Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing , China. *

Similar documents
Megan Lawless. Journal Club. January 20 th, 2011

New Insights into Mechanism of Action

NIH Public Access Author Manuscript Diabetologia. Author manuscript; available in PMC 2014 February 01.

Current Status of Bariatric Surgery in Asia

Treating Type 2 Diabetes with Bariatric Surgery. Goal of Treating T2DM. Remission of T2DM with Bariatric

Roux-and-Y Gastric Bypass and its Metabolic Effects

type 2 diabetes is a surgical disease

New insights in metabolic surgery

Int J Clin Exp Med 2015;8(3): /ISSN: /IJCEM

Appetite, Glycemia and Entero-Insular Hormone Responses Differ Between Oral, Gastric-Remnant and Duodenal Administration of a Mixed Meal Test After

Chief of Endocrinology East Orange General Hospital

CME Post Test. D. Treatment with insulin E. Age older than 55 years

-1(IRS-1) 10. [DOI] /j.issn

The Mediterranean Diet: HOW and WHY It Works So Well for T2DM

Bariatric Surgery MM /11/2001. HMO; PPO; QUEST 05/01/2012 Section: Surgery Place(s) of Service: Outpatient; Inpatient

New and Emerging Therapies for Type 2 DM

Type 2 DM in Adolescents: Use of GLP-1 RA. Objectives. Scope of Problem: Obesity. Background. Pathophysiology of T2DM

Div. of Endocrinology and Metabolism Konyang University, School of Medicine. Byung-Joon Kim M.D., Ph.D.

The enteroinsular axis in the pathogenesis of prediabetes and diabetes in humans

Jianxiang Niu, Wanxiang Wang, Junjing Zhang, Jianjun Ren, Xingkai Meng

MULTI-CENTER, PROSPECTIVE, CONTROLLED TRIAL OF THE DUODENAL JEJUNAL BYPASS LINER FOR THE TREATMENT OF TYPE 2 DIABETES IN OBESE PATIENTS

BARIATRIC SURGERY AND TYPE 2 DIABETES MELLITUS

INJECTABLE THERAPIES IN DIABETES. Barbara Ann McKee Diabetes Specialist Nurse

Research Article Effect of Modified Roux-en-Y Gastric Bypass Surgery on GLP-1, GIP in Patients with Type 2 Diabetes Mellitus

Discussion & Conclusion

Scope. History. History. Incretins. Incretin-based Therapy and DPP-4 Inhibitors

What is Metabolic About Metabolic Surgery? The New ADA Recommendations

Inflammation & Type 2 Diabetes Prof. Marc Y. Donath

Bariatric Procedures and Mechanisms of Weight Loss. September 22 nd, 2018 Aryan Modasi MD MSc FRCSC

Management of diabetes with Bariatric surgery. Muhammad Jawad MD FACS FASMBS

Intestinal Hypertrophy and Increased Glucose Transporter Expression after Gastric Bypass. Senior Thesis. Presented to

Control of Glucose Metabolism

Duodenal-Jejunal Bypass and Jejunectomy Improve Insulin Sensitivity in Goto-Kakizaki Diabetic Rats Without Changes in Incretins or Insulin Secretion

Effect of macronutrients and mixed meals on incretin hormone secretion and islet cell function

Gastric Emptying Time after Laparoscopic Sleeve Gastrectomy

Letter to the Editor. Association of TCF7L2 and GCG Gene Variants with Insulin Secretion, Insulin Resistance, and Obesity in New-onset Diabetes *

Diabetes in Chronic Pancreatitis: When is it type 3c? Melena Bellin, MD Associate Professor, Pediatrics & Surgery Schulze Diabetes Institute

Astragaloside IV ameliorates 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced

Comparison Between Laparoscopic Sleeve Gastrectomy and Laparoscopic Adjustable Gastric Banding for Morbid Obesity: a Meta-analysis

Elevated Serum Levels of Adropin in Patients with Type 2 Diabetes Mellitus and its Association with

Preserved insulin secretory capacity and weight loss are the predominant predictors

Other Ways to Achieve Metabolic Control

Type 2 diabetes and metabolic surgery:

Bariatric surgery, such as Roux-en-Y gastric bypass

Practical Strategies for the Clinical Use of Incretin Mimetics CME/CE. CME/CE Released: 09/15/2009; Valid for credit through 09/15/2010

Therapeutic strategy to reduce Glucagon secretion

Diabetes control and lessened cerebral cardiovascular risks after gastric bypass surgery in Asian Taiwanese with a body mass index <35 kg/m2

SOCIETA ITALIANA DI NUTRIZIONE UMANA Milano novembre Geltrude Mingrone. Istituto di Clinica Medica Università Cattolica S.

Abstract. Effect of sitagliptin on glycemic control in patients with type 2 diabetes. Introduction. Abbas Mahdi Rahmah

NOTES: The Digestive System (Ch 14, part 2)

Diabetes: What is the scope of the problem?

UNIVERSITY OF CALGARY

Gastric Artery Embolization for Weight Loss: Rationale

ABDOMEN - GI. Duodenum

Endoscopic Duodenal Jejunal Bypass Liner Rapidly Improves Type 2 Diabetes

6/23/2011. Bariatric Surgery: What the Primary Care Provider Should Know. Case Presentation: Rachelle

The Small Intestine. The pyloric sphincter at the bottom of the stomach opens, squirting small amounts of food into your small intestine.

Role of Malabsorptive Endoscopic Procedures in Obesity Treatment

General Laboratory methods Plasma analysis: Gene Expression Analysis: Immunoblot analysis: Immunohistochemistry:

Associations among Body Mass Index, Insulin Resistance, and Pancreatic ß-Cell Function in Korean Patients with New- Onset Type 2 Diabetes

Endocrine mechanisms mediating remission of diabetes after gastric bypass surgery

Technique. Matthew Bettendorf, MD Essentia Health Duluth Clinic. Laparoscopic approach One 12mm port, Four 5mm ports

Laparoscopy-assisted D2 radical distal subtotal gastrectomy

Figure S1. Body composition, energy homeostasis and substrate utilization in LRH-1 hep+/+ (white bars) and LRH-1 hep-/- (black bars) mice.

Current Trends in Bariatric Surgery

THE ROLE OF THE PROXIMAL SMALL INTESTINE IN IMPROVEMENTS IN DIABETES RESOLUTION AND INSULIN SENSITIVITY FOLLOWING BARIATRIC

A Central Role of MG53 in Metabolic Syndrome. and Type-2 Diabetes

Diabesita : integrazione tra terapia medica e terapia chirurgica Prof. Monica Nannipieri Dip. Medicina Clinica e Sperimentale Università di Pisa

An integrated glucose homeostasis model of glucose, insulin, C-peptide, GLP-1, GIP and glucagon in healthy subjects and patients with type 2 diabetes

Understanding the Effects of Roux-en-Y Gastric Bypass (RYGB) Surgery on Type 2 Diabetes Mellitus

Obesity Management Workshop for Health Professionals

Laparoscopic Roux-en-Y Gastric Bypass for the Treatment of Type II Diabetes Mellitus in Chinese Patients with Body Mass Index of 25 35

4. Mechanisms Mediating Weight Loss and Diabetes Remission After Bariatric/ Metabolic Surgery

Choosing the right phytase to improve sodium pump function, reduce the catabolism of amino acids and increase protein and glucose uptake

Zhong Chen, Xiaoying Fu, Jian Kuang *, Ju Chen, Hongmei Chen, Jianhao Pei and Huazhang Yang

ASSOCIATION OF BMI WITH INSULIN RESISTANCE IN TYPE 2 DIABETES MELLITUS -A STUDY IN LOCAL TELANGANA POPULATION

GLP-1 agonists. Ian Gallen Consultant Community Diabetologist Royal Berkshire Hospital Reading UK

ENERGY FROM INGESTED NUTREINTS MAY BE USED IMMEDIATELY OR STORED

Correlation between estrogen receptor β expression and the curative effect of endocrine therapy in breast cancer patients

Gastric By-Pass and Remission of Type II Diabetes in Obese Adults

Obesity and Insulin Resistance According to Age in Newly Diagnosed Type 2 Diabetes Patients in Korea

ROLE OF TASTE IN METABOLIC CONTROL OF TYPE 1 DIABETES MELLITUS

Sitagliptin: first DPP-4 inhibitor to treat type 2 diabetes Steve Chaplin MSc, MRPharmS and Andrew Krentz MD, FRCP

NUTRIENT DIGESTION & ABSORPTION

Totally laparoscopic distal gastrectomy reconstructed by Rouxen-Y with D2 lymphadenectomy and needle catheter jejunostomy for gastric cancer

Abstract. Effect of Duodenal-Jejunal Bypass on Skeletal Muscle Insulin Signaling in Goto- Kakizaki Rats. by Ruben Carnell Sloan, III.

Benefits of Bariatric Surgery

Il blocco del cotrasportatore. della terapia antiiperglicemica. Anna Solini

Drug Class Monograph

Gut hormones KHATTAB

The clathrin adaptor Numb regulates intestinal cholesterol. absorption through dynamic interaction with NPC1L1

22 Emerging Therapies for

DPP-4 inhibitor. The new class drug for Diabetes

Blood glucose concentrations in healthy humans

Distribution of Pancreatic Polypeptide in the Head of the Human Pancreas. By Andrew Kringas

Bariatric surgery: Impact on Co-morbidities and Weight Loss Expectations ALIYAH KANJI, MD FRCSC MIS AND BARIATRIC SURGERY SEPTEMBER 22, 2018

Cordoba 01/02/2008. Slides Professor Pierre LEFEBVRE

Sweetness and Glycaemic Regulation. (A focus on gut related and physiological aspects) John McLaughlin Manchester University

Organic consequences of ileal transposition in rats with diet-induced obesity.

Transcription:

Int J Clin Exp Pathol 2014;7(7):4136-4142 www.ijcep.com /ISSN:1936-2625/IJCEP0000640 Original Article Ileal interposition reduces blood glucose levels and decreases insulin resistance in a type 2 diabetes mellitus animal model by up-regulating glucagon-like peptide-1 and its receptor Xu Sun 1*, Meizhu Zheng 2*, Maomin Song 3, Rixing Bai 3, Shi Cheng 3, Ying Xing 3, Huisheng Yuan 3, Pilin Wang 3 1 Department of Gastrointestinal Tumor Surgery, Shandong Cancer Hospital, 440 Jiyan Road, Jinan 250117, Shandong, China; 2 Breast Cancer Center, Shandong Cancer Hospital, 440 Jiyan Road, Jinan 250117, Shandong, China; 3 Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China. * Equal contributors. Received April 27, 2014; Accepted June 23, 2014; Epub June 15, 2014; Published July 1, 2014 Abstract: This study is to explore the possible mechanism of ileal interposition (IT) treatment of glycemic control of the type 2 diabetes mellitus (T2DM) by establishing an IT animal model. Twelve T2DM rats (GK rats) of 8-week old were divided into GK IT surgery group (GK-IT) and GK sham group (GK-Sham). Six Wistar rats were used as the non- T2DM sham group (WS-Sham). Enzyme-linked immunosorbent assay was used to detect plasma insulin concentration and fasting pancreas glucagon-like peptide-1 (GLP-1) concentration changes. Homeostasis model assessment of insulin resistance was used to quantitatively measure insulin resistance. Glucagon-like peptide-1 receptor (GLP- 1R) expression was detected by Western blotting. IT significantly decreased fasting blood glucose level and the oral glucose tolerance, and reduced insulin resistance of GK rats by increasing GLP-1 concentration and GLP-1R levels. The postoperative pancreatic β-cell apoptosis rate of GK-Sham group was significantly higher than those in the GK-IT group and the WS-Sham group. IT significantly reduces blood glucose and decreases insulin resistance by upregulating GLP-1 concentrations and GLP-1R levels, which may contribute to insulin secretion of pancreatic β-cells and decreases apoptosis of pancreatic β-cell. Keywords: Ileal interposition, type 2 diabetes mellitus (T2DM), glucagon-like peptide-1 (GLP-1), glucagon-like peptide-1 receptor (GLP-1R) Introduction The type 2 diabetes mellitus (T2DM) is one of the chronic diseases that seriously threaten human health. However, etiology and pathogenesis of T2DM is still unclear. From some obesity surgeries, it is found that in addition to effective control of patient weight, their T2DMs are also effectively controlled by the surgeries [1, 2]. Recently, gastrointestinal tract reconstruction for treatment of T2DM is a research hotspot. It is considered that the Roux-en Y gastric bypass (RYGB) surgery is shown to be effective in the treatment of T2DM [3]. Although there are two types of hypotheses related to RYGB, termed as foregut hypothesis [4] and hindgut hypothesis [5], respectively, the exactly mechanism is still unclear. In this study, ileal interposition (IT) was performed to explore the potential possibility of hindgut hypothesis. IT may induce changes in the gastrointestinal hormone concentration in the blood [6, 7]. The incretin hormones are the polypeptide secreted by intestinal. GLP-1 is a particularly important incretin hormone, since it enhances glucosestimulated insulin secretion, controls absorption of oral sugar, protein and fat [8], and promotes regeneration of pancreatic β cells [9]. GLP-1 is secreted by L cells of distal intestine and colon [8]. GLP-1 functions by binding to specific G protein-coupled receptor of β cells in pancreas (GLP-1R) [8].

Figure 1. Changes in the body weight values and food intakes among the three groups (WS-Sham, GK-Sham, and GK-IT). A. The rat body weight was measured one week before and every week after operation in each group. *, P < 0.05 compared with GK-Sham group and WS-Sham group. B. The rat food intake rates were measured 1 week before and every week after operation in each group. *, P < 0.05 compared with the GK-Sham group and WS-Sham groups. In this study, the GK rat model of T2DM was used to investigate the effects of IT on glycemic controlling, insulin resistance, GLP-1, GLP-1R, and β cell apoptosis, in order to further elucidate its possible mechanisms. Materials and methods Animals and grouping Twelve male GK rats of 8-week old (provided by Slac Laboratory Animal Co., Shanghai Laboratory Animal Center, Chinese Academy of Sciences) were randomly divided into 2 groups: IT group (GK-IT group) and sham group (GK-Sham group). Each group contains of 6 rats. Six Wistar rats of the same age were used as the non-diabetes sham group (WS-Sham group). Rats were fed for one week to adapt to the environment with 12-hour light/dark cycle, and free access to standard feed and water. All animal experiments were carried out in accordance with the requirements of Capital Medical University Animal Committee. Surgical procedures After fasting of 12 hours before surgery, 10% chloral hydrate (0.3 ml/100 g body weight) was injected intraperitoneally for anesthesia. IT was performed according to Culnan et al [10] as following. Firstly, midline longitudinal incision of abdominal was taken. Then, 10 cm of ileum in distance of 5-15 cm to ileocecum was intercepted with mesenteric reserved. The 10 cm of ileum was moved to jejunum 5 cm from the flexor ligament, and end-to-end anastomosis was performed. The 5-0 silk threads were used for anastomosis. Bleeding and intestinal leakage was checked and 3-0 threads were used for abdomen closure. In the GK-sham and WS-sham groups, enterotomy and intestinal anastomosis were performed in the corresponding sites. Postoperative treatment was in accordance with Li et al [11]. Experiments Rats body weight and food intake were recorded one week before surgery, and weekly after surgery in four weeks. Blood glucose meter was used to detect the fasting (fasting for 12-14 hours usually) blood glucose levels one week before surgery and the levels of the weekly fasting blood glucoses within 4 weeks post-surgery. Glucose meter SureStep Plus was purchased from Lifescan, Johnson & Johnson (Lifescan, Johnson & Johnso, New Brunswick, New Jersey, USA). Oral glucose tolerance test (OGTT) was performed before surgery and the fourth week after surgery. After fasting for 12-14 hours, rats were given orally glucose solution gavage in sober state (1 g/kg). Glucose values were measured before gavage (fasting) and at 30 min, 60 min, or 120 min after gavage. Trapezoidal method was used to calculate the area under 4137 Int J Clin Exp Pathol 2014;7(7):4136-4142

Table 1. Fasting glucose values of all groups one week pre-surgery and weekly post-surgery (mg/dl) GK-Sham GK-IT WS-Sham 1 week pre-surgery 118 ± 4* 123 ± 6* 93 ± 3 1 week post-surgery 115 ± 5* 97 ± 5*, ** 88 ± 6 2 weeks post-surgery 120 ± 6* 101 ± 7*, ** 95 ± 2 3 weeks post-surgery 124 ± 10* 96 ± 4** 89 ± 7 4 weeks post-surgery 130 ± 11* 94 ± 5** 89 ± 8 Note: *, P < 0.05 compared with the WS-Sham group; **, P < 0.05 compared with the GK-Sham group. least 3 times. Image quantifications were performed using ImageQuant software. Statistical analysis Data are expressed as mean ± standard deviation (SD). One-way ANOVA was analyzed by SPSS13.0 software. P < 0.05 was considered statistically significant. Results the curve (AUC). Plasma insulin concentration and fasting GLP-1 concentration were determined by ELISA assay before gavage (fasting) and at 30 min after gavage one week before surgery and weekly after surgery in four weeks. Rat insulin enzyme-linked immunosorbent (ELISA) kit and rat GLP-1 ELISA kit was purchased from IBL Company (Hamburg, Germany). According to the fasting glucose and fasting insulin, insulin resistance levels were measured by homeostasis model assessment value (HOMA-IR), which is calculated as fasting glucose (mmol/l) fasting insulin (mu/ml)/22.5. Rats were sacrificed after 28 days, and pancreas tissues were taken along the longitudinal axis of incision for GLP-1R protein expression by Western blotting. Some pancreas tissue was sliced and detected in accordance with apoptosis detection kit. In situ end labeling (TUNEL) was used for detection of pancreatic β-cell apoptosis. Apoptosis Detection Kit was purchased from Roche Company (Branford, CT, USA). TUNEL result is apoptosis rate in five high fields of each slice ( 400) = (number of apoptosis cells/number of total cells) 100%. Western blotting Total proteins were harvested and separated on 10% SDS/PAGE gels, and then subjected to immunoblot analyses. PVDF membrane was purchased from Millipore Corporation (Billerica, MA, USA). Rabbit anti-mouse GLP-1R polyclonal antibody was purchased from Abcam Company, UK (Cambridge, UK). Rabbit anti-mouse β-actin polyclonal antibody was purchased from Tianjin Saier Biotechnology Company (Tianjin, China). Bound antibodies were detected using the ECL system (Perkin Elmer, NEL100001EA). The immunoblot experiments were repeated at Surgery reduces body weight and food intake in the animal model Rats in all three groups (the GK-Sham group, WS-Sham group, and GK-IT group) survived after surgery and four weeks of experiment. One week before surgery, there was no significant difference of rat body weight and food intake per kilogram between groups (P > 0.05). Average body weight decreased after surgery in all groups. In the GK-Sham group and WS-Sham group, body weight slowly increased one week after surgery. In the GK-IT group, body weight decreased two weeks after surgery and then rebounded. Postoperatively, food intake was significantly reduced, and gradually increased afterwards. In 2, 3, and 4 weeks after surgery, the body weights (Figure 1A) and food intakes (Figure 1B) of the GK-IT group were significantly reduced compared with those in the GK-Sham group and the WS-Sham group (P < 0.05). IT decreases glucose tolerance and insulin resistance The influence of IT on fasting glucose and oral glucose tolerance of type 2 diabetes rats is shown in Table 1. One week before surgery, fasting glucose of GK rats was significantly higher than that of Wistar rats (P < 0.05). After IT, fasting blood glucose of the GK-IT group was significantly decreased compared with the GK-Sham group (P < 0.05). Three week after surgery, the fasting glucose of GK-IT rats were reduced to levels similar to those WS sham group level (P > 0.05). To further study the influence of IT on glucose tolerance, oral glucose tolerance test (OGTT) was detected one week before surgery and weekly after surgery for four weeks in each group (Table 2). The AUC of OGTT in GK-IT and GK-Sham group was significantly greater than 4138 Int J Clin Exp Pathol 2014;7(7):4136-4142

Table 2. OGTT and AUC values of all groups one week pre-surgery and four weeks post-surgery Pre-surgery 0 min 30 min 60 min 120 min AUC [mg/dl(min)] GK-IT 123 ± 6 253 ± 9 224 ± 13 155 ± 4 24,215 ± 795* GK-Sham 118 ± 4 242 ± 11 217 ± 13 151 ± 4 23,378 ± 791* WS-Sham 93 ± 3 142 ± 8 107 ± 10 95 ± 15 13,302 ± 869 Post-suegery GK-IT 94 ± 5 162 ± 19 136 ± 7 110 ± 18 15,696 ± 689*, **,# GK-Sham 130 ± 11 260 ± 22 232 ± 17 136 ± 13 24,260 ± 1544* WS-Sham 89 ± 8 140 ± 4 108 ± 10 95 ± 7 13,221±640 Note: *, P < 0.05 compared with WS-Sham; **, P < 0.05 compared with GK-Sham; #, P < 0.05 compared with pre-operation. Table 3. GLP-1 concentration changes of all groups pre-surgery and four weeks post-surgery (pmol/l) Pre-surgery 4 weeks post-surgery GK-IT 20.89 ± 2.56* 53.53 ± 1.87**,# GK-Sham 20.06 ± 2.00* 19.70 ± 2.25* WS-Sham 50.39 ± 3.08 50.36 ± 3.14 Note: *, P < 0.05 compared with WS-Sham group; **, P < 0.05 compared with GK-Sham group; #, P < 0.05 compared to the values of the relative pre-operative group. that in WS-Sham group (P < 0.05). Fourth weeks after surgery, OGTT was also detected in all groups, and oral glucose tolerance AUC of GK-IT group was significantly less than that in GK-Sham group (P < 0.05) with 35% decrease compared with that before surgery. It showed IT effectively increased oral glucose tolerance in GK rats. Our results also showed that glucosestimulated insulin secretion of GK-IT rats after surgery increased significantly compared with GK-Sham (data not shown). Four weeks after surgery, HOMA-IR value of GK-IT group was significantly lower than that in the GK-Sham group (data not shown). IT increases pancreas GLP-1R expression levels of GK rats To further explore the mechanism of IT treatment for diabetes, plasma GLP-1 concentration was detected one week before and 4 weeks after surgery. Rats were sacrificed four weeks after surgery, and pancreas GLP-1R protein expression was determined. One week before surgery, plasma GLP-1 concentration of diabetes rats was significantly lower than that of WS-Sham group (P < 0.05) (Figure 2). It showed that IT increased plasma GLP-1 concentrations and pancreas GLP-1 receptor expression. The increase of GLP-1 concentration and GLP-1R level is the possible mechanism of IT treatment for improved glucose tolerance and type 2 diabetes. IT decreases β-cell apoptosis rates of GK-IK rats Four weeks after operation, TUNEL apoptosis kit was used to detect rat β-cell apoptosis. The apoptosis rate was 7.18 ± 0.49% in GK-IT group, 25.02 ± 1.57% in GK-Sham group, 5.98 ± 0.87% in WS-sham group. After IT, β-cell apoptosis rate of GK-IT rat was significantly lower than that in the GK-Sham group (P < 0.05), and there was no significant difference when compared levels in the WS-Sham group (Figure 3A and 3B). Discussion Wistar rats (P < 0.05). Four weeks after surgery, plasma GLP-1 concentration of GK-IT group was significantly increased, but there was no significant difference with WS-Sham group (Table 3). After IT, pancreas GLP-1R expression of GK rats was significantly increased and significantly higher than those in the GK-Sham group and In this study, body weight and food intake of GK rats after IT were gradually reduced, and fasting glucose also decreased. Fasting glucose decreased to the same level of non-diabetic rats in WS-Sham group level three weeks after surgery. Though body weight and food intake of GK-IT rats increased three weeks after surgery, fasting glucose remained same. It indicated that after IT, improved glucose tolerance was not associated with body weight and food intake, which is consistent with the previous studies [12, 13]. Therefore, decreased fasting glucose is independent of weight loss after IT. Four weeks after surgery, OGTT AUC of GK-IT 4139 Int J Clin Exp Pathol 2014;7(7):4136-4142

Figure 2. Expression of GLP-1R protein in rats of the three groups. A. Western blot analysis of the expression of GLP- 1R protein. The GLP-1R protein was isolated from tissues four weeks after surgery. β-actin was used as a loading control. B. The expression levels of GLP-1R protein relative to β-actin in the three groups were calculated. *, P < 0.05 compared with WS-Sham group; #, P < 0.05 compared with the GK-IT group. Figure 3. Apoptosis assay in pancreas β cells. A. The β-cell apoptosis pancreas paraffin sections ( 400) detected by TU- NEL assay four weeks after surgery. Apoptotic pancreas β cells are stained brown. B. Apoptotic index in 28-day postoperation. The change of apoptotic index caused by IT was shown. *, P < 0.05 compared with the GK-Sham group. group was significantly lower than that in GK-Sham group, which indicated surgery effectively controlled blood glucose [13, 14]. Before and 4 weeks after surgery, there was no significant difference of fasting plasma insulin levels in GK-IT group and GK-Sham group. After IT, insulin resistance of GK-IT group was significantly improved than that in GK-Sham group. Four weeks after surgery, plasma insulin concentration of GK-IT group was significantly higher than that in GK-Sham group 30 minutes after gavage, indicating that IT can reduce insulin resistance of type 2 diabetes in rats and promote pancreas β-cell insulin secretion after meal. GLP-1 is polypeptide with 30 amino acids secreted by L cells in distal small intestine and colon [15]. GLP-1 increases glucose-induced insulin secretion, inhibits glucagon secretion, reduces food absorption, improves insulin sensitivity, promotes β-cell regeneration, and reduces its apoptosis [6]. When excessive chyme contacts with the distal small intestine, gastrointestinal hormone secretion was increased [13]. IT inserts distal ileum into proximal intestine, so food that is not completely digested or undigested would prematurely reach the ileum, thus stimulates intestinal L cells to increase GLP-l secretion. In our study, it shows that 4 weeks after surgery, plasma GLP-1 concentration increased to normal levels. GLP-1 is 4140 Int J Clin Exp Pathol 2014;7(7):4136-4142

effective only after binding to its receptor. Therefore four weeks after surgery, pancreas GLP-1R protein expression was detected. GLP- 1R is membrane protein located on pancreas β cells [16, 17]. It shows that, four weeks after surgery, GLP-1R protein expression level in IT group was higher than that in GK-Sham group and WS-Sham group. Apoptosis is the main reason for reduced pancreas β-cell in type 2 diabetes [18], it is found that β-cell apoptosis was less in GK-IT group than that in GK-Sham group. Reduced pancreas β-cell apoptosis by IT is one reason for the increased plasma insulin concentration after glucose gavage. GLP-1 regulates the expression of apoptosis-related gene Bcl-2/Bax, reduces β-cell apoptosis and increases β-cell number to increase insulin secretion for control of diabetes [19]. Improved GLP-1 concentration and GLP-1R level may be the possible mechanism to reduce β-cell apoptosis. In conclusion, it shows that IT effectively reduced the blood glucose levels, improved insulin resistance and increased insulin concentrations after glucose gavage of diabetes GK rats. The increased plasma GLP-1 concentration and pancreas GLP-1R protein expression, as well as reduced pancreas β-cell apoptosis rate, may be the possible mechanism of IT for the treatment of type 2 diabetes. Acknowledgements This work was supported by Capital Medical Development Scientific Research Fund of China (No. 2009-3104). Disclosure of conflict of interest None. Address correspondence to: Dr. Maomin Song, Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China. Tel: 86 10 67096589; Fax: 86 10 67096593; E-mail: songmaom@163.com References [1] Brolin RE. Update: NIH consensus conference. Gastrointestinal surgery for severe obesity. Nutrition 1996; 12: 403-404. [2] Rubino F. Bariatric surgery: effects on glucose homeostasis. Curr Opin Clin Nutr Metab Care 2006; 9: 497-507. [3] Meneghini LF. Impact of bariatric surgery on type 2 diabetes. Cell Biochem Biophys 2007; 48: 97-102. [4] Cummings DE, Overduin J, Foster-Schubert KE. Gastric bypass for obesity: mechanisms of weight loss and diabetes resolution. J Clin Endocrinol Metab 2004; 89: 2608-2615. [5] Rubino F, Marescaux J. Effect of duodenal-jejunal exclusion in a non-obese animal model of type 2 diabetes: a new perspective for an old disease. Ann Surg 2004; 239: 1-11. [6] Cummings BP, Strader AD, Stanhope KL, Graham JL, Lee J, Raybould HE, Baskin DG, Havel PJ. Ileal interposition surgery improves glucose and lipid metabolism and delays diabetes onset in the UCD-T2DM rat. Gastroenterology 2010; 138: 2441-2446. [7] Ikezawa F, Shibata C, Kikuchi D, Imoto H, Miura K, Naitoh T, Ogawa H, Sasaki I, Tsuchiya T. Effects of ileal interposition on glucose metabolism in obese rats with diabetes. Surgery 2012; 151: 822-830. [8] Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132: 2131-2157. [9] Drucker DJ. The biology of incretin hormones. Cell Metab 2006; 3: 153-165. [10] Culnan DM, Albaugh V, Sun M, Lynch CJ, Lang CH, Cooney RN. Ileal interposition improves glucose tolerance and insulin sensitivity in the obese Zucker rat. Am J Physiol Gastrointest Liver Physiol 2010; 299: G751-G760. [11] Li SQ, Zhou Y, Wang Y, Liu Y, Geng DH, Liu JG. Upregulation of IRS-1 expression in Goto-Kakizaki rats following Roux-en-Y gastric bypass surgery: resolution of type 2 diabetes? Tohoku J Exp Med 2011; 225: 179-186. [12] Liu S, Zhang G, Wang L, Sun D, Chen W, Yan Z, Sun Y, Hu S. The entire small intestine mediates the changes in glucose homeostasis after intestinal surgery in goto-kakizaki rats. Ann Surg 2012; 256: 1049-1058. [13] Ikezawa F, Shibata C, Kikuchi D, Imoto H, Miura K, Naitoh T, Ogawa H, Sasaki I, Tsuchiya T. Effects of ileal interposition on glucose metabolism in obese rats with diabetes. Surgery 2012; 151: 822-830. [14] Patriti A, Facchiano E, Annetti C, Aisa MC, Galli F, Fanelli C, Donini A. Early improvement of glucose tolerance after ileal transposition in a non-obese type 2 diabetes rat model. Obes Surg 2005; 15: 1258-1264.. [15] Mojsov S, Heinrich G, Wilson IB, Ravazzola M, Orci L, Habener JF. Preproglucagon gene expression in pancreas and intestine diversifies at the level of post-translational processing. J Biol Chem 1986; 261: 11880-11889. [16] Tornehave D, Kristensen P, Romer J, Knudsen LB, Heller RS. Expression of the GLP-1 recep- 4141 Int J Clin Exp Pathol 2014;7(7):4136-4142

tor in mouse, rat, and human pancreas. J Histochem Cytochem 2008; 56: 841-851. [17] 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. [18] Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC. Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 2003; 52: 102-110. [19] Liu Y, Zhou Y, Wang Y, Geng D, Liu J. Roux-en-Y gastric bypass-induced improvement of glucose tolerance and insulin resistance in type 2 diabetic rats are mediated by glucagon-like peptide-1. Obes Surg 2011; 21: 1424-1431. 4142 Int J Clin Exp Pathol 2014;7(7):4136-4142