• Non ci sono risultati.

Adipose Tissue Glucose Uptake

N/A
N/A
Protected

Academic year: 2021

Condividi "Adipose Tissue Glucose Uptake "

Copied!
134
0
0

Testo completo

(1)

Introduction

In this thesis I have evaluated the postprandial effects on glucose metabolism of a GLP-1 receptor analogue (Exenatide) in subjects with increased postprandial hyperglycemia (ie IGT or newly diagnosed diabetes at screening OGTT). By using tracers infusion combined with Positron Emission Tomography (PET) and 18F-2- fluoro-2-deoxy-D-glucose (FDG), I have evaluated the effect of a single injection of Exeatide vs Placebo on hepatic glucose production, oral glucose absorption, peripheral glucose clearance as well as organ metabolism, eg postprandial brain and liver glucose metabolism.

Diabetes mellitus is a common condition and a serious global health problem and it is important to understand the pathophysiology of this disease in order to prevent the onset of disease as well as further associated complications. Over 382 million people worldwide are estimated to have diabetes that represents about 8.3% of adults (1).

This trend demonstrates that by 2035, some 592 million people, or one adult in 10, will have diabetes. It is demonstrated that diabetes is the leading cause of cardiovascular diseases (angina, myocardial infarction, stroke, peripheral artery disease, and congestive heart failure), blindness, kidney failure, and lower-limb amputation strictly associated and caused by peripheral neuropathy. IDF estimates that as many as 175 million people worldwide, or close to half of all people with diabetes, are unaware of their disease (1). Most of these cases are type 2 diabetes and many studies have found that people with undiagnosed diabetes already have complications, such as chronic kidney disease and heart failure, retinopathy and neuropathy (2-4).

Prediabetes is a condition of high-risk for diabetes and is defined by impaired glucose tolerance (IGT), along with impaired fasting glucose (IFG), that are intermediate states in the transition in glucose tolerance from normal to overt diabetes. IFG was originally introduced by the American Diabetes Association to be analogous to IGT (5). IFG is defined by a fasting glucose between 100 and 126 mg/dl (5.6 and 7.0 mmol/l) while IGT is detected as a postprandial glucose level between 140 and 200 mg/dl (7.8 and 11.1 mmol/l) after a standard glucose load of 75 g oral glucose tolerance testing (OGTT). The future risk of type 2 diabetes associated with IFG and IGT is additive. Thus, the risk of type 2 diabetes in subjects with IFG plus IGT is twofold greater compared with subjects with either state alone (6-11). Furthermore,

(2)

(12) IFG and IGT are associated with a modestly increased risk of cardiovascular disease.

Among the causes of impaired glucose tolerance there are increased insulin resistance and impaired insulin secretion (13). Moreover subjects with prediabetes have an impaired response of splanchnic hormones after meal ingestion, ie glucagon like peptide 1 (GLP-1) and GIP (14-18). Several clinical trials involed prediabetic individuals have been conducted to prevent or delay the overt type 2 diabetes either employing lifestyle intervention (19-25) or antihyperglycemic treatment (21,26-28).  

GLP-1 receptor analogues are a new class of antihyperglycemic agents that have been shown to be effective in treating diabetic hyperglycemia.After discovering GLP-1 and GLP-1-receptors in the brain (29), subsequent studies have shown metabolic effects of GLP-1 intracerebroventricular administration (30). A recent study (31) employing 15O-water Positron Emission Tomography (PET), showed that the postprandial GLP-1 response is associated with activation of the left dorsolateral prefrontal cortex and the hypothalamus. PET in combination with FDG is considered the gold-standard method in the quantification of regional brain glucose uptake. The effect of GLP-1 on brain glucose uptake across the blood brain barrier was recently investigated using this method (32). The results of the study indicated a neuroprotective effect of GLP-1 in maintaining glucose balance and limiting glucose transport across the blood brain barrier. Another study reported that intravenous GLP- 1 infusion in non-diabetic male subjects under conditions of hyperglycemic pancreatic-pituitary clamp, the intracerebral glucose concentration was reduced significantly in multiple brain regions although the glucose clearance across the blood brain barrier, net glucose clearance and cerebral glucose metabolic rate were increased (33).

These studies although interesting, do not answer important questions, such as the effect of GLP-1 on cerebral glucose metabolism in postprandial state since GLP-1 is secreted in response to a meal while previously subjects were studied under euglycemic or hyperglycemic condition obtained with hyperinsulinemic or pancreatic clamp with the suppression of counter-regolatory hormonal secretion by somatostatin.

Thus the primary aim of this study was to measure brain glucose metabolism during an oral glucose load in subjects at risk of developing T2DM using [18F]FDG-PET.

There have been increasing experimental and clinical evidences suggesting that hypothalamic dysregulation may be one of the underlying mechanisms of abnormal

(3)

peripheral glucose metabolism. It has been recently shown that the intravenous continuous infusion of Exenatide in a group of obese non-diabetic subjects, was associated to a increase of functional hypothalamic connectivity (studied with fMRI) with the rest of the areas involved in the glucose homeostasis (34). Therefore the second aim of this study was to evaluate the hypothalamic glucose metabolism in postprandial state and its relation with peripheral glucose metabolism.

It has been suggested that brain GLP-1 signalling mediates peripheral glucose metabolism (35). Recently it has been shown that physiological manifestations of the GLP-1 incretin effect are potentiation of insulin secretion and glucose sensing(36), with a positive relationship between the appearance of oral glucose in peripheral blood and the release of GLP-1(36).

However, the above experiments did not allow to distinguish whether GLP-1 acts on glucose absorption or hepatic extraction. Thus, the third aim of the study was to relate changes in cerebral glucose metabolism with other effects of Exenatide injection, such as postprandial changes in glucose metabolism and in insulin and glucagon secretion. By using glucose stable isotope tracers we measured hepatic glucose production and extraction, oral glucose absorption and peripheral clearance. Hepatic glucose extraction, glucose production and oral absorption, peripheral clearance were assessed by coinfusing stable isotopes and [18F]FDG using a method recently developed (37). Finally we explored if changes in cerebral and peripheral glucose metabolism induced by Exenatide are partly explained by reduction in whole body lipolysis.

The thesis is organized in six chapters. In the first chapter I described the metabolic abnormalities that characterize the large spectrum that leads a normal glucose tolerance individuals to develop prediabetes and then overt type 2 diabetes. In the second chapter I evaluated the postprandial effects of a single Exenatide administration in prediabetic individuals following a standard oral glucose load. In the third chapter the role of the brain in the glucose homeostasis is described highlighting the importance of the gut-to-brain GLP-1 dependent axis and its role in the regulation of food intake and energy expenditure. In the forth chapter the Positron Emission Tomography (PET) technique is fully examined including a detailed description of the methods used to process and analyze PET images obtained during this study. In the fifth chapter I have evaluated the effects of a single Exenatide administration on postprandial cerebral glucose metabolism in prediabetic individuals. In the last

(4)

chapter I have examine the role of Exenatide in the hepatic glucose metabolism suggesting a cross-talk hypothesis between the liver and the brain. An appendix is included to provide a complete description of the study design and methods used during the study.

This intervention study was carried out at the University of Texas, Health Science Center, Research Imaging Institute at San Antonio, TX, USA.

                                                                       

(5)

 

References  

1. International Diabetes Federation. IDF Diabetes Atlas, 6th edn. Brussels, Belgium: International Diabetes Federation, 2013. Available at http://www.idf.org/diabetesatlas

2. Plantinga Lc, crews Dc, coresh J, et al. Prevalence of chronic kidney disease in US adults with undiagnosed diabetes or prediabetes. clin J Am Soc nephrol 2010; 5 (4): 673-682

3. Flores-Le Roux JA, comin J, Pedro-botet J, et al. Seven-year mortality in heart failure patients with undiagnosed diabetes: an observational study. Cardiovasc Diabetol 2011; 10: 39

4. Spijkerman AMW, Dekker JM, nijpels G, et al. Microvascular complications at time of diagnosis of type 2 diabetes are similar among diabetic patients detected by targeted screening and patients newly diagnosed in general practice: the hoorn screening study. Diabetes care 2003; 26 (9): 2604-2608 5. The Expert Committee on the Diagnosis and Classification of Diabetes

Mellitus. Report of the expert committee on the diagnosis and classifica- tion of diabetes mellitus. Diabetes Care 1997;20:1183–1197

6. Gerstein HC, Santaguida P, Raina P, et al. Annual incidence and relative risk of diabetes in people with various categories of dysglycemia: a systematic overview and meta-analysis of prospective studies. Diabetes Res Clin Pract 2007;78:305–312

7. Unwin N, Shaw J, Zimmet P, Alberti KGMM. Impaired glucose tolerance and impaired fasting glycaemia: the current status on definition and in- tervention.

Diabet Med 2002;19:708–723

8. Dankner R, Abdul-Ghani MA, Gerber Y, Chetrit A, Wainstein J, Raz I.

Predicting the 20-year diabetes incidence rate. Diabetes Metab Res Rev 2007;23:551–558

9. Shaw JE, Zimmet PZ, de Courten M, et al. Impaired fasting glucose or impaired glucose tolerance. What best predicts future diabetes in Maur- itius?

Diabetes Care 1999;22:399–402

10. Gabir MM, Hanson R, Dabelea D, et al. Plasma glucose and prediction of microvascular disease and mortality: evaluation of 1997 American Diabetes

(6)

Association and 1999 World Health Organization criteria for diagnosis of diabetes. Diabetes Care 2000;23:1113–1118

11. de Vegt F, Dekker JM, Stehouwer CD, Nijpels G, Bouter LM, Heine RJ. The 1997 American Diabetes Association criteria versus the 1985 World Health Organization criteria for the diagnosis of abnormal glucose tolerance: poor agreement in the Hoorn Study. Diabetes Care 1998;21:1686–1690

12. Ford ES, Zhao G, Li C. Prediabetes and the risk for cardiovascular disease: a systematic review of the evidence. J Am Coll Cardiol 2010;55:1310–7

13. Ferrannini E. Med Clin North Am. 2011;95(2):327-39

14. Toft-Nielsen MB, Damholt MB, Madsbad S, Hilsted LM, Hughes TE, Michelsen BK, Holst JJ. Determinants of the impaired secretion of glucagon- like peptide-1 in type 2 diabetic patients. J Clin Endocrinol Metab 2001,86:3717-2

15. Nauck MA, Vardarli I, Deacon CF, Holst JJ, Meier JJ. Secretion of glucagon- like peptide-1 (GLP-1) in type 2 diabetes: what is up, what is down?

Diabetologia 2011, 54:10-18

16. Rask E, Olsson T, Soderberg S, Holst JJ, Tura A, Pacini G, Ahren B. Insulin secretion and incretin hormones after oral glucose in non-obese subjects with impaired glucose tolerance. Metabolism 2004, 53:624-631

17. Byrne MM, Gliem K, Wank U, Anrold R, Katschinski M, Polonsky KS, Goke B Glucagon-like peptide 1 improves the ability of the beta-cell to sense and respond to glucose in subjects with impaired glucose tolerance. Diabetes 1998, 47:1259-1265

18. Ahren B, Larsson H, Holst. JJ. Reduced gastric inhibitory polypeptide but normal glucagon-like peptide 1 response to oral glucose in postmenopausal women with impaired glucose tolerance. Eur J Endocrinol 1997, 137:127-131 19. Pan XR, Li GW, Hu YH, et al. Effects of diet and exercise in preventing

NIDDM in people with impaired glucose tolerance. The Da Qing IGT and Diabetes Study. Diabetes Care 1997; 20: 537–44

20. Tuomilehto J, Lindström j, Eriksson JG, et al. Prevention of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired glucose tolerance. N Engl J Med. 2001

(7)

21. Knowler WC, Connor E, Fowler SE, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002; 346:

393–403

22. Ramachandran A, Snehalatha C, Mary S, Mukesh B, Bhaskar AD, Vijay V.

The Indian Diabetes Prevention Programme shows that lifestyle modification and metformin prevent type 2 diabetes in Asian Indian subjects with impaired glucose tolerance (IDPP-1). Diabetologia 2006; 49: 289–97

23. Lindahl B, Nilsson TK, Borch-Johnsen K, Roder ME, Soderberg S, Widman L. A randomized lifestyle intervention with 5-year follow-up in subjects with impaired glucose tolerance: pronounced short-term impact but long-term adherence problems. Scand J Public Health 2009; 37: 434–42

24. Bo S, Ciccone G, Baldi C, et al. Effectiveness of a lifestyle intervention on metabolic syndrome. A randomized controlled trial. J Gen Intern Med 2007;

22: 1695–703

25. Roumen C, Feskens EJM, Corpeleijn E, Mensink M, Saris WHM, Blaak EE.

Predictors of lifestyle intervention outcome and dropout: the SLIM study. Eur J Clin Nutr 2011; 65: 1141–7

26. The Diabetes Prevention Program Research Group. Prevention of type 2 diabetes with troglitazone in the diabetes prevention program. Diabetes 2005, 54:1150-1156

27. Dream Trial Investigators. Effect of rosiglitazone on the frequency of diabetes in patients with impaired glucose tolerance or impaired fasting glucose: a randomized controlled trial. Lancet 2006, 368:1096-1105

28. DeFronzo RA, Tripathy D,Schwenke DC, Banerji MA, Bray GA, Buchanan TA, , Clement SC, Henry RR, Hodis HN, Kitabchi AE, Mack WJ, Mudaliar S, Ratner RE, Williams K, Stentz FB, Musi N, Reaven PD for the ACT NOW Study. Pioglitazone for diabetes prevention in impaired glucose tolerance.

NEJM 2011, 364:1104-1115

29. Hamilton A, Holscher C. Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuroreport 2009;20(13):1161–6

30. Turton MD, O'Shea D, Gunn I, Beak SA, Edwards CM, Meeran K, Choi SJ, Taylor GM, Heath MM, Lambert PD, Wilding JP, Smith DM, Ghatei MA,

(8)

Herbert J, Bloom SR. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379(6560):69-72

31. Pannacciulli N, Le DS, Salbe AD, Chen K, Reiman EM, Tataranni PA, Krakoff J. Postprandial glucagon-like peptide-1 (GLP-1) response is positively associated with changes in neuronal activity of brain areas implicated in satiety and food intake regulation in humans. Neuroimage. 2007 Apr 1;35(2):511-7

32. Lerche S, Brock B, Rungby J, Bøtker HE, Møller N, Rodell A, Bibby BM, Holst JJ, Schmitz O, Gjedde A. Glucagon-like peptide-1 inhibits blood-brain glucose transfer in humans. Diabetes. 2008;57(2):325-31

33. Gejl M, Egefjord L, Lerche S, Vang K, Bibby BM, Holst JJ, Mengel A, Møller N, Rungby J, Brock B, Gjedde A. Glucagon-like peptide-1 decreases intracerebral glucose content by activating hexokinase and changing glucose clearance during hyperglycemia. J Cereb Blood Flow Metab 2012;32(12):2146-52

34. Schlögl H, Kabisch S, Horstmann A, Lohmann G, Müller K, Lepsien J, Busse- Voigt F, Kratzsch J, Pleger B, Villringer A, Stumvoll M. Exenatide-induced reduction in energy intake is associated with increase in hypothalamic connectivity. Diabetes Care. 2013;36(7):1933-40

35. Lockie SH. Glucagon-like peptide-1 receptor in the brain: role in neuroendocrine control of energy metabolism and treatment target for obesity.

J Neuroendocrinol. 2013;25(7):597-604

36. Muscelli E, Mari A, Casolaro A, Camastra S, Seghieri G, Gastaldelli A, Holst JJ, Ferrannini E. Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 2008 May;57(5):1340-8

37. Iozzo P, Jarvisalo MJ, Kiss J, Borra R, Naum GA, Viljanen A, Viljanen T, Gastaldelli A, Buzzigoli E, Guiducci L, Barsotti E, Savunen T, Knuuti J, Haaparanta-Solin M, Ferrannini E, Nuutila P. Quantification of liver glucose metabolism by positron emission tomography: validation study in pigs.

Gastroenterology. 2007 Feb;132(2):531-42  

   

(9)

Chapter 1

Metabolic abnormalities from prediabetes toward overt type 2 diabetes

1.1 Introduction

The glucose system is highly homeostatic and in normal glucose tolerance state, plasma glucose concentration fluctuations rarely exceed 54 mg/dl. As the plasma glucose concentration represent a tight balance between the glucose entry into and the exit from the circulation via tissue metabolism or excretion, any excessive release and/or defective removal will result in increasing glucose levels. The knowledge of the mechanisms that underlie prediabetes development provides an extremely useful tool for improving a pharmacological treatment approach focused on T2DM prevention. Insulin resistance is a core defect in both prediabetes and T2DM (1-4) and involves several organs as liver (5) muscle (6), and adipose tissue(7)and it precedes the development of glucose intolerance and overt T2DM (8-10). IFG individuals have moderate hepatic insulin resistance and impaired early insulin response (0–30 min) during the OGTT (11-12). As such, they are characterized by elevated FPG and an excessive early rise (0–60 min) in PG during OGTT (13). However, because the late plasma insulin response (60–120 min) is intact and muscle insulin sensitivity is normal/near-normal, the 2-h PG returns to its starting FPG level (14). In contrast, IGT individuals have moderate-severe muscle insulin resistance and impaired early (0–30 min) and late (60–120 min) plasma insulin responses during the OGTT (14). Thus, although the FPG is not elevated, there is a progressive, sustained rise in PG during the OGTT and the 2-h PG remains well above the FPG level. Impaired first phase insulin secretion is characteristic of both IGT and IFG. However, they are distinguished by intact second phase insulin secretion in IFG and the tissues (liver in IFG vs. muscle in IGT) responsible for insulin resistance. In both states skeletal muscle is the main responsible for impaired insulin-mediated glucose uptake (it typically represents 40% if body weight (15). However, adipose tissue has a significant contribution to whole body glucose disposal (16) especially because adipocytes insulin resistance directly causes an excessive NEFA release into the

(10)

bloodstream that directly generate their preferential uptake over that of circulating glucose to insulin target tissue (Randle cycle) (17).

1.2 The role of β-cell dysfunction and insulin resistance

Although insulin resistance is a cardinal feature of IGT, ultimately, β-cell failure is responsible for the development of IGT and its progression to T2DM (18-19). In all ethnic groups the low plasma insulin response during the OGTT associated with the impairment in first phase insulin secretion (0-10 minute time period following an intravenous glucose challenge) are strong predictors of the progression from IGT to T2DM (20-21). When the FPG exceeds 90 mg/dl, first phase insulin secretion begins to deteriorate, and, when the FPG exceeds 110 mg/dl, first phase insulin secretion is almost completely lost (22). Genetic and acquired factors [glucotoxicity (23), lipotoxicity (24), incretin deficiency/resistance (25-27)] play a major role in the progressive deterioration of β-cell function. Individuals with IGT are maximally/near maximally insulin resistant and have lost 70–80% of β-cell function and even a small further decline in insulin secretion will result in a marked increase in fasting/postprandial blood glucose levels. Therefore, any intervention effective in retard/prevent β-cell failure will result in reverse/delay of the progression from IGT to T2DM.

1.3 The role of Incretin System

The incretin effect is characterized by a 2-3 fold increase in plasma insulin response following a glucose ingestion as compared to the same level of hyperglycemia obtained by an intravenous glucose administration (28-29). Together, GLP-1, released by L cells, and glucose-dependent insulinotropic polypeptide (GIP), released by K cells, account for 90% of the incretin effect (28-29). Both GLP-1 and GIP are potent insulin secretagogues. In addition to augmenting insulin secretion, GLP-1 also inhibits glucagon secretion, delays gastric emptying, and promotes weight loss by suppressing appetite and decreasing food intake (28,30).Because both GLP-1 and GIP are rapidly cleaved (half-life = 1–2 min) by dipeptidyl peptidase-IV, both peptides are inappropriate for therapeutic use in T2DM and IGT subjects. Liraglutide and exenatide are GLP-1 receptor agonists (GLP-1rA) that mimic the actions of GLP-

(11)

1 and are resistant to dipeptidyl peptidase-IV degradation (31-32). Similar to endogenous GLP-1, both agonists are potent insulin secretagogues (28-29)inhibit glucagon secretion, promote weight loss and effectively reduce plasma glucose levels in T2DM (29). In a long-term (3 years) extension study, exenatide produced a durable HbA1c reduction, improved β-cell function, and caused progressive weight loss in type 2 diabetic individuals(33).Hypoglycemia is uncommon with GLP-1 analogues because they only augment insulin secretion in the presence of hyperglycemia. Both GLP-1 and GIP mediate their β-cell effects via a mechanism that is completely distinct from hyperglycemia. Following binding to their respective receptors, GLP-1 and GIP activate adenylate cyclase which convert ATP to cAMP, which, in turn,

“amplifies” an insulin secretory system that has been activated by hyperglycemia (34). In the absence of hyperglycemia, neither GLP-1 nor GIP increases insulin secretion.

Subjects with IGT and T2DM are characterized by severely impaired β-cell function (35-36) and markedly reduced incretin effect in response to meal/glucose ingestion (29).Conversely most studies have reported no change or slightly impaired total GLP- 1 response to a mixed meal in IGT (25,37-38) although the early (0–10 min) GLP-1 response is diminished, indicating a phasic defect in GLP-1 secretion; GIP secretion is normal or slightly increased in prediabetes (29). However, GLP-1 analogs represent a candidates for type 2 diabetes prevention. It has been shown that the primary defect in IGT and T2DM is an inability of the β-cell to respond to glucose (18) and this β-cell

“blindness” to glucose, at least in part, can be restored by the incretin hormones (39- 40). These results indicate that pharmacological levels of plasma GLP-1 are capable of correcting β-cell “blindness” to glucose and restoring normal β-cell function to glucose in IGT and T2DM individuals. Although the stimulatory effect of the GLP-1 analogues on beta cell function weans rapidly upon washing out the drug, a recent study reported that, if exenatide is continued for 3 years, its ability to enhance β-cell responsiveness to glucose is partially retained (41). For the reasons, GLP-1 analogs may represent an ideal treatment option to delay or prevent the progression to T2DM in individuals with prediabetes.

1.4 The role of endogenous glucose production in postabsorptive state

In the postabsorptive state, the liver of healthy subjects produces glucose at the rate of

(12)

2.0 mg/kg/min (4,42). This glucose efflux is important to meet the needs of the brain and other neural tissues, which use glucose at a constant rate of approximately 1.0 to 1.2 mg/kg/min (43). Under fasting conditions, brain glucose uptake is insulin- independent and accounts for approximately 50% to 60% of glucose disposal (Figure 1).

Figure 1. : Percentages of contribution to glucose homeostasis after an 18 hours fast in human metabolism.

Also brain glucose uptake occurs at the same rate during absorptive and postabsorptive periods, and seems to be not altered in type 2 diabetes. As shown in the figure 1 the rest of the glucose uptake occurs principally in the skeletal muscle, adipose tissue, liver and kidneys while the majority of endogenous glucose production occur in the liver. The kidney possesses all of the gluconeogenesis enzymes required to produce glucose, and estimates of the renal contribution to total endogenous glucose production have ranged from 10% to 25% (44-45). One unconfirmed study suggests that the rate of renal gluconeogenesis is increased in type 2 diabetics with fasting hyperglycemia (46), but studies using the hepatic vein catheter technique have shown that all of the increase in total body endogenous glucose production (measured with [3-3H]glucose) in type 2 diabetics can be accounted for by increased hepatic glucose output (measured by the hepatic vein catheter technique) (47). In type 2 diabetics with mild fasting hyperglycemia (≤140 mg/dL), the postabsorptive level of hyperinsulinemia is sufficient to overcome the hepatic insulin resistance and maintain a normal basal rate of hepatic glucose output (4). In diabetic subjects with mild to moderate fasting hyperglycemia (140– 200 mg/dL, 7.8–11.1 mmol/L), however, basal

(13)

hepatic glucose production is increased by approximately 0.5 mg/kg/min (4,42). The increase in basal hepatic glucose production (HGP) is closely correlated with the severity of fasting hyperglycemia and this has been demonstrated in numerous studies (48). In conclusion, in type 2 diabetics with overt fasting hyperglycemia (≥140 mg/dL, 7.8 mmol/L), an excessive rate of hepatic glucose output is the major abnormality responsible for the elevated FPG concentration.

1.5 The role of hepatic insulin resistance

Under postabsorptive conditions, the fasting plasma insulin concentration in type 2 diabetics is 2- to 4-fold greater than in nondiabetic subjects (49). Given that hyperinsulinemia is a potent inhibitor of HGP, an hepatic resistance to the action of insulin must be present to explain the excessive output of glucose by the liver.

Because hyperglycemia per se exerts a powerful suppressive action on HGP, the liver also must be resistant to the inhibitory effect of hyperglycemia on hepatic glucose output. The dose response relationship between hepatic glucose production and the plasma insulin concentration has been examined with the euglycemic insulin clamp technique and radioisotopic glucose (Figure 2)(42).

Figure 2. Dose-response curve relating the plasma insulin concentration to the suppression of hepatic glucose production in control (Ÿ) and type 2 diabetic (o) subjects with moderately severe fasting hyperglycemia. * P < 0.05; ** P < 0.01 versus control subjects. (Reproduced form Groop LC, Bonadonna RC, DelPrato S, Ratheiser K, Zyck K, Ferrannini E, DeFronzo RA. Glucose and free fatty acid metabolism in non-insulin-dependent diabetes mellitus. Evidence for multiple sites of insulin resistance. J Clin Invest. 1989;84(1):205–13)

The figure 2 shows that the dose-response curve relating inhibition of HGP to the

(14)

plasma insulin level is very steep and in type 2 diabetics it is shifted to the right, indicating resistance to the inhibitory effect of insulin on hepatic glucose production.

Elevation of the plasma insulin concentration to the high physiologic range (≈100 UI/mL), however, can overcome the hepatic insulin resistance and cause a near normal suppression of HGP, also the severity of the hepatic insulin resistance is related to the level of glycemic control. In type 2 diabetics with mild fasting hyperglycemia, an increment in plasma insulin concentration of 100 UI/mL causes a complete suppression of HPG; however, in diabetic subjects with more severe fasting hyperglycemia, the ability of the same plasma insulin concentration to suppress HGP is impaired. These observations indicate that there is an acquired component of hepatic insulin resistance, which becomes progressively worse as the diabetic state decompensates over time. Hepatic glucose production can be derived from either glycogenolysis or gluconeogenesis (50)(Figure 3 ).

Figure 3. Hepatic and renal gluconeogenesis in healthy individuals (CT) and type 2 diabetic subjects (T2D) (Reproduced from Gastaldelli A, Baldi S, Pettiti M, Toschi E, Camastra S, Natali A, et al. Influence of gluconeogenesis and glucose output in humans:

Recent studies using 13C-labeled magnetic resonance imaging (51-52) have confirmed that in the liver the majority of endogenous glucose production can be accounted by gluconeogenesis while the rest is accounted by glycogenolysis. In the kidneys the totality of glucose production is accounted by gluconeogenesis. A variety of mechanisms has been shown to contribute to the increase in hepatic

Hepa%c'and'Renal'Gluconeogenesis'

0' 3' 6' 9' 12' 15' 18'

CT'lean' CT'obese' GLYCOG' GNG'

0' 3' 6' 9' 12' 15' 18'

T2D'lean' T2D'obese' GLYCOG' GNG'

EGP$'(umol/min$kgffm)'

*' *'

*' *'

Blood$glucose$

Hepa%c'GNG'

22C43%'of'EGP' Renal'GNG' 10C25%'of'EGP' Glycogenolysis''

38C53%'of'EGP' Glycogenolysis'' 0%'of'EGP'

JE'Gerich'Diabe%c'Medicine,'2010;'Gastaldelli'A'et'al'Diabetes'2000'

(15)

gluconeogenesis, including hyperglucagonemia, enhanced sensitivity to glucagon, increased circulating levels of gluconeogenic precursors (lactate, alanine, glycerol), increased FFA oxidation, and decreased sensitivity to insulin.

1.6 The role of Splanchnic (hepatic) glucose uptake and Glucagon

The splanchnic tissues, like the brain, are largely insensitive to insulin with respect to the stimulation of glucose uptake. Also there are no differences between diabetic and control subjects in the amount of glucose taken up by the splanchnic tissues as demonstrated in study performed using the hepatic vein catheterization technique in combination with the euglycemic insulin clamp (53). Under conditions of euglycemic hyperinsulinemia, very little infused glucose is taken up by the splanchnic (and therefore hepatic) tissues. Under postabsorptive conditions, approximately 50% of total hepatic glucose output is dependent on the maintenance of normal basal glucagon levels, and inhibition of basal glucagon secretion with somatostatin causes a reduction in hepatic glucose production and plasma glucose concentration (Figure 4).

Figure 4. Interrelation between plasma insulin and glucagon levels during postprandial state and postabsorbitive state.

As reported in the figure 4, in fasting state glucagon levels participate to glucose homeostasis by stimulating glycogenolysis, gluconeogenesis and ketogenesis. After a glucose load, glucagon secretion is inhibited by hyperinsulinemia, and the resultant

(16)

hypoglucagonemia contributes to the suppression of hepatic glucose production and maintenance of normal postprandial glucose tolerance. The route of glucose entry into the body also plays an important role in the distribution of administered glucose and overall glucose homeostasis (50, 54-55).Several studies have shown that basal plasma glucagon concentration is elevated in type 2 diabetic individuals (56-58). The important contribution of elevated fasting plasma glucagon levels to the increased basal rate of HGP in type 2 diabetic individuals was provided by Baron (59).

Compared with control subjects, diabetic individuals had a markedly elevated rate of basal HGP, which correlated closely with the increase in fasting plasma glucagon concentration. However, following a somatostatin infusion, plasma glucagon levels declined by 44% in association with a 58% decrease in basal HGP. These results conclusively demonstrate the pivotal role of hyperglucagonemia in the pathogenesis of fasting hyperglycemia in type 2 diabetes.

1.7 The role of adipocytes and fat metabolism

A deranged adipocyte metabolism and altered fat topography has an important role in the development of glucose intolerance in type 2 diabetes(24,60-61). Fat cells are resistant to insulin’s antilipolytic effect, leading to day-long elevation in the plasma NEFA concentration (62). Chronically increased plasma NEFA levels stimulate gluconeogenesis (63), induce hepatic/muscle insulin resistance (64), and impair insulin secretion (60,65). These NEFA-induced disturbances are referred to as lipotoxicity. Dysfunctional fat cells produce excessive amounts of insulin resistance–

inducing, inflammatory, and atherosclerotic provoking adipocytokines and fail to secrete normal amounts of insulin-sensitizing adipocytokines such as adiponectin (24). Enlarged fat cells are insulin resistant and have diminished capacity to store fat (66). When adipocyte storage capacity is exceeded, lipid “over-flows” into muscle, liver, and beta cells, causing muscle/ hepatic insulin resistance and impaired insulin secretion. In both type 2 diabetic and obese nondiabetic subjects, the ability of insulin to suppress the plasma FFA concentration and inhibit FFA turnover is significantly impaired compared with lean normal glucose tolerant control subjects at all plasma insulin concentrations (62).Many studies have shown that a physiological elevation in the plasma NEFA concentration stimulates HGP and impairs insulin-stimulated glucose uptake in liver (67) and muscle (68-69). Also NEFA have been shown to have

(17)

independent effects to inhibit glycogen synthase (70) and both glucose transport and glucose phosphorylation (71). Collectively, these findings provide strong support for lipotoxicity and adipocyte insulin resistance in the pathogenesis of type 2 diabetes.

1.8 Conclusions

Individuals with prediabetes manifest almost all the metabolic abnormalities that are present in type 2 diabetes. Insulin resistance (primarily affecting skeletal muscle, liver and adipose tissue) and islet dysfunction mostly characterized by β-cell failure and hyperglucagonemia play an important role for the progression from NGT to IGT to T2DM. The principal consequences of insulin resistance in metabolically important organs, as such as liver and adipose tissue, are characterized by an increase in hepatic insulin resistance and derangement of adipocyte metabolism. The net results is an increased endogenous glucose production (primarily from liver) and lipid “over- flows” from adipose tissue into muscle, liver, and beta cells, which in turn causes muscle/ hepatic insulin resistance and impaired insulin secretion, suggesting a typical vicious cycle. The incretin system is an important player for the control of glucose homeostasis and acts within the continuum of metabolic abnormalities that lead to overt type 2 diabetes. It has been shown that incretin effect is abnormal in prediabetic individual and GLP-1 administration can, at least in part, correct the metabolic abnormalities that characterize the glucose tolerance deterioration. In particular it has been shown that GLP-1 analogs can improve β-cell function by restoring beta cell glucose sensitivity, promote weight loss, reduce appetite and food intake and do not cause hypoglycemia. Therefore GLP-1 analogs are likely to be an ideal agent for decreasing the progression of IFG/IGT to T2DM and reverting glucose tolerance to normal. However, the effect of GLP-1 analogues on the progression of IGT to T2DM has yet to be tested in a large scale in long term outcome studies.

(18)

References

1. DeFronzo RA. Banting Lecture. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes. Diabetes 2009, 58:773-795 2. DeFronzo RA, Ferrannini E. Insulin resistance: a multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and ASCVD.

Diabetes Care 1991, 14:173-194

3. Reaven G. Banting Lecture. Role of insulin resistance in human disease.

Diabetes 1988, 37:1595-1607

4. DeFronzo RA, Ferrannini E, Simonson DC. Fasting hyperglycemia in non- insulin dependent diabetes mellitus: contributions of excessive hepatic glucose production and impaired tissue glucose uptake. Metabolism 1989 38:387-395 5. DeFronzo RA, Ferrannini E, Simonson DC. Fasting hyperglycemia in non- insulin dependent diabetes mellitus: contributions of excessive hepatic glucose production and impaired tissue glucose uptake. Metabolism 1989, 38:387-395 6. Groop LC, Bonadonna RC, DelPrato S, Ratheiser K, Zyck K, Ferrannini E, DeFronzo RA. Glucose and free fatty acid metabolism in non-insulin- dependent diabetes mellitus. Evidence for multiple sites of insulin resistance. J Clin Invest. 1989, 84:205-13

7. DeFronzo RA. Banting Lecture. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes. Diabetes 2009, 58:773-795 8. Ferrannini E, Gastaldelli A, Miyazaki Y, Matsuda M, Mari A, DeFronzo RA.

Beta cell function in subjects spanning the range from normal glucose tolerance to overt diabetes mellitus: A new analysis. J Clin Endocrinol Metab 2005, 90:493-500

9. Weyer C, Tataranni PA, Bogardus C, Pratley RE. Insulin resistance and insulin secretory dysfunction are independent predictors of worsening of glucose tolerance during each stage of type 2 diabetes development. Diabetes Care 2000, 24:89-94

10. Saad MF, Knowler WC, Pettitt DJ, Nelson RG, Mott DM, Bennett PH. The natural history of impaired glucose tolerance in the Pima Indians. N Engl J Med 1988, 319:1500-1505

(19)

11. Abdul-Ghani MA, Matsuda M, and DeFronzo RA. Strong association between insulin resistance in liver and skeletal muscle in non-diabetic subjects.

Diabetes Medicine 2008, 25:1289-94

12. Jani R, Molina M, Matsuda M, Balas B, Chavez A, DeFronzo RA and Abdul- Ghani MA. Decreased non-insulin-dependent glucose clearance contributes to the rise in fasting plasma glucose in the nondiabetic range. Diabetes Care.

2008, 31:311-5

13. Abdul-Ghani MA, Lyssenko V, Tuomi T, DeFronzo RA and Groop L. The shape of glucose curve predicts the risk for future type 2 diabetes. Diabete Metab. Res Rev 2010, 26:280-6

14. Meyer C, Pimenta W, Woerle HJ, Van Haeften T, Szoke E, Mitrakou A, Gerich J. Different mechanisms for impaired fasting glucose and impaired postprandial glucose tolerance in humans. Diabetes Care 2006, 29:1909-14 15. Ferrannini E, Mari A. How to measure insulin sensitivity. J Hypertens

1998;16: 895–906

16. VirtanenKA, IozzoP, HallstenK, et al.Increased fat mass compensates for insulin resistance in abdominal obesity and type 2 diabetes: a positronemitting tomography study. Diabetes 2005;54:2720–6

17. Randle PJ. Regulatory interactions between lipids and carbohydrates: the glucose fatty acid cycle after 35 years. Diabetes Metab Rev 1998;14:263–8 18. Ferrannini E, Gastaldelli A, Miyazaki Y, Matsuda M, Mari A, DeFronzo RA

Beta cell function in subjects spanning the range from normal glucose tolerance to overt diabetes mellitus: A new analysis. J Clin Endocrinol Metab 2005 90:493-500

19. Abdul-Ghani M, Tripathy D, DeFronzo RA. Contribution of beta cell dysfunction and insulin resistance to the pathogenesis of impaired glucose tolerance and impaired fasting glucose. Diabetes Care 2006, 29:1130-1139 20. Pimenta W, Korytkowski M, Mitrakou A, Jenssen T, Yki-Jarvinen H, Evron

W, Dailey G, Gerich J Pancreatic beta-cell dysfunction as the primary genetic lesion in NIDDM: Evidence from studies in normal glucose-tolerant individuals with a first-degree NIDDM relative. JAMA 1995, 273: 1855-1861 21. Abdul-Ghani MA, Williams K, DeFronzo RA, Sterm M. What is the best

predictor of future type 2 diabetes? Diabetes Care 2007, 30:1544-48.

(20)

22. Godsland IF, Jeffs JA, Johnston DG Loss of beta cell function as fasting glucose increases in the non-diabetic range. Diabetologia 2004, 47:1157-66 23. Rossetti L, Giaccari A, DeFronzo RA. Glucose toxicity. Diabetes Care-

Reviews 1990, 13:610-630

24. Bays H, Mandarino L, DeFronzo RA. Role of the adipocytes, FFA, and ectopic fat in the pathogenesis of type 2 diabetes mellitus: PPAR agonists provide a rational therapeutic approach. J Clin Endocrinol Metab 2004, 89:463-478

25. Toft-Nielsen MB, Damholt MB, Madsbad S, Hilsted LM, Hughes TE, Michelsen BK, Holst JJ. Determinants of the impaired secretion of glucagon- like peptide-1 in type 2 diabetic patients. J Clin Endocrinol Metab 2001, 86:3717-2

26. Højberg PV, Vilsbøll T, Rabøl R, Knop FK, Bache M, Krarup T, Holst JJ, Madsbad S. Four weeks of near-normalisation of blood glucose improves the insulin response to glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide in patients with type 2 diabetes. Diabetologia 2009, 52:199-207

27. Vilsbøll T, Krarup T, Madsbad S, Holst JJ. Defective amplification of the late phase insulin response to glucose by GIP in obese Type II diabetic patients.

Diabetologia 2002, 45:1111-1119

28. Holst JJ. Glucagon-like peptide-1: from extract to agent. The Claude Bernard Lecture. Diabetologia 2005, 49:253-260

29. Nauck MA, Vardarli I, Deacon CF, Holst JJ, Meier JJ. Secretion of glucagon- like peptide-1 (GLP-1) in type 2 diabetes: what is up, what is down?

Diabetologia 2011,54:10-18

30. Triplitt C, DeFronzo RA. Exenatide: first-in-class incretin mimetic for the treatment of type 2 diabetes mellitus. Expert Rev Endocrinol Metab 2006, 1:329-341

31. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (synthetic exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes mellitus. Diabetes Care 2005, 28:1092-1100

32. Nauck M, Frid A, Hermansen K, Shah NS, Tankova T, Mitha IH, Zdravkovic M, During M, Matthews DR; LEAD-2 Study Group. Efficacy and safety

(21)

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 2009, 32:84-90

33. Klonoff DC, Buse JB, Nielsen LL, Guan X, Bowlus CL, Holcombe JH, Wintle ME, Maggs DG. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008, 24:275-86

34. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP.

Gastroenterology 2007, 132:2131-2157

35. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, Nathan DM; Diabetes Prevention Program Research Group. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin.

NEJM 2002, 346:393-403

36. Abdul-Ghani M, Tripathy D, DeFronzo RA. Contribution of beta cell dysfunction and insulin resistance to the pathogenesis of impaired glucose tolerance and impaired fasting glucose. Diabetes Care 2006, 29:1130-1139 37. Rask E, Olsson T, Soderberg S, Holst JJ, Tura A, Pacini G, Ahren B. 2004

Insulin secretion and incretin hormones after oral glucose in non-obese subjects with impaired glucose tolerance. Metabolism 53:624-631

38. Ahren B, Larsson H, Holst. JJ. 1997 Reduced gastric inhibitory polypeptide but normal glucagon-like peptide 1 response to oral glucose in postmenopausal women with impaired glucose tolerance. Eur J Endocrinol 137:127-131

39. Vilsbøll T. The effects of glucagon-like peptide-1 on the beta cell. Diabetes Obes Metab. 2009;11 Suppl 3:11-8

40. Byrne MM, Gliem K, Wank U, Anrold R, Katschinski M, Polonsky KS, Goke B. Glucagon-like peptide 1 improves the ability of the beta-cell to sense and respond to glucose in subjects with impaired glucose tolerance. Diabetes 1998, 47:1259-1265

41. Bunck MC, Cornér A, Eliasson B, Heine RJ, Shaginian RM, Taskinen MR, Smith U, Yki-Järvinen H, Diamant M Effects of exenatide on measures of β- cell function after 3 years in metformin-treated patients with type 2 diabetes.

Diabetes Care 2011, 34:2041-7

(22)

42. Groop LC, Bonadonna RC, Del Prato S, Ratheiser K, Zych K, Ferrannini E, DeFronzo RA. Glucose and free fatty acid metabolism in non-insulin dependent diabetes mellitus. Evidence for multiple sites of insulin resistance. J Clin Invest 1989;84:205–15

43. Huang SC, Phelps ME, Hoffman EJ, Sideris K, Selin CJ, Kuhl DE. Non- invasive determination of local cerebral metabolic rate of glucose in man. Am J Physiol 1980;238:E69–82

44. Gerich JE, Meyer C, Woerle HJ, Stumvoll M. Renal gluconeogenesis. Its importance in human glucose homeostasis. Diabetes Care 2001;24:382–91 45. Ekberg K, Landau BR, Wajngot A, Chandramouli V, Efendic S, Brunengraber

H, et al. Contributions by kidney and liver to glucose production in the postabsorptive state and after 60 h of fasting. Diabetes 1999;48:292–8

46. Meyer C, Stumvoll M, Nadkarni V, Dostou J, Mitrakou A, Gerich J.

Abnormal renal and hepatic glucose metabolism in type 2 diabetes mellitus. J Clin Invest 1998;102:619–24

47. DeFronzo RA, Gunnarsson R, Bjorkman O, Olsson M, Wahren J. Effects of insulin on peripheral and splanchnic glucose metabolism in non-insulin dependent diabetes mellitus. J Clin Invest 1985;76:149–55

48. Fery F. Role of hepatic glucose production and glucose uptake in the pathogenesis of fasting hyperglycemia in Type 2 diabetes: normalization of glucose kinetics by short-term fasting. J Clin Endocrinol Metab 1994;78:536–

42

49. DeFronzo RA. Pathogenesis of type 2 diabetes mellitus: metabolic and molecular implications for identifying diabetes genes. Diabetes 1997;5:177–

269

50. Cherrington AD. Control of glucose uptake and release by the liver in vivo.

Diabetes 1999;48:1198–214

51. Magnusson I, Rothman D, Katz L, Shulman R, Shulman G. Increased rate of gluconeogenesis in type II diabetes: a 13C nuclear magnetic resonance study.

J Clin Invest 1992;90:1323–7

52. Gastaldelli A, Baldi S, Pettiti M, Toschi E, Camastra S, Natali A, et al.

Influence of obesity and type 2 diabetes on gluconeogenesis and glucose output in humans: a quantitative study. Diabetes 2000;49:1367–73

(23)

53. DeFronzo RA, Ferrannini E, Hendler R, Felig P, Wahren J. Regulation of splanchnic and peripheral glucose uptake by insulin and hyperglycemia.

Diabetes 1983;32:35–45

54. DeFronzo RA, Ferrannini E, Hendler R, Wahren J, Felig P. Influence of hyper- insulinemia, hyperglycemia, and the route of glucose administration on splanchnic glucose exchange. Proc Natl Acad Sci USA 1978;75:5173–7.

55. Ferrannini E, Wahren J, Felig P, DeFronzo RA. Role of fractional glucose extraction in the regulation of splanchnic glucose metabolism in normal and diabetic man. Metabolism 1980;29:28–35.

56. Consoli A, Nurjhan N, Reilly JJ Jr, Bier DM, Gerich JE. Mechanism of increased gluconeogenesis in noninsulin-dependent diabetes mellitus: role of alterations in systemic, hepatic, and muscle lactate and alanine metabolism. J Clin Invest 1990;86:2038–2045

57. Matsuda M, DeFronzo RA, Glass L, Consoli A, Giordano M, Bressler P, DelPrato S. Glucagon dose response curve for hepatic glucose production and glucose disposal in type 2 diabetic patients and normal individuals.

Metabolism 2002;51:1111–1119

58. Reaven GM, Chen YD, Golay A, Swislocki AL, Jaspan JB. Documentation of hyperglucagonemia throughout the day in nonobese and obese patients with noninsulin-dependent diabetes mellitus. J Clin Endocrinol Metab 1987;64:106–110

59. Baron AD, Schaeffer L, Shragg P, Kolterman OG. Role of hyperglucagonemia in maintenance of increased rates of hepatic glucose output in type II diabetics. Diabetes 1987;36:274–283

60. Kashyap S, Belfort R, Gastaldelli A, Pratipanawatr T, Berria R, Pratipana- watr W, Bajaj M, Mandarino L, DeFronzo RA, Cusi K. A sustained increase in plasma free fatty acids impairs insulin secretion in non-diabetic subjects genetically predisposed to develop type 2 diabetes. Diabetes 2003;52:2461–

2474

61. Bays HE, Gonzalez-Campoy JM, Bray GA, Kitabchi AE, Bergman DA, Schorr AB, Rodbard HW, Henry RR. Pathogenic potential of adipose tissue and metabolic consequences of adipocyte hypertrophy and in- creased visceral adiposity. Expert Rev Cardio Ther 2008;6:343–368

(24)

62. Groop L, Saloranta C, Shank M, Bonadonna RC, Ferrannini E, DeFronzo RA.

The role of free fatty acid metabolism in the pathogensis of insulin resistance in obesity and non-insulin dependent diabetes mellitus. J Clin Endocrinol Metab 1991;72:96–107

63. Bevilacqua S, Bonadonna R, Buzzigoli G, Boni C, Ciociaro D, Maccari F, Giorico MA, Ferrannini E. Acute elevation of free fatty acid levels leads to hepatic insulin resistance in obese subjects. Metabolism 1987;36:502–506 64. Roden M, Price TB, Perseghin G, Petersen KF, Rothman DL, Cline GW,

Shulman GI. Mechanism of free fatty acid-induced insulin resistance in humans. J Clin Invest 1996;97:2859–2865

65. Carpentier A, Mittelman SD, Bergman RN, Giacca A, Lewis GF. Prolonged elevation of plasma free fatty acids impairs pancreatic beta-cell function in obese nondiabetic humans but not in individuals with type 2 diabetes.

Diabetes 2000;49:399–408

66. Bray GA, Glennon JA, Salans LB, Horton ES, Danforth E Jr, Sims EA.

Spontaneous and experimental human obesity: effects of diet and adipose cell size on lipolysis and lipogenesis. Metabolism 1977;26:739–747

67. Bajaj M, Pratipanawatr T, Berria R, Pratipanawatr W, Kashyap S, Cusi K, Mandarino L, DeFronzo RA. Free fatty acids reduce splanchnic and peripheral glucose uptake in patients with type 2 diabetes. Diabetes 2002;51:3043–3048 68. Itani SI, Ruderman NB, Schmieder F, Boden G. Lipid-induced insulin

resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IkB-β. Diabetes 2002;51:2005–2011

69. Richardson DK, Kashyap S, Bajaj M, Cusi K, DeFronzo RA, Jenkinson CP, Mandarino LJ. Lipid infusion induces an inflammatory/fibrotic response and decreases expression of nuclear encoded mitochondrial genes in human skeletal muscle. J Biol Chem 2005;280:10290–10297

70. Johnson AB, Argyraki M, Thow JC, Cooper BG, Fulcher G, Taylor R. Effect of increased free fatty acid supply on glucose metabolism and skeletal muscle glycogen synthase activity in normal man. Clin Science 1992;82:219 –226 71. Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, Cline GW, Slezak

LA, Andersen DK, Hundal RS, Rothman DL, Petersen KF, Shulman GI.

Effects of free fatty acids on glucose transport and IRS-1-associated phosphatidylinositol 3-kinase activity. J Clin Invest 1999;103:253–259

(25)

Chapter 2

The acute effects of Exenatide on postprandial glucose metabolism in prediabetic individuals

2.1 Introduction

It has been shown that the effect of Exenatide on postprandial glucose metabolism is mainly acute and it is due mainly to the delay in gastric emptying (1-2). The acute effect of Exenatide in subjects with prediabetes or newly diagnosed type 2 diabetes, i.e., when the beta cell function is not completely lost, has not been studied previously. In the present study we evaluated the effect of acute Exenatide subcutaneous administration on postprandial glucose metabolism in 15 individuals of which 12 with prediabetes and 3 newly diagnosed type 2 diabetes at screening using oral glucose tolerance test (OGTT, 75g of glucose per os) combined with the infusion of a deuterated glucose tracer i.v. and 13C glucose added to the glucose load.

It has been established that Ex decreases postprandial hyperglycemia mainly by delaying gastric emptying. We have previously shown that this effect of Exenatide is acute, since after 14 days of treatment gastric emptying was delayed only when subjects were given Ex before the meal (1-2).The double tracer OGTT allows to fully evaluate the role of Ex in the modulation of postprandial glucose metabolism via the evaluation of the rate of glucose absorption, the endogenous glucose production and the peripheral glucose disposal.

Moreover we combined the OGTT with the injection of 2-deoxy-2-(18F)fluoro-D- glucose (FDG) and PET screening to study if the acute injection of exenatide could affect postprandial hepatic glucose uptake.

2.2 Protocol and Characteristics of the study subjects

We studied fifteen male subjects that had at screening OGTT impaired glucose tolerance (n=12) or newly diagnosed with type 2 diabetes mellitus (n=3).  In this study we included 12 subjects with prediabetes and 3 patients with type 2 diabetes that was newly diagnosed at the time of the screening visit. We decided to include these

(26)

subjects in the study because their HbA1c was lower than prediabetic subjects (5.54%

vs. 5.7%) and fasting plasma glucose was similar. Moreover, these 3 diabetic subjects provided a previous recent OGTT done before screening visit and the beginning of the study in which a prediabetes was diagnosed. Individuals with prediabetes had FPG=111±3 mg/dl, 2h-PG=157±4 mg/dl, HbA1c=5.7±0.1% while those with newly diagnosed type 2 diabetes had FPG=116±4 mg/dl, 2h-PG=230±2 mg/dl, HbA1c=5.5±0.2%. Each enrolled subject underwent two study sessions combining oral glucose tolerance test (OGTT, 75g of glucose per os) with the infusion of a deuterated glucose tracer i.v. and 13C glucose added to the glucose load. Moreover at OGTT we injected of 5 uCi of 2-deoxy-2-(18F)fluoro-D-glucose (FDG) and PET screening for the quantification of regional organ glucose uptake (in this case liver). A 6,6-2H2-glucose infusion was commenced at T=-120 min and administered continuously for 6h throughout the study. Exenatide (Ex) (5mcg) or placebo (PLC) was administered in random double blind order 30min before the 75 g oral glucose load. Glucose enriched with U-13C6-glucose (1.5g) was added to the 75g of oral glucose that was administered at T=0 together with the FDG bolus i.v. infusion.

Figure 1 reports the diagram of the study protocol.

Figure 1: Protocol Design

Liver/Heart*scan!

Brain*scan!

Posi1oning*

+*Pre*scan! Post*FDG*scan*

(Heart*+*Brain)!

75*grams*glucose*load*with*13!C!!D!glucose!to*! quan1fy*glucose*absorp1on!

D!180!min! D30!min! 0*min! 180!min!

[!18F]FDG! !

injec1on!

60*min! 120*min!

PET*scan! Exena1de!or*!

Placebo*! injec1on!

240!min!

75 grams glucose load with 13C-glucoseto quantify glucose absorption

-180min -20min 0 min 180min

[18F]FDG injection

60 min 120 min PET scan

Exenatideor Placebo injection

240min Liver scan

Brain scan Positioning

+ Pre scan Post FDG scan

(Liver + Brain)

Primed-continuous 2H Glucose and 2H Glycerol infusion to quantify glucose production, glucose clearance and whole body lipolysis

Blood drawn for glucose, insulin, c-peptide and NEFA

(27)

The parameters of postprandial glucose metabolism were obtained from stable isotope tracer infusion, ie plasma glucose clearance (from 6,6-2H-glucose co-infusion) as an index of insulin sensitivity, endogenous glucose production and oral glucose absorption (from modelling analysis of 6,6-2H-glucose and U-13C-glucose tracer kinetics during OGTT) (Details are provided in the Appendix). Table 1 shows the characteristic of study subjects.

Table 1. Characteristic of study subjects. Data are presented as mean ± standard error mean (SEM).

Range indicates the minimum and the maximum of the parameter

2.3 The effect of Exenatide on Postprandial glucose metabolism

After Ex or PLC injection but before glucose load (ie, between -30 min and 0 min) plasma glucose concentrations were almost superimposable (mean plasma glucose:

105±8 vs. 105±3 mg/dl in Ex vs. PLC respectively; p=0.11; Figure 2A). Following the glucose load glucose concentration rose only in PLC until 120 min as a

Characteristics of participants (n=15)

Mean ±SEM Range

Age (years) 56±8 47-65

Glucose tolerance status 2 IGT- 10 IFG/IGT – 3 T2DM

BMI (Kg/m2) 29.4±0.9 24.8-37.6

Waist (cm) 103±3 91-128

FPG (mg/dl) 112±3 95-124

HbA1c (%) 5.7±0.1 5.2-6.7

Fasting Plasma Insulin (mUI/l) 9.6±1.8 3.1-18.3

Fasting Plasma Glucagon (pg/ml) 34.6±5.3 13-99

Fasting Plasma C-peptide (ng/ml) 3.1±1.0 1.4-5.2

Fasting NEFA (mEq/L) 0.77±0.27 0.53-1.51

Creatinine (mg/dl) 0.94±0.03 0.7-1.11

ALT (U/l) 33±4 17-67

AST (U/l) 28±3 18-58

Alkaline Phosphatase (U/l) 78±5 41-131

Total Cholesterol (mg/dl) 181±8 140-250

LDL cholesterol (mg/dl) 112±7 63-178

HDL cholesterol (mg/dl) 40±1 31-48

Triglycerides (mg/dl) 146±20 45-316

(28)

consequence of gut glucose absorption (Figure 2, panel A-B) and then slowly decreased reaching the fasting levels at 240 min. This triggered insulin release (Figure 3A) that increased peripheral glucose clearance (Figure 2C) and inhibited hepatic glucose production (Figure 2D). As consequence of insulin release, hepatic glucose production was significantly inhibited in both groups reaching stable values after 60 min (Figure 2D). In Ex plasma glucose remained close to the fasting levels for 240 min after the glucose load although minimal variations were observed during the first and the last part of the OGTT (slightly decreased at 0-30 min and increased at 150- 240 min compared to fasting level). EGP in Ex also decreased but to a lesser extent compared to PLC. As shown in the Figure 2B the rate of oral glucose absorption increased significantly in PLC reaching the maximum between 60 and 120 min and then decreased during the last part of the OGTT. The single Exenatide injection significantly delayed the postprandial glucose absorption (Figure 2B) two hours as compared to PLC although the rate of glucose absorption slightly increased during the last part of the OGTT. For this reason glucose concentrations in Ex remained close to the fasting levels for the four hours after the glucose load.

The Figure 2C depicts the glucose clearance in both groups. In PLC glucose clearance constantly increased after the glucose load although an initial minor reduction below the fasting level was observed during the first 30 min of the OGTT. In Ex glucose clearance constantly increased from the beginning of the OGTT reaching the highest values between 180 and 210 min.

Table 2A-B illustrates in details the glucose fluxes in fasting and postprandial states in Exenatide vs. Placebo. Fasting glucose levels were similar in both groups, however the area under the curve (AUC) was significantly lower in Ex as compared to PLC throughout the OGTT. The rate of oral glucose appearance was significantly lower in Ex vs. PLC between 0 and 120 min (p=0.0004), however it was similar between 120 and 240 min. Finally the AUC between 0 and 240 min remained significantly lower in Ex as compared to PLC. The total rate of glucose appearance which includes endogenous as well as exogenous glucose, was similar in fasting state and it was significantly reduced in Ex as compared to PLC as demonstrated by the AUC 0-120 and AUC 0-240.

(29)

Figure 2: Effect of Exenatide (close circle) vs. Placebo (open circle) on postprandial glucose metabolism. A. Plasma glucose concentrations B. Rate of oral glucose absorption C. Glucose clearance D. Endogenous glucose production. Data are presented as mean±SEM

The rate of glucose disappearance was similar at fasting state in both groups and it was significantly lower in Ex vs. PLC throughout the study. However, if the glucose disposal is expressed as glucose clearance, a significant reduction brought by Ex injection vs. PLC was observed only in the last part of OGTT (AUC 120-240:

55.2±4.8 vs. 66.6±3.1 ml/Kg*min; p=0.03 respectively) whereas it was similar during at 0-120 min and collectively at 0-240 min. The endogenous glucose production that was equal to the rate of glucose disappearance in the fasting state, was similar at fasting and it was similarly suppressed in both Ex and PLC at 0-120 after glucose load. At 120-240 endogenous glucose production was more suppressed in PLC than Ex causing an greater overall endogenous glucose suppression in PLC as compared to EX.

mg/dl

0 40 80 120 160 200 240

-30 0 30 60 90 120 150 180 210 240

mol/(Kg ! min)]

0 5 10 15 20 25

0 30 60 90 120 150 180 210 240

0,0 0,5 1,0 1,5 2,0 2,5 3,0 3,5

0 30 60 90 120 150 180 210 240

[ml/(Kg ! min)]

Time (min)

0 2 4 6 8 10 12

0 30 60 90 120 150 180 210 240

Placebo Exenatide

mol/(Kg ! min)]

Time (min)

A B

C D

Plasma Glucose Rate of Oral Glucose Appearance

Endogenous Glucose Production Glucose Clearance

(30)

Table 2A. Fasting and postprandial metabolic parameters in Exenatide and Placebo . Data are presented as mean ± standard error mean (SEM).

Parameter Placebo Exenatide p

Plasma Glucose (mg/dl)

Fasting 107±3 104±2 0.34

AUC 0-120 min 18353±1038 12607±760 0.001

AUC 120-240 min 5532±501 4859±502 0.001

AUC 0-240 min 23884±1204 17466±902 0.0007

Rate of Oral Glucose Appearance (RaOr) (µmol/ Kg Ÿ min)

Fasting Ra Or 0 0 -

AUC 0-120 1885±148 660±167 <0.001

AUC 120-240 1210±64 1256±198 0.54

AUC 0-240 3096±138 1917±277 0.001

Total Rate of Glucose Appearance (Ratot) (µmol/Kg Ÿ min)

Fasting Ra tot 10.5±0.3 10.2±0.3 0.26

AUC 0-120 2400±122 1385±152 <0.001

AUC 120-240 1487±70 1782±185 0.12

AUC 0-240 3887±143 3167±271 0.03

Rate of Glucose Disappearance (µmol/(Kg Ÿ min)

Fasting Rd 10.5±0.4 10.2±0.3 0.48

AUC 0-120 1835±108 1357±116 0.01

AUC 120-240 2281±90 1741±176 0.008

AUC 0-240 4116±163 3099±265 0.002

Riferimenti

Documenti correlati

Recent findings of sporadic autochthonous canine infections and the presence of phlebotomine vectors in some provinces of north-eastern Italy have stimulated risk assessment for

In this sample of hypertensive subjects, the level of adherence to current guidelines on HBPM was not univocal: some of the recommended practices for HBPM were followed by a

The objective of IDEC is to promote the use of geographic information (GI) by making data more easily available to public and private sector users, and to the general public. Its

L’individuazione delle varianti genetiche associate con il rischio di sviluppare tali tumori permetterebbe l’identificazione, nella popolazione generale, degli individui ad

While misuse malware detection is based on an abstract representation of the inner working of a malicious code viewed as a single entity, the propagation monitoring approach is based

Expected energy balance (upper figure) and scheduled power requested from grid. Operating cost results – i.e., not including plant installation -are shown in Fig. 17; they are

The lead in this extension of the European refugee regime was taken by the Schengen states in 1991 with the conclusion of a first model readmission agreement

Se nella storia della mendicante sono in gioco, anche se trasfigurati, alcuni elementi dell’esperienza biografica di Duras e del suo rapporto con la madre, quello che la