldehydes and formation of protein and nucleotide advanced glycation endproducts in tissue damage. Novartis Found Symp 285, 229–243, discussion 243–246 (2007).
    1. Sheader E. A., Benson R. S. P. & Best L. Cytotoxic action of methylglyoxal on insulin-secreting cells. Biochem Pharmacol 61, 1381–1386 (2001).
    1. Dhar A., Dhar I., Jiang B., Desai K. M. & Wu L. Chronic methylglyoxal infusion by minipump causes pancreatic beta-cell dysfunction and induces type 2 diabetes in Sprague-Dawley rats. Diabetes 60, 899–908 (2011).
    1. Hui H., Nourparvar A., Zhao X. & Perfetti R. Glucagon-like peptide-1 inhibits apoptosis of insulin-secreting cells via a cyclic 5-adenosine mono- phosphate-dependent protein kinase A and a phosphatidylinositol 3-kinase-dependent pathway. Endocrinology 144, 1444–1455 (2003).
    1. Farilla L. et al. Glucagon-like peptide-1 promotes islet cell growth and inhibits apoptosis in Zucker diabetic rats. Endocrinology 143, 4397–4408 (2002).
    1. Mellado-Gil J. M. & Aguilar-Diosdado M. High glucose potentiates cytokine and streptozotocin-induced apoptosis of rat islet cells: effect on apoptosis-related genes. J Endocrinol 183, 155–162 (2004).
    1. Li Y. et al. Glucagon-like peptide-1 receptor signaling modulates beta cell apoptosis. J Biol Chem 278, 471–478 (2003).
    1. Fan R., Li X., Gu X., Chan J. C. N. & Xu G. Exendin-4 protects pancreatic beta cells from human islet amyloid peptide-induced cell damage: potential involvement of AKT and mitochondria biogenesis. Diabetes Obes Metab 12, 815–824 (2010).
    1. Tews D. et al. Anti-apoptotic action of exandin-4 in INS-1 beta cells: comparative protein pattern analysis of isolated mitochondria. Horm Metab Res 41, 294–301 (2009).
    1. Sharma M. K., Jalewa J. & Hölscher C. Neuroprotective and anti-apoptotic effects of liraglutide on SH-SY5Y cells exposed to methylglyoxal stress. J Neurochem 128, 459–471 (2014).
    1. Kimura R. et al. Epidermal growth factor receptor transactivation is necessary for glucagon-like peptide-1 to protect PC12 cells from apoptosis. Neuroendocrinology 97, 300–308 (2013).
    1. American Diabetes Association. Approaches to glycemic treatment. Diabetes Care 38 Suppl 1, S41–S48 (2015).
    1. Zhou G. et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 108, 1167–1174 (2001).
    1. Simon-Szabó L., Kokas M., Mandl J., Kéri G. & Csala M. Metformin attenuates palmitate-induced endoplasmic reticulum stress, serine phosphorylation of IRS-1 and apoptosis in rat insulinoma cells. PLoS One 6, e97868 (2014).
    1. Masini M. et al. Prevention by metformin of alterations induced by chronic exposure to high glucose in human islet beta cells is associated with preserved ATP/ADP ratio. Diabetes Res Clin Pract 104, 163–170 (2014).
    1. Tewari M., et al. Yama/CPP32β, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose) polymerase. Cell 81, 801–809 (1995).
    1. Hui H., Nourparvar A., Zhao X. & Perfetti R. Glucagon-like peptide-1 inhibits apoptosis of insulin-secreting cells via a cyclic 5′-adenosine monophosphate-dependent protein kinase A- and a phosphatidylinositol 3-kinase-dependent pathway. Endocrinology 144, 1444–1455 (2003).
    1. Buteau J., et al. Glucagon-like peptide-1 prevents beta cell glucolipotoxicity. Diabetologia 47, 806–815 (2004).
    1. Wang Q., et al. Glucagon-like peptide-1 regulates proliferation and apoptosis via activation of protein kinase B in pancreatic INS-1 beta cells. Diabetologia 47, 47: 478–487 (2004).
    1. Hardie D. G., Salt I. P., Hawley S. A. & Davies S. P. AMP-activated protein kinase: an ultrasensitive system for monitoring cellular energy charge. Biochem J 338, 717–722 (1999).
    1. Rosca M. G., Monnier V. M., Szweda L. I. & Weiss M. F. Alterations in renal mitochondrial respiration in response to the reactive oxoaldehyde methylglyoxal. Am J Physiol 283, F52–F59 (2002).
    1. Shangari N. & O’Brien P. J. The cytotoxic mechanism of glyoxal involves oxidative stress. Biochem Pharmacol 68, 1433–1442 (2004).
    1. de Arriba S. G. et al. Methylglyoxal impairs glucose metabolism and leads to energy depletion in neuronal cells—protection by carbonyl scavengers. Neurobiol Aging 28, 1044–1050 (2007).
    1. Wang H., Liu J. & Wu L. Methylglyoxal-induced mitochondrial dysfunction in vascular smooth muscle cells. Biochem Pharmacol 77, 1709–1716 (2009).
    1. Kennedy E. D., Maechler P. & Wollheim C. B. Effects of depletion of mitochondrial DNA in metabolism secretion coupling in INS-1 cells. Diabetes 47, 374–380 (1998).
    1. Simmons R. A., Suponitsky-Kroyter I. & Selak M. A. Progressive accumulation of mitochondrial DNA mutations and decline in mitochondrial function lead to beta-cell failure. J Biol Chem 280, 28785–28791 (2005).
    1. Tsuboi T. et al. Glucagon-like peptide-1 mobilizes intracellular Ca2+ and stimulates mitochondrial ATP synthesis in pancreatic MIN6 β-cells. Biochem J 369, 287–299 (2003).
    1. Wu J., Wu J. J., Yang L. J., Wei L. X. & Zou D. J. Rosiglitazone protects against palmitate-induced pancreatic beta-cell death by activation of autophagy via 5′-AMP-activated protein kinase modulation. Endocrine 44, 87–98 (2013).
    1. Meares G. P. et al. AMP-activated protein kinase attenuates nitric oxide-induced β-cell death. J Biol Chem 285, 3191–3200 (2010).
    1. Kefas B. A. et al. AICA-riboside induces apoptosis of pancreatic beta cells through stimulation of AMP-activated protein kinase. Diabetologia 46, 250–254 (2003).
    1. Cai Y. et al. Increased oxygen radical formation and mitochondrial dysfunction mediate beta cell apoptosis under conditions of AMP-activated protein kinase stimulation. Free Radic Biol Med 42, 64–78 (2007).
    1. Miao X. Y. et al. The human glucagon-like peptide-1 analogue liraglutide regulates pancreatic beta-cell proliferation and apoptosis via an AMPK/mTOR/P70S6K signaling pathway. Peptides 39, 71–79 (2013).
    1. Riboulet-Chavey A., Diraison F., Siew L. K., Wong F. S. & Rutter G. A. Inhibition of AMP-activated protein kinase protects pancreatic β-cells from cytokine-mediated apoptosis and CD8 + T-cell–induced cytotoxicity. Diabetes 57, 415–423 (2008).
    1. Kim Y. K. et al. Protective role of glucagon-like peptide-1 against glucosamine-induced cytotoxicity in pancreatic beta cells. Cell Physiol Biochem 25, 211–220 (2010).
    1. Horman S. et al. Activation of AMP-activated protein kinase leads to the phosphorylation of elongation factor 2 and an inhibition of protein synthesis. Curr Biol 12, 1419–1423 (2002).
    1. Inoki K., Zhu T. & Guan K. TSC2 mediates cellular energy response to control cell growth and survival. Cell 115, 577–590 (2003).
    1. Igata M. et al. Adenosine monophosphate-activated protein kinase suppresses vascular smooth muscle cell proliferation through the inhibition of cell cycle progression. Circ Res 97, 837–844 (2005).
    1. Jones R. G. et al. AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol Cell 18, 283–293 (2005).
    1. Kefas B. A. et al. AMP-activated protein kinase can induce apoptosis of insulin-producing MIN6 cells through stimulation of c-Jun-N-terminal kinase. J Mol Endocrinol 30, 151–161 (2003).
    1. Jambal P. et al. Cytokine-mediated down-regulation of the transcription factor cAMP-response element-binding protein in pancreatic beta-cells. J Biol Chem 278, 23055–23065 (2003).
    1. Kim W. H. et al. AICAR potentiates ROS production induced by chronic high glucose: roles of AMPK in pancreatic beta-cell apoptosis. Cell Signal 19, 791–805 (2007).
    1. Del Prato S., Bianchi C. & Marchetti P. Beta-cell function and anti-diabetic pharmacotherapy. Diabetes Metab Res Rev 23, 518–527 (2007).
    1. Tahrani A. A., Bailey C. J., Del Prato S. & Barnett A. H. Management of type 2 diabetes: new and future developments in treatment. Lancet 378, 182–197 (2011).
    1. Moreno-Navarrete J. M. et al. OCT1 Expression in adipocytes could contribute to increased metformin action in obese subjects. Diabetes 60, 168–176 (2011).
    1. Nauck M. et al. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care 32, 84–90 (2009).
    1. The RISE Consortium. Restoring Insulin Secretion (RISE): design of studies of β –cell preservation in prediabetes and early type 2 diabetes across the life span. Diabetes Care 37, 780–788 (2014).
    1. Ali Salim L. Z. et al. Thymoquinone inhibits murine leukemia WEHI-3 cells in vivo and in vitro. PLoS One 9, e115340 (2014).
    1. Huang T. S., Kuo M. L., Shew J. Y., Chou Y. W. & Yang W. K. Distinct p53-mediated G1/S checkpoint responses in two NIH3T3 subclone cells following treatment with DNA-damaging agents. Oncogene 13, 625–632 (1996).

Source: PubMed

3
Abonnieren