Type 2 diabetes is increasing at alarming rates in the U.S., in Westernized countries, and in the third world. The increasing costs in terms of human suffering as well as economics are well recognized (1). Improved understanding of the pathogenesis of diabetes should lead to better approaches to predict, forestall, or even prevent diabetes and to treat extant cases. Yet, the precise causes of this disease remain to be totally explained. There is little question that obesity per se is a primary contributor, but the causes of the so-called “obesity epidemic” are in debate (2), as are the mechanisms by which obesity itself is linked to diabetes (3,4).

For some years, our laboratory has been engaged in efforts to understand causation of type 2 diabetes in terms of insulin secretion and resistance and to attempt to account for the role of obesity. The Banting Lecture provided me with a rare opportunity to describe our research efforts done over a prolonged history in a single presentation. It was my goal to address the several areas of diabetes research we have done. At first glance these different areas may appear unrelated. But these lines of work are closely linked within an overall effort to understand diabetes. I am deeply grateful to the members of the American Diabetes Association for the great privilege of making this presentation. I hope I will be able to emphasize the wonderful opportunity that I and my colleagues have had to pursue original research in a highly deserving and important cause. One can scarcely ask for more in one's professional life.

For several decades it has been popular to pursue a “reductionist” approach to studying disease (Fig. 1). The underlying concept is that understanding of disease will result from describing events at increasingly more microscopic levels: (organism→organ→receptor→organelle→substrates→pathway→enzyme→gene).

This reductionist approach emerged naturally from the revolutionary development of molecular biology and molecular genetics. A plethora of animal models of obesity and/or hyperglycemia have been produced (58). It is possible to sample tissues from such animal models and study function in vitro. From the reductionist approach has emerged the well-described signaling pathways by which insulin binds to cells and performs multiple functions, including stimulating glucose disposal, reducing lipolysis, enhancing lipogenesis, and enhancing protein synthesis (912). Equally important have been studies of the molecular events leading to the synthesis and secretion of insulin and other important glucoregulatory peptides (glucagon, glucagon-like peptide-1 [GLP-1], glucose-dependent insulinotropic peptide [GIP], and catecholamines). But it is becoming increasingly clear that the massive amount of information provided by the molecular approach may not, in itself, allow us to explain disease. Approaches to integrate this massively increasing body of material using computer models are emerging with the moniker “systems biology” (13), but principles of systems biology have long been applied to physiological regulation. Has it been possible to apply concepts of systems biology to help us understand the causes of diabetes?

Using the systems approach, mathematical or computer models are constructed based upon known intra- and/or inter-organ signaling patterns. The models are tested by experiment, validated, or altered, and the parameters of the model, which represent real physiological processes, can be measured from experimental data. Such an approach can potentially lead to an understanding of the importance of inter-organ communication with blood glucose regulation and can tell us how a breakdown in this communication breeds disease. But how should such a model be constructed?

At this juncture it remains a daunting challenge to convert the mountain of data emerging from the new technologies to yield deep understanding of diabetes. In the course of dealing with these newer technologies, it will be important to reduce the data to a simplified comprehensive portrait amenable to clinical interpretation. It is critical to remember the dictates of William of Occam (“Occam's Razor”), “entia non sunt multiplicanda praeter necessitatem,” which translates to “entities should not be multiplied beyond necessity” (14). Albert Einstein reminds us to represent physical reality in its simplest form—but not too simple (!!).

Some years ago we attempted to apply the principle of “Occam's Razor” to describe glucose control (15). Having measured glucose and insulin levels in blood minute by minute after intravenous glucose, we selected a deceptively simple, but physiologically based, model that could totally and accurately account for the glucose and insulin kinetics measured after intravenous glucose injection (1619). This “minimal model” cannot be said to be the only representation that could be acceptable, but it has survived several decades of investigation and probing by a significant number of independent investigators, including mathematicians (2022). Every model is a hypothesis and is therefore imperfect by definition. But, given its survival, we may conclude that the minimal model is a reasonably accurate representation of glucose regulation.

The minimal model.

The minimal model is represented in Fig. 2A; Fig. 2B presents a diagrammatic representation of the model revealing the underlying assumptions—the minimal assumptions required to describe glucose and insulin kinetics after injection. The required processes included the following:

  • Under overnight fasting conditions, endogenous hepatic (and renal) glucose production is balanced by basal glucose utilization by brain (50% of total) and other tissues.

  • Prandial elevation in glycemia is renormalized in part by glucose's ability to enhance its own disposal and suppress liver production (16,2325) independent of the plasma insulin response. This “glucose effectiveness” is reduced in type 2 diabetes (26).

  • Intake of carbohydrate elicits a prompt insulin response critical to glucose renormalization. Importantly, the renormalizing effects of insulin on glucose production and uptake are not manifest immediately, but modeling required a significant temporal delay in insulin's actions. Trying to explain this temporal delay in insulin's actions taught us much about insulin action in vivo (see below).

Has the minimal model been useful? Its original publication was hardly a barnstormer in that the model was hardly cited in the literature for 5 years after appearing. What appears to have increased its significance since are the following. 1) The model was validated by several independent laboratories (2729). 2) The model, coupled with experimental work, helped increase understanding of physiology and pathophysiology. 3) The model has led to clinical tools useful in the clinic and in epidemiological and genetic studies (3032).

Emergent research.

We can identify three main research “branches” which represent offspring of the minimal model (Fig. 3):

  1. Applying the model resulted in the simultaneous measurement of insulin sensitivity and insulin response from a single clinically applicable protocol. These measurements led us to define the “disposition index,” an important index of β-cell functionality and predictor of impaired glucose tolerance and diabetes (1618,33).

  2. Studying the aforementioned insulin action delay revealed the importance of insulin movement across the capillary endothelium (and possibly capillary recruitment) in insulin action and insulin resistance.

  3. An unexpected temporal delay in insulin suppression of liver glucose production evoked the role of free fatty acids as a mediator of insulin action on the liver.

The present exposition will follow the branches of the metaphorical tree shown in Fig. 3. I will describe our journeys to follow up the clues that the minimal model has provided us to climb branches that hopefully have increased to a measurable extent our understanding of what causes type 2 diabetes and what may be done to try to predict, prevent, and eventually cure it.

After years of often contentious debate, a consensus has emerged regarding the pathogenesis of type 2 diabetes (online appendix Fig. A1 [available in an online appendix at http://dx.doi.org/10.2337/db07-9903]) (34,35). Insulin resistance is an important risk factor, necessary but not sufficient to cause the disease. There may be a genetic component to resistance (36), but many nongenetic factors can contribute, including obesity (particularly truncal obesity), lack of exercise, onset of puberty, pregnancy, and a group of conditions and therapies such as polycystic ovarian syndrome, infection, and treatment for HIV. We are now aware that a reduced compensatory response of insulin secretion in the face of insulin resistance is necessary for the development of frank type 2 diabetes. To understand the pathogenesis of diabetes, both insulin resistance and β-cell function must be assessed. One approach to assessment is the intravenous glucose tolerance test, which is described as follows.

In a healthy individual, upon glucose injection, glucose rises immediately to an early peak due to distribution in the extracellular fluid (online appendix Fig. A2). Immediately after, glucose begins to return toward basal levels due to glucose's own effect to enhance cellular uptake (due to mass action as well as glucose transporter mobilization to the membrane) (37) and glucose's ability to suppress hepatic glucose output (“autoregulation”) (23,24). We termed these two latter effects of glucose per se “glucose effectiveness” (16). The attendant hyperglycemia simultaneously provokes the β-cells to release insulin; soon the secreted insulin migrates from the plasma to the interstitial space of skeletal muscle, binding to insulin-sensitive cells and enhancing glucose disposal. The latter insulin effects accelerate the renormalization of glucose; in sensitive individuals glucose may slide below basal before renormalization between 3–4 h. Accounting for the measured time courses of glucose and insulin data with a digital computer (online appendix Fig. A2) yields estimates of the physiological coefficients of the model. Such fitting can be done even for a single individual. With Ray Boston and colleagues at the University of Pennsylvania, we have provided software, “MINMOD Millennium” (copyright R.N. Bergman) (38), that allows for this computer “fitting” process in a friendly computer environment. The resulting metabolic profile allows us to distinguish among different individuals, groups, or populations in terms of metabolic function (31,3942).

Physiologic parameters from the minimal model.

What are the physiological coefficients that emerge from fitting the minimal model to real data from an individual? The most significant ones, along with normal glucose tolerance, impaired glucose tolerance, and diabetes values, are listed in Table 1.

Applications of the minimal model.

The model has been widely applied to physiological and pathophysiologic situations, in animals and in human subjects. A cursory search of the literature reveals nearly 900 minimal model publications about widely varying subjects, ranging from arcane mathematical treatments (2022) to studies in animals (23,43,44) and humans (29,31,45). Many of these applications have employed the frequently sampled intravenous glucose tolerance test to yield insulin sensitivity and secretory response. The use of the minimal model is continuing. It yields a useful metabolic profile that can be applied to physiologic and pathophysiologic studies, comparison of therapeutic regimens, population dynamics, and population genetics.

Disposition index.

We hypothesized that in a normal individual, insulin resistance would be compensated by increased insulin secretion and that this compensation explains why normal glucose tolerance can be maintained in the face of even large changes in insulin sensitivity due to environmental or other nongenetic factors. This remarkable ability of the β-cells to precisely compensate is indicated in online appendix Fig. A3, in which a high-fat diet in the normal dog model induces severe insulin resistance and frank hyperinsulinemia but absolutely no change in either fasting or 24-h glucose excursions.

One significant outcome of minimal model studies was an ability to express in quantitative terms the compensatory relationship between insulin resistance and insulin response. Again we were inspired by an engineering concept, that of “closed loop gain.” The regulation of the blood glucose concentration represents a classic closed-loop endocrine feedback system: nutrient ingestion increases glucose, leading to a reflexive stimulation of insulin release, which counters the tendency to hyperglycemia. The ability of this closed-loop system to normalize glucose quickly can be expressed as the product of the release of the signal, plasma insulin response, and the effect of that signal, insulin sensitivity. We therefore defined the disposition index (DI) as a measure of the overall ability of the glucose regulating system to renormalize glycemia after perturbation by nutrient intake (16). The DI was defined as follows:

\[\mathrm{DI}{=}\mathrm{AIR}_{\mathrm{GLUCOSE}}{\times}S_{\mathrm{I}}\]

where AIR is the acute insulin response. There is strong evidence for increased plasma insulin in the face of insulin resistance, at least in normal individuals (46). Therefore, the DI can be interpreted as the ability of the glucose regulating system to compensate for insulin resistance by increasing plasma insulin. We hypothesized that in normal individuals, a reduction in SI (insulin resistance) would be compensated by an equivalent increase in plasma insulin response, such that the DI remains relatively constant (Fig. 4, ↓ SI compensated by ↑ in AIRGLUCOSE). The latter concept, implemented as Eq. 1, has been referred to as the “Hyperbolic Law of Glucose Tolerance” (47). Online appendix Fig. A4 shows the constancy of the DI in the face of insulin resistance.

Interpretation of DI.

Confusion regarding the relative importance of β-cell dysfunction to type 2 diabetes resulted from the fact that hyperinsulinemia often accompanies insulin resistance, even in the face of a latent β-cell defect. Thus, an individual with reduced DI may increase plasma insulin in the face of insulin resistance (Fig. 4), but the degree of compensation may be less than expected if the β-cells were healthy. Thus, while impaired glucose tolerance is often associated with increased risk for diabetes, and with β-cell dysfunction, the degree of dysfunction is represented by a reduced DI compared with healthy individuals. Therefore, it is reasonable to suggest that reduced DI is a harbinger of type 2 diabetes. In the Pima Indians, Weyer and colleagues identified the DI as the strongest predictor of conversion from normal glucose tolerance to type 2 diabetes in a longitudinally observed population over a period of 5 years (48), and a similar result emerged from the Malmo Prospective Study of pre-diabetes (49).

There is an emerging consensus that reduced β-cell function as reflected in the DI is the strongest predictor of type 2 diabetes in at-risk populations. But does this reduced islet function have a genetic component?

Genetics of DI.

Type 2 diabetes has higher concordance in monozygotic versus dizygotic twins (50), suggesting a strong genetic basis for this disease. Numerous studies have attempted to identify the specific inherited metabolic function(s) or gene(s) that might be responsible. Heritability calculations have supported indices of β-cell function as being important. The heritability of DI has been estimated to be as high as 0.73 in an African-American group (36). Poulsen and colleagues used state-of-the-art methodology to measure a variety of metabolic parameters in monozygotic and dizygotic Danish twins (50). They reported heritabilities for the DI of 0.75 in younger and identical twins but much lower values in younger fraternal sets (0.30). These data strongly support the inheritance of β-cell function and suggest that it is inheritance of reduced DI, which may contribute to increased genetic risk for type 2 diabetes.

It is not known what gene or genes may explain inheritance of DI. One clue has emerged from two disparate population studies—the FUSION Study of type 2 diabetes in Finland and the IRAS Family Study of several ethnicities in the U.S. (51,52). Both studies have identified a locus on chromosome 11 related to diabetes risk and linked with the DI. Given the heritability of DI and similarity of these loci—logarithm of odds score of 4.80 at 80 cM for linkage to the DI in the IRAS Family Study and predisposition to type 2 diabetes at a similar locus in the FUSION Study—it is tempting to hypothesize that there may be a gene for DI in this region of chromosome 11 that predisposes to diabetes. Until the putative gene for DI and/or diabetes on chromosome 11 is identified, it will remain unknown what is the true significance of this interesting region for diabetes inheritance. Very recently, several groups have reported 10 genes emerging from genome-wide association studies that increase risk for type 2 diabetes (5355). While the function of the genes are not all known, several appear to be related to β-cell function.

What is the mechanistic explanation for the “Hyperbolic Law of Glucose Tolerance”?

It would seem obvious that the increase in insulin response that attends onset of insulin resistance might be explained by the sequence of events of online appendix Fig. A5; that is, insulin resistance leads to a small reduction in glucose tolerance, mild hyperglycemia, and resultant increase in β-cell mass and/or sensitivity to glucose stimulation. The glycemic hypothesis has been strongly supported in a recent publication (56). However, there is evidence that glycemia per se may not play a primary role. In fact, in the face of obesity induced by either elevated fat-eucaloric or elevated fat-hypercaloric diets, we observed the expected increased body weight, insulin resistance, and hyperinsulinemia and relatively constant DI (online appendix Fig. A4) (43,57). Surprisingly, we did not observe any increase in fasting hyperglycemia. Not only were fasting glucose levels either constant or slightly reduced by the fat intake regimen, but there was absolutely no change in 24-h glucose values (online appendix Fig. A6) (58). Neither did we observe increases in other putative signals, such as cortisol or active GLP-1. These data indicate that in the conscious dog model, glucose cannot be the signal responsible for β-cell upregulation in the face of fat-feeding/obesity-induced insulin resistance. Neither can steroids or the gut peptide GLP-1 play this important role. As will be discussed below, there is stronger evidence that free fatty acids (FFAs) may play the central role in accounting for the increase in insulin secretory response in the face of nutrient-induced insulin resistance. However, at this moment in time, despite extensive evidence for the hyperbolic relationship between insulin sensitivity and insulin secretion (17,45,59,60), the precise signaling accounting for this fundamental physiological relationship remains unexplained! Needless to say, we continue the search for the mechanism(s) that may explain the “Hyperbolic Law of Glucose Tolerance.”

As discussed above, one requisite assumption in the minimal model is the sluggish or delayed effect of insulin to stimulate glucose disappearance. This model result recapitulated conclusions from Andres, Sherwin, and colleagues (61) from the first clinical glucose clamp studies, showing that the rapid increase in plasma insulin by injection resulted in a slow enhancement of glucose disposal, which did not reach steady state before 3 h. Online appendix Fig. A7 compares the rapid effect of insulin in vitro to stimulate glucose uptake by adipose cells, with the sluggish effect in vivo. We were interested in understanding the physiologic basis underlying this slow insulin effect, as well as understanding its possible role in insulin resistance and diabetes pathogenesis.

Once secreted, insulin follows a tortuous path to stimulate glucose uptake. Entering the bloodstream from the pancreatic veins, it must survive passage through the liver (at least 50% of secreted insulin does not) and travel from the venous system to muscle or adipose capillaries rather impermeable to large proteins. The large protein molecule must either pass through capillary endothelial cells or between them through paracellular routes to enter the interstitium, diffuse to target cells, bind, and act via the insulin pathway. The delay in insulin action might be due to slow biochemical steps after receptor binding; alternatively, it may be the delivery of insulin to the sensitive cells that is slow (62). To examine the transendothelial transport process, and to compare the time course of plasma insulin with that of interstitial insulin and glucose disposal, we exploited the use of lymph sampling to access the interstitial fluid. Our studies showed clearly that there is a strong “hand-in-glove” temporal relationship between interstitial insulin and glucose disposal but a weak relationship between plasma insulin and glucose uptake (63,64). We concluded that the delay in insulin action observed in the glucose clamp experiments and implemented in the minimal model is due to sluggish movement of insulin across capillary endothelium between plasma and interstitium; but, once at the cell surface, insulin binds and acts almost immediately (as observed in vitro, Fig. 5).

Implications of sluggish transendothelial transport of insulin.

When stimulated by rapid changes in plasma glucose, insulin release from the β-cells is biphasic. This biphasic pattern, originally described by Grodsky and colleagues (65,66), has been much studied and reflects two pools of releasable insulin within the β-cells, which have recently been imaged directly (67). It is interesting to contemplate the possible significance of biphasic release with regards to overall glucose regulation. Is it possible that biphasic secretion evolved to compensate for slow transendothelial insulin transport, which temporally limits insulin's access to skeletal muscle and adipose tissues?

Lisa Getty, when in our laboratory, injected insulin intravenously to simulate the first-phase insulin release (online appendix Fig. A8). A quick but short first-phase insulin pattern obviates the usual delay in insulin action (seen during glucose clamp experiments, for example), resulting in a rapid and profound increase in glucose disposal (68). It is tempting to hypothesize that the first phase of insulin release was naturally selected to accord the most rapid glucose disposal after nutrient ingestion and to limit the postprandial glycemia. Such a supposition can never be proved, but it does provide a potentially satisfying explanation for the evolution of a rapid first-phase insulin response.

A very important but still unanswered question relates to the putative importance of transendothelial transport in pathogenesis of insulin resistance. Clear biochemical defects have been identified in insulin-resistant animal models and humans, including receptor downregulation, phosphatidylinositol 3-kinase defects, reduction in mitochondrial function, and reduced numbers of glucose transporters. But, is there a defect in delivery of insulin to the sensitive cells?

Martin Ellmerer, in our laboratory, worked with Joyce Richey and used compartmental modeling to analyze distribution of insulin in animals rendered obese with a high-fat diet (69). Their analysis suggested that obesity limited access of insulin to skeletal muscle, accounting for about one-half of the insulin resistance caused by high-fat feeding. Very recently, to examine the possible effects of obesity on insulin kinetics more directly, Jenny Chiu worked with Richey to examine the movement of insulin within the interstitial compartment of muscle tissue. These investigators injected insulin directly into muscle and measured insulin in the interstitium (lymph) and glucose disposal by the hindlimb. We found that glucose uptake by skeletal muscle in normal animals is much more sensitive to the hormone than is suggested by systemic measurements and glucose clamps (Vmax ∼22 mg · min−1 · leg−1 and an ED50 of ∼120 pmol/l). The latter result implies that availability of insulin to the sensitive tissue is limited by either transport across endothelium or distribution of blood flow between “nutritive” versus “non-nutritive” tissues, as suggested by Clark, Barrett, and their colleagues (70,71). Even more interesting is the recent result of Chiu and colleagues (69) that infusion of the lipid emulsion Liposyn, which raises peripheral FFA levels, appears to absent insulin from skeletal muscle tissue—when Liposyn is infused, insulin appears to exit skeletal muscle rapidly, as there is virtually no increase in interstitial insulin despite direct injection of the hormone into muscle tissue. The latter result suggests rapid washout of insulin from skeletal muscle, which could suggest a great increase in permeability of capillary endothelium to the insulin molecule. It is clear that availability of insulin to the sensitive tissues (muscle and adipose) is a major factor determining insulin sensitivity. Thus, conclusions regarding insulin action from measurements of cellular and subcellular function alone cannot reproduce insulin's action in vivo, and the availability of insulin at skeletal muscle deserves further study.

The slow action of insulin, revealed by minimal modeling and glucose clamp experiments, have led to interesting and potentially important insights regarding the action of insulin and mechanisms of insulin resistance. Given that insulin resistance is an important factor, albeit not the only factor in pathogenesis of type 2 diabetes, further study of insulin access to skeletal muscle and adipose tissue is justified.

In normal individuals, several factors guarantee that the blood glucose level is normalized rapidly after meals. In dogs in particular, this normalization process is remarkable, as there is little increase in the blood glucose after glucose ingestion, despite the large increase in glucose turnover (72). Rizza and his colleagues (73) have examined the plethora of factors that play a role in this normalization, which involves suppression of endogenous glucose output (primarily by liver) and enhancement of glucose disposal (primarily by skeletal muscle). One of the most interesting factors is the gut peptide GLP-1, which has several effects to normalize glucose: slowing of gastric emptying, enhancement of the plasma insulin response, and suppression of glucagon (74). Viorica Ionut, in our laboratory, and others have exciting results pointing to an additional and potentially potent effect of GLP-1 to increase glucose effectiveness (7577), possibly via GLP-1 receptors in the porta hepatic circulation, via the central nervous system (CNS) to muscle (Fig. 6). Evidence emanating from Rossetti's laboratory that glucoreception in the CNS may control hepatic glucose output by liver directly (78) adds to the concept that the CNS is more important than has been previously thought (in glucoregulation).

Intravenous administration of glucose (as in the frequently sampled intravenous glucose tolerance test, for example) bypasses effects of gastrointestinal peptides, and yet glucose is rapidly normalized. The euglycemic glucose clamp examines effects of insulin per se on normalization and, as discussed, reveals slow activation of glucose uptake (online appendix Fig. A9). Traditional thinking presumed that suppression of glucose output, unlike activation of disposal, would be very rapid, as insulin secreted by the β-cells has immediate access to the liver via the portal vein. In addition, portohepatic vessels have large fenestrations, allowing insulin entering the liver from the portal vein to access and bind to hepatocytes almost immediately after appearance in the liver. Scintigraphy confirms very rapid concentration of injected insulin molecules in liver tissue (79). Yet, in modeling we were able to account for glucose kinetics by making the counterintuitive assumption that insulin action to suppress endogenous glucose output was sluggish, similar to the effect of stimulation of glucose disposal. This latter assumption was ultimately confirmed by David Bradley in our laboratory using euglycemic glucose clamps (80)—stimulation of glucose uptake and suppression of production were equally slow, with almost identical kinetics (i.e., similar t1/2). This latter result led to the counterintuitive hypothesis that the effect of insulin to suppress glucose output by the liver was an indirect effect of the hormone (i.e., mediated by one or more insulin-dependent extrahepatic signals).

Search for the signal mediating insulin action.

Several signals seemed possible candidates for mediating the apparent indirect insulin effect (online appendix Fig. A10). Further investigation utilizing the glucose clamp revealed a remarkable similarity between insulin suppression of glucose output and insulin suppression of lipolysis from adipose tissue (81,82). It was known that 1) the delay in insulin effect on disposal was due to slow transendothelial transport. From early studies of Scow and colleagues (83), it was also known that 2) access of insulin to adipocytes (similar to skeletal muscle access) was relatively slow. We attempted to explain these results by the “single gateway hypothesis” (84) (online appendix Fig. A11). It was posited that the slow effects of insulin on production and disposal were explained by slow transport into muscle and adipose tissue. The former slowed increasing glucose disposal, and the latter slowed suppression of FFA, hence slowing the suppression of glucose output. There is still debate regarding what precise fraction of suppression of glucose output is secondary to suppression of FFA (85). However, there is consensus that FFA suppression accounts for at least part of the reduction in gluconeogenesis and glycogenolysis that accompanies nutrient intake. What was important to our laboratory was the realization that FFAs play a more important role than previously appreciated in glucose homeostasis (7). A similar message was emanating from other laboratories (86). Adding to this understanding was epidemiologic evidence for the importance of adiposity—especially truncal adiposity—in the development of insulin resistance and as a risk factor for type 2 diabetes and cardiovascular disease. To further examine the role of FFA, and adiposity per se, we developed a canine model of obesity.

The role of FFA in pathogenesis of the metabolic syndrome.

It is a truism that obesity is increasing in the U.S. and other Western countries and is also increasing in less–well-developed nations (87). Obesity is an important risk factor for type 2 diabetes due to its close relationship with insulin resistance. Yet, the mechanistic relationships among obesity, insulin resistance, and diabetes are not totally clarified. There is strong epidemiologic evidence that central adiposity, in particular, carries risk. Experimental studies are needed to explain the causal relationships between central adipose depots and risk.

There are particular advantages in studying obesity in large animal models. Insulin resistance and obesity are characterized by communication among different tissues such as adipose and liver, and in the dog model it is possible to access the abdominal portal vein reflecting visceral fat signaling to the liver. In addition, in the canine model it is possible to study the development of obesity longitudinally (or reversal thereof) while making repetitive metabolic measurements. One such study followed the time course of insulin resistance, insulin secretion, and insulin clearance in the canine model fed a diet with elevated fat content (online appendix Fig. A3) (43). It is noted that there was a reduction in insulin sensitivity that was followed by a slow increase in insulin response, which reached a peak at 6 weeks. Complimenting increased insulin response was a reduction in first-pass clearance of insulin by liver, which accounted for at least as much hyperinsulinemia as increased insulin release (43,88). As discussed, despite these significant changes, there was no detectable change in DI. What mechanisms account for this well-orchestrated response to lipid intake without changes in glucose tolerance in the normal animal?

Nuclear magnetic resonance confirmed a significant deposition of lipid in both the central (omental) and subcutaneous fat depots during fat feeding (online appendix Fig. A12). Very recent data obtained by Morvarid Kabir in our laboratory confirms that while there are increases in adipose deposition in visceral as well as subcutaneous depots in fat-fed dogs, the visceral depot is unique. As previously reported (89,90), we noted that visceral fat tissue is more sensitive to adrenergic stimulation of lipolysis (ED50 of 1.31 × 10−7 mol/l compared with 2.77 × 10−7 mol/l for subcutaneous adipose tissue). Very interesting is the appearance in the enlarged omental fat depot of a unique population of “new” smaller adipocytes, suggesting conversion of preadipocytes to fully developed adipocytes preferentially in the visceral depot. Euglycemic clamp results revealed that most of the developing insulin resistance after feeding the eucaloric high-fat diet was due to resistance of the liver (91). We observed failure of insulin to suppress endogenous glucose output before significant changes in peripheral insulin sensitivity, measured as insulin's action to stimulate glucose disposal. Assessment of expression of genes in visceral tissues as well as liver revealed a pattern that favored increased turnover of omental fat, as well as enhanced hepatic gluconeogenesis (92)—changes that favor an effect of FFA, released from the visceral adipose depot, to cause hepatic insulin resistance (online appendix Fig. A13). However, in this model of modest obesity, we did not observe changes in expression of genes for “adipokines” including tumor necrosis factor-α, interleukin-6, leptin, or adiponectin. Therefore, central obesity induced by 6 weeks of elevated fat causes liver insulin resistance and hyperinsulinemia with a pattern supporting increased flux of FFA from visceral depot to liver but without measurable changes in expression of adipokines in visceral adipose tissue. These data support an important role for FFA release from the visceral fat in the pathogenesis of insulin resistance associated with increased truncal lipid deposition.

Access to the portal vein allowed us to measure the rate of release of FFA from the visceral depot. To our surprise, we found that visceral FFA release was oscillatory, with a burst observed about every 9–11 min (online appendix Fig. A14) (93,94). That the release was lipolytic was supported by a similar and coordinated pattern of glycerol release. Pulsatile visceral lipolysis was totally suppressed by bupranolol, a high-affinity antagonist to β-3 adrenergic receptors, located in the visceral fat depot of the dog. Recent data obtained by Isabel Hsu in our laboratory have supported the concept that oscillations imposed in the portal vein of the conscious animal have profound effects to enhance insulin resistance of the liver.

Our data has led us to support the original concept of Landsberg (95) that the sympathetic nervous system plays a central role in the development of insulin resistance, secondary to deposition of visceral fat. We suggest that fat feeding results in deposition of visceral fat. Adrenergic signals from the CNS induce phasic lipolysis of the visceral depot, which bathes the liver with FFA at regular intervals (online appendix Fig. A15). This FFA barrage induces insulin resistance in the liver by upregulating gluconeogenic enzymes. Further studies are being performed to address the source of the central adrenergic signals, how they may depend upon metabolic state, and what role the signals may play in other insulin-resistant conditions.

Hyperinsulinemic compensation for insulin resistance: putative role of nocturnal FFAs.

As discussed, it remains unexplained why in the face of insulin resistance, plasma insulin response increases and insulin clearance decreases in a well-coordinated manner such that, in normal animals, glucose intolerance does not invariably result in the insulin-resistant state. How do the β-cells of the pancreatic islets “know” the appropriate enhancement of insulin release? How does the insulin-degrading mechanism of the liver establish the appropriate downregulation of first-pass liver insulin clearance? As discussed above, careful investigation of glycemia failed to explain insulinemic upregulation. Is there a possibility that FFAs are involved in this highly regulated inter-organ orchestration?

We considered a group of blood-borne signals that could be put forth as mediating hyperinsulinemia in the face of fat-diet–induced insulin resistance (58). Among these candidates were GLP-1, known to cause pancreatic islet-cell proliferation in rodents, and growth hormone and cortisol, each of which can result in increased insulin. After 6 weeks of a high-fat, hypercaloric diet, resulting in significant weight gain in the dog model, we observed zero evidence for any increase in 24-h glycemia (online appendix Fig. A6). Even more surprising, we measured a paradoxical reduction in 24-h active GLP-1 levels and no increases in either 24-h patterns of cortisol or growth hormone. On the contrary, and to our surprise, we observed a striking and powerful increase in plasma FFA levels in the middle of the night. Comparing the fat-fed animal with the lean model, FFAs are increased beginning in the late afternoon and continue to elevate with a maximal elevation at 3 a.m. What is the significance of the nighttime FFA increase, and does it relate to our sympathetic hypothesis of the causation of the metabolic syndrome?

We are attempting to understand the significance of the nighttime FFA increase. We would propose that it represents lipolysis, stimulated by the sympathetic nervous system. If so, the increasing FFA may be oscillatory, and adrenergic blocking agents should suppress them specifically. While it is indeed tedious to do rapid sampling to establish oscillatory lipolysis, students in our laboratory are doing these studies, led by Stella Kim, Isabel Hsu, Jenny Chiu, and Karyn Catalano. We plan to test whether the lipolysis can be blocked with adrenergic blocking agents, and if so, whether this will reverse the putative effects of elevated nocturnal FFA on insulin secretion and action.

Hypothesis integrating the role of FFAs in the metabolic syndrome.

Our working hypothesis for the role of the visceral fat depot in the development of the metabolic syndrome (Fig. 7) is as follows: We suggest that the adrenergic nervous system plays a central role in the development of the syndrome. We suggest that fat feeding increases the signaling from the brain to the adipose depot and that this signal is particularly evident at night, maximized at 3:00 a.m. We further suggest that the sympathetic signal results in bursts of FFA via the portal vein, bathing the liver in lipid and rendering it insulin resistant. The resistance, at least in the short term, does not appear to involve changes in adipokines, although it is certainly likely that adipokines are important over the longer term. The resistance is associated with increase of expression of enzymes that favor lipolysis and enhancement of gluconeogenesis. It is possible that interfering with this pattern, for example by suppressing lipolysis during the nighttime, may break this pattern and reverse the insulin resistance of the metabolic state.

I have been a very lucky person. With my wonderful colleagues (see acknowledgments), we have been able to begin with an arcane idea—development of a minimal model of glucose kinetics—and to follow this “acorn” where it led us, hopefully to some greater understanding of the pathogenesis of diabetes and associated metabolic diseases. Trees will not flourish in the wilderness without rich soil and fertilizer. First is my wife Ronni and my kids, Doug and Beth, their spouses, Nancy and Guy, and my grandchildren, Emily, Jessica, Samantha, and Hannah.

I am a lousy tree climber. I received very necessary “legs up” from many wonderful people and institutions: My early mentors included Oscar Hechter, I. Arthur Mirsky, and John Urquhart. Dan Porte, Jr., Mladen Vranic, and Larry Phillips were kind enough to appreciate our early research efforts and enabled us to test our ideas in the clinic. My wonderful peers Claudio Cobelli and Diane Finegood were there at the tree's conception, birth, and early life, and Marilyn Ader, Richard Watanabe, Tom Buchanan, Joyce Richey, and Andrea Dunaif were present during the Cambrian adolescence and adulthood. I am tremendously grateful to the wonderful friends/colleagues who chose to work with me on our journey, listed by necessity in the acknowledgments. Finally, I thank the American Diabetes Association, the National Institutes of Health, and the University of Southern California, all of whom translated their confidence in our work to the tangible resources without which our work not have been performed.

Richard Watanabe, Diane Finegood, Giovanni Pacini, Claudio Cobelli, Jay Taborsky, Steve Mittelman, Martin Ellmerer, Dave Bradley, Aage Volund, Joyce Richey, Gianna Toffolo, Kerstin Rebrin, Katrin Hücking, Idit Liberty, Vivi Ionut, Mori Kabir, Chester Ni, Pat Crane, Marianthe Hamilton-Wessler, Lisa Getty, Dave Cohen, Melvin Dea, Jang Youn, Andrea Hevener, Gregg Van Citters, Casey Donovan, Garry Steil, Y. Ziya Ider, Ray Boston, Charlie Bowden, Renee Poulin, Lise Kjems, Stella Kim, Karyn Catalano, Isabel Hsu, Jenny Chiu, Darko Stefanovski, Maya Lottati, Nicki Harrison, Orison Woolcott, Dan Zheng, Elza Demirchyan, Rita Thomas, Ed Zuniga, Erlinda Kirkman, IRAS Investigators, FUSION family, Anne Sumner, Steve Kahn, Michael Goran, Mike Schwartz, and Jim Best.

I am deeply grateful to Dr. Marilyn Ader for helping me with this manuscript and for many years of fruitful collaboration and camaraderie.

FIG. 1.

Reductionist vs. holistic approaches to study carbohydrate homeostasis. Elegant molecular and genetic approaches have elucidated cellular pathways of hormone action, including insulin (left) (ref. 12). Alternative approach (right) is to test the system in vivo and compare predictions with computer model to hone hypotheses of hormone/substrate interactions.

FIG. 1.

Reductionist vs. holistic approaches to study carbohydrate homeostasis. Elegant molecular and genetic approaches have elucidated cellular pathways of hormone action, including insulin (left) (ref. 12). Alternative approach (right) is to test the system in vivo and compare predictions with computer model to hone hypotheses of hormone/substrate interactions.

Close modal
FIG. 2.

The minimal model of glucose kinetics. A: Insulin in plasma [I(t)] crosses the endothelial barrier to enter interstitial fluid (I’). Production of glucose (G) by liver and glucose disposal in periphery are controlled by “remote” (i.e., interstitial) insulin concentration. Later studies confirmed that slow effect of insulin on liver is due to indirect effect of the hormone to suppress glucose production (via free fatty acids; “single gateway hypothesis,” ref. 83). Adapted from ref. 16. B: Diagrammatic representation of the minimal model. Fasting blood glucose level is determined by a balance between liver production and insulin-independent (brain) and insulin-dependent (skeletal muscle) mechanisms. Carbohydrate intake elicits insulin signal; insulin slowly crosses capillary endothelium to enhance glucose disposal to muscle and suppress lipolysis in adipose, which in turn contributes to reduction in liver glucose output.

FIG. 2.

The minimal model of glucose kinetics. A: Insulin in plasma [I(t)] crosses the endothelial barrier to enter interstitial fluid (I’). Production of glucose (G) by liver and glucose disposal in periphery are controlled by “remote” (i.e., interstitial) insulin concentration. Later studies confirmed that slow effect of insulin on liver is due to indirect effect of the hormone to suppress glucose production (via free fatty acids; “single gateway hypothesis,” ref. 83). Adapted from ref. 16. B: Diagrammatic representation of the minimal model. Fasting blood glucose level is determined by a balance between liver production and insulin-independent (brain) and insulin-dependent (skeletal muscle) mechanisms. Carbohydrate intake elicits insulin signal; insulin slowly crosses capillary endothelium to enhance glucose disposal to muscle and suppress lipolysis in adipose, which in turn contributes to reduction in liver glucose output.

Close modal
FIG. 3.

History of research in the Bergman Laboratory. Minimal model assumptions were tested experimentally, leading to the DI (Disposition Index) concept, the role of transendothelial insulin transport, and role of FFAs in the “Metabolic Xyndrome.”

FIG. 3.

History of research in the Bergman Laboratory. Minimal model assumptions were tested experimentally, leading to the DI (Disposition Index) concept, the role of transendothelial insulin transport, and role of FFAs in the “Metabolic Xyndrome.”

Close modal
FIG. 4.

The Hyperbolic Law of Glucose Tolerance. Insulin resistance (abscissa) is normally compensated by a reflexive hyperinsulinemia, to maintain glucose tolerance. The law states that in normal individuals, product of insulin sensitivity and secretion is constant—the DI. In at-risk individuals, DI may be reduced (lower hyperbolic curve). In such individuals, hyperinsulinemia for a given reduction in insulin sensitivity may be inadequate (lower DI). Lower DI may portent eventual type 2 diabetes.

FIG. 4.

The Hyperbolic Law of Glucose Tolerance. Insulin resistance (abscissa) is normally compensated by a reflexive hyperinsulinemia, to maintain glucose tolerance. The law states that in normal individuals, product of insulin sensitivity and secretion is constant—the DI. In at-risk individuals, DI may be reduced (lower hyperbolic curve). In such individuals, hyperinsulinemia for a given reduction in insulin sensitivity may be inadequate (lower DI). Lower DI may portent eventual type 2 diabetes.

Close modal
FIG. 5.

Proposed events in insulin stimulation of glucose disposal. Insulin crosses from capillary to interstitial compartment slowly; delay may be due to intercellular or transcellular transport. Once in interstitium, insulin binds to receptors and rapidly stimulates glucose uptake.

FIG. 5.

Proposed events in insulin stimulation of glucose disposal. Insulin crosses from capillary to interstitial compartment slowly; delay may be due to intercellular or transcellular transport. Once in interstitium, insulin binds to receptors and rapidly stimulates glucose uptake.

Close modal
FIG. 6.

Possible indirect effect of gastrointestinal peptide GLP-1. GLP-1 degrades rapidly in plasma, suggesting that it may have a site of action close to its site of secretion (L-cells of gut). Putative receptors in the portal vein (or liver) are sensitive to GLP-1 and glucose; interaction with receptors may send afferent signal to brain with efferent signals to enhance glucose utilization (e.g., increased insulin secretion, reduced glucagon, and/or increase in insulin-independent glucose uptake). Extant data show enhanced glucose disposal secondary to portal glucose/GLP-1 administration (ref. 73).

FIG. 6.

Possible indirect effect of gastrointestinal peptide GLP-1. GLP-1 degrades rapidly in plasma, suggesting that it may have a site of action close to its site of secretion (L-cells of gut). Putative receptors in the portal vein (or liver) are sensitive to GLP-1 and glucose; interaction with receptors may send afferent signal to brain with efferent signals to enhance glucose utilization (e.g., increased insulin secretion, reduced glucagon, and/or increase in insulin-independent glucose uptake). Extant data show enhanced glucose disposal secondary to portal glucose/GLP-1 administration (ref. 73).

Close modal
FIG. 7.

Events resulting in the “Metabolic Xyndrome.” CNS and other factors result in lipolysis from visceral and subcutaneous fat depots. Visceral fat depots in particular cause flux of FFA to liver and hepatic insulin resistance; more stored fat causes insulin resistance of skeletal muscle. In our experiments, compensation for insulin resistance (hyperinsulinemia) occurs in the absence of elevated glucose. FFAs themselves may be the compensating signal that causes the hyperinsulinemia by stimulating insulin release, increasing β-cell size or number, and reducing liver clearance of insulin. Hyperinsulinemia is adequate to compensate for insulin resistance, except in the face of a latent β-cell defect, wherein hyperglycemia ensues and type 2 diabetes results.

FIG. 7.

Events resulting in the “Metabolic Xyndrome.” CNS and other factors result in lipolysis from visceral and subcutaneous fat depots. Visceral fat depots in particular cause flux of FFA to liver and hepatic insulin resistance; more stored fat causes insulin resistance of skeletal muscle. In our experiments, compensation for insulin resistance (hyperinsulinemia) occurs in the absence of elevated glucose. FFAs themselves may be the compensating signal that causes the hyperinsulinemia by stimulating insulin release, increasing β-cell size or number, and reducing liver clearance of insulin. Hyperinsulinemia is adequate to compensate for insulin resistance, except in the face of a latent β-cell defect, wherein hyperglycemia ensues and type 2 diabetes results.

Close modal
TABLE 1

Physiologic parameters emerging from the minimal model (data from refs. 39,42)

Parameters emerging from fitting the minimal modelNormalIGTType 2 diabetes
SG Glucose effectiveness (min−1): This parameter reflects the effect of glucose per se to enhance glucose disposal and suppress glucose output, at basal insulin concentration. 0.021 ± 0.008 0.016 ± 0.007 0.015 ± 0.011 
AIRGLUCOSE Insulin response (μU/ml × min): This can be limited to first-phase release (0–10 min above basal) but can also yield second-phase response, depending upon which injection protocol is used 59.6 ± 54.8 42.4 ± 42.6 6.7 ± 18.5 
SI Insulin sensitivity (× 10−4 min−1 per μU/ml): Probably the most important parameter is the insulin sensitivity index (SI). This index reflects to ability of insulin in blood to augment glucose's ability to activate its own disappearance and to suppress glucose output. The SI is quantitative. It is normalized to the size of the glucose distribution volume, making SI values comparable among individuals, between genders, between ethnic groups, and even between species. 2.62 ± 2.21 1.27 ± 1.20 0.57 ± 0.82 
DI Disposition index: The DI is the product of SI and AIRGLUCOSE. It represents the ability of the b-cells to compensate for changes in insulin sensitivity (see text). 1,249 ± 1,559 430 ± 594 30 ± 95 
Parameters emerging from fitting the minimal modelNormalIGTType 2 diabetes
SG Glucose effectiveness (min−1): This parameter reflects the effect of glucose per se to enhance glucose disposal and suppress glucose output, at basal insulin concentration. 0.021 ± 0.008 0.016 ± 0.007 0.015 ± 0.011 
AIRGLUCOSE Insulin response (μU/ml × min): This can be limited to first-phase release (0–10 min above basal) but can also yield second-phase response, depending upon which injection protocol is used 59.6 ± 54.8 42.4 ± 42.6 6.7 ± 18.5 
SI Insulin sensitivity (× 10−4 min−1 per μU/ml): Probably the most important parameter is the insulin sensitivity index (SI). This index reflects to ability of insulin in blood to augment glucose's ability to activate its own disappearance and to suppress glucose output. The SI is quantitative. It is normalized to the size of the glucose distribution volume, making SI values comparable among individuals, between genders, between ethnic groups, and even between species. 2.62 ± 2.21 1.27 ± 1.20 0.57 ± 0.82 
DI Disposition index: The DI is the product of SI and AIRGLUCOSE. It represents the ability of the b-cells to compensate for changes in insulin sensitivity (see text). 1,249 ± 1,559 430 ± 594 30 ± 95 

IGT, impaired glucose tolerance.

Additional information for this article can be found in an online appendix at http://dx.doi.org/10.2337/db07-9903.

R.B.'s work has been supported by the National Institutes of Health (DK 29867 and DK 27619) and the American Diabetes Association (Mentor Award).

1
Zimmet P: The burden of type 2 diabetes: are we doing enough?
Diabetes Res
29
:
6S
9–6S18,
2003
2
Keith SW, Redden DT, Katzmarzyk PT, Boggiano MM, Hanlon EC, Benca RM, Ruden D, Pietrobelli A, Barger JL, Fontaine KR, Wang C, Aronne LJ, Wright SM, Baskin M, Dhurandhar NV, Lijoi MC, Grilo CM, DeLuca M, Westfall AO, Allison DB: Putative contributors to the secular increase in obesity: exploring the roads less traveled.
Int J Obes
30
:
1585
–1594,
2006
3
Boden G, Laakso M: Lipids and glucose in type 2 diabetes: what is the cause and effect?
Diabetes Care
27
:
2253
–2259,
2004
4
Trujillo ME, Scherer PE: Adipose tissue-derived factors: impact on health and disease.
Endocr Rev
27
:
762
–778,
2006
5
Unger RH: The hyperleptinemia of obesity-regulator of caloric surpluses.
Cell
117
:
145
–146,
2004
6
Shafrir E, Ziv E, Mosthaf L: Nutritionally induced insulin resistance and receptor defect leading to beta-cell failure in animal models.
Ann N Y Acad Sci
892
:
223
–246,
1999
7
Bergman RN, van Citters GW, Mittelman SD, Dea MK, Hamilton-Wessler M, Kim SP, Ellmerer M: Central role of the adipocyte in the metabolic syndrome.
J Investig Med
49
:
119
–126,
2001
8
Kahn CR: Knockout mice challenge our concepts of glucose homeostasis and the pathogenesis of diabetes.
Exp Diabesity Res
4
:
169
–182,
2003
9
Musi N, Goodyear LJ: Insulin resistance and improvements in signal transduction.
Endocrine
29
:
73
–80,
2006
10
Wang X, Proud CG: The mTOR pathway in the control of protein synthesis.
Physiology
21
:
362
–369,
2006
11
Kusunoki J, Kanatani A, Moller DE: Modulation of fatty acid metabolism as a potential approach to the treatment of obesity and the metabolic syndrome.
Endocrine
29
:
91
–100,
2006
12
Giri L, Mutalik VK, Venkatesh KV: A steady state analysis indicates that negative feedback regulation of PTP1B by Akt elicits bistability in insulin-stimulated GLUT4 translocation.
Theor Biol Med Model
1
:
2
,
2004
13
Palsson BO:
Systems Biology: Properties of Reconstructed Networks.
New York, Cambridge University Press,
2006
14
Thorburn WM: Occam's Razor. In
Mind.
p.
287
–288,
1915
15
Bergman RN: Integrated control of hepatic glucose metabolism.
Fed Proc
36
:
265
–270,
1977
16
Bergman RN, Ider YZ, Bowden CR, Cobelli C: Quantitative estimation of insulin sensitivity.
Am J Physiol
236
:
E667
–E677,
1979
17
Bergman RN, Phillips LS, Cobelli C: Physiologic evaluation of factors controlling glucose tolerance in man: measurement of insulin sensitivity and B-cell glucose sensitivity from the response to intravenous glucose.
J Clin Invest
68
:
1456
–1467,
1981
18
Bergman RN: The minimal model of glucose regulation: a biography. In
Mathematical Modeling in Nutrition and Health.
Novotny J, Green M, Boston R, Eds. London, Kluwer Academic/Plenum Publishers,
2001
19
Ader M: Physiologic principles underlying glucose effectiveness. In
The Minimal Model Approach and Determinants of Glucose Tolerance.
Pennington Center Nutrition Series, vol. 7. Bergman RN, Lovejoy JC, Eds. Baton Rouge, LA, Louisiana State University Press,
1997
, p.
159
–186
20
Godsland IF, Agbaje OF, Hovorka R: Evaluation of nonlinear regression approaches to estimation of insulin sensitivity by the minimal model with reference to Bayesian hierarchical analysis.
Am J Physiol
291
:
E167
–E174,
2006
21
Zheng Y, Zhao M: Modified minimal model sing a single-step fitting process for the intravenous glucose tolerance test in type 2 diabetes and healthy humans.
Comput Methods Programs Biomed
79
:
73
–79,
2005
22
Araujo-Vilar D, Rega-Liste CA, Garcia-Estevez DA, Sarmiento-Escalona F, Mosquera-Tallon V, Cabezas-Cerrato J: Minimal model of glucose metabolism: modified equations and its application in the study of insulin sensitivity in obese subjects.
Diabetes Res Clin Pract
39
:
129
–141,
1998
23
Ader M, Ni T-C, Bergman RN: Glucose effectiveness assessed under dynamic and steady state conditions: comparability of uptake versus production components.
J Clin Invest
99
:
1187
–1199,
1997
24
Glinsmann WH, Hern EP, Lynch A: Intrinsic regulation of glucose output by rat liver.
Am J Physiol
216
:
698
–703,
1969
25
Cherrington AD, Williams PE, Harris MS: Relationship between the plasma glucose level and glucose uptake in the conscious dog.
Metabolism
27
:
787
–791,
1978
26
Basu A, Caumo A, Bettini F, Gelisio A, Alzaid A, Cobelli C, Rizza RA: Impaired basal glucose effectiveness in NIDDM: contribution of defects in glucose disappearance and production, measured using an optimized minimal model independent protocol.
Diabetes
46
:
421
–432,
1997
27
Korytkowski MT, Berga SL, Horwitz MJ: Comparison of the minimal model and the hyperglycemic clamp for measuring insulin sensitivity and acute insulin response to glucose.
Metabolism
44
:
1121
–1125,
1995
28
Bergman RN, Prager R, Volund A, Olefsky JM: Equivalence of the insulin sensitivity index in man derived by the minimal model method and the euglycemic glucose clamp.
J Clin Invest
79
:
790
–800,
1987
29
Dunaif A, Finegood DT: β-Cell dysfunction independent of obesity and glucose intolerance in the polycystic ovary syndrome.
J Clin Endocrinol Metab
81
:
942
–947,
1996
30
Wagenknecht LE, Meyer EJ, Rewers M, Haffner S, Selby J, Borok GM, Henkin L, Howard G, Savage PJ, Saad MF, Bergman RN, Hamman R: The Insulin Resistance Atherosclerosis Study (IRAS): objectives, design, and recruitment results.
Ann Epidemiol
5
:
464
–472,
1996
31
Flanagan DE, Vaile JC, Petley GW, Phillips DI, Godsland IF, Owens P, Moore VM, Cockington RA, Robinson JS: Gender differences in the relationship between leptin, insulin resistance and the autonomic nervous system.
Regul Pept
140
:
37
–42,
2007
32
Watanabe RM, Ghosh S, Langefeld CD, Valle T, Hauser ER, Magnuson VL, Mohlke KL, Silander K, Ally DS, Chines P, Blaschak-Harvan J, Douglas JA, Duren WL, Epstein MP, Fingerlin TE, Kaleta HS, Lange EM, Li C, McEachin RC, Stringham HM, Trager E, White PP, Balow J Jr, Birznieks G, Chang J, Eldridge W, Erdos MR, Karanjawala ZE, Knapp JI, Kudelko K, Martin C, Morales-Mena A, Musick A, Musick T, Pfahl C, Porter R, Rayman JB, Rha D, Segal L, Shapiro S, Sharaf R, Shurtleff B, So A, Tannenbaum J, Te C, Tovar J, Unni A, Welch C, Whiten R, Witt A, Kohtamaki K, Ehnholm C, Eriksson J, Toivanen L, Vidgren G, Nylund SJ, Tuomilehto-Wolf E, Ross EH, Demirchyan E, Hagopian WA, Buchanan TA, Tuomilehto J, Collins FS, Boenke M: The Finland-United States investigation of non-insulin-dependent diabetes mellitus genetics (FUSION) study. II. An autosomal genome scan for diabetes-related quantitative-trait loci.
Am J Hum Genet
67
:
1186
–1200,
2000
33
Bergman RN, Ader M, Huecking K, van Citters GW: Accurate assessment of β-cell function: the hyperbolic correction.
Diabetes
51 (Suppl. 1)
:
S212
–S220,
2001
34
Kahn CR: Insulin action, diabetogenes, and the cause of type II diabetes (Banting Lecture).
Diabetes
43
:
1066
–1084,
1994
35
Porte D Jr: Mechanisms for hyperglycemia in the metabolic syndrome: the key role of beta-cell dysfunction.
Ann N Y Acad Sci
892
:
73
–83,
1999
36
Rich SS, Bowden DW, Haffner SM, Norris JM, Saad MF, Mitchell BD, Rotter JI, Langefeld CD, Wagenknecht LE, Bergman RN, Insulin Resistance Atherosclerosis Study Family Study: Identification of quantitative trait loci for glucose homeostasis: the Insulin Resistance Atherosclerosis Study (IRAS) Family Study.
Diabetes
53
:
1866
–1875,
2004
37
Cushman SW, Wardzala LJ: Potential mechanism of insulin action of glucose transport in the isolated rat adipose cell: apparent translocation of intracellular transport systems to the plasma membrane.
J Biol Chem
255
:
4758
–4762,
1980
38
Boston RC, Stefanovski D, Moate PJ, Sumner AE, Watanabe RM, Bergman RN: MINMOD Millennium: a computer program to calculate glucose effectiveness and insulin sensitivity from the frequently sampled intravenous glucose tolerance test.
Diabetes Technol Therapeut
5
:
1003
–1015,
2003
39
Haffner SM, Howard G, Mayer E, Bergman RN, Savage PJ, Rewers M, Mykkanen L, Karter AJ, Hamman R, Saad MF: Insulin sensitivity and acute insulin response in African-Americans, non-Hispanic whites, and Hispanics with NIDDM.
Diabetes
46
:
63
–69,
1997
40
Clausen JO, Hein HO, Suadicani P, Winther K, Gyntelberg F, Pedersen O: Lewis phenotypes and the insulin resistance syndrome in young healthy white men and women.
Am J Hypertens
8
:
1060
–1066,
1995
41
Snitker S, Watanabe RM, Ani I, Xiang A, Marroquin A, Ochoa C, Goico J, Shuldiner AR, Buchanan TA: Troglitazone in Prevention of Diabetes (TRIPOD) study: Changes in insulin sensitivity in response to troglitazone do not differ between subjects with and without the common, functional Pro12Ala peroxisome proliferator-activated receptor-gamma2 gene variant: results from the Troglitazone in Prevention of Diabetes (TRIPOD) study.
Diabetes Care
27
:
1365
–1368,
2004
42
Haffner SM, D'Agostino RD, Saad MF, Rewers M, Mykkanen L, Selby J, Howard G, Savage PJ, Hamman R, Wagenknecht LE, Bergman RN: Increased insulin resistance and insulin secretion in non-diabetic African-Americans and Hispanics compared to non-Hispanic whites: the Insulin Resistance and Atherosclerosis Study.
Diabetes
45
:
742
–748,
1996
43
Mittelman SD, van Citters GW, Kim SP, Davis DA, Dea MK, Bergman RN: The longitudinal compensation for fat induced insulin resistance includes reduced insulin clearance and enhanced B-cell response.
Diabetes
49
:
2116
–2125,
2000
44
Pacini G, Thomaseth K, Ahren B: Contribution to glucose tolerance of insulin-independent vs. insulin-dependent mechanisms in mice.
Am J Physiol
281
:
E693
–E703,
2001
45
Xiang A, Peters RK, Trigo E, Kjos SL, Lee WP, Buchanan TA: Multiple metabolic defects during late pregnancy in women at high risk for type 2 diabetes mellitus.
Diabetes
48
:
848
–854,
1999
46
Modan M, Halkin H, Almog S, Lusky A, Eshkol A, Shefi M, Shitrit A, Fuchs Z: Hyperinsulinemia: a link between hypertension, obesity, and glucose intolerance.
J Clin Invest
75
:
809
–817,
1985
47
Stumvoll M, Tataranni PA, Bogardus C: The hyperbolic law: a 25-year perspective.
Diabetologia
48
:
207
–209,
2005
48
Weyer C, Hanson K, Bogardus C, Pratley RE: Long-term changes in insulin action and insulin secretion associated with gain, loss, regain and maintenance of body weight.
Diabetologia
43
:
36
–46,
2000
49
Lyssenko V, Almgren P, Anevski D, Perfekt R, Lahti K, Nissen M, Isomaa B, Forsen B, Homstrom N, Saloranta C, Taskinen MR, Groop L, Tuomi T, for the Botnia Study Group: Predictors of and longitudinal changes in insulin sensitivity and secretion preceding onset of type 2 diabetes.
Diabetes
54
:
166
–174,
2005
50
Poulsen P, Levin K, Petersen I, Christensen K, Beck-Nielsen H, Vaag A: Heritability of insulin secretion, peripheral and hepatic insulin action, and intracellular glucose partitioning in young and old Danish twins.
Diabetes
54
:
275
–283,
2005
51
Silander K, Scott LJ, Valle TT, Mohlke KL, Stringham HM, Wiles KR, Duren WL, Doheny KF, Pugh EW, Chines P, Narisu N, White PP, Fingerlin TE, Jackson AU, Li C, Ghosh S, Magnuson VL, Colby K, Erdos MR, Hill JE, Hollstein P, Humphreys KM, Kasad RA, Lambert J, Lazaridis KN, Lin G, Morales-Mena A, Patzkowski K, Pfahl C, Porter R, Rha D, Segal L, Suh YD, Tovar J, Unni A, Welch C, Douglas JA, Epstein MP, Hauser ER, Hagopian W, Buchanan TA, Watanabe RM, Bergman RN, Tuomilehto J, Collins FS, Boehnke M, Finland-United States Investigation of NIDDM Genetics (FUSION): A large set of Finnish affected sibling pair families with type 2 diabetes suggests susceptibility loci on chromosomes 6, 11, and 14.
Diabetes
53
:
821
–829,
2004
52
Palmer JP, Langefeld CD, Campbell JK, Williams AH, Saad M, Norris JM, Haffner SM, Rotter JI, Wagenknecht LE, Bergman RN, Rich SS, Bowden DW: Genetic mapping of disposition index and acute insulin response loci on chromosome 11q: the Insulin Resistance Atherosclerosis Study (IRAS) Family Study.
Diabetes
55
:
911
–918,
2006
53
Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI, Chen H, Roix JJ, Kathiresan S, Hirschhorn JN, Daly MJ, Hughes TE, Groop L, Altshuler D, Almgren P, Florez JC, Meyer J, Ardlie K, Bengtsson K, Isomaa B, Lettre G, Lindblad U, Lyon HN, Melander O, Newton-Cheh C, Nilsson P, Orho-Melander M, Rastam L, Speliotes EK, Taskinen MR, Tuomi T, Guiducci C, Berglund A, Carlson J, Gianniny L, Hackett R, Hall L, Holmkvist J, Laurila E, Sjogren M, Sterner M, Surti A, Svensson M, Tewhey R, Blumenstiel B, Parkin M, Defelice M, Barry R, Bordeur W, Camarata J, Chia N, Vava M, Gibbons J, Handsaker B, Healy C, Nguyen K, Gates C, Sougnez C, Gage D, Nizzari M, Gabriel SB, Chirn GW, Ma Q, Parikh H, Richardson D, Ricke D, Purcell S: Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels.
Science.
26 April 2007 [Epub ahead of print]
54
Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos MR, Stringham HM, Chines PS, Jackson AU, Prokunina-Olsson L, Ding CJ, Swift AJ, Narisu N, Hu T, Pruim R, Xiao R, Li XY, Conneely KN, Riebow NL, Sprau AG, Tong M, White PP, Hetrick KN, Barnhart MW, Bark CW, Goldstein JL, Watkins L, Xiang F, Saramies J, Buchanan TA, Watanabe RM, Valle TT, Kinnunen L, Abecasis GR, Pugh EW, Doheny KF, Bergman RN, Tuomilehto J, Collins FS, Boehnke M: A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants.
Science.
26 April 2007 [Epub ahead of print]
55
Zeggini E, Weedon MN, Lindgren CM, Frayling TM, Elliott KS, Lango H, Timpson NJ, Perry JR, Rayner NW, Freathy RM, Barrett JC, Shields B, Morris AP, Ellard S, Groves CJ, Harries LW, Marchini JL, Owen KR, Knight B, Cardon LR, Walker M, Hitman GA, Morris AD, Doney AS, McCarthy MI, Hattersly AT: Replication of genome-wide association signals in U.K. samples reveals risk loci for type 2 diabetes.
Science.
26 April 2007 [Epub ahead of print]
56
Weir GC, Bonner-Weir S: A dominant role for glucose in beta cell compensation of insulin resistance.
J Clin Invest
117
:
81
–83,
2007
57
Stefanovski D, Woolcott O, Lottati M, Zheng D, Harrison LN, Ionut V, Kim SP, Hsu I, Bergman RN, Richey JM: Maintenance of glucose tolerance in diet-induced insulin resistant dogs (Abstract).
FASEB J
21
:
737.2
,
2007
58
Kim SP, Catalano KJ, Hsu IR, Chui JD, Richey JM, Bergman RN: Nocturnal free fatty acids are uniquely elevated in the longitudinal development of diet-induced insulin resistance and hyperinsulinemia.
Am J Physiol
30 January 2007 [Epub ahead of print]
59
Pacini G: The hyperbolic equilibrium between insulin sensitivity and secretion.
Nutr Metab Cardiovasc Dis
16
:
522
–527,
2006
60
McManus RM, Cunningham I, Watson A, Harker L, Finegood DT: Beta-cell function and visceral fat in lactating women with a history of gestational diabetes.
Metabolism
50
:
715
–719,
2001
61
Insel PA, Liljenquist JE, Tobin JD, Sherwin RS, Watkins P, Andres R, Berman M: Insulin control of glucose metabolism in man.
J Clin Invest
55
:
1057
–1066,
1975
62
Bergman RN, Bradley DC, Ader M: On insulin action in vivo: the single gateway hypothesis.
Adv Exp Med Biol
334
:
181
–198,
1993
63
Yang YJ, Hope ID, Ader M, Bergman RN: Insulin transport across capillaries is rate limiting for insulin action in dogs.
J Clin Invest
84
:
1620
–1628,
1989
64
Yang YJ, Hope ID, Ader M, Poulin RA, Bergman RN: Dose response relationship between lymph insulin and glucose uptake reveals enhanced insulin sensitivity of peripheral tissues.
Diabetes
41
:
241
–253,
1992
65
Bergman RN, Urquhart J: The pilot gland approach to the study of insulin secretory dynamics.
Rec Progr Horm Res
27
:
583
–605,
1971
66
Grodsky GM: A threshold distribution hypothesis for packet storage of insulin and its mathematical modeling.
J Clin Invest
51
:
2047
–2059,
1972
67
Michael DJ, Ritzel RA, Haataja L, Chow RH: Pancreatic β-cells secrete insulin in fast- and slow-release forms.
Diabetes
55
:
600
–607,
2006
68
Getty L, Hamilton-Wessler M, Ader M, Dea MK, Bergman RN: Biphasic insulin secretion during intravenous glucose tolerance test promotes optimal interstitial insulin profile.
Diabetes
47
:
1941
–1947,
1998
69
Ellmerer M, Hamilton-Wessler M, Kim SP, Huecking K, Kirkman E, Chiu J, Richey J, Bergman RN: Reduced access to insulin-sensitive tissues in dogs with obesity secondary to increased fat intake.
Diabetes
55
:
1769
–1775,
2006
70
Vincent MA, Clerk LH, Rattigan S, Clark MG, Barrett EJ: Active role for the vasculature in the delivery of insulin to skeletal muscle.
Clin Exp Pharmacol Physiol
32
:
302
–307,
2005
71
Clerk LH, Vincent MA, Lindner JR, Clark MG, Rattigan S, Barrett EJ: The vasodilatory actions of insulin on resistance and terminal arterioles and their impact on muscle glucose uptake.
Diabete Metab Res Rev
20
:
3
–12,
2004
72
Strack TR, Poussier P, Marliss EB, Albisser AM: Glucose turnover after a mixed meal in dogs: glucoregulation without change in arterial glycemia.
Am J Physiol
266
:
R889
–R895,
1994
73
Vella A, Camilleri M, Rizza RA: The gastrointestinal tract and glucose tolerance.
Curr Opin Clin Nutr Metab Care
7
:
479
–484,
2004
74
Drucker DJ, Nauck MA: The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes.
Lancet
368
:
1696
–1705,
2006
75
Burcelin R: The incretins: a link between nutrients and well-being.
Br J Nutr
93 (Suppl. 1)
:
S147
–S156,
2005
76
Ionut V, Hucking K, Liberty IF, Bergman RN: Synergistic effect of portal glucose and glucagon-like peptide-1 to lower systemic glucose and stimulate counter-regulatory hormones.
Diabetologia
48
:
967
–975,
2005
77
Ionut V, Liberty IF, Hucking K, Lottati M, Stefanovski D, Zheng D, Bergman RN: Exogenously imposed postprandial-like rises in systemic glucose and GLP-1 do not produce an incretin effect, suggesting an indirect mechanism of GLP-1 action.
Am J Physiol
291
:
E779
–E785,
2006
78
Lam TKT, Gutierrez-Juarez R, Pocai A, Rossetti L: Regulation of blood glucose by hypothalamic pyruvate metabolism.
Science
309
:
943
–947,
2005
79
Jensen I, Kruse V, Larsen UD: Scintigraphic studies in rats: kinetics of insulin analogues covering wide range of receptor affinities.
Diabetes
40
:
628
–632,
1991
80
Bradley DC, Poulin RA, Bergman RN: Dynamics of hepatic and peripheral insulin effects suggest common rate-limiting step in vivo.
Diabetes
42
:
296
–306,
1992
81
Rebrin K, Steil GM, Getty L, Bergman RN: Free fatty acid as a link in the regulation of hepatic glucose output by peripheral insulin.
Diabetes
44
:
1038
–1045,
1995
82
Rebrin K, Steil GM, Mittelman S, Bergman RN: Causal linkage between insulin regulation of lipolysis and liver glucose output.
J Clin Invest
98
:
741
–749,
1996
83
Chernick SS, Gardiner RJ, Scow RO: Restricted passage of insulin across capillary endothelium in perfused rat adipose tissue.
Am J Physiol
253
:
E475
–E480,
1987
84
Bergman RN: New concepts in extracellular signaling for insulin action: the single gateway hypothesis.
Rec Progr Horm Res
42
:
359
–385,
1997
85
Chu CA, Sherck SM, Igawa K, Sindelar DK, Neal DW, Emshwiller M, Cherrington AD: Effects of free fatty acids on hepatic glycogenolysis and gluconeogenesis in conscious dogs.
Am J Physiol
282
:
E402
–E411,
2002
86
Boden G, Shulman GI: Free fatty acids in obesity and type 2 diabetes: defining their role in the development of insulin resistance and beta-cell dysfunction.
Eur J Clin Invest
32 (Suppl. 3)
:
14
–23,
2002
87
Yoon KH, Lee JH, Kim JW, Cho JH, Choi YH, Ko SH, Zimmet P, Son HY: Epidemic obesity and type 2 diabetes in Asia.
Lancet
368
:
1681
–1688,
2006
88
Kim SP, Ellmerer M, Kirkman EL, Bergman RN: Beta-cell “rest” accompanies reduced first-pass hepatic insulin extraction in the insulin resistant, fat-fed canine model.
Am J Physiol
6 Febrary 2007 [Epub ahead of print]
89
Arner P: Differences in lipolysis between human subcutaneous and omental adipose tissues.
Ann Med
27
:
435
–438,
1995
90
Tchernof A, Belanger C, Morisset AS, Richard C, Mailloux J, Laberge P, Dupont P: Regional differences in adipose tissue metabolism in women: minor effect of obesity and body fat composition.
Diabetes
55
:
1353
–1360,
2006
91
Kim SP, Ellmerer M, van Citters GW, Bergman RN: Primacy of hepatic insulin resistance in the development of the metabolic syndrome induced by an isocaloric, moderate fat diet in the dog.
Diabetes
52
:
2453
–2460,
2003
92
Kabir M, Catalano KC, Ananthnarayan S, Kim SP, van Citters GW, Dea MK, Bergman RN: Molecular evidence supporting the portal theory: a causative link between visceral adiposity and hepatic insulin resistance.
Am J Physiol
288
:
E454
–E461,
2005
93
Getty L, Panteleon AE, Mittelman SD, Dea MK, Bergman RN: Rapid oscillations in omental lipolysis are independent of changing insulin levels in vivo.
J Clin Invest
106
:
421
–430,
2000
94
Hucking K, Hamilton-Wessler M, Ellmerer M, Bergman RN: Burst-like control of lipolysis by the sympathetic nervous system in vivo.
J Clin Invest
111
:
247
–264,
2003
95
Landsberg L: Role of the sympathetic adrenal system in the pathogenesis of the insulin resistance syndrome.
Ann N Y Acad Sci
892
:
84
–90,
1999

Supplementary data