Journal of Biomedical Translational Research
Research Institute of Veterinary Medicine, Chungbuk National University
Original Article

Effect of glucagon-like peptide 1 on salivary gland hypofunction in diabetic db/db mice

Hyung Rae Kim1https://orcid.org/0000-0003-0239-3170, Woo Kwon Jung1https://orcid.org/0009-0005-9604-4773, Junghyun Kim1,*https://orcid.org/0000-0002-9003-2158
1Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju 54896, Korea
*Corresponding author: Junghyun Kim, Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju 54896, Korea, Tel: +82-63-270-4032, E-mail: dvmhyun@jbnu.ac.kr

© Research Institute of Veterinary Medicine, Chungbuk National University.

Received: Nov 29, 2023; Revised: Dec 13, 2023; Accepted: Dec 14, 2023

Abstract

Salivary gland dysfunction is a common complication of diabetes. Decreased saliva production and changes in saliva composition may cause oral diseases. Reactive oxygen species (ROS) generation in the salivary glands results in the loss of acinar cells and decreased saliva secretion. Glucagon-like peptide 1 (GLP-1) is the incretin hormone that regulates blood glucose level and can suppress ROS production and inflammation through its antioxidant effects. Dipeptidyl peptidase-4 (DPP-4) is an enzyme that breaks down GLP-1. In this study, we evaluated the pathological role of DPP-4 and GLP-1 on salivary gland dysfunction in type 2 diabetic db/db mice. We observed reduced salivary secretion and histopathological alteration of salivary glands in the db/db mice. The increased DPP-4 and decreased GLP-1 levels in the salivary glands were also detected in the db/db mice. Furthermore, the db/db mice had increased apoptosis and oxidative injury in salivary glands. There was an accumulation of advanced glycation end products and mucus in the salivary glands of the db/db mice. In conclusion, these results showed the possible involvement of DPP-4 and GLP-1, leading to increased ROS-induced apoptosis in diabetes-related salivary gland dysfunction. DPP-4 and GLP-1 may be a pharmacological target for patients with diabetes-related salivary gland dysfunction.

Keywords: diabetes; dipeptidyl peptidase 4; glucagon-like peptide 1; xerostomia; salivary hypofunction

INTRODUCTION

Saliva is essential for digestion, taste, cleansing, and antimicrobial action. Salivary gland dysfunction can be caused by various factors such as systemic diseases, medications, radiation, and aging [1-3]. Diabetes has long been known to be associated with oral diseases [4, 5]. Oral dryness is a common symptom in diabetic patients, and changes in the salivary glands and their functions are linked to hyperglycemia Patients with diabetes report symptoms of dry mouth and decreased salivary secretion [6-9]. Inadequate saliva production can damage tissue, making it more susceptible to infections [10]. Current treatments for impaired salivary secretion offer only temporary relief and fail to address the underlying cause. Therefore, research into the mechanisms and treatments of salivary gland dysfunction is necessary to improve salivary gland function [11, 12].

Glucagon-like peptide 1 (GLP-1) is the major incretin hormone released from L cells that regulates blood sugar by stimulating glucose-dependent insulin secretion and suppressing glucagon secretion [13, 14]. Dipeptidyl peptidase-4 (DPP-4) is an enzyme responsible for breaking down GLP-1. DPP-4 inhibitors, such as sitagliptin, vildagliptin, and saxagliptin, prevent the inactivation of endogenously released incretin hormones, prolonging their physiological actions. DPP-4 inhibitors have roles beyond glucose regulation; they have cytoprotective effects against various organ damages DPP-4 inhibitors have demonstrated protective effects against various diabetes complications, including non-alcoholic fatty liver disease, impaired lipid function due to obesity, pancreatic beta cell atrophy, cardiovascular complications, and neuropathy [15]. Saxagliptin suppressed the increased insulin signaling impairment and oxidative stress in a D-galactose-induced aging model [16]. Linagliptin treatment improves diabetic retinopathy by reducing oxidative stress and decreasing the formation of advanced glycation end products (AGEs) [17]. Linagliptin also reduced AGE-mediated oxidative stress in cultured endothelial cells and lowered AGE levels, the expression of the receptor for AGE (RAGE), and oxidative stress in diabetic rats [18]. In animal studies, Vildagliptin prevented diabetes-related vascular damage by reducing AGEs, RAGE, and oxidative marker levels [19]. Recently, the DPP-4 inhibitor, alogliptin, was found to block the AGE-RAGE axis and reduce albuminuria in patients with type 2 diabetes [20]. Moreover, gemigliptin improved salivary gland function in streptozotocin-induced diabetic rats by exerting antioxidant and antiapoptotic effects [21]. Building upon previous findings, this study aimed to verify the pathological roles of DPP-4 and GLP-1 in diabetes-induced salivary gland dysfunction.

MATERIALS AND METHODS

Animals

Seven male db/db type 2 diabetic mice and seven male wild-type C57BL/6 mice, five weeks of age, were supplied by Central Lab Animal (Seoul, Korea). The mice were then randomly divided into the C57BL/6 control group (n = 7) and the diabetic db/db model group (n = 7). The animals were housed in a temperature (20°C–25°C) and humidity-controlled (40%–70%) barrier system with a 12/12 hr light/dark cycle. After five weeks, salivary glands were isolated from each mouse at necropsy. The animal experiments were conducted following the procedures approved by the Institutional Animal Care and Use Committee of Jeonbuk National University Laboratory Animal Center (IACUC, Approved No.: JBNU 2021-087).

Collection of saliva

The mice were anesthetized, and pilocarpine hydrochloride (1.5 mg/kg, IP) was administered. We then placed cotton balls in the mouths of the mice for 15 minutes to collect saliva. After 15 minutes, we measured the weight of the cotton balls to determine the amount of saliva absorbed. We compared and analyzed salivary secretion by calculating the difference in weight between the cotton balls before and after saliva absorption.

Transferase dUTP nick-end labeling staining

We conducted cell apoptosis analysis within the salivary gland tissue of mice. The process involved fixing the samples with formaldehyde, dehydrating them, and embedding them in paraffin blocks. Subsequently, we sectioned the paraffin blocks to 4 μm. Next, we deparaffinized the tissue sections twice in xylene (5 minutes each) and rehydrated them in ethanol. The terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay was performed according to the DeadEndTM Fluorometric TUNEL System instructions from Promega (Madison, WI, USA). Cell nuclei were counterstained with DAPI mounting solution (4',6-diamidino-2-phenylindole, Santa Cruz Biotechnology, Santa Cruz, CA, USA), and TUNEL-positive cells were quantified. TUNEL-positive apoptotic cells were visualized with green fluorescence using a BX51 microscope (Olympus, Tokyo, Japan). Five high-power fields (× 200 magnification) were randomly selected for each group, and the number of TUNEL-positive cells was calculated using ImageJ software (NIH, Bethesda, MD, USA).

Histopathological examination

The sections, fixed in 10% formalin and embedded in paraffin, were deparaffinized and rehydrated. Hematoxylin and Eosin (H&E) staining was performed for histological evaluation. To assess the accumulation of mucins (neutral mucopolysaccharides dyed pink), we stained the sections with Periodic Acid-Schiff (PAS) and performed a hematoxylin counterstain. Slide observations were made using an optical microscope at × 200 magnification (BX51, Olympus).

Immunohistochemistry

The 4 μm thick sections, fixed in 10% formalin and subsequently embedded in paraffin, underwent deparaffinization and rehydration through an ethanol gradient. Antigen retrieval was performed using a microwave to treat the sections to Tris-EDTA Buffer (10 mM Tris Base, 1 mM EDTA Solution, 0.05% Tween 20, pH 9.0). Endogenous peroxidase activity was neutralized with 3% hydrogen peroxide, followed by a 30-minute blocking step using 2.5% normal horse serum. The following primary antibodies were used: 8-hydroxy-2'-deoxyguanosine (8-OHdG, Abcam, Waltham, MA, USA) and AGEs (TransGenic, Fukuoka, Japan). Primary antibody signals were detected using the VECTASTAIN Elite ABC Universal Kit (Vector Laboratories, Burlingame, CA, USA), and visualization was achieved with the DAB peroxidase substrate kit (Vector Laboratories). Counterstaining was performed with hematoxylin, and the samples were mounted. Tissue sections were incubated with nonimmune animal serum instead of the primary antibodies as a negative control. Observations were made using an optical microscope at 200 × magnification (BX51, Olympus). Quantification was done using ImageJ software to measure the average optical density (mm2) per unit area in five randomly selected areas at 200 × magnification (NIH).

Measurement of dipeptidyl peptidase-4 and glucagon-like peptide 1

The DPP4 activity and GLP-1 level were measured using the mouse DPP4 (MyBioSource, San Diego, CA, USA) and GLP-1 ELISA Kit (MyBioSource), following the manufacturer’s instructions. The results were measured using the Spark® Multimode Microplate Reader (Tecan, Männedorf, Switzerland).

Measurement of advanced glycation end products

Following the manufacturer’s instructions, we detected AGE levels in salivary gland tissue and serum using the mouse AGEs ELISA kits (MyBioSource). Optical density measurements were taken using a Spark® Multimode Microplate Reader (Tecan).

Statistical analysis

Statistical analysis was performed by performing an unpaired t-test between two groups using Prism 8.0 software (GraphPad, San Diego, CA, USA).

RESULTS

Salivary gland dysfunction and histopathological alterations in db/db mice

Body weight was higher in the db/db mice at the endpoint, and blood glucose levels also increased in the db/db mice (Fig. 1A and B). Additionally, salivation decreased significantly in the db/db mice (Fig. 1C). The weight of the salivary gland reduced considerably in the db/db mice (Fig. 1D). H&E staining was performed to observe morphological changes in the salivary gland. The ductal area to serous acini area ratio increased in the salivary gland. In contrast, the location of serous acinar cells decreased in the db/db mice (Fig. 2A and B).

jbtr-24-4-139-g1
Fig. 1. Salivary gland dysfunction in db/db mice. (A) Body weights. (B) Blood glucose levels. (C) Amount of salivary secretion. (D) Salivary gland weight. All data are presented as the mean ± S.D. (n = 7). *p<0.05.
Download Original Figure
jbtr-24-4-139-g2
Fig. 2. Histopathological alteration of salivary glands in db/db mice. (A) Representative H&E images of salivary glands. Scale bar = 200 μm. (B) Quantitative evaluation of acinar cell area and duct area. All data are presented as the mean ± S.D. (n = 7). *p<0.05. H&E, Hematoxylin and Eosin.
Download Original Figure
Dipeptidyl peptidase-4 activity and glucagon-like peptide 1 levels in the salivary gland and serum

There was a significant increase in both salivary gland and serum DPP-4 activity in db/db mice compared to the normal mice (Fig. 3A and B). In contrast, GLP-1 levels in salivary glands and serum significantly decreased in the db/db mice compared to the normal mice (Fig. 3C and D). This could potentially have implications for DPP-4 and GLP-1 in salivary gland dysfunction.

jbtr-24-4-139-g3
Fig. 3. DPP-4 activity and GLP-1 levels in salivary glands and serum of db/db mice. (A) DPP-4 activity in salivary glands. (B) DPP-4 activity in Serum. (C) GLP-1 levels in salivary glands. (D) GLP-1 levels in serum. All data are presented as the mean ± S.D. (n = 7). *p<0.05. DPP-4, dipeptidyl peptidase-4; GLP-1, glucagon-like peptide 1.
Download Original Figure
Apoptosis and oxidative injury in the salivary glands of db/db mice

TUNEL assay was conducted to confirm apoptosis in the salivary glands. TUNEL-positive apoptotic cells were significantly increased in the db/db mice (Fig. 4A and B). Immunohistochemistry was conducted to assess the extent of 8-OHdG accumulation as a marker of cellular damage induced by oxidative stress. 8-OHdG presence was confirmed in the salivary glands. Accumulation of 8-OHdG increased in the db/db mice (Fig. 4C and D).

jbtr-24-4-139-g4
Fig. 4. Apoptosis and oxidative injury in salivary glands of db/db mice. (A) TUNEL staining of apoptotic cells in salivary gland tissue sections. Scale bar = 200 μm. (B) Quantitative analysis of TUNEL positive cells per unit area. (C) Immunohistochemical staining for 8-OHdG. Scale bar = 200 μm. (D) Quantification of the 8-OHdG signal intensity. All data are presented as the mean ± S.D. (n = 7). *p<0.05. TUNEL, transferase dUTP nick-end labeling.
Download Original Figure
Accumulation of advanced glycation end products and mucus in the salivary glands of db/db mice

The accumulation of AGEs in the salivary glands was examined using immunohistochemistry. There was an increased accumulation of AGEs in the db/db mice (Fig. 5A and B). AGE levels were measured in salivary gland lysate and serum. The levels of AGEs increased in both salivary glands and serum in the db/db mice (Fig. 5C and D).

jbtr-24-4-139-g5
Fig. 5. AGEs accumulation in salivary glands of db/db mice. (A) Immunohistochemical staining for AGEs. Scale bar = 200 μm. (B) Quantification of the AGEs signal intensity. (C) AGEs levels in salivary glands. (D) AGEs levels in serum. All data are presented as the mean ± S.D. (n = 7). *p<0.05. AGEs, advanced glycation end products.
Download Original Figure

PAS staining was performed to confirm changes in mucin accumulation in salivary gland acinar cells. In the normal group, the acini showed moderate staining with PAS reagents. However, in the db/db mice, there was a noticeable increase in staining intensity, indicating a higher accumulation of mucin within the acini (Fig. 6A and B).

jbtr-24-4-139-g6
Fig. 6. Mucin accumulation in salivary gland of db/db mice. (A) Periodic acid-Schiff (PAS) staining of salivary glands. Scale bar = 200 μm. (B) Quantification of PAS stained signal intensity. All data are presented as the mean ± S.D. (n = 7). *p<0.05.
Download Original Figure

DISCUSSION

Saliva is essential in maintaining oral health homeostasis [1-3]. Without adequate saliva production, oral tissues become damaged and vulnerable to infection, which can lead to oral diseases [10]. The generation of reactive oxygen species (ROS) in the salivary glands results in acinar cell loss and decreased saliva secretion [9]. In this study, we investigated the role of DPP-4 and GLP-1 on salivary gland function in db/db diabetic mice.

DPP-4, also known as adenosine deaminase complexing protein 2 or CD26, degrades incretin hormones such as GLP-1 [22, 23]. DPP-4 inhibition improves metabolic control by enhancing GLP-1-mediated pancreatic insulin secretion and suppressing hepatic glucose production [24]. GLP-1 has a very short half-life and is rapidly metabolized by DPP-4 [25-27]. GLP-1 is an incretin hormone secreted from the gut lumen that enhances insulin secretion [28-30]. Increasing endogenous levels of GLP-1 may improve glycemic control in individuals with type 2 diabetes and could potentially serve as a treatment for diabetes-related complications. We first confirmed increased DPP-4 activity and decreased GLP-1 levels in the salivary glands and serum of db/db mice, demonstrating their function within the glands.

Studies have shown that hyperglycemia can increase intracellular ROS through glycation reactions of amino acids or proteins, glutathione depletion, and inactivation of ROS scavenger enzymes. This can result in mitochondrial dysfunction and apoptosis [31-35]. Hyperglycemia-induced oxidative stress can be blocked by antioxidants, suggesting that ROS plays a vital role in hyperglycemia-induced cell death [36, 37]. We showed apoptosis in salivary glands with elevated levels of ROS. Increased ROS may be caused by increased DPP-4 and a decrease in GLP-1. GLP-1 plays a role in glucose-stimulated insulin secretion and gluconeogenesis, improving glycemic control in individuals with type 2 diabetes [24]. GLP-1, acting as an antioxidant, can suppress ROS production and inflammation in endothelial cells during hyperglycemia and in animal models of diabetes [38, 39]. DPP-4 increases ROS production in endothelial cells, and DPP-4 deficiency inhibits ROS production in cardiomyocytes [40, 41]. DPP-4 inhibitors reduce ROS production, prevent mitochondrial dysfunction, improve endothelial function, and reduce vascular inflammation and oxidative stress in animal models of diabetes and its complications [42-45]. A DPP-4 inhibitor, gemigliptin, has been reported to improve liver disease and diabetes by alleviating mitochondrial dysfunction and reducing ROS production [46, 47]. Based on previous studies and our results, DPP-4 and GLP-1 are attributed to the increase of ROS.

In addition, there was increased accumulation of AGEs in the salivary glands of db/db mice. In a previously reported study, AGEs formation inhibition by increased GLP-1 in DPP-4-deficient mice improved diabetes and diabetic complications [48, 49]. Additionally, numerous existing studies have verified the antiglycation effect of many DPP-4 inhibitors [17-20, 50]. Further, gemigliptin prevented a decline in salivary gland function through its antioxidant effect in diabetic rats [21]. We observed increased DPP4 activity, decreased GLP-1 levels, and elevated AGE accumulation in db/db mice. These factors subsequently induced ROS and, ultimately, apoptosis in salivary gland cells.

We observed morphological differences in the ratio of acinar cells to ductal cells in the salivary glands. Db/db mice showed decreased acinar cells and increased ductal cells in their salivary glands relative to the control group. Similar results have been observed in diverse salivary gland dysfunction models. The diabetes-related reduction in secretory cells in salivary glands is thought to result from decreased cell proliferation or increased cell death [38]. We measured salivary flow after administering pilocarpine, a parasympathetic agonist on muscarinic acetylcholine receptors in salivary glands [42]. The increase in ROS-induced apoptosis caused by diabetes may lead to salivary gland dysfunction. Oxidative stress, caused by ROS accumulation, altered secretion functions, such as saliva secretion, salivary amylase, and calcium levels, in rodent salivary glands [45]. These findings suggest that ROS-related oxidative stress can lead to salivary secretion disorders.

In our study, strong PAS staining demonstrated increased mucin accumulation in the acini in the db/db mice. Mucin plays a crucial role in mucosal lubrication, serving as one of the primary functions of saliva. Mucin can hold significant water, preventing mucosal dehydration [51, 52]. Mucins are vital components of saliva, lubricating and facilitating microbial adhesion. Mucus accommodates microbial communities and forms a protective coating on epithelial surfaces, playing a significant role in host defense [53]. Our results showed that mucin accumulated in acini with salivary gland dysfunction. Previous reports have shown that stronger PAS staining density in acini is associated with the accumulation of mucin in diabetic salivary glands [54-56]. Mucin composition in salivary glands can change due to salivary gland dysfunction [57]. Huang et al. also showed that acini of submandibular glands were moderately stained by PAS reagents in db/m mice but strong staining was observed in db/db mice. Stronger PAS staining densities of acini in db/db mice demonstrated that more mucins accumulated in the acini of diabetic salivary glands [54]. Therefore, mucus accumulation under diabetic conditions provides evidence that the secretory function of the salivary glands is impaired .

The limitation of this study is that we only showed the impaired DPP-4 activiry and GLP-1 levels under diabetic conditions. To better understand diabetes-related salivary dysfunction, additional investigation is required to elucidate the effect of GLP-1 recovery by DPP-4 inhibition using a selective DPP-4 inhibitor.

In conclusion, we have shown the possible involvement of DPP-4 and GLP-1 in increased ROS-induced apoptosis in diabetes-related salivary gland dysfunction. The pathological changes induced by DPP-4 and GLP-1 in salivary glands decrease the amount salivary secretion and increase the accumulation of mucins in the salivary glands due to decreased secretory. DPP-4 and GLP-1 could be a potent pharmacological target for patients with diabetes-related salivary gland dysfunction.

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Acknowledgements

This research was carried out with the support of the National Research Foundation of Korea (NRF) grant funded by the Korea government (MEST) (No. NRF-2023R1A2C1005120) and a Research Promotion Program of Jeonbuk National University.

Ethics Approval

All of the experimental procedures using animals were performed under the supervision of our Institutional Animal Care and Use Committee (IACUC No. 2021-087).

References

1.

Dodds MWJ, Johnson DA, Yeh CK. Health benefits of saliva: a review. J Dent 2005;33:223-233.

2.

Farsi NMA. Signs of oral dryness in relation to salivary flow rate, pH, buffering capacity and dry mouth complaints. BMC Oral Health 2007;7:1-6.

3.

Amerongen AN, Veerman E. Saliva: the defender of the oral cavity. Oral Dis 2002;8:12-22.

4.

Mauri-Obradors E, Estrugo-Devesa A, Jané-Salas E, Viñas M, López-López J. Oral manifestations of diabetes mellitus. A systematic review. Med Oral Patol Oral Cir Bucal 2017;22: e586-e594.

5.

Oliver RC, Tervonen T. Diabetes: a risk factor for periodontitis in adults? J Periodontol 1994; 65:530-538.

6.

Moore PA, Guggenheimer J, Etzel KR, Weyant RJ, Orchard T. Type 1 diabetes mellitus, xerostomia, and salivary flow rates. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2001; 92:281-291.

7.

Manfredi M, McCullough MJ, Vescovi P, Al-Kaarawi ZM, Porter SR. Update on diabetes mellitus and related oral diseases. Oral Dis 2004;10:187-200.

8.

Nicolau J, de Matos JA, de Souza DN, Neves LB, Lopes AC. Altered glycogen metabolism in the submandibular and parotid salivary glands of rats with streptozotocin-induced diabetes. J Oral Sci 2005;47:111-116.

9.

von Bültzingslöwen I, Sollecito TP, Fox PC, Daniels T, Jonsson R, Lockhart PB, Wray D, Brennan MT, Carrozzo M, Gandera B, Fujibayashi T, Navazesh M, Rhodus NL, Schiødt M. Salivary dysfunction associated with systemic diseases: systematic review and clinical management recommendations. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2007;103:S57.E1-S57.E15.

10.

D’Aiuto F, Gable D, Syed Z, Allen Y, Wanyonyi KL, White S, Gallagher JE. Evidence summary: the relationship between oral diseases and diabetes. Br Dent J 2017;222:944-948.

11.

Saleh J, Figueiredo MAZ, Cherubini K, Salum FG. Salivary hypofunction: an update on aetiology, diagnosis and therapeutics. Arch Oral Biol 2015;60:242-255.

12.

Visvanathan V, Nix P. Managing the patient presenting with xerostomia: a review. Int J Clin Pract 2010;64:404-407.

13.

Kim SH, Jung E, Yoon MK, Kwon OH, Hwang DM, Kim DW, Kim J, Lee SM, Yim HJ. Pharmacological profiles of gemigliptin (LC15-0444), a novel dipeptidyl peptidase-4 inhibitor, in vitro and in vivo. Eur J Pharmacol 2016;788:54-64.

14.

Kim SH, Lee SH, Yim HJ. Gemigliptin, a novel dipeptidyl peptidase 4 inhibitor: first new anti-diabetic drug in the history of Korean pharmaceutical industry. Arch Pharm Res 2013; 36:1185-1188.

15.

Bae EJ. DPP-4 inhibitors in diabetic complications: role of DPP-4 beyond glucose control. Arch Pharm Res 2016;39:1114-1128.

16.

Kenawy S, Hegazy R, Hassan A, El-Shenawy S, Gomaa N, Zaki H, Attia A. Involvement of insulin resistance in D-galactose-induced age-related dementia in rats: protective role of metformin and saxagliptin. PLOS ONE 2017;12:e0183565.

17.

Dietrich N, Kolibabka M, Busch S, Bugert P, Kaiser U, Lin J, Fleming T, Morcos M, Klein T, Schlotterer A, Hammes HP. The DPP4 inhibitor linagliptin protects from experimental diabetic retinopathy. PLOS ONE 2016;11:e0167853.

18.

Nakashima S, Matsui T, Takeuchi M, Yamagishi SI. Linagliptin blocks renal damage in type 1 diabetic rats by suppressing advanced glycation end products-receptor axis. Horm Metab Res 2014;46:717-721.

19.

Matsui T, Nishino Y, Takeuchi M, Yamagishi S. Vildagliptin blocks vascular injury in thoracic aorta of diabetic rats by suppressing advanced glycation end product–receptor axis. Pharmacol Res 2011;63:383-388.

20.

Sakata K, Hayakawa M, Yano Y, Tamaki N, Yokota N, Eto T, Watanabe R, Hirayama N, Matsuo T, Kuroki K, Sagara S, Mishima O, Koga M, Nagata N, Nishino Y, Kitamura K, Kario K, Takeuchi M, Yamagishi S. Efficacy of alogliptin, a dipeptidyl peptidase-4 inhibitor, on glucose parameters, the activity of the advanced glycation end product (AGE): receptor for AGE (RAGE) axis and albuminuria in Japanese type 2 diabetes. Diabetes Metab Res Rev 2013;29: 624-630.

21.

Kang WS, Jung WK, Park SB, Kim HR, Kim J. Gemigliptin suppresses salivary dysfunction in streptozotocin-induced diabetic rats. Biomed Pharmacother 2021;137:111297.

22.

Kameoka J, Tanaka T, Nojima Y, Schlossman SF, Morimoto C. Direct association of adenosine deaminase with a T cell activation antigen, CD26. Science 1993;261:466-469.

23.

Drucker DJ. Dipeptidyl peptidase-4 inhibition and the treatment of type 2 diabetes: preclinical biology and mechanisms of action. Diabetes Care 2007;30:1335-1343.

24.

Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006;368:1696-1705.

25.

Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, Willms B, Holst JJ. Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type II diabetic patients and in healthy subjects. Diabetes 1995;44:1126-1131.

26.

Mentlein R, Gallwitz B, Schmidt WE. Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1 (7–36) amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur J Biochem 1993;214:829-835.

27.

Kieffer TJ, McIntosh CH, Pederson RA. Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV. Endocrinology 1995;136:3585-3596.

28.

Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007;132: 2131-2157.

29.

Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 2013;17:819-837.

30.

MacDonald PE, El-kholy W, Riedel MJ, Salapatek AMF, Light PE, Wheeler MB. The multiple actions of GLP-1 on the process of glucose-stimulated insulin secretion. Diabetes 2002;51: S434-S442.

31.

Yim HS, Kang SO, Hah YC, Chock PB, Yim MB. Free radicals generated during the glycation reaction of amino acids by methylglyoxal: a model study of protein-cross-linked free radicals. J Biol Chem 1995;270:28228-28233.

32.

Oya T, Hattori N, Mizuno Y, Miyata S, Maeda S, Osawa T, Uchida K. Methylglyoxal modification of protein: chemical and immunochemical characterization of methylglyoxal-arginine adducts. J Biol Chem 1999;274:18492-18502.

33.

Lee C, Yim MB, Chock PB, Yim HS, Kang SO. Oxidation-reduction properties of methylglyoxal-modified protein in relation to free radical generation. J Biol Chem 1998;273:25272-25278.

34.

Chan CM, Huang DY, Huang YP, Hsu SH, Kang LY, Shen CM, Lin WW. Methylglyoxal induces cell death through endoplasmic reticulum stress-associated ROS production and mitochondrial dysfunction. J Cell Mol Med 2016;20:1749-1760.

35.

Seo K, Ki SH, Shin SM. Methylglyoxal induces mitochondrial dysfunction and cell death in liver. Toxicol Res 2014;30:193-198.

36.

Saito Y, Nishio K, Ogawa Y, Kimata J, Kinumi T, Yoshida Y, Noguchi N, Niki E. Turning point in apoptosis/necrosis induced by hydrogen peroxide. Free Radic Res 2006;40:619-630.

37.

Chan WH, Wu HJ. Methylglyoxal and high glucose co-treatment induces apoptosis or necrosis in human umbilical vein endothelial cells. J Cell Biochem 2008;103:1144-1157.

38.

Ishibashi Y, Matsui T, Takeuchi M, Yamagishi S. Glucagon-like peptide-1 (GLP-1) inhibits advanced glycation end product (AGE)-induced up-regulation of VCAM-1 mRNA levels in endothelial cells by suppressing AGE receptor (RAGE) expression. Biochem Biophys Res Commun 2010;391:1405-1408.

39.

Oeseburg H, de Boer RA, Buikema H, van der Harst P, van Gilst WH, Silljé HHW. Glucagon-like peptide 1 prevents reactive oxygen species–induced endothelial cell senescence through the activation of protein kinase A. Arterioscler Thromb Vasc Biol 2010;30:1407-1414.

40.

Ishibashi Y, Matsui T, Maeda S, Higashimoto Y, Yamagishi S. Advanced glycation end products evoke endothelial cell damage by stimulating soluble dipeptidyl peptidase-4 production and its interaction with mannose 6-phosphate/insulin-like growth factor II receptor. Cardiovasc Diabetol 2013;12:1-9.

41.

Ku HC, Chen WP, Su MJ. DPP4 deficiency exerts protective effect against H2O2 induced oxidative stress in isolated cardiomyocytes. PLOS ONE 2013;8:e54518.

42.

Pintana H, Apaijai N, Chattipakorn N, Chattipakorn SC. DPP-4 inhibitors improve cognition and brain mitochondrial function of insulin-resistant rats. J Endocrinol 2013;218:1-11.

43.

Chinda K, Palee S, Surinkaew S, Phornphutkul M, Chattipakorn S, Chattipakorn N. Cardioprotective effect of dipeptidyl peptidase-4 inhibitor during ischemia–reperfusion injury. Int J Cardiol 2013;167:451-457.

44.

Shah Z, Pineda C, Kampfrath T, Maiseyeu A, Ying Z, Racoma I, Deiuliis J, Xu X, Sun Q, Moffatt-Bruce S. Acute DPP-4 inhibition modulates vascular tone through GLP-1 independent pathways. Vascul Pharmacol 2011;55:2-9.

45.

Barbieri M, Rizzo MR, Marfella R, Boccardi V, Esposito A, Pansini A, Paolisso G. Decreased carotid atherosclerotic process by control of daily acute glucose fluctuations in diabetic patients treated by DPP-IV inhibitors. Atherosclerosis 2013;227:349-354.

46.

Nguyen G, Park SY, Do DV, Choi DH, Cho EH. Gemigliptin alleviates succinate-induced hepatic stellate cell activation by ameliorating mitochondrial dysfunction. Endocrinol Metab 2022;37:918-928.

47.

Jung E, Kim J, Ho Kim S, Kim S, Cho MH. Gemigliptin improves renal function and attenuates podocyte injury in mice with diabetic nephropathy. Eur J Pharmacol 2015;761:116-124.

48.

Sarker MK, Lee JH, Lee DH, Chun KH, Jun HS. Attenuation of diabetic kidney injury in DPP4-deficient rats; role of GLP-1 on the suppression of AGE formation by inducing glyoxalase 1. Aging 2020;12:593-610.

49.

Matsui T, Nakashima S, Nishino Y, Ojima A, Nakamura N, Arima K, Fukami K, Okuda S, Yamagishi S. Dipeptidyl peptidase-4 deficiency protects against experimental diabetic nephropathy partly by blocking the advanced glycation end products-receptor axis. Lab Invest 2015;95:525-533.

50.

Wongchai K, Schlotterer A, Lin J, Humpert PM, Klein T, Hammes HP, Morcos M. Protective effects of liraglutide and linagliptin in C. elegans as a new model for glucose-induced neurodegeneration. Horm Metab Res 2016;48:70-75.

51.

Pedersen AM, Bardow A, Jensen SB, Nauntofte B. Saliva and gastrointestinal functions of taste, mastication, swallowing and digestion. Oral Dis 2002;8:117-129.

52.

Tabak LA. In defense of the oral cavity: structure, biosynthesis, and function of salivary mucins. Annu Rev Physiol 1995;57:547-564.

53.

Frenkel ES, Ribbeck K. Salivary mucins in host defense and disease prevention. J Oral Microbiol 2015;7:29759.

54.

Huang Y, Mao QY, Shi XJ, Cong X, Zhang Y, Wu LL, Yu GY, Xiang RL. Disruption of tight junctions contributes to hyposalivation of salivary glands in a mouse model of type 2 diabetes. J Anat 2020;237:556-567.

55.

Kim HR, Jung WK, Park SB, Ryu HY, Kim YH, Kim J. Polydatin alleviates diabetes-induced hyposalivation through anti-glycation activity in db/db mouse. Pharmaceutics 2021;14:51.

56.

Zhang S, Li J, Nong X, Zhan Y, Xu J, Zhao D, Ma C, Wang Y, Li Y, Li Z, Li J. Artesunate combined with metformin ameliorate on diabetes-induced xerostomia by mitigating superior salivatory nucleus and salivary glands injury in type 2 diabetic rats via the PI3K/AKT pathway. Front Pharmacol 2021;12:774674.

57.

High AS, Sutton J, Hopper AH. A morphometric study of submandibular salivary gland changes in streptozotocin-induced diabetic rats. Arch Oral Biol 1985;30:667-671.