Abstract
Historically, pancreatic islet beta cells have been viewed as principal regulators of glycemia, with type 2 diabetes (T2D) resulting when insulin secretion fails to compensate for peripheral tissue insulin resistance. However, glycemia is also regulated by insulin-independent mechanisms that are dysregulated in T2D. Based on evidence supporting its role both in adaptive coupling of insulin secretion to changes in insulin sensitivity and in the regulation of insulin-independent glucose disposal, the central nervous system (CNS) has emerged as a fundamental player in glucose homeostasis. Here, we review and expand upon an integrative model wherein the CNS, together with the islet, establishes and maintains the defended level of glycemia. We discuss the implications of this model for understanding both normal glucose homeostasis and T2D pathogenesis and highlight centrally targeted therapeutic approaches with the potential to restore normoglycemia to patients with T2D.
Keywords: glucose homeostasis, diabetes, central nervous system, autonomic nervous system, insulin-independent glucose disposal, glucose effectiveness, therapeutics
1. INTRODUCTION
The circulating level of glucose is determined by the balance between rates of appearance (via synthesis, release from storage, or ingestion) and disappearance (via utilization, uptake, or excretion). This balance is influenced by the ongoing energetic demand of every organ that requires adequate glucose provision to ensure reproductive fitness and survival across ever-changing environmental conditions. While many regulatory checkpoints have evolved to safeguard glucose storage and allocation, the central nervous system (CNS) has the unique capacity to compare the energetic needs of the body with the predicted effects of possible behaviors. By integrating environmental cues of food availability with interoceptive signals of stored and circulating fuel, the CNS works cooperatively with pancreatic islets to adjust glucose production, storage, and utilization to establish the biologically defended level of glycemia (BDLG) (1) (Figure 1). In type 2 diabetes (T2D), the BDLG is pathologically elevated such that plasma glucose is maintained at a higher level (2, 3), and accumulating evidence implicates both brain and islet dysfunction in this process. Failure to address these pathological processes increasing the BDLG is a fundamental shortcoming of current antidiabetic medications premised on augmenting insulin action—plasma glucose lowering will be short-lived if competing homeostatic mechanisms that increase glycemia in T2D are not also targeted. This shortcoming is borne out in the disappointing results of current T2D pharmacotherapy: Despite the introduction of more than 40 new US Food and Drug Administration (FDA)-approved drugs over the past decade, the fraction of T2D patients falling short of glycemic targets remains at approximately 50% (4).
Figure 1.
Mechanisms governing the biologically defended level of glycemia (BDLG). The BDLG is determined by the balance between rates of glucose appearance into and disappearance from the circulation, and imbalance in these rates contributes to the elevated BDLG in type 2 diabetes (T2D). In health, acute deviations from the BDLG are counteracted by both insulin-dependent and insulin-independent mechanisms that restore blood glucose levels into the normal range. Responses to rising blood glucose levels (dashed arrows) include increased glucose-stimulated insulin secretion (GSIS) by the pancreatic beta cell, which, together with the ability of glucose to independently facilitate its own disposal [termed glucose effectiveness (GE)], increases glucose uptake by peripheral tissues and inhibits hepatic glucose production (HGP). Conversely, a fall in blood glucose levels triggers adaptive neuroendocrine and autonomic counterregulatory responses (CRRs, solid arrows) that collectively increase glucose appearance into the circulation and decrease its removal. These responses include increased secretion of glucagon, cortisol, and epinephrine (which stimulate HGP), while insulin secretion is inhibited to prevent a further fall in blood glucose. In T2D, the lower boundary of the BDLG is increased (as reflected by a higher glycemic threshold for inducing CRRs), and the same is true of the upper boundary of the BDLG, as evidenced by diminished rates of glucose disappearance (owing to reduced GSIS, insulin resistance, and reduced GE) and failure to suppress HGP. The net outcome is a persistently elevated BDLG in T2D. Figure adapted from images created with BioRender.com.
Current approaches to T2D treatment are based on an islet-centered view of glucose homeostasis that has dominated the field since insulin was discovered over 100 years ago. According to this view, T2D results when pancreatic beta cells fail to compensate for progressive insulin resistance (5), usually associated with obesity (6). While we agree with this model of T2D pathogenesis, we note that it is incomplete without considering the role of insulin-independent mechanisms of glucose disposal that are also impaired in T2D (see Section 3) (7, 8). Insulin-independent glucose disposal contributes roughly 50% to intravenous glucose tolerance (9), accounts for up to 80% of glucose disposal in the basal state (i.e., in the absence of exogenous nutrients) (9, 10), and is among the first deficits detected in individuals who develop T2D (11, 12). The CNS can potently modulate insulin-independent glucose disposal (13) and synchronously adjust insulin secretion and sensitivity during physiological challenges to glucose homeostasis (14). As T2D is associated with anomalies affecting brain areas and neurocircuits that regulate these processes, a role for the CNS in T2D pathogenesis warrants careful scrutiny.
In this review, we summarize contributions made by both pancreatic islets and insulin-independent mechanisms to overall glucose disposal and T2D pathogenesis, discuss the role of the CNS in these processes, and evaluate the potential of CNS-targeted strategies for future treatment of this common disease.
2. THE ISLET-CENTERED MODEL OF GLUCOSE HOMEOSTASIS
January 12, 2022, commemorates the centennial anniversary of the first administration of insulin to a human (15). The discovery that a crude pancreatic extract could resolve hyperglycemia in type 1 diabetes (T1D), a previously fatal condition, constitutes a landmark achievement in medicine. Insulin’s discovery triggered a paradigm shift in the field of diabetes research: It sparked the field of islet biology, led to the identification of insulin-producing beta cells, and heralded a new islet-centered model of glucose homeostasis. Though headlines at the time of insulin’s discovery proclaimed the cure to diabetes was imminent, 100 years later, this prediction remains to be fully realized. In this section, we review this islet-centered model of glucose homeostasis and present evidence that invokes a prominent, collaborative role for CNS mechanisms in both normal and abnormal glucose homeostasis.
The islet-centered model suggests that stability of the BDLG depends largely upon the beta cell’s capacity to adjust insulin secretion to changes in circulating glucose levels. When blood glucose levels rise after a meal, for instance, glucose-stimulated insulin secretion (GSIS) returns blood glucose to its preprandial level by both suppressing further glucose production by the liver and facilitating glucose uptake into peripheral tissues. Impairment of either GSIS or insulin action reduces glucose disposal and incompletely suppresses hepatic glucose production (HGP), yielding hyperglycemia. In health, normal fluctuations in insulin sensitivity occurring daily (e.g., sleep or exercise) and seasonally (e.g., puberty, pregnancy, or aging) are counteracted by adaptive changes in insulin secretion (6). In obesity, associated with persistently reduced insulin sensitivity, a compensatory increase of beta cell function can still preserve glucose homeostasis. In genetically susceptible individuals, failure of beta cells to meet this heightened demand for insulin secretion is proposed to drive the BDLG out of the normal range, producing T2D (6) (Figure 1).
Most therapies consequently aim to support or supplant beta cell function by augmenting insulin secretion or sensitivity or by supplementing exogenous insulin itself. Designed to transiently lower blood glucose levels, these approaches can prevent or delay T2D complications, but with the accompanying risk of hypoglycemia and side effect of weight gain. More importantly, strategies designed to simply lower the blood glucose level do not address the underlying pathological processes that elevate the BDLG and therefore fail to induce sustained diabetes remission.
A core issue not addressed by current therapeutic modalities is whether beta cell dysfunction in T2D is a primary disorder or a secondary consequence of severe metabolic derangements that accompany the disease. Indeed, it is possible that both are true. The following discussion addresses this fundamental question.
2.1. Cell-Autonomous and Non-Cell-Autonomous Mechanisms Governing Insulin Secretion
The pancreatic beta cell is a highly specialized cell type that senses and responds to glucose in a cell-autonomous manner, and impaired GSIS is a cardinal feature of T2D (16). Beta cell function is also strongly influenced by factors external to the islet, including input from the CNS. Beta cell regulation by the brain involves both indirect (e.g., via neuroendocrine hormone secretion) and direct [via the autonomic nervous system (ANS) and spinal sensory innervation (Figure 2)] mechanisms (for reviews, see 17-19). Despite slight differences between humans and rodents in the anatomy of islet innervation (20, 21), autonomic stimulation affects islet secretion similarly across species. Sympathetic nervous system (SNS) outflow to the beta cell suppresses both glucose-stimulated and basal insulin secretion, while parasympathetic nervous system (PNS) outflow enhances GSIS (17, 22). By contrast, both SNS and PNS outflow to the islet increase glucagon secretion (17). Evidence from multiple species (23, 24) indicates that the PNS is critical for meal-associated GSIS, as muscarinic cholinergic receptor blockade with atropine suppresses postprandial insulin secretion. Interestingly, in humans, atropine suppresses GSIS following an oral, but not intravenous, glucose challenge, suggesting an interaction between the PNS and gut-derived incretin peptides that augment insulin secretion, such as glucagon-like peptide 1 (GLP1) (Supplemental Appendix 1) (24). Because vagal (PNS) fibers extensively innervate the gastrointestinal tract and other organs in addition to the pancreas (for detailed reviews, see 25, 26), atropine’s effects on islet function may involve gut-vagal-brain hormone sensing (27, 28) or gastric motility, in addition to direct effects on beta cells.
Figure 2.
Anatomy of glycemic control: central and peripheral mechanisms. (①) After a meal, nutrients digested by the GI tract are delivered into the blood via the hepatic portal vein and circulated throughout the body. Postprandial elevations in blood glucose activate GSIS. Effects of postprandial GSIS (purple circles) include suppression of HGP (directly via activation of hepatic insulin receptors and indirectly via inhibition of glucagon release from pancreatic alpha cells) and stimulation of glucose uptake by insulin-sensitive tissues, including skeletal muscle and adipose tissue. Nutrient ingestion also stimulates endocrine cells lining the GI tract to release the incretin hormones, GLP1 and GIP. Incretins augment insulin secretion (yellow circle) and, via binding their cognate receptors expressed by sensory afferents innervating the GI tract, convey information to the brain about the size and composition of ingested nutrients (for reviews, see 26, 47, 48). (②) Spinal afferents (dark green) have their cell bodies within the dorsal root ganglia, which express molecules such as the ion channel TRPV1 (activated by noxious heat) that processes nociceptive signals conveyed to the brain via the spinothalamic tract. Spinal afferents also can exert effects by releasing inflammatory peptides such as SP and CGRP. Vagal afferents (light green) also express SP/CGRP and can sense the local microenvironment through receptors that include the serotonin receptor (5HTR3) in the pancreas and incretin receptors in the GI tract. Cell bodies of vagal sensory neurons are contained within the nodose ganglion and convey sensory information to the CNS via projections to the NTS. In addition to surveilling peripheral tissue function, the CNS can influence peripheral glucose effector function via neuroendocrine systems, including the HPA axis (③) and the ANS. In response to stressful stimuli, activation of the hypothalamic-pituitary-adrenal axis causes the adrenal cortex to secrete cortisol, which stimulates glucose production by the liver. (④) The SNS and PNS branches of the ANS innervate tissues throughout the body to influence glycemia. Stimulation of the SNS (blue circles in ①) suppresses GSIS and increases glucagon release from the islets while stimulating increased HGP from the liver via releasing the neurotransmitter NE, which binds to and activates adrenergic receptors on pancreatic islet cells and hepatocytes. Stimulation of the PNS (orange circles in ①) increases both insulin and glucagon secretion via binding of the neurotransmitter ACh to muscarinic cholinergic receptors. In the postprandial state, the responses to PNS activation promote glucose uptake and storage. Abbreviations: ACh, acetylcholine; ACTH, adrenocorticotropic hormone; ANS, autonomic nervous system; CGRP, calcitonin gene-related peptide; CNS, central nervous system; GI, gastrointestinal; GIP, glucose-stimulated insulinotropic polypeptide; GLP1, glucagon-like peptide 1; GSIS, glucose-stimulated insulin secretion; HGP, hepatic glucose production; HPA, hypothalamic-pituitary-adrenal; NE, norepinephrine; NPY, neuropeptide Y; NTS, nucleus of the solitary tract; PACAP, pituitary adenylate cyclase-activating polypeptide; PNS, parasympathetic nervous system; SNS, sympathetic nervous system; SP, substance P; TRPV1, transient receptor potential vanilloid 1. Figure adapted from images created with BioRender.com.
Islets are also innervated by spinal (dorsal root ganglion) sensory neurons. Deletion of TRPV1-expressing spinal afferents (Figure 2) suppresses islet inflammation and preserves GSIS and beta cell mass in several diabetic models (29-31). Like vagal afferents, however, spinal afferents innervate multiple tissues, so indirect effects of TRPV1-mediated ablation on GSIS are possible. Target-specific approaches (e.g., via retrograde viral tagging, as in References 32 and 33) are needed to clarify whether sensory neurons directly modulate islet activity and/or sense the islet secretory state. These studies will also help interpret recent work showing increases in nerve density per islet, number of innervated islets, and size of intrapancreatic ganglia in humans with obesity and T2D (21, 34).
While identifying the cause of perturbed islet innervation and its functional consequences for glycemic control requires further study, these observations raise the possibility that aberrant neural input contributes or predisposes to beta cell dysfunction in T2D. Among individuals who develop T2D, beta cells may have an underlying genetic susceptibility to the deleterious impact of influences extrinsic to the islet itself. This view is compatible with clear evidence that beta cell dysfunction can result from sustained hyperglycemia (35) [referred to as glucotoxicity (36)] and obesity-associated metabolic impairment (e.g., hyperlipidemia, systemic inflammation) (37, 38). For these reasons, it remains uncertain the extent to which defective GSIS in T2D involves a primary beta cell defect or instead is secondary to one or more beta cell–extrinsic factors.
2.2. Insights from Genome-Wide Association Studies
A primary beta cell defect in T2D pathogenesis is also inferred from genome-wide association studies (GWAS) data revealing over 400 gene variants associated with T2D risk, many of which are expressed in beta cells (39). However, these variants collectively account for only about 20% of T2D risk (39), and neither the causal role in T2D played by GWAS-identified transcripts (39) nor the significance of their expression in beta cells versus other tissues is known. In fact, many T2D-associated gene variants expressed in the islet are also expressed in the CNS, including genes encoding the glucose phosphorylating enzyme glucokinase (GCK) (40) and the ATP-sensitive potassium (KATP) channel subunits SUR1 (ABCC8) and Kir6.2 (KCNJ11) (41). The proteins encoded by these genes play important roles in cellular glucose sensing in both the CNS and beta cells (see Section 4). These observations collectively support a model in which individuals that go on to develop T2D have a genetic predisposition to beta cell dysfunction that is aggravated by external challenges to the beta cell, yielding progressive impairment of insulin secretion characteristic of T2D.
2.3. Islet Transplantation
In humans with T1D, implanting healthy donor islets is sufficient to restore normoglycemia (42). Thus, the islet can function independently of CNS input to regulate glucose homeostasis in this pathological setting. However, removing islets from neural control creates a nonphysiological state that does not inform how glucose homeostasis normally works in natural conditions or how it goes awry in T2D. In light of growing evidence that the brain, like the islet, can powerfully impact the BDLG via both islet-dependent and islet-independent mechanisms (detailed below), we infer that the BDLG is determined by cooperative interactions between brain and islet. This interaction is exemplified by the adaptive coupling of insulin secretion to increased insulin sensitivity during cold exposure, in which the SNS plays a pivotal role.
2.4. Dynamic Coupling of Insulin Secretion and Insulin Sensitivity During Cold Exposure: A Paradigm for Understanding the Brain’s Role in Glucose Homeostasis
To maintain core body temperature in the cold, thermogenic tissues rapidly increase glucose utilization to fuel heat production, including thermogenesis by brown adipose tissue and shivering by skeletal muscle. Enhanced glucose utilization is facilitated in part by increased insulin sensitivity in these tissues (43), which is essential to preserve core temperature in the cold (44). To prevent blood glucose levels from falling during the resultant increase of glucose utilization, an adaptive decrease in insulin secretion occurs with such rapidity and precision that blood glucose levels remain stable throughout the transition to a cold environment. The effect of cold to suppress insulin secretion therefore cannot be attributed to a change in glucose stimulation of beta cells, since glucose levels do not change. These observations illustrate how adaptive coupling of insulin secretion to insulin sensitivity preserves normoglycemia when the energetic demands of tissues change.
Based on the assumption that defective feedback between insulin-sensitive tissues and beta cells underlies glucose intolerance in T2D (6), intensive effort has been devoted to identifying this coupling signal, with candidates that include free fatty acids, intrinsic beta cell glucose metabolism, and sensitivity to incretins (6, 45). While the role played by these factors remains uncertain, a clear role for the SNS has been demonstrated in adaptive coupling during cold exposure. Increased SNS outflow not only drives heat production by thermogenic tissues (44, 46, 47) but also suppresses insulin secretion (47). Blocking SNS outflow (with α-adrenergic receptor antagonists) prevents both the cold-induced suppression of insulin secretion and the increase in insulin sensitivity (14). Collectively, the observations above highlight the difficulty in distinguishing the intertwined roles played by the islet and the CNS in glucose homeostasis—cooperative interactions between them are essential.
2.5. Is Impaired Beta Cell Function a Cause or Consequence of T2D?
This question may have no correct answer and is perhaps not the most scientifically relevant or clinically meaningful question. We view T2D as a heterogenous disorder involving multiple peripheral and central defects, where interactions between them create a vicious cycle that progressively impairs both insulin-dependent and insulin-independent glucose disposal. Impaired beta cell function—whether primary or secondary—plays a key role, but the gradual increase in the BDLG implicates defective CNS control mechanisms as well.
3. INSULIN-INDEPENDENT MECHANISMS OF GLUCOSE DISPOSAL
Our understanding of insulin-independent glucose disposal has been limited by two hurdles: (a) the perception that it is a passive, unregulated process with limited relevance to glucose homeostasis, and (b) it is hard to measure. The former has been abandoned in light of growing evidence that this process can be rapidly and potently activated by the CNS (see Section 4), and the latter was addressed by the development and validation of Bergman’s minimal model of glucose kinetics (Supplemental Figure 1), which allowed this variable to be measured. In the original conception of that model—derived by mathematical modeling of frequently sampled intravenous glucose tolerance (FSIGT) test data—glucose effectiveness (designated Sg in minimal model analyses) was a parameter that measured “the quantitative enhancement of glucose disappearance due to an increase in the plasma glucose concentration” that was not attributable to the action of insulin (48, p. E673). That is, Sg was a measure of the effect of glucose to promote its own disposal down a concentration gradient after an intravenous load (49). However, studies applying the minimal model approach in humans revealed that Sg can change during acute experimental intervention (50-52) and in disease (53-55). These seminal findings showed that this underappreciated variable (and the various insulin-independent mechanisms that are represented by Sg) contributes significantly to individual variance in glucose tolerance and T2D risk. A notable limitation of Sg (glucose effectiveness as a measure of insulin-independent glucose disposal) is that since it is based on the response to a glucose load (i.e., in FSIGT tests), it offers limited insight into insulin-independent glucose disposal in the basal state, where it is quantitatively more important than the insulin-mediated contribution (9, 52).
3.1. Reduced Insulin-Independent Glucose Disposal Predicts T2D Risk
To enable minimal modeling of glucose kinetics in T2D patients, Welch et al. (55) developed an FSIGT protocol using exogenous insulin to compensate for diminished GSIS characteristic of T2D (minimal modeling of FSIGT data requires a dynamic interaction between changing plasma insulin and glucose levels). In addition to the expected reduction in insulin sensitivity, Sg was reduced by approximately 40% in T2D patients. Two subsequent studies (11, 56) implicate this defect as causal in T2D pathogenesis.
In the first study (11), 18 lean and obese nondiabetic subjects underwent FSIGT testing and were subdivided into two groups based on quantified glucose tolerance (Kg), with body weight tending to segregate, though not exclusively, with Kg. Minimal modeling of the FSIGT data revealed that subjects with lower Kg had not only the expected lower disposition index (DI, a composite measure of insulin-dependent glucose disposal; (Supplemental Figure 1) but also twofold lower Sg. Glucose intolerance in nondiabetic subjects, therefore, appears to involve reductions in the handling of glucose by both insulin-dependent and insulin-independent mechanisms.
To investigate whether either of these impairments is predictive of future T2D, a prospective cohort study was conducted in 155 normoglycemic offspring of parents who both had T2D (56). FSIGT and minimal model analysis were performed at baseline, and subjects were followed for up to 25 years. In the 25 subjects that developed T2D, reductions of both Sg and insulin sensitivity (Si) were manifest at least 10 years prior to developing T2D. Remarkably, none of the subjects with Sg and Si above the median developed T2D, whereas those with Sg and Si values below the median had a 76% cumulative incidence of T2D over 20 years, with reduced Sg imparting the greatest relative risk—15 times greater risk of developing T2D comparing the lowest and highest quintiles of Sg. These studies provide compelling evidence linking impaired insulin-independent glucose disposal to T2D pathogenesis. It was only years later that the potent capacity of the brain to regulate insulin-independent glucose disposal came to light (Section 4).
3.2. Reconciling Insulin-Dependent and Insulin-Independent Mechanisms of T2D Pathogenesis
The hypothesis that T2D arises when beta cells fail to compensate for insulin resistance (57) has extensive support, including cross-sectional and longitudinal studies implying that an early beta cell defect develops as a consequence of insulin resistance (for detailed reviews, see 6, 58). Whether assessing groups at high risk for developing dysglycemia (16, 59, 60) or populations that are phenotypically not commonly obese (61, 62), these studies consistently suggest that an early impairment of beta cell function—measured by insulin secretion and/or DI (Supplemental Figure 1)—is predictive of T2D.
Unexpectedly, recent evidence suggests that insulin hypersecretion, rather than beta cell dysfunction, identifies otherwise normal individuals at risk for T2D. In a recent prospective cohort study conducted by Ferrannini and colleagues (63), 1,168 healthy adults with normal glucose tolerance were stratified according to their level of insulin secretion, and advanced statistical methods were used to ensure virtually identical insulin sensitivity between strata. After a period of 3 years, subjects with higher insulin secretion were more likely to progress to impaired glucose tolerance or T2D than those with lower insulin secretion. This result, which is consistent with other human (64-68) and animal studies (69-73), suggests that primary insulin hypersecretion is a triggering event in T2D pathogenesis (74), possibly by precipitating subsequent insulin resistance (69, 70, 75-79).
A paradoxical finding in the study by Ferrannini and colleagues (63) is that insulin hypersecretors had worse glucose tolerance despite having equivalent insulin sensitivity. Notably, insulin hypersecretion, normal insulin sensitivity, and worsened glucose tolerance simply cannot coexist unless the other key determinant of glucose tolerance—insulin-independent glucose lowering—is reduced. This interpretation is consistent with evidence cited above linking reduced insulin-independent glucose disposal to T2D pathogenesis (11, 56). Although additional work is required to explain the reduced glucose tolerance in this cohort (63), reduced hepatic glucose uptake [regulated largely by insulin-independent mechanisms (81, 82) discussed below] may contribute, as this process is known to be impaired in patients with T2D (83, 84).
3.3. Insulin-Independent Glucose Disposal and Hepatic Glucose Flux
As the recipient of portal venous blood from the gastrointestinal tract, the liver is uniquely placed to monitor glucose appearance after a meal (1). Following meal ingestion, the glucose level entering the liver through the portal system is much greater than that of the arterial circulation. First described in dogs by Cherrington and colleagues (81, 85), this negative arterial-portal glucose gradient (termed the portal signal) is specific to the postprandial setting and produces a robust, insulin-independent increase in net hepatic glucose uptake (NHGU), in effect directing the liver to switch rapidly from glucose output in the fasted state to glucose uptake following a meal. Cherrington and colleagues (86, 87) provide evidence that (a) in the fasted state, basal sympathetic tone to the liver blocks NHGU, and the portal signal suppresses sympathetic activity to produce a sharp rise in NHGU; (b) within hepatocytes, this response involves increased activity of glucokinase and glycogen synthase, which increase glucose uptake and glycogen synthesis, respectively (88); (c) chronic high-fat diet (HFD)/high-fructose diet feeding abolishes portal signal induction of NHGU together with reductions in glucokinase content and activity, glycogen synthase activity, and glucose tolerance (89, 90); and (d) sympathetic denervation along the common hepatic artery in the HFD/high-fructose diet-fed dog partially restores NHGU, independent of insulin, thereby improving glucose tolerance (91).
This work thus suggests that overnutrition heightens sympathetic tone to the liver and blocks the ability of the portal signal to drive the normal meal-induced increase of NHGU. This interpretation is supported by earlier work in dogs in which long-term epinephrine infusion, to mimic effects of chronic sympathetic activation, induces a nearly two-thirds drop in Sg, with no difference in Si or insulin secretion (92). How might these findings impact our understanding of T2D pathogenesis?
3.4. Evidence for Autonomic Dysfunction in T2D Pathogenesis
Several studies suggest that autonomic dysfunction and excessive sympathetic tone are early precursors of T2D (93). Prospective cohort studies have shown that both baseline norepinephrine levels and sympathetic reactivity predict future T2D risk (94-96). Two large prospective cohort studies in ethnically distinct patient populations recently demonstrated that reduced heart rate variability, a biomarker of dysautonomia with sympathetic-parasympathetic imbalance (favoring sympathetic tone), was associated with significantly increased T2D risk (97, 98). Cross-sectional studies show increased resting muscle sympathetic nerve activity in T2D patients compared to patients with impaired glucose tolerance (99) or obesity alone (100). Other human (101-104) and animal (105) studies suggest that diet-induced obesity (DIO) is associated with chronically elevated sympathetic nerve activity.
Thus, in addition to the role played by beta cell dysfunction, both reduced Sg and dysautonomia (characterized by increased sympathetic tone) are consistently predictive of increased risk for T2D. Since insulin-independent mechanisms predominate in the control of basal glycemia, explaining up to 80% of overall glucose disposal (9, 52), and since reduced Sg is an expected consequence of increased sympathetic tone to the liver, we propose that an individual’s propensity to develop T2D may depend on primary or acquired defects in these centrally controlled parameters.
4. AN INTEGRATIVE MODEL FOR CNS CONTROL OF GLUCOSE HOMEOSTASIS AND T2D PATHOGENESIS
Many brain regions have the capacity to influence glycemia, including several nuclei within the mediobasal hypothalamus [such as the ventromedial nucleus (VMN), arcuate nucleus (ARC), paraventricular nucleus, and lateral hypothalamus] and the hindbrain (including the nucleus of the solitary tract) (Figures 2 and 3). Neurons in these regions are anatomically well positioned to influence rates of glucose production and disposal by polysynaptic relays involving both limbs of the ANS, as well as via neuroendocrine secretion (26, 106) (Figure 2). Many neuronal subsets in these brain regions also express the molecular machinery necessary for intrinsic glucose sensing (see the sidebar titled Cellular Mechanisms of Glucose Sensing) and receptors for hormones and nutrients, raising the possibility that their effects on glucose homeostasis are regulated by circulating cues of energetic status, in addition to afferent neural input.
Figure 3.
Hypothalamic neurocircuits in glycemic control. The hypothalamus is a forebrain structure situated ventral to the thalamus and dorsal to the pituitary gland and median eminence. It is an important component of the hypophyseal portal system. The mediobasal hypothalamus exhibits reciprocal connectivity with hindbrain structures, including the nucleus of the solitary tract (NTS), as well as with neighboring hypothalamic nuclei (arrows indicate connectivity in hypothalamus inset). The median eminence is a circumventricular organ with fenestrated capillaries that increase vascular permeability (indicated by dashed red lines in circular insets) and that receives dense innervation from hypothalamic neurons [including from parvocellular neurons of the paraventricular nucleus (PVN)], which release hypophysiotropic hormones (e.g., corticotropin-releasing factor) into the pituitary portal system, thereby directing pituitary secretions. Heightened vascular permeability of the median eminence exposes neurons in the adjacent arcuate nucleus (ARC) to higher levels of circulating nutrients and hormones than seen by brain regions located behind the blood-brain barrier (bold red line), such as the ventromedial nucleus (VMN), PVN, and lateral hypothalamic area (LHA). The ARC is also adjacent to the third ventricle (3V), permitting access to factors circulating in the cerebrospinal fluid, and receives afferent neuronal input from multiple brain regions conveying information about energetic demands, environmental cues of time and food availability, and cue-reward associations. These dynamic and varied inputs are integrated by pro-opiomelanocortin (POMC)- and agouti-related peptide (AgRP)-expressing neurons in the ARC that are regulated in a reciprocal manner and drive opposing effects on glycemia and energy balance, in part, by engaging melanocortin receptor signaling in downstream neurons. Hypothalamic network activity is also shaped by interactions with neighboring glia, including astrocytes and microglia. In addition to providing structural support for neurons and their projections, astrocytes and microglia play critical roles in neurovascular coupling, neurotransmitter uptake, synaptic pruning, immune surveillance, and inflammatory signaling (for a detailed review, see 116). In the setting of overnutrition, these cell types engage in a hypothalamic-specific reactive gliosis—characterized by cellular morphological changes, including enlarged, extended processes and heightened release of proinflammatory cytokines—that contributes to the metabolic consequences of diet-induced obesity. Figure adapted from images created with BioRender.com.
CELLULAR MECHANISMS OF GLUCOSE SENSING.
As recently reviewed (107-110), glucose-excited and glucose-inhibited neurons increase their electrical activity in response to a rise or fall, respectively, in ambient glucose levels. Glucose-excited neurons and pancreatic beta cells employ analogous glucose-sensing strategies: After GLUT2-mediated glucose entry and phosphorylation by glucokinase, glycolysis and oxidative phosphorylation of glucose increase cytosolic ATP levels, triggering the closure of ATP-sensitive potassium (KATP) channels and cell depolarization. In contrast, glucose-inhibited neurons are activated when glucose is low via a mechanism involving reduced glucose phosphorylation by glucokinase, elevation of AMP:ATP levels, AMP kinase activation, and closure of membrane chloride channels. Whether neuronal glucose sensing is required for normal neuronal activity and central nervous system control of glycemia is debated. Although brain glucose levels parallel circulating levels, brain glucose entry is tightly controlled by the blood-brain barrier (BBB), and glucose concentrations in brain interstitium (0.7–2.5 mmol/L) are considerably lower than in the circulation (3.9–5.6 mmol/L in health) (109, 111). Thus, direct neuronal glucose sensing is more likely to be relevant in specialized brain regions, called circumventricular organs, not protected by the BBB. Alternatively, neuronal glucose sensing may involve indirect mechanisms such as the metabolism of lactate released from neighboring glia (112) and/or afferent neural input from peripheral glucose sensors (26, 113-115).
From a teleological perspective, a neural-based glucoregulatory system makes sense given the brain’s substantial energetic demands and its dependence upon glucose as a fuel. Phylogenetic evidence supports the notion that glucoregulatory functions, including glucose sensing and insulin secretion, originated in neural cell types that preceded the evolution of pancreatic islets (117). Interestingly, the beta cell shares many morphological, transcriptional, and electrophysiological properties with nerve cells (118) despite originating from a distinct embryonic germ layer (endoderm). These observations have spurred the hypothesis that specialized endodermal cells adopted a neuronal transcription program during beta cell evolution, while glucoregulatory systems in the brain were largely preserved (117). This concept, though speculative, fits well with accruing evidence of the central control of glycemia and provides a framework for understanding the complex contribution of the mammalian CNS to glucoregulatory functions delegated to the islets.
We hypothesize that glucose homeostasis depends upon the ability of the CNS to (a) reliably detect interoceptive signals regarding the circulating glucose level, (b) rapidly and precisely compute the anticipated homeostatic consequences of possible behaviors, and (c) regulate the balance between rates of glucose production and glucose utilization by adaptively adjusting autonomic and neuroendocrine outflow to glucose effector organs, including the pancreas and liver. Combined with evidence that normal islets can compensate for progressive impairment of glucose effectiveness or insulin sensitivity, we infer that normal CNS and islet function may be permissive for normal glucose homeostasis, with islet compensation (e.g., insulin hypersecretion) limiting the consequences of central glucoregulatory dysfunction (e.g., impaired Sg) when it is mild but not when it is more advanced. Defects in both central control and islet function may therefore be required for disease progression. Here, we consider the cause(s) of central glucoregulatory dysfunction with the potential to raise the BDLG in T2D.
4.1. Does Reduced Brain Glucose Sensing Contribute to T2D Pathogenesis? Lessons from the Defense Against Hypoglycemia
One possible explanation for the increased BDLG in T2D is that it is a CNS response to a perceived glucose deficiency. The brain response to a fall in blood glucose is well characterized, involving highly coordinated neuroendocrine and autonomic counterregulatory responses (CRRs) that collectively restore normoglycemia (for detailed reviews, see 114, 119). These responses include secretion of epinephrine, corticosterone/cortisol, and glucagon, which, combined with suppression of insulin secretion and elevation of HGP, rapidly return low blood glucose levels back to the BDLG (Figure 1). Multiple research groups have identified subsets of VMN neurons whose activity is required for full CRR generation during systemic hypoglycemia, implicating the VMN as a central node in the underlying neurocircuitry (120–123). If the same neurons are activated experimentally when glucose levels are not limiting (i.e., during normoglycemia by optogenetic or chemogenetic stimulation), diabetes-range hyperglycemia is rapidly elicited by activating CRRs normally reserved for the response to hypoglycemia—including, paradoxically, suppression of GSIS despite profound hyperglycemia (120-122, 124). As diminished GSIS, hyperglucagonemia, and elevated HGP are features common to both CRRs and T2D, pathological overactivation of neurocircuits (e.g., VMN) that function to mobilize glucose may contribute to diabetic hyperglycemia. This is a provocative idea, but both direct and indirect evidence support this possibility.
The glycemic threshold for triggering CRRs (presumably, the level at which the brain perceives a glucose deficit) is elevated by 40% or more in people with T2D compared to nondiabetic individuals (125, 126). Despite this elevated threshold for CRR activation, the magnitude of CRRs (and their ability to raise blood glucose) is preserved, implying that adaptive responses to glucose deficiency are intact, but the degree of hypoglycemia needed to mount them has lessened. This effect is even more pronounced in monogenic diabetes resulting from glucokinase mutations (126), suggesting that defective glucose metabolism (and putatively cellular glucose sensing) raises the glycemic threshold for CRR initiation. Although glucokinase is expressed in multiple tissues, including brain and islets, a specific role for central glucokinase activity is supported by rodent studies in which increasing glucokinase activity in the ARC improves GSIS and glucose tolerance (127). Together, these findings support a model of T2D pathogenesis in which the central representation of glucose deficiency occurs at a blood glucose level higher than the normal hypoglycemia threshold (Figure 1).
Additional support stems from evidence that brain glucose uptake is reduced in humans with obesity and T2D (128, 129). While the underlying mechanisms are poorly understood, a role for overnutrition in hypothalamic dysfunction has been proposed. In rodents, 3 days of HFD feeding is sufficient to induce hypothalamic gliosis (130) (reactive astrocyte formation and proinflammatory microglial infiltration with deleterious consequences for resident neurocircuits; for a detailed review, see 116) (Figure 3). Further, obesity is attenuated in mice fed a HFD by interventions that block this microglial inflammatory activation (131). HFD feeding also suppresses brain glucose uptake via endothelial downregulation of the glucose transporter, GLUT1 (132); over time, these responses are associated with hypothalamic angiogenesis, which may reflect compensatory vascular remodeling (132, 133). Studies in humans have similarly shown that both hypothalamic gliosis (134) and the number of hypothalamic arterioles (133) are increased in obesity and T2D. Addressing whether and how these vascular changes affect brain glucose sensing, neurocircuit function, and T2D pathogenesis are priorities for future studies.
4.2. Hypothalamic Glucoregulatory Neurocircuits: the Melanocortin System
Among the brain’s distributed network of glucoregulatory neurons, those in the hypothalamic ARC have received the most attention. By virtue of their privileged anatomical position nestled between the circumventricular median eminence and the third ventricle (Figure 3), ARC neurons can sense nutrients and hormones circulating in both the blood and cerebrospinal fluid (for reviews, see 135, 136). ARC neurons not only adjust their activity according to energetic state (137), feed-forward cues of nutrient availability (137, 138), and metabolic demand (139) but also exert strong effects on energy (140, 141) and glucose (41, 142) homeostasis, many of which involve the central melanocortin system.
The melanocortin system, a highly conserved cellular signaling pathway, directs catabolic and anabolic metabolism depending upon the balance of melanocortin 4 receptor (MC4r) binding to two opposing ligands, each expressed by a distinct subset of ARC neurons (143). The anabolic arm is activated by agouti-related peptide (AgRP), an inverse agonist of MC4r that is released in response to caloric insufficiency. By reducing MC4r signaling, AgRP promotes increased food intake, reduced energy expenditure, and weight gain (1, 143). Conversely, the catabolic arm is driven by the MC4r agonist α-melanocyte stimulating hormone [α-MSH, derived from its precursor pro-opiomelanocortin([POMC)] in response to caloric excess. MC4r activation by α-MSH suppresses appetite and raises energy expenditure (143). Loss-of-function MC4r mutations in rodents and humans result in profound early-onset obesity; polymorphisms at the POMC gene locus are associated with T2D risk (144-146). Growing evidence suggests that reduced melanocortin signaling due to AgRP and POMC neuron dysfunction predisposes to glycemic abnormalities characteristic of T2D (147, 148).
4.3. CNS Regulation of Insulin-Independent Glucose Disposal
Among the most impressive examples of central glucoregulatory capacity is the finding that intracerebroventricular infusion of the adipocyte-derived hormone leptin restores normoglycemia to rodent models of insulin-deficient T1D [induced by streptozotocin (STZ), a beta cell toxin] (for a review, see 149). Leptin-mediated glucose normalization is associated with increased autonomic outflow to the liver (150), suppressed gluconeogenesis and HGP (151), reduced hyperglucagonemia (151, 152), and increased brain and peripheral tissue glucose uptake (151). These findings raise the question, how can leptin action in the brain ameliorate diabetes induced by beta cell destruction unless the brain plays a role in the hyperglycemia of insulin deficiency (2)?
To address this question, we consider recent evidence that inactivation of select glucoregulatory VMN neurons (whose activity promotes CRRs during hypoglycemia) greatly diminishes STZ-mediated hyperglycemia (123). This suggests that hyperglycemia elicited by severe insulin deficiency requires activation of these neurons. Earlier evidence similarly implicated VMN activity in diabetic hyperglycemia, as intra-VMN delivery of brain-derived neurotrophic factor (153) or leptin (154) comparably resolves STZ-mediated hyperglycemia via insulin-independent mechanisms. However, given that leptin deficiency develops as a consequence of insulin deficiency and fat loss in STZ-treated mice, whether centrally driven hyperglycemia in this setting is due to insulin versus leptin deficiency per se is unclear.
Evidence suggests that both may contribute. For example, HGP is suppressed by central administration of either insulin or leptin (151, 155), primarily via KATP-mediated neuronal hyperpolarization (41, 155). Consistent with this notion is recent evidence of an important role for KATP channels in the control of ARC AgRP neuron activity in relation to glucose homeostasis. Specifically, (a) genetic deletion of KATP channels in AgRP neurons elicits their hyperactivity, leading in turn to hyperglycemia, insulin resistance, and obesity (41), and (b) central administration of KATP channel openers (resulting in neuronal silencing) reduces blood glucose by suppression of HGP (156). In humans, systemic delivery of KATP channel antagonists blunts hyperglycemia-induced suppression of HGP by nearly 50%, suggesting that similar brain mechanisms may contribute to glycemic control in humans (13). These findings highlight hypothalamic regulation of HGP as a promising target for glucose lowering in T2D.
4.4. CNS Control of Insulin Sensitivity
The CNS is capable of driving rapid, parallel changes in insulin secretion and sensitivity during cold exposure, suggesting that aberrant neural activity could conceivably underlie defective coupling of these determinants of glycemia during T2D progression. Multiple brain regions have the capacity to drive rapid and reversible changes in peripheral insulin sensitivity (157), and daily changes in insulin sensitivity appear to be orchestrated by the central circadian system (158). Magnetic resonance imaging (MRI)-based evidence in humans has linked obesity-induced hypothalamic gliosis to systemic insulin resistance and glucose intolerance, suggesting that a hypothalamic defect contributes to obesity-associated metabolic impairment (134). Metabolic improvement induced by bariatric surgery in women with T2D is associated with an improved MRI signature of hypothalamic gliosis (159), suggesting that the underlying neuropathological process may be reversible. Future studies that clarify the contribution of gliosis and associated mechanisms driving aberrant neurocircuit activity to insulin resistance and T2D pathogenesis are a scientific priority.
4.5. CNS Regulation of Beta Cell Mass
As discussed in Section 2, the CNS has the capacity to surveil and regulate islet activity during acute challenges to glucose homeostasis such as meal intake, insulin-induced hypoglycemia, and cold exposure. Beyond these functions, the ANS also affects the development and plasticity of the endocrine pancreas (160). Animal models with reduced parasympathetic activity during postnatal development exhibit diminished beta cell mass and GSIS in adulthood (161), whereas beta cell proliferation is increased when cultured islets are treated with muscarinic agonists. Since adrenergic stimulation has the opposite effect, available evidence points to direct and counteracting roles for the two limbs of the ANS in regulating beta cell mass (162-165). Manipulating the balance between sympathetic and parasympathetic outflow, therefore, may present a promising therapeutic approach to the preservation of both beta cell mass and function in T2D.
4.6. Targeting the Brain to Induce Sustained Diabetes Remission
Preclinical studies have demonstrated the ability of multiple centrally administered peptides, in addition to leptin, to elicit diabetes remission in rodent T2D models. Peptides in the fibroblast growth factor (FGF) family have received the most attention, based on evidence that central administration of FGF19 (166), FGF21 (167), and FGF1 (168-171) promotes insulin-independent glucose lowering in rodent models of T2D via activation of FGF receptors in the mediobasal hypothalamus. Strikingly, the glucose-lowering effects of FGF1 are quite long lived: A single intracerebroventricular administration of FGF1 normalizes glycemia for months in the leptin-deficient (ob/ob) mouse model of T2D (168) via an action in the ARC (170). Unlike conventional antidiabetic medications that elicit glucose lowering, FGF1 treatment does not increase the risk of hypoglycemia, nor does it alter insulin secretion or sensitivity. Instead, FGF1 appears to mediate its effects by restoring the BDLG to normal (168). Investigation into mechanisms underlying this FGF1 effect has identified extracellular matrix remodeling (172) and sustained inhibition of AgRP neurons coupled with increased melanocortin signaling (173) as likely targets for this effect. Clarifying where, how, and why FGF1 works to normalize BDLG in T2D is a priority for future work.
5. CNS-TARGETED THERAPEUTIC STRATEGIES FOR T2D: LOOKING TO THE FUTURE
Growing evidence that CNS-targeted therapies can normalize the BDLG in preclinical T2D models (151, 168) underscores their untapped potential as future T2D treatments. Supplemental Table 1 lists currently approved antidiabetic drug classes (and examples) with CNS-based metabolic effects. Below we discuss the most promising of the approved agents, the GLP1R agonists (Supplemental Appendix 1), to highlight the potential of centrally mediated drug action even when the drug’s development was not predicated on central action. We then propose future therapeutic strategies targeting both central and peripheral glucoregulatory neural control mechanisms with the potential to enhance both insulin-dependent and insulin-independent glucose disposal as a path toward T2D remission/disease modulation.
5.1. GLP1R Agonists
Long-acting GLP1R agonists are among the more promising new drugs for T2D. Semaglutide, a once-weekly subcutaneously administered GLP1R agonist, was evaluated in the SUSTAIN clinical trial program in over 8,000 patients across the T2D spectrum. These trials demonstrated superior glycemic control and weight loss with semaglutide versus all comparators (including insulin, other GLP1R agonists, and dipeptidyl peptidase inhibitors) (174, 175). Semaglutide also has shown efficacy for clinically meaningful (~15%) weight loss in obese patients without diabetes (176).
GLP1R agonists elicit multiple glucose-lowering effects that include augmentation of GSIS (via a direct effect on beta cells) and reductions of food intake and body weight that are mediated centrally. Identifying contributions of various subnetworks of GLP1R+ neurocircuits (Supplemental Appendix 1) to the pleiotropic effects of GLP1 is an active area of research. Another active area of research involves the development and validation of intermixed, unimolecular, multiagonist peptides involving combinations of GLP1, glucagon, and/or glucose-stimulated insulinotropic polypeptide (GIP) (177). The goal of this strategy is to enhance GLP1’s activity by broadening tissue targets and synergizing within tissues (e.g., brain and islet) that express multiple receptors. Several preclinical (178, 179) and Phase I and II clinical studies (180, 181) have demonstrated that these drugs may outperform single agonists in sustained glycemic improvement and weight loss and provide a promising path forward for GLP1-related agents (177). The roles of the brain and the islets as targets for these drugs are an active area of study, and it is likely that both contribute.
5.2. Future Prospects for CNS-Targeted T2D Remission
Perhaps the most important lesson from preclinical models of T2D is that CNS-targeted therapies have the potential to induce T2D remission by restoring BDLG to normal (unlike current non-surgical treatment options). Two notable examples are intracerebroventricular injection of FGF1 in rodent models of T2D (168) and of leptin in rodent models of T1D (151). In both cases, BDLG normalization appears to require suppression of abnormally hyperactive ARC AgRP neurons, possibly coupled with POMC neuron activation, thereby increasing melanocortin signaling (41, 173). FGF1 is ineffective in normalizing BDLG in T2D models with deficient melanocortin signaling (173). Leptin is ineffective in normalizing BDLG in T1D models in which leptin receptors are deleted from AgRP neurons (41).
How does one pursue translation of these preclinical models? A first step would be to test the safety and efficacy of the agents themselves in large animal models. One such study is underway for FGF1 in a nonhuman primate model of T2D. Safety and efficacy of centrally administered leptin has been tested in nonhuman primates (182, 183). A pilot study of peripheral administration of metreleptin (modified to increase half-life, which is approved for use in lipodystrophic diabetes) has been completed in T1D patients (184). While glycemia was not significantly improved in this study, preclinical findings suggest that leptin must be administered directly into the brain for its antidiabetic effects to be observed (151). A pilot study to test the safety and efficacy of central administration of leptin in T1D patients is therefore a priority. Such a strategy has the potential to elicit effective and sustained normalization of glucose without the risks of hypoglycemia (185) and weight gain (186).
Parallel efforts aimed at translation may further dissect the cellular and molecular mechanisms of promising agents. At multiple time points after intracerebroventricular FGF1 injection into obese diabetic mice, a recent comprehensive transcriptomic analysis across mediobasal hypothalamic cell types revealed other potentially druggable pathways (173). Notably, the transcriptomic response of astrocytes displayed prominent neuroprotective and neuron-interacting phenotypes, with evidence suggesting increased astrocytic coverage of AgRP neuron synapses. Some of these cellular signatures are common to other neurological disease processes, including ischemic stroke, and therefore may highlight the potential of drug candidates co-opted from other disease states.
A critical consideration for any centrally targeted pharmaceutical will be the pharmacokinetic nuances of drug delivery to the brain. This highlights the therapeutic potential of strategies that increase hypothalamic melanocortin signaling for treatment of T2D and associated metabolic disorders. The close proximity of AgRP neurons to the median eminence, which has a fenestrated blood-brain barrier (BBB), suggests that drugs aimed at silencing these neurons might access them via the circulation. In other cases, however, centrally targeted therapies that are administered systemically will have to traverse the BBB and contend with unique pharmacokinetic properties of CNS drug delivery (187). Promising alternative possibilities include intranasal (188) or catheter-based intrathecal delivery (189) (Figure 4).
Figure 4.
Future prospects for central nervous system (CNS)-targeted type 2 diabetes (T2D) remission/disease modulation. Device-based, CNS-targeted approaches to T2D disease modulation include (❶) vagal nerve stimulation targeting fascicles innervating liver, portal vein, and pancreatic islets, (❷) spinal cord stimulation with electrodes placed epidurally to target preganglionic sympathetic nerves in the intermediolateral cell column (blue cell bodies in inset) innervating the celiac ganglion (~T5–T9 levels), and (❸) intrathecal catheter-based targeted drug delivery (e.g., leptin, fibroblast growth factor analogs) with infusion via an implanted subcutaneous reservoir or pump. Figure adapted from images created with BioRender.com.
In addition to pharmaceuticals, other therapeutic modalities for restoring brain neural circuit disorders include (a) deep brain stimulation, currently FDA-approved for Parkinson’s disease, essential tremor, dystonia, obsessive compulsive disorder, and epilepsy (190), and (b) CNS gene therapy via antisense oligonucleotides (191) or stereotactic virally delivered gene therapy (192), which are being investigated in Huntington’s and Parkinson’s diseases, respectively, with promising results.
Deep brain stimulation relies on modulating neural circuit activity using controlled electrical fields emitted from chronically implanted electrodes. While future iterations of this technique may provide a strategy for modulating glucoregulatory neural circuits, a current limitation of this approach is that the electric fields generated are rather large compared to the relatively small and cellularly heterogenous nuclei of the mediobasal hypothalamus. CNS gene therapy is an increasingly viable strategy in select neurodegenerative disorders, with improved techniques for gene delivery and coverage of the brain region of interest and an understanding of the genetic targets that are most tractable (i.e., therapeutic gain versus loss of gene function) (192). The potential of this approach in T2D is intriguing in light of preclinical studies demonstrating improved glucose tolerance with ARC-specific transduction of the glucokinase gene (127), conferring improved neuronal glucose sensing, and the aforementioned evidence of reduced brain glucose sensing in T2D pathogenesis. Further investigation and refinement of the safety and efficacy of CNS gene therapy for severe neurodegenerative diseases should ultimately lay the groundwork in the potential application of this approach to chronic neural circuit disorders, including T2D.
5.3. Antidiabetic Strategies Targeting the Autonomic Nervous System
In addition to developing T2D therapies directed at the brain, the ANS presents an attractive target based on its role as an essential relay between central neurocircuits and peripheral glucose effectors. Modulating the PNS and/or SNS branch of the ANS inT2D may facilitate the balancing of mechanisms governing BDLG. Vagal efferents innervating beta cells not only stimulate GSIS (193, 194) but can also promote beta cell proliferation and expansion (163-165), thereby offering potential T2D disease-modifying treatment. Although uncertainty remains regarding the extent to which the liver parenchyma is innervated by vagal efferent fibers (195), activation of these fibers offers a potentially viable approach to inhibit expression of gluconeogenic genes, gluconeogenesis, and HGP (196).
Conversely, vagal afferents are well documented to innervate the hepatic portal vein and are a critical component of the glucose-sensing machinery that provides input relevant to the circulating glucose level to central neurocircuits (e.g., for the response to hypoglycemia or the portal glucose signal) (115, 197-201). Recent work (115) characterizes a vago-vagal circuit, originating with vagal afferents innervating the portal vein wall, directly contacting vagal efferents in the hindbrain dorsal motor nucleus of the vagus (which in turn modulates hepatic and islet function). Therapeutic strategies targeting these neurons, perhaps in combination with reduced SNS tone at the liver and pancreas, may have the potential to lower the BDLG in patients with T2D.
While cholinergic/anticholinergic drugs may be limited by off-target effects on other organs, device-based neuromodulation using implantable vagal nerve stimulators (VNSs) offers a more targeted approach. For example, if the vago-vagal circuit supplying the hepatic portal vein could be specifically targeted with a VNS device, the circuit could potentially be dialed up or down as needed to optimize the BDLG. Considerations in the development of this strategy will include optimal placement of the electrode along the central-peripheral axis of the nerve (to reduce the number of vagal fascicles destined for nonglucoregulatory organs) and the stimulation parameters (i.e., pulse width/frequency/duration to engage the highest fraction of desirable efferent/afferent glucoregulatory vagal fascicles) (202-204). VNS is already FDA approved for use in epilepsy and depression, and early proof-of-principle studies in patients with obesity and T2D show promising results (205, 206) (Figure 4).
Alternatively, therapeutic strategies could target SNS modulation of the BDLG. As discussed above, increased sympathetic tone on beta cells suppresses GSIS (14), while stimulating glycogenolysis and gluconeogenesis in hepatocytes, thus increasing HGP (26, 208, 209); conversely, sympathetic denervation of the liver in DIO dogs improves glucose tolerance (91). Similarly, catheter-based renal sympathetic denervation in patients—developed for treatment-resistant hypertension—surprisingly appears to reduce whole-body sympathetic nerve activity (210) and in small early studies showed reduced fasting plasma glucose and insulin levels, suggesting improved insulin sensitivity (211). Larger studies of this approach for T2D have not been conducted, but meta-analyses of small studies failed to show that the antidiabetic effect is reliable (212). Inconsistent results from this approach extend to its initial indication—hypertension—with randomized, controlled studies showing both positive and negative outcomes for its efficacy (caveats cited for negative studies included variability in denervation technique across study sites and patient selection criteria) (213).
Analogous to the considerations discussed for VNS above, effective SNS modulation may be better achieved with more targeted, device-based neuromodulation. Implantable epidural spinal cord stimulators are currently FDA approved and widely used as a targeted treatment for chronic neuropathic pain (214). In Europe, targeted spinal cord stimulators are also used for ischemic pain (e.g., angina) related to arterial insufficiency and have been shown to produce sympathetic blockade as part of their mechanism of action (215, 217). By extension, spinal cord stimulation (SCS) modulation of SNS innervation of the islet and liver, by way of preganglionic sympathetic nerves located in the intermediolateral cell column and projecting to the celiac ganglion, may provide a tunable strategy for normalizing the BDLG in T2D. Furthermore, in patients with neuropathic pain, SCS appears to modulate dorsal horn–dorsal column spinal sensory pathways that include direct inputs from TRPV1+ dorsal root ganglion neurons (216), subsets of which innervate the islets at the upper thoracic levels as discussed above (29-31). SCS thus offers multiple potential mechanisms by which to modulate neural activity to and from peripheral glucose effectors.
Development of new therapeutic strategies targeting CNS control of glucose homeostasis will be a formidable challenge. However, growing and undeniable evidence that the CNS plays a fundamental role in normal and abnormal glucose metabolism is a sign of an emerging era of CNS-centered research and drug discovery in T2D.
Supplementary Material
SUMMARY POINTS.
Insulin-independent glucose disposal mechanisms predominate in the control of the basal glucose level.
Type 2 diabetes (T2D) is associated with impaired insulin-independent glucose disposal. Evidence that this impairment is primary in T2D pathogenesis is as compelling as the evidence of a causal role played by a beta cell defect.
In rodent models of type 1 diabetes and T2D, the brain can be targeted to normalize the biologically defended level of glycemia (BDLG) via mechanisms that are largely, if not exclusively, insulin independent.
The mediobasal hypothalamus is the only known brain area capable of normalizing the BDLG in preclinical models of T2D, with increased melanocortin signaling engaged by neurons in the arcuate nucleus likely playing a key role.
In both humans and preclinical models, obesity and T2D are associated with hypothalamic pathology, including activation of glial cells (reactive gliosis) in areas involved in glucose homeostasis; this pathological response may contribute to elevation of the BDLG by reducing neuronal sensing of (or responsiveness to) the circulating glucose level.
Neuronal subsets in the hypothalamic ventromedial nucleus that normally orchestrate counterregulatory responses to glucose deficiency may aberrantly promote the elevated BDLG in T2D; inactivation of these neurons in rodent T2D models elicits insulin-independent glucose lowering.
Future therapeutic strategies and drug discovery focused on central nervous system control of both insulin-independent and insulin-dependent glucose disposal offer significant untapped potential yet to be addressed by current T2D treatments.
ACKNOWLEDGMENTS
The authors gratefully acknowledge Dr. Daniel Porte Jr. for his careful reading of and comments on the manuscript and Dr. Alan Watts for his insights into functional anatomy of the autonomic nervous system. This work was supported by the NIH National Institute of Diabetes and Digestive and Kidney Diseases grants DP2-DK128802 (Z.M.), T32-DK007247 (C.L.F.), R01 DK124238 and R01 DK83042 (M.W.S.); a Department of Defense CDMRP/PRMRP grant W81XWH2010250 (Z.M.); and a Barrow Neurological Foundation grant 18-0025-30-05 (Z.M.).
Glossary
- CNS
central nervous system
- BDLG
biologically defended level of glycemia
- T2D
type 2 diabetes
- T1D
type 1 diabetes
- GSIS
glucose-stimulated insulin secretion
- HGP
hepatic glucose production
- ANS
autonomic nervous system
- SNS
sympathetic nervous system
- PNS
parasympathetic nervous system
- GLP1
glucagon-like peptide 1
- FSIGT
frequently sampled intravenous glucose tolerance test
- Sg
glucose effectiveness derived from minimal model of glucose kinetics
- Kg
glucose disappearance constant derived from FSIGT test
- Si
insulin sensitivity index derived from minimal model of glucose kinetics
- NHGU
net hepatic glucose uptake, or net hepatic glucose balance
- VMN
ventromedial nucleus
- ARC
arcuate nucleus
- CRR
counterregulatory response
Footnotes
DISCLOSURE STATEMENT
The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.
LITERATURE CITED
- 1.Grayson BE, Seeley RJ, Sandoval DA. 2013. Wired on sugar: the role of the CNS in the regulation of glucose homeostasis. Nature 14(1):24–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Schwartz MW, Seeley RJ, Tschöp MH, Woods SC, Morton GJ, et al. 2013. Cooperation between brain and islet in glucose homeostasis and diabetes. Nature 503(7474):59–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Turner RC, Holman RR, Matthews D, Hockaday TDR, Peto J. 1979. Insulin deficiency and insulin resistance interaction in diabetes: estimation of their relative contribution by feedback analysis from basal plasma insulin and glucose concentrations. Metabolis 28(11):1086–96 [DOI] [PubMed] [Google Scholar]
- 4.Edelman SV, Polonsky WH. 2017. Type 2 diabetes in the real world: the elusive nature of glycemic control. Diabetes Care 40(11):1425–32 [DOI] [PubMed] [Google Scholar]
- 5.Kahn SE, Cooper ME, Prato SD. 2014. Pathophysiology and treatment of type 2 diabetes: perspectives on the past, present, and future. Lancet 383(9922):1068–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kahn SE, Hull R, Utzschneider K. 2006. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444:840–46 [DOI] [PubMed] [Google Scholar]
- 7.Esterson YB, Carey M, Boucai L, Goyal A, Raghavan P, et al. 2016. Central regulation of glucose production may be impaired in type 2 diabetes. Diabetes 65(9):2569–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hawkins M, Gabriely I, Wozniak R, Reddy K, Rossetti L, Shamoon H. 2002. Glycemic control determines hepatic and peripheral glucose effectiveness in type 2 diabetic subjects. Diabetes 51(7):2179–89 [DOI] [PubMed] [Google Scholar]
- 9.Kahn SE, Prigeon RL, McCulloch DK, Boyko EJ, Bergman RN, et al. 1994. The contribution of insulin-dependent and insulin-independent glucose uptake to intravenous glucose tolerance in healthy human subjects. Diabetes 43(4):587–92 [DOI] [PubMed] [Google Scholar]
- 10.Baron AD, Brechtel G, Wallace P, Edelman SV. 1988. Fasting decreases rates of noninsulin-mediated glucose uptake in man. J. Clin. Endocrinol. Metab 67(3):532–40 [DOI] [PubMed] [Google Scholar]
- 11.Bergman RN, Phillips LS, Cobelli C. 1981. Physiologic evaluation of factors controlling glucose tolerance in man: measurement of insulin sensitivity and beta-cell glucose sensitivity from the response to intravenous glucose. J. Clin. Investig 68(6):1456–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Martin BC, Warram JH, Krolewski AS, Soeldner JS, Kahn CR, et al. 1992. Role of glucose and insulin resistance in development of type 2 diabetes mellitus: results of a 25-year follow-up study. Lancet 340(8825):925–29 [DOI] [PubMed] [Google Scholar]
- 13.Carey M, Lontchi-Yimagou E, Mitchell W, Reda S, Zhang K, et al. 2020. Central KATP channels modulate glucose effectiveness in humans and rodents. Diabetes 69(6):1140–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Morton GJ, Muta K, Kaiyala KJ, Rojas JM, Scarlett JM, et al. 2017. Evidence that the sympathetic nervous system elicits rapid, coordinated, and reciprocal adjustments of insulin secretion and insulin sensitivity during cold exposure. Diabetes 66(4):823–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Banting FG, Best CH, Collip JB, Campbell WR, Fletcher AA. 1922. Pancreatic extracts in the treatment of diabetes mellitus. Can. Med. Assoc. J 12(3):141–46 [PMC free article] [PubMed] [Google Scholar]
- 16.Cnop M, Vidal J, Hull RL, Utzschneider KM, Carr DB, et al. 2007. Progressive loss of β-cell function leads to worsening glucose tolerance in first-degree relatives of subjects with type 2 diabetes. Diabetes Care 30(3):677–82 [DOI] [PubMed] [Google Scholar]
- 17.Ahrén B. 2000. Autonomic regulation of islet hormone secretion—implications for health and disease. Diabetologia 43(4):393–410 [DOI] [PubMed] [Google Scholar]
- 18.Rodriguez-Diaz R, Caicedo A. 2014. Neural control of the endocrine pancreas. Best Pract. Res. Clin. Endocrinol. Metab 28(5):745–56 [DOI] [PubMed] [Google Scholar]
- 19.Faber CL, Deem JD, Campos CA, Taborsky GJ, Morton GJ. 2020. CNS control of the endocrine pancreas. Diabetologia 63(10):2086–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chien H-J, Chiang T-C, Peng S-J, Chung M-H, Chou Y-H, et al. 2019. Human pancreatic afferent and efferent nerves: mapping and 3-D illustration of exocrine, endocrine, and adipose innervation. Am. J. Physiol. Gastrointest. Liver Physiol 317(5):G694–706 [DOI] [PubMed] [Google Scholar]
- 21.Tang S-C, Baeyens L, Shen C-N, Peng S-J, Chien H-J, et al. 2018. Human pancreatic neuro-insular network in health and fatty infiltration. Diabetologia 61(1):168–81 [DOI] [PubMed] [Google Scholar]
- 22.Gautam D, Han S-J, Hamdan FF, Jeon J, Li B, et al. 2006. A critical role for β cell M3 muscarinic acetylcholine receptors in regulating insulin release and blood glucose homeostasis in vivo. Cell Metab. 3(6):449–61 [DOI] [PubMed] [Google Scholar]
- 23.D’Alessio DA, Kieffer TJ, Taborsky GJ, Havel PJ. 2001. Activation of the parasympathetic nervous system is necessary for normal meal-induced insulin secretion in rhesus macaques. J. Clin. Endocrinol. Metab 86(3):1253–59 [DOI] [PubMed] [Google Scholar]
- 24.Henderson JR, Jefferys DB, Jones RH, Stanley D. 1976. The effect of atropine on the insulin release caused by oral and intravenous glucose in human subjects. Acta. Endocrinol 83(4):772–80 [DOI] [PubMed] [Google Scholar]
- 25.de Lartigue G. 2016. Role of the vagus nerve in the development and treatment of diet-induced obesity. J. Physiol 594(20):5791–815 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lin EE, Scott-Solomon E, Kuruvilla R. 2021. Peripheral innervation in the regulation of glucose homeostasis. Trends Neurosci. 44(3):189–202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Cheung GWC, Kokorovic A, Lam CKL, Chari M, Lam TKT. 2009. Intestinal cholecystokinin controls glucose production through a neuronal network. Cell Metab. 10(2):99–109 [DOI] [PubMed] [Google Scholar]
- 28.Varin EM, Mulvihill EE, Baggio LL, Koehler JA, Cao X, et al. 2019. Distinct neural sites of GLP-1R expression mediate physiological versus pharmacological control of incretin action. Cell Rep. 27(11):3371–84.e3 [DOI] [PubMed] [Google Scholar]
- 29.Razavi R, Chan Y, Afifiyan FN, Liu XJ, Wan X, et al. 2006. TRPV1+ sensory neurons control β cell stress and islet inflammation in autoimmune diabetes. Cell 127(6):1123–35 [DOI] [PubMed] [Google Scholar]
- 30.Akiba Y, Kato S, Katsube K, Nakamura M, Takeuchi K, et al. 2004. Transient receptor potentia lvanilloid subfamily 1 expressed in pancreatic islet β cells modulates insulin secretion in rats. Biochem. Biophys. Res. Commun 321(1):219–25 [DOI] [PubMed] [Google Scholar]
- 31.Gram DX, Ahrén B, Nagy I, Olsen UB, Brand CL, et al. 2007. Capsaicin-sensitive sensory fibers in the islets of Langerhans contribute to defective insulin secretion in Zucker diabetic rat, an animal model for some aspects of human type 2 diabetes. Eur. J. Neurosci 25(1):213–23 [DOI] [PubMed] [Google Scholar]
- 32.Bai L, Mesgarzadeh S, Ramesh KS, Huey EL, Liu Y, et al. 2019. Genetic identification of vagal sensory neurons that control feeding. Cell 179(5):1129–43.e23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Makhmutova M, Weitz J, Tamayo A, Pereira E, Boulina M, et al. 2021. Pancreatic β-cells communicate with vagal sensory neurons. Gastroenterology 160(3):875–88.e11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Alvarsson A, Jimenez-Gonzalez M, Li R, Rosselot C, Tzavaras N, et al. 2020. A 3D atlas of the dynamic and regional variation of pancreatic innervation in diabetes. Sci. Adv 6(41):eaaz9124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Eizirik DL, Korbutt GS, Hellerström C. 1992. Prolonged exposure of human pancreatic islets to high glucose concentrations in vitro impairs the beta-cell function. J. Clin. Investig 90(4):1263–68 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kaneto H. 2015. Pancreatic β-cell glucose toxicityin type 2 diabetes mellitus. Curr. Diabetes Rev. 11(1):2–6 [DOI] [PubMed] [Google Scholar]
- 37.Nicol LE, Grant WF, Grant WR, Comstock SM, Nguyen ML, et al. 2013. Pancreatic inflammation and increased islet macrophages in insulin-resistant juvenile primates. J. Endocrinol 217(2):207–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Dula SB, Jecmenica M, Wu R, Jahanshahi P, Verrilli GM, et al. 2010. Evidence that low-grade systemic inflammation can induce islet dysfunction as measured by impaired calcium handling. Cell Calcium 48(2–3):133–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Krentz NAJ, Gloyn AL. 2020. Insights into pancreatic islet cell dysfunction from type 2 diabetes mellitus genetics. Nat. Rev. Endocrinol 16(4):202–12 [DOI] [PubMed] [Google Scholar]
- 40.Stanley S, Domingos AI, Kelly L, Garfield A, Damanpour S, et al. 2013. Profiling of glucose-sensing neurons reveals that GHRH neurons are activated by hypoglycemia. Cell Metab. 18(4):596–607 [DOI] [PubMed] [Google Scholar]
- 41.Xu J, Bartolome CL, Low CS, Yi X, Chien C-H, et al. 2018. Genetic identification of leptin neural circuits in energy and glucose homeostases. Nature 556(7702):505–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Foster ED, Bridges ND, Feurer ID, Eggerman TL, Hunsicker LG, et al. 2018. Improved health-related quality of life in a phase 3 islet transplantation trial in type 1 diabetes complicated by severe hypoglycemia. Diabetes Care 41(5):1001–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Vallerand AL, Perusse F, Bukowiecki LJ. 1987. Cold exposure potentiates the effect of insulin on in vivo glucose uptake. Am. J. Physiol 253(2):E179–86 [DOI] [PubMed] [Google Scholar]
- 44.Maickel RP, Matussek N, Stern DN, Brodie BB. 1967. The sympathetic nervous system as a homeostatic mechanism. I. Absolute need for sympathetic nervous function in body temperature maintenance of cold-exposed rats. J. Pharmacol. Exp. Ther 157(1):103–10 [PubMed] [Google Scholar]
- 45.Stefanovski D, Richey JM, Woolcott O, Lottati M, Zheng D, et al. 2011. Consistency of the disposition index in the face of diet induced insulin resistance: potential role of FFA. PLOS ONE 6(3):e18134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Morrison SF 2016. Central neural control of thermoregulation and brown adipose tissue. Auton. Neurosci 196:14–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Young JB, Landsberg L. 1979. Effect of diet and cold exposure on norepinephrine turnover in pancreas and liver. Am. J. Physiol 236(5):E524–33 [DOI] [PubMed] [Google Scholar]
- 48.Bergman RN, Ider YZ, Bowden CR, Cobelli C. 1979. Quantitative estimation of insulin sensitivity. Am. J. Physiol 236(6):E667–77 [DOI] [PubMed] [Google Scholar]
- 49.Shulman GI, Liljenquist JE, Williams PE, Lacy WW, Cherrington AD. 1978. Glucose disposal during insulinopenia in somatostatin-treated dogs. J. Clin. Investig 62(2):487–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kahn SE, Klaff LJ, Schwartz MW, Beard JC, Bergman RN, et al. 1990. Treatment with a somatostatin analog decreases pancreatic B-cell and whole body sensitivity to glucose. J. Clin. Endocrinol. Metab 71(4):994–1002 [DOI] [PubMed] [Google Scholar]
- 51.D’Alessio DA, Kahn SE, Leusner CR, Ensinck JW. 1994. Glucagon-like peptide 1 enhances glucose tolerance both by stimulation of insulin release and by increasing insulin-independent glucose disposal. J. Clin. Investig 93(5):2263–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Baron AD, Brechtel G, Wallace P, Edelman SV. 1988. Rates and tissue sites of non-insulin- and insulin-mediated glucose uptake in humans. Am. J. Physiol 255(6):E769–74 [DOI] [PubMed] [Google Scholar]
- 53.Ward GM, Weber KM, Walters IM, Aitken PM, Lee B, et al. 1991. A modified minimal model analysis of insulin sensitivity and glucose-mediated glucose disposal in insulin-dependent diabetes. Metabolis 40(1):4–9 [DOI] [PubMed] [Google Scholar]
- 54.Finegood DT, Hramiak IM, Dupre J. 1990. A modified protocol for estimation of insulin sensitivity with the minimal model of glucose kinetics in patients with insulin-dependent diabetes. J. Clin. Endocrinol. Metab 70(6):1538–49 [DOI] [PubMed] [Google Scholar]
- 55.Welch S, Gebhart SSP, Bergman RN, Phillips LS. 1990. Minimal model analysis of intravenous glucose tolerance test-derived insulin sensitivity in diabetic subjects. J. Clin. Endocrinol. Metab 71(6):1508–18 [DOI] [PubMed] [Google Scholar]
- 56.Martin BC, Warram JH, Krolewski AS, Soeldner JS, Kahn CR, et al. 1992. Role of glucose and insulin resistance in development of type 2 diabetes mellitus: results of a 25-year follow-up study. Lancet 340(8825):925–29 [DOI] [PubMed] [Google Scholar]
- 57.DeFronzo RA. 2009. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 58(4):773–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Esser N, Utzschneider KM, Kahn SE. 2020. Early beta cell dysfunction versus insulin hypersecretion as the primary event in the pathogenesis of dysglycaemia. Diabetologia 63(10):2007–21 [DOI] [PubMed] [Google Scholar]
- 59.Pimenta W, Korytkowski M, Mitrakou A, Jenssen T, Yki-Jarvinen H, et al. 1995. 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 273(23):1855–61 [PubMed] [Google Scholar]
- 60.van Haeften TW, Dubbeldam S, Zonderland ML, Erkelens DW. 1998. Insulin secretion in normal glucose-tolerant relatives of type 2 diabetic subjects: assessments using hyperglycemic glucose clamps and oral glucose tolerance tests. Diabetes Care 21(2):278–82 [DOI] [PubMed] [Google Scholar]
- 61.Utzschneider KM, Prigeon RL, Faulenbach MV, Tong J, Carr DB, et al. 2009. Oral disposition index predicts the development of future diabetes above and beyond fasting and 2-h glucose levels. Diabetes Care 32(2):335–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Staimez LR, Weber MB, Ranjani H, Ali MK, Echouffo-Tcheugui JB, et al. 2013. Evidence of reduced β-cell function in Asian Indians with mild dysglycemia. Diabetes Care 36(9):2772–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tricò D, Natali A, Arslanian S, Mari A, Ferrannini E. 2018. Identification, pathophysiology, and clinical implications of primary insulin hypersecretion in nondiabetic adults and adolescents. JCI Insight 3(24):e124912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Brøns C, Jensen CB, Storgaard H, Hiscock NJ, White A, et al. 2009. Impact of short-term high-fat feeding on glucose and insulin metabolism in young healthy men. J. Physiol 587(10):2387–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Ferrannini E, Natali A, Bell P, Cavallo-Perin P, Lalic N, Mingrone G. 1997. Insulin resistance and hypersecretion in obesity. J. Clin. Investig 100(5):1166–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Loves S, van Groningen L, Filius M, Mekking M, Brandon T, et al. 2018. High-dose, diazoxide-mediated insulin suppression boosts weight loss induced by lifestyle intervention. J. Clin. Endocrinol Metab 103(11):4014–22 [DOI] [PubMed] [Google Scholar]
- 67.Dankner R, Chetrit A, Shanik MH, Raz I, Roth J. 2009. Basal-state hyperinsulinemia in healthy normoglycemic adults is predictive of type 2 diabetes over a 24-year follow-up. Diabetes Care 32(8):1464–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Rizza RA, Mandarino LJ, Genest J, Baker BA, Gerich JE. 1985. Production of insulin resistance by hyperinsulinaemia in man. Diabetologia 28(2):70–75 [DOI] [PubMed] [Google Scholar]
- 69.Martin C, Desai KS, Steiner G. 1983. Receptor and postreceptor insulin resistance induced by in vivo hyperinsulinemia. Can. J. Physiol. Pharm 61(8):802–7 [DOI] [PubMed] [Google Scholar]
- 70.Kobayashi M, Olefsky JM. 1978. Effect of experimental hyperinsulinemia on insulin binding and glucose transport in isolated rat adipocytes. Am. J. Physiol 235(1):E53–62 [DOI] [PubMed] [Google Scholar]
- 71.Roth Flach RJ, Danai LV, DiStefano MT, Kelly M, Menendez LG, et al. 2016. Protein kinase mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) promotes obesity-induced hyperinsulinemia. J. Biol. Chem 291(31):16221–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Templeman NM, Clee SM, Johnson JD. 2015. Suppression of hyperinsulinaemia in growing female mice provides long-term protection against obesity. Diabetologia 58(10):2392–402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mehran AE, Templeman NM, Brigidi GS, Lim GE, Chu K-Y, et al. 2012. Hyperinsulinemia drives diet-induced obesity independently of brain insulin production. Cell Metab. 16(6):723–37 [DOI] [PubMed] [Google Scholar]
- 74.Czech MP. 2017. Insulin action and resistance in obesity and type 2 diabetes. Nat. Med 23(7):804–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sbraccia P, D’Adamo M, Leonetti F, Caiola S, Iozzo P, et al. 1996. Chronic primary hyperinsulinaemia is associated with altered insulin receptor mRNA splicing in muscle of patients with insulinoma. Diabetologia 39(2):220–25 [DOI] [PubMed] [Google Scholar]
- 76.Pontiroli AE, Alberetto M, Capra F, Pozza G. 1990. The glucose clamp technique for the study of patients with hypoglycemia: insulin resistance as a feature of insulinoma. J. Endocrinol. Investig 13(3):241–45 [DOI] [PubMed] [Google Scholar]
- 77.Prato SD, Leonetti F, Simonson DC, Sheehan P, Matsuda M, DeFronzo RA. 1994. Effect of sustained physiologic hyperinsulinaemia and hyperglycaemia on insulin secretion and insulin sensitivity in man. Diabetologia 37(10):1025–35 [DOI] [PubMed] [Google Scholar]
- 78.Alemzadeh R, Langley G, Upchurch L, Smith P, Slonim AE. 1998. Beneficial effect of diazoxide in obese hyperinsulinemic adults. J. Clin. Endocrinol. Metab 83(6):1911–15 [DOI] [PubMed] [Google Scholar]
- 79.Gavin JR, Roth J, Neville DM, Meyts PD, Buell DN. 1974. Insulin-dependent regulation of insulin receptor concentrations: a direct demonstration in cell culture. PNAS 71(1):84–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Deleted in proof
- 81.Pagliassotti MJ, Holste LC, Moore MC, Neal DW, Cherrington AD. 1996. Comparison of the time courses of insulin and the portal signal on hepatic glucose and glycogen metabolism in the conscious dog. J. Clin. Investig 97(1):81–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.O-Sullivan I, Zhang W, Wasserman DH, Liew CW, Liu J, et al. 2015. FoxO1 integrates direct and indirect effects of insulin on hepatic glucose production and glucose utilization. Nat. Commun 6(1):7079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Basu A, Basu R, Shah P, Vella A, Johnson CM, et al. 2000. Effects of type 2 diabetes on the ability of insulin and glucose to regulate splanchnic and muscle glucose metabolism: evidence for a defect in hepatic glucokinase activity. Diabetes 49(2):272–83 [DOI] [PubMed] [Google Scholar]
- 84.Basu A, Basu R, Shah P, Vella A, Johnson CM, et al. 2001. Type 2 diabetes impairs splanchnic uptake of glucose but does not alter intestinal glucose absorption during enteral glucose feeding: additional evidence for a defect in hepatic glucokinase activity. Diabetes 50(6):1351–62 [DOI] [PubMed] [Google Scholar]
- 85.Pagliassotti MJ, Myers SR, Moore MC, Neal DW, Cherrington AD. 1991. Magnitude of negative arterial-portal glucose gradient alters net hepatic glucose balance in conscious dogs. Diabetes 40(12):1659–68 [DOI] [PubMed] [Google Scholar]
- 86.DiCostanzo CA, Dardevet DP, Neal DW, Lautz M, Allen E, et al. 2006. Role of the hepatic sympathetic nerves in the regulation of net hepatic glucose uptake and the mediation of the portal glucose signal. Am. J. Physiol. Endocrinol. Metab 290(1):E9–16 [DOI] [PubMed] [Google Scholar]
- 87.Adkins-Marshall B, Pagliassotti MJ, Asher JR, Connolly CC, Neal DW, et al. 1992. Role of hepatic nerves in response of liver to intraportal glucose delivery in dogs. Am. J. Physiol 262(5):E679–86 [DOI] [PubMed] [Google Scholar]
- 88.Coate KC, Kraft G, Irimia JM, Smith MS, Farmer B, et al. 2013. Portal vein glucose entry triggers a coordinated cellular response that potentiates hepatic glucose uptake and storage in normal but not high-fat/high-fructose-fed dogs. Diabetes 62(2):392–400 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Coate KC, Scott M, Farmer B, Moore MC, Smith M, et al. 2010. Chronic consumption of a high-fat/high-fructose diet renders the liver incapable of net hepatic glucose uptake. Am. J. Physiol 299(6):E887–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Coate KC, Smith MS, Shiota M, Irimia JM, Roach PJ, et al. 2013. Hepatic glucose metabolism in late pregnancy: normal versus high-fat and -fructose diet. Diabetes 62(3):753–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Kraft G, Vrba A, Scott M, Allen E, Edgerton DS, et al. 2019. Sympathetic denervation of the common hepatic artery lessens glucose intolerance in the fat- and fructose-fed dog. Diabetes 68(6):1143–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Martin IK, Weber KM, Boston RC, Alford FP, Best JD. 1988. Effects of epinephrine infusion on determinants of intravenous glucose tolerance in dogs. Am. J. Physiol 255(5):E668–73 [DOI] [PubMed] [Google Scholar]
- 93.Thorp AA, Schlaich MP. 2015. Relevance of sympathetic nervous system activation in obesity and metabolic syndrome. J. Diabetes Res 2015:341583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Masuo K, Mikami H, Ogihara T, Tuck ML. 1997. Sympathetic nerve hyperactivity precedes hyperinsulinemia and blood pressure elevation in a young, nonobese Japanese population. Am. J. Hypertens 10(1):77–83 [DOI] [PubMed] [Google Scholar]
- 95.Flaa A, Aksnes TA, Kjeldsen SE, Eide I, Rostrup M. 2008. Increased sympathetic reactivity may predict insulin resistance: an 18-year follow-up study. Metabolis 57(10):1422–27 [DOI] [PubMed] [Google Scholar]
- 96.Carnethon MR, Golden SH, Folsom AR, Haskell W, Liao D. 2003. Prospective investigation of autonomic nervous system function and the development of type 2 diabetes. Circulation 107(17):2190–95 [DOI] [PubMed] [Google Scholar]
- 97.Coopmans C, Zhou TL, Henry RMA, Heijman J, Schaper NC, et al. 2020. Both prediabetes and type 2 diabetes are associated with lower heart rate variability: the Maastricht study. Diabetes Care 43(5):1126–33 [DOI] [PubMed] [Google Scholar]
- 98.Lee DY, Lee MY, Cho JH, Kwon H, Rhee E-J, et al. 2020. Decreased vagal activity and deviation in sympathetic activity precedes development of diabetes. Diabetes Care 43(6):1336–43 [DOI] [PubMed] [Google Scholar]
- 99.Straznicky NE, Grima MT, Sari CI, Eikelis N, Lambert EA, et al. 2012. Neuroadrenergic dysfunction along the diabetes continuum: a comparative study in obese metabolic syndrome subjects. Diabetes 61(10):2506–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Huggett RJ, Scott EM, Gilbey SG, Stoker JB, Mackintosh AF, Mary DASG. 2003. Impact of type 2 diabetes mellitus on sympathetic neural mechanisms in hypertension. Circulation 108(25):3097–101 [DOI] [PubMed] [Google Scholar]
- 101.Straznicky NE, Lambert EA, Lambert GW, Masuo K, Esler MD, Nestel PJ. 2005. Effects of dietary weight loss on sympathetic activity and cardiac risk factors associated with the metabolic syndrome. J. Clin. Endocrinol. Metab 90(11):5998–6005 [DOI] [PubMed] [Google Scholar]
- 102.Vaz M, Jennings G, Turner A, Cox H, Lambert G, Esler M. 1997. Regional sympathetic nervous activity and oxygen consumption in obese normotensive human subjects. Circulation 96(10):3423–29 [DOI] [PubMed] [Google Scholar]
- 103.Grassi G, Seravalle G, Cattaneo BM, Bolla GB, Lanfranchi A, et al. 1995. Sympathetic activation in obese normotensive subjects. Hypertension 25(4):560–63 [DOI] [PubMed] [Google Scholar]
- 104.Grassi G, Dell’Oro R, Facchini A, Trevano FQ, Bolla GB, Mancia G. 2004. Effect of central and peripheral body fat distribution on sympathetic and baroreflex function in obese normotensives. J. Hypertens 22(12):2363–69 [DOI] [PubMed] [Google Scholar]
- 105.Muntzel MS, Al-Naimi OAS, Barclay A, Ajasin D. 2018. Cafeteria diet increases fat mass and chronically elevates lumbar sympathetic nerve activity in rats. Hypertension 60(6):1498–1502 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Rosario W, Singh I, Wautlet A, Patterson C, Flak J, et al. 2016. The brain-to-pancreatic islet neuronal map reveals differential glucose regulation from distinct hypothalamic regions. Diabetes 65(9):2711–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Yoon NA, Diano S. 2021. Hypothalamic glucose-sensing mechanisms. Diabetologia 64:985–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Thorens B. 2015. GLUT2, glucose sensing and glucose homeostasis. Diabetologia 58(2):221–32 [DOI] [PubMed] [Google Scholar]
- 109.Routh VH, Hao L, Santiago AM, Sheng Z, Zhou C. 2014. Hypothalamic glucose sensing: making ends meet. Front. Syst. Neurosci 8:236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Backer ID, Hussain SS, Bloom SR, Gardiner JV. 2016. Insights into the role of neuronal glucokinase. Am. J. Physiol. Endocrinol. Metab 311(1):E42–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Abi-Saab WM, Maggs DG, Jones T, Jacob R, Srihari V, et al. 2002. Striking differences in glucose and lactate levels between brain extracellular fluid and plasma in conscious human subjects: effects of hyperglycemia and hypoglycemia. J. Cereb. Blood Flow Metab 22(3):271–79 [DOI] [PubMed] [Google Scholar]
- 112.Brooks GA. 2018. The science and translation of lactate shuttle theory. Cell Metab. 27(4):757–85 [DOI] [PubMed] [Google Scholar]
- 113.Fujita S, Bohland M, Sanchez-Watts G, Watts AG, Donovan CM. 2007. Hypoglycemic detection at the portal vein is mediated by capsaicin-sensitive primary sensory neurons. Am. J. Physiol. Endocrinol. Metab 293(1):E96–101 [DOI] [PubMed] [Google Scholar]
- 114.Donovan CM, Watts AG. 2014. Peripheral and central glucose sensing in hypoglycemic detection. Physiology 29(5):314–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Garcia-Luna C, Sanchez-Watts G, Arnold M, de Lartigue G, DeWalt N, et al. 2021. The medullary targets of neurally conveyed sensory information from the rat hepatic portal and superior mesenteric veins. eNeuro 8(1):ENEURO.0419-20.2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Douglass JD, Dorfman MD, Thaler JP. 2017. Glia: silent partners in energy homeostasis and obesity pathogenesis. Diabetologia 60(2):226–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Arntfield ME, van der Kooy D. 2011. β-Cell evolution: how the pancreas borrowed from the brain. Bioessays 33(8):582–87 [DOI] [PubMed] [Google Scholar]
- 118.Rorsman P, Ashcroft FM. 2018. Pancreatic β-cell electrical activity and insulin secretion: of mice and men. Physiol. Rev 98(1):117–214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Zhou C, Teegala SB, Khan BA, Gonzalez C, Routh VH. 2018. Hypoglycemia: role of hypothalamic glucose-inhibited (GI) neurons in detection and correction. Front Physiol. 9:192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Meek TH, Nelson JT, Matsen ME, Dorfman MD, Guyenet SJ, et al. 2016. Functional identification of a neurocircuit regulating blood glucose. PNAS 113(14):E2073–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Faber CL, Matsen ME, Velasco KR, Damian V, Phan BA, et al. 2018. Distinct neuronal projections from the hypothalamic ventromedial nucleus mediate glycemic and behavioral effects. Diabetes 67(12):db180380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Stanley SA, Kelly L, Latcha KN, Schmidt SF, Yu X, et al. 2016. Bidirectional electromagnetic control of the hypothalamus regulates feeding and metabolism. Nature 531(7596):647–50 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Flak JN, Goforth P, Dell’Orco J, Sabatini PV, Li C, et al. 2020. Ventromedial hypothalamic nucleus neuronal subset regulates blood glucose independently of insulin. J. Clin. Investig 130(6):2943–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Khodai T, Nunn N, Worth AA, Feetham CH, Belle MDC, et al. 2018. PACAP neurons in the ventromedial hypothalamic nucleus are glucose inhibited and their selective activation induces hyperglycaemia. Front. Endocrinol 9:632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Spyer G, Hattersley AT, MacDonald I, Amiel S, MacLeod KM. 2000. Hypoglycaemic counter-regulation at normal blood glucose concentrations in patients with well controlled type-2 diabetes. Lancet 356(9246):1970–74 [DOI] [PubMed] [Google Scholar]
- 126.Chakera AJ, Hurst PS, Spyer G, Ogunnowo-Bada EO, Marsh WJ, et al. 2018. Molecular reductions in glucokinase activity increase counter-regulatory responses to hypoglycemia in mice and humans with diabetes. Mol. Metab 17:17–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ma Y, Ratnasabapathy R, Izzi-Engbeaya C, Nguyen-Tu M, Richardson E, et al. 2018. Hypothalamic arcuate nucleus glucokinase regulates insulin secretion and glucose homeostasis. Diabetes Obes. Metab 20(9):2246–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Hwang JJ, Jiang L, Hamza M, Rangel ES, Dai F, et al. 2017. Blunted rise in brain glucose levels during hyperglycemia in adults with obesity and T2DM. JCI Insight 2(20):e95913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Li W, Risacher SL, Huang E, Saykin AJ. 2016. Type 2 diabetes mellitus is associated with brain atrophy and hypometabolism in the ADNI cohort. Neurology 87(6):595–600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Thaler JP, Yi C-X, Schur EA, Guyenet SJ, Hwang BH, et al. 2012. Obesity is associated with hypothalamic injury in rodents and humans. J. Clin. Investig 122(1):153–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Valdearcos M, Douglass JD, Robblee MM, Dorfman MD, Stifler DR, et al. 2017. Microglial inflammatory signaling orchestrates the hypothalamic immune response to dietary excess and mediates obesity susceptibility. Cell Metab. 26(1):185–97.e3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Jais A, Solas M, Backes H, Chaurasia B, Kleinridders A, et al. 2016. Myeloid-cell-derived VEGF maintains brain glucose uptake and limits cognitive impairment in obesity. Cell 165(4):882–95 [DOI] [PubMed] [Google Scholar]
- 133.Yi C-X, Gericke M, Krüger M, Alkemade A, Kabra DG, et al. 2012. High calorie diet triggers hypothalamic angiopathy. Mol. Metab 1(1–2):95–100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Schur EA, Melhorn SJ, Oh S, Lacy JM, Berkseth KE, et al. 2015. Radiologic evidence that hypothalamic gliosis is associated with obesity and insulin resistance in humans. Obesity 23(11):2142–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.García-Cáceres C, Balland E, Prevot V, Luquet S, Woods SC, et al. 2019. Role of astrocytes, microglia, and tanycytes in brain control of systemic metabolism. Nat. Neurosci 22(1):7–14 [DOI] [PubMed] [Google Scholar]
- 136.Prevot V, Dehouck B, Sharif A, Ciofi P, Giacobini P, Clasadonte J. 2018. The versatile tanycyte: a hypothalamic integrator of reproduction and energy metabolism. Endocr. Rev 39(3):333–68 [DOI] [PubMed] [Google Scholar]
- 137.Mandelblat-Cerf Y, Ramesh RN, Burgess CR, Patella P, Yang Z, et al. 2015. Arcuate hypothalamic AgRP and putative POMC neurons show opposite changes in spiking across multiple timescales. eLife 4:e07122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Chen Y, Lin Y-C, Kuo T-W, Knight ZA. 2015. Sensory detection of food rapidly modulates arcuate feeding circuits. Cell 160(5):829–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Deem J, Faber CL, Pedersen C, Phan BA, Larsen SA, et al. 2020. Cold-induced hyperphagia requires AgRP-neuron activation in mice. eLife 9:e58764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Aponte Y, Atasoy D, Sternson SM. 2011. AGRP neurons are sufficient to orchestrate feeding behavior rapidly and without training. Nat. Neurosci 14(3):351–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Krashes MJ, Koda S, Ye C, Rogan SC, Adams AC, et al. 2011. Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J. Clin. Investig 121(4):1424–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Steculorum SM, Ruud J, Karakasilioti I, Backes H, Ruud LE, et al. 2016. AgRP neurons control systemic insulin sensitivity via myostatin expression in brown adipose tissue. Cell 165(1):125–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Krashes MJ, Lowell BB, Garfield AS. 2016. Melanocortin-4 receptor-regulated energy homeostasis. Nat. Neurosci 19(2):206–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Lee YS, Challis BG, Thompson DA, Yeo GSH, Keogh JM, et al. 2006. A POMC variant implicates β-melanocyte-stimulating hormone in the control of human energy balance. Cell Metab. 3(2):135–40 [DOI] [PubMed] [Google Scholar]
- 145.Mencarelli M, Zulian A, Cancello R, Alberti L, Gilardini L, et al. 2012. A novel missense mutation in the signal peptide of the human POMC gene: a possible additional link between early-onset type 2 diabetes and obesity. Eur. J. Hum. Genet 20(12):1290–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Dubern B, Lubrano-Berthelier C, Mencarelli M, Ersoy B, Frelut M-L, et al. 2008. Mutational analysis of the pro-opiomelanocortin gene in French obese children led to the identification of a novel deleterious heterozygous mutation located in the α-melanocyte stimulating hormone domain. Pediatr. Res 63(2):211–16 [DOI] [PubMed] [Google Scholar]
- 147.Üner AG, Keçik O, Quaresma PGF, Araujo TMD, Lee H, et al. 2019. Role of POMC and AgRP neuronal activities on glycaemia in mice. Sci. Rep 9(1):13068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Parton LE, Ye CP, Coppari R, Enriori PJ, Choi B, et al. 2007. Glucose sensing by POMC neurons regulates glucose homeostasis and is impaired in obesity. Nature 449(7159):228–32 [DOI] [PubMed] [Google Scholar]
- 149.da Silva AA, do Carmo JM, Hall JE. 2020. CNS regulation of glucose homeostasis: role of the leptin-melanocortin system. Curr. Diabetes Rep 20(7):29. [DOI] [PubMed] [Google Scholar]
- 150.Tanida M, Yamamoto N, Morgan DA, Kurata Y, Shibamoto T, Rahmouni K. 2015. Leptin receptor signaling in the hypothalamus regulates hepatic autonomic nerve activity via phosphatidylinositol 3-kinase and AMP-activated protein kinase. J. Neurosci 35(2):474–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.German JP, Thaler JP, Wisse BE, Oh-I S, Sarruf DA, et al. 2011. Leptin activates a novel CNS mechanism for insulin-independent normalization of severe diabetic hyperglycemia. Endocrinology 152(2):394–404 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Fujikawa T, Chuang J-C, Sakata I, Ramadori G, Coppari R. 2010. Leptin therapy improves insulin-deficient type 1 diabetes by CNS-dependent mechanisms in mice. PNAS 107(40):17391–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Meek TH, Wisse BE, Thaler JP, Guyenet SJ, Matsen ME, et al. 2013. BDNF action in the brain attenuates diabetic hyperglycemia via insulin-independent inhibition of hepatic glucose production. Diabetes 62(5):1512–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Meek TH, Matsen ME, Dorfman MD, Guyenet SJ, Damian V, et al. 2013. Leptin action in the ventromedial hypothalamic nucleus is sufficient, but not necessary, to normalize diabetic hyperglycemia. Endocrinology 154(9):3067–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Obici S, Zhang BB, Karkanias G, Rossetti L. 2002. Hypothalamic insulin signaling is required for inhibition of glucose production. Nat. Med 8(12):1376–82 [DOI] [PubMed] [Google Scholar]
- 156.Pocai A, Lam TKT, Gutierrez-Juarez R, Obici S, Schwartz GJ, et al. 2005. Hypothalamic KATP channels control hepatic glucose production. Nature 434(7036):1026–31 [DOI] [PubMed] [Google Scholar]
- 157.Ruud J, Steculorum SM, Brüning JC. 2017. Neuronal control of peripheral insulin sensitivity and glucose metabolism. Nat. Commun 8(1):15259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Stenvers DJ, Scheer FAJL, Schrauwen P, la Fleur SE, Kalsbeek A. 2019. Circadian clocks and insulin resistance. Nat. Rev. Endocrinol 15(2):75–89 [DOI] [PubMed] [Google Scholar]
- 159.vande Sande-Lee S, Melhorn SJ, Rachid B, Rodovalho S, De-Lima-Junior JC, et al. 2020. Radiologic evidence that hypothalamic gliosis is improved after bariatric surgery in obese women with type 2 diabetes. Int. J. Obesity 44(1):178–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Thorens B. 2011. Brain glucose sensing and neural regulation of insulin and glucagon secretion. Diabetes Obes. Metab 13(S1):82–88 [DOI] [PubMed] [Google Scholar]
- 161.Tarussio D, Metref S, Seyer P, Mounien L, Vallois D, et al. 2014. Nervous glucose sensing regulates postnatal β cell proliferation and glucose homeostasis. J. Clin. Investig 124(1):413–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Moullé VS, Tremblay C, Castell A-L, Vivot K, Ethier M, et al. 2019. The autonomic nervous system regulates pancreatic β-cell proliferation in adult male rats. Am. J. Physiol. Endocrinol. Metab 317(2):E234–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Lausier J, Diaz WC, Roskens V, LaRock K, Herzer K, et al. 2010. Vagal control of pancreatic β-cell proliferation. Am. J. Physiol. Endocrinol. Metab 299(5):E786–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Edvell A, Lindström P. 1998. Vagotomy in young obese hyperglycemic mice: effects on syndrome development and islet proliferation. Am. J. Physiol. Endocrinol. Metab 274(6):E1034–39 [DOI] [PubMed] [Google Scholar]
- 165.Kiba T, Tanaka K, Numata K, Hoshino M, Misugi K, Inoue S. 1996. Ventromedial hypothalamic lesion-induced vagal hyperactivity stimulates rat pancreatic cell proliferation. Gastroenterology 110(3):885–93 [DOI] [PubMed] [Google Scholar]
- 166.Morton GJ, Matsen ME, Bracy DP, Meek TH, Nguyen HT, et al. 2013. FGF19 action in the brain induces insulin-independent glucose lowering. J. Clin. Investig 123(11):4799–808 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Xu J, Stanislaus S, Chinookoswong N, Lau YY, Hager T, et al. 2009. Acute glucose-lowering and insulin-sensitizing action of FGF21 in insulin-resistant mouse models—association with liver and adipose tissue effects. Am. J. Physiol. Endocrinol. Metab 297(5):E1105–14 [DOI] [PubMed] [Google Scholar]
- 168.Scarlett JM, Rojas JM, Matsen ME, Kaiyala KJ, Stefanovski D, et al. 2016. Central injection of fibroblast growth factor 1 induces sustained remission of diabetic hyperglycemia in rodents. Nat. Med 22(7):800–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Scarlett JM, Muta K, Brown JM, Rojas JM, Matsen ME, et al. 2018. Peripheral mechanisms mediating the sustained anti-diabetic action of FGF1 in the brain. Diabetes 68(3):db180498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Brown JM, Scarlett JM, Matsen ME, Nguyen HT, Secher A, et al. 2019. The hypothalamic arcuate nucleus-median eminence is a target for sustained diabetes remission induced by fibroblast growth factor 1. Diabetes 68(5):1054–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Tennant KG, Lindsley SR, Kirigiti MA, True C, Kievit P. 2019. Central and peripheral administration of fibroblast growth factor 1 improves pancreatic islet insulin secretion in diabetic mouse models. Diabetes 68(7):1462–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Alonge KM, Mirzadeh Z, Scarlett JM, Logsdon AF, Brown JM, et al. 2020. Hypothalamic perineuronal net assembly is required for sustained diabetes remission induced by fibroblast growth factor 1 in rats. Nat. Metab 2:1025–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Bentsen MA, Rausch DM, Mirzadeh Z, Muta K, Scarlett JM, et al. 2020. Transcriptomic analysis links diverse hypothalamic cell types to fibroblast growth factor 1-induced sustained diabetes remission. Nat. Commun 11(1):4458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Aroda VR, Rosenstock J, Terauchi Y, Altuntas Y, Lalic NM, et al. 2019. PIONEER 1: randomized clinical trial of the efficacy and safety of oral semaglutide monotherapy in comparison with placebo in patients with type 2 diabetes. Diabetes Care 42(9):1724–32 [DOI] [PubMed] [Google Scholar]
- 175.Nauck MA, Meier JJ. 2019. Management of endocrine disease: Are all GLP-1 agonists equal in the treatment of type 2 diabetes? Eur. J. Endocrinol 181(6):R211–34 [DOI] [PubMed] [Google Scholar]
- 176.Wilding JPH, Batterham RL, Calanna S, Davies M, Gaal LFV, et al. 2021. Once-weekly semaglutide in adults with overweight or obesity. New Engl. J. Med 384:989–1002 [DOI] [PubMed] [Google Scholar]
- 177.Capozzi ME, DiMarchi RD, Tschöp MH, Finan B, Campbell JE. 2018. Targeting the incretin/glucagon system with triagonists to treat diabetes. Endocr. Rev 39(5):719–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Sachs S, Niu L, Geyer P, Jall S, Kleinert M, et al. 2021. Plasma proteome profiles treatment efficacy of incretin dual agonism in diet-induced obese female and male mice. Diabetes Obes. Metab 23(1):195–207 [DOI] [PubMed] [Google Scholar]
- 179.Finan B, Yang B, Ottaway N, Smiley DL, Ma T, et al. 2015. A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents. Nat. Med 21(1):27–36 [DOI] [PubMed] [Google Scholar]
- 180.Sheetz M, Bray R, Tham LS, Jones C, Kinsella J, Violante-Ortiz R. 2017. Results of a phase 2 study of the oxyntomodulin (OXM) analogue LY2944876 in patients with type 2 diabetes (T2DM). Paper presented at the 77th Scientific Session of the American Diabetes Association, San Diego, CA [Google Scholar]
- 181.Posch M, Tillner J, Hüser A, Teichert L, Einig C, et al. 2017. Safety, pharmacokinetics, and pharmacodynamics of the novel dual GLP-1/glucagon agonist SAR425899 in healthy subjects and diabetes patients. Paper presented at the 77th Scientific Session of the American Diabetes Association, San Diego, CA [Google Scholar]
- 182.Ramsey JJ, Kemnitz JW, Colman RJ, Cunningham D, Swick AG. 1998. Different central and peripheral responses to leptin in rhesus monkeys: brain transport may be limited. J. Clin. Endocrinol. Metab 83(9):3230–35 [DOI] [PubMed] [Google Scholar]
- 183.Tang-Christensen M, Havel PJ, Jacobs RR, Larsen PJ, Cameron JL. 1999. Central administration of leptin inhibits food intake and activates the sympathetic nervous system in rhesus macaques. J. Clin. Endocrinol. Metab 84(2):711–17 [DOI] [PubMed] [Google Scholar]
- 184.Vasandani C, Clark GO, Adams-Huet B, Quittner C, Garg A. 2017. Efficacy and safety of metreleptin therapy in patients with type 1 diabetes: a pilot study. Diabetes Care 40(5):694–97 [DOI] [PubMed] [Google Scholar]
- 185.Gubitosi-Klug RA, Braffett BH, White NH, Sherwin RS, Service FJ, et al. 2017. Risk of severe hypoglycemia in type 1 diabetes over 30 years of follow-up in the DCCT/EDIC study. Diabetes Care 40(8):1010–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Boucher-Berry C, Parton EA, Alemzadeh R. 2016. Excess weight gain during insulin pump therapy is associated with higher basal insulin doses. J. Diabetes Metab. Disord 15(1):47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Banks WA. 2016. From blood-brain barrier to blood-brain interface: new opportunities for CNS drug delivery. Nat. Rev. Drug Discov 15(4):275–92 [DOI] [PubMed] [Google Scholar]
- 188.Pires A, Fortuna A, Alves G, Falcão A. 2009. Intranasal drug delivery: How, why and what for? J. Pharm. Pharm. Sci 12(3):288–311 [DOI] [PubMed] [Google Scholar]
- 189.Smith HS, Deer TR, Staats PS, Singh V, Sehgal N, Cordner H. 2008. Intrathecal drug delivery. Pain Phys 11(Suppl. 2):S89–104 [PubMed] [Google Scholar]
- 190.Youngerman BE, Chan AK, Mikell CB, McKhann GM, Sheth SA. 2016. A decade of emerging indications: deep brain stimulation in the United States. J. Neurosurg 125(2):461–71 [DOI] [PubMed] [Google Scholar]
- 191.Tabrizi SJ, Leavitt BR, Landwehrmeyer GB, Wild EJ, Saft C, et al. 2019. Targeting huntingtin expression in patients with Huntington’s disease. New Engl. J Med 380(24):2307–16 [DOI] [PubMed] [Google Scholar]
- 192.Christine CW, Bankiewicz KS, Van Laar AD, Richardson RM, Ravina B, et al. 2019. Magnetic resonance imaging-guided phase 1 trial of putaminal AADC gene therapy for Parkinson’s disease. Ann. Neurol 85(5):704–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Ahrén B, Taborsky GJ, Porte D. 1986. Neuropeptidergic versus cholinergic and adrenergic regulation of islet hormone secretion. Diabetologia 29(12):827–36 [DOI] [PubMed] [Google Scholar]
- 194.Gautam D, Han S-J, Hamdan FF, Jeon J, Li B, et al. 2006. A critical role for β cell M3 muscarinic acetylcholine receptors in regulating insulin release and blood glucose homeostasis in vivo. Cell Metab. 3(6):449–61 [DOI] [PubMed] [Google Scholar]
- 195.Liu K, Yang L, Wang G, Liu J, Zhao X, et al. 2021. Metabolic stress drives sympathetic neuropathy within the liver. Cell Metab. 33(2):666–75.e4 [DOI] [PubMed] [Google Scholar]
- 196.Pocai A, Obici S, Schwartz GJ, Rossetti L. 2005. A brain-liver circuit regulates glucose homeostasis. Cell Metab. 1(1):53–61 [DOI] [PubMed] [Google Scholar]
- 197.Niijima A. 1983. Glucose-sensitive afferent nerve fibers in the liver and their role in food intake and blood glucose regulation. J. Autonom. Nerv. Syst 9(1):207–20 [DOI] [PubMed] [Google Scholar]
- 198.Donovan CM, Hamilton-Wessler M, Halter JB, Bergman RN. 1994. Primacy of liver glucosensors in the sympathetic response to progressive hypoglycemia. PNAS 91(7):2863–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Saberi M, Bohland M, Donovan CM. 2008. The locus for hypoglycemic detection shifts with the rate of fall in glycemia: the role of portal-superior mesenteric vein glucose sensing. Diabetes 57(5):1380–86 [DOI] [PubMed] [Google Scholar]
- 200.Berthoud H-R. 2004. Anatomy and function of sensory hepatic nerves. Anat. Rec. A Discov. Mol. Cell Evol. Biol 280(1):827–35 [DOI] [PubMed] [Google Scholar]
- 201.Niijima A. 1984. Reflex control of the autonomic nervous system activity from the glucose sensors in the liver in normal and midpontine-transected animals. J. Autonom. Nerv. Syst 10(3–4):279–85 [DOI] [PubMed] [Google Scholar]
- 202.Payne SC, Ward G, MacIsaac RJ, Hyakumura T, Fallon JB, Villalobos J. 2020. Differential effects of vagus nerve stimulation strategies on glycemia and pancreatic secretions. Physiol. Rep 8(11):e14479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Yin J, Ji F, Gharibani P, Chen JD. 2019. Vagal nerve stimulation for glycemic control in a rodent model of type 2 diabetes. Obes. Surg 29(9):2869–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Meyers EE, Kronemberger A, Lira V, Rahmouni K, Stauss HM. 2016. Contrasting effects of afferent and efferent vagal nerve stimulation on insulin secretion and blood glucose regulation. Physiol. Rep 4(4):e12718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Shikora S, Toouli J, Herrera MF, Kulseng B, Zulewski H, et al. 2013. Vagal blocking improves glycemic control and elevated blood pressure in obese subjects with type 2 diabetes mellitus. J. Obes 2013:245683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Shikora SA, Toouli J, Herrera MF, Kulseng B, Brancatisano R, et al. 2016. Intermittent vagal nerve block for improvements in obesity, cardiovascular risk factors, and glycemic control in patients with type 2 diabetes mellitus: 2-year results of the VBLOC DM2 study. Obes. Surg 26(5):1021–28 [DOI] [PubMed] [Google Scholar]
- 207.Deleted in proof
- 208.Shimazu T. 1996. Innervation of the liver and glucoregulation: roles of the hypothalamus and autonomic nerves. Nutrition 12(1):65–66 [DOI] [PubMed] [Google Scholar]
- 209.Burcelin R, Uldry M, Foretz M, Perrin C, Dacosta A, et al. 2004. Impaired glucose homeostasis in mice lacking the α1b-adrenergic receptor subtype. J. Biol. Chem 279(2):1108–15 [DOI] [PubMed] [Google Scholar]
- 210.Hering D, Lambert EA, Marusic P, Walton AS, Krum H, et al. 2013. Substantial reduction in single sympathetic nerve firing after renal denervation in patients with resistant hypertension. Hypertension 61(2):457–64 [DOI] [PubMed] [Google Scholar]
- 211.Mahfoud F, Schlaich M, Kindermann I, Ukena C, Cremers B, et al. 2011. Effect of renal sympathetic denervation on glucose metabolism in patients with resistant hypertension. Circulation 123(18):1940–46 [DOI] [PubMed] [Google Scholar]
- 212.Zhang Z, Liu K, Xiao S, Chen X. 2021. Effects of catheter-based renal denervation on glycemic control and lipid levels: a systematic review and meta-analysis. Acta Diabetol. 58(5):603–14 [DOI] [PubMed] [Google Scholar]
- 213.Kiuchi MG, Ho JK, Gavidia LML, Schlaich MP. 2019. Shaping the future of renal denervation-the relevance of sham-controlled randomized trials and recent meta-analyses. Cardiovasc. Diagnosis Ther 9(6):601–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Kapural L, Yu C, Doust MW, Gliner BE, Vallejo R, et al. 2016. Comparison of 10-kHz high-frequency and traditional low-frequency spinal cord stimulation for the treatment of chronic back and leg pain: 24-month results from a multicenter, randomized, controlled pivotal trial. Neurosurgery 79(5):667–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Linderoth B, Herregodts P, Meyerson BA. 1994. Sympathetic mediation of peripheral vasodilation induced by spinal cord stimulation. Neurosurgery 35(4):711–19 [DOI] [PubMed] [Google Scholar]
- 216.Lee KY, Bae C, Lee D, Kagan Z, Bradley K, et al. 2020. Low-intensity, kilohertz frequency spinal cord stimulation differently affects excitatory and inhibitory neurons in the rodent superficial dorsal horn. Neuroscience 428:132–39 [DOI] [PubMed] [Google Scholar]
- 217.Norrsell H, Eliasson T, Mannheimer C, Augustinsson LE, Bergh CH, et al. 1997. Effects of pacing-induced myocardial stress and spinal cord stimulation on whole body and cardiac norepinephrine spillover. Eur. Heart J 18:1890–96 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.




