Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Dec 1.
Published in final edited form as: Physiol Behav. 2015 Jun 3;152(0 0):381–388. doi: 10.1016/j.physbeh.2015.05.036

Physiological mechanisms by which non-nutritive sweeteners may impact body weight and metabolism

Mary V Burke a,b, Dana M Small a,b,c,d,e
PMCID: PMC4661139  NIHMSID: NIHMS706096  PMID: 26048305

Abstract

Evidence linking sugar-sweetened beverage (SSB) consumption to weight gain and other negative health outcomes has prompted many individuals to resort to artificial, non-nutritive sugar (NNS) substitutes as a means of reducing SSB intake. However, there is a great deal of controversy regarding the biological consequences of NNS use, with accumulating evidence suggesting that NNS consumption may influence feeding and metabolism via a variety of peripheral and central mechanisms. Here we argue that NNSs are not physiologically inert compounds and consider the potential biological mechanisms by which NNS consumption may impact energy balance and metabolic function, including actions on oral and extra-oral sweet taste receptors, and effects on metabolic hormone secretion, cognitive processes (e.g. reward learning, memory, and taste perception), and gut microbiota.

Keywords: Non-nutritive sweeteners, Feeding, Obesity, Metabolism, Body weight, Taste perception, Energy balance, Sweetening agents, Gut, Brain

1. Introduction

Soft drinks and other sugar-sweetened beverages (SSBs) represent the largest source of added dietary sugars and discretionary calories for both children and adults in the United States [1]. SSB consumption is consistently identified as a major contributor to weight gain, obesity, type 2 diabetes and metabolic syndrome (for reviews, see [26]), and compelling evidence from large epidemiological studies and randomized controlled trials linking excessive sugar consumption to adverse health consequences has prompted leading healthcare professionals to recommend population-wide reductions in the intake of added sugars [7]. One approach to promote adherence to these recommendations is to substitute non-nutritive sweeteners (NNSs) for caloric sweeteners in foods and beverages. NNSs, also referred to as artificial sweeteners, non-caloric sweeteners, and high-intensity sweeteners, are highly potent sugar substitutes that permit reductions in the energy density of foods and beverages while maintaining high palatability. However, there is a great deal of controversy regarding the health consequences of NNS consumption. Numerous reviews and meta-analyses of epidemiological and experimental data have failed to reach a consensus on this matter, concluding that NNSs have potentially beneficial [810], harmful [11,12], or trivial [13,14] effects. Here we argue that NNSs are not inert compounds and we review potential physiological mechanisms by which NNS consumption may impact energy balance and metabolic function.

2. Oral Mechanisms

The sense of taste facilitates the detection of nutrients and toxins in potential food sources [15]. Carbohydrates are an essential energy source often equated with sweet taste, the detection of which reliably elicits acceptance responses across many species. Attraction to sweetness appears to have an innate basis; newborn mammals, including humans, respond positively to the presence of sweet taste in the mouth – even in the absence of prior experience – and will ingest, rather than reject the substance [16]. However, there is some evidence that these innate responses are susceptible to conditioning, and may be molded by pre- and post-natal experiences [16,17]. With the advent of NNSs, sweet taste is now frequently experienced in the absence of an energy source. What consequence, if any, does consumption of these engineered tastants have on the biology of taste?

2.1. Prandial orosensory stimulation

Perception of sweet taste is initiated in the oral cavity through the binding of a sweet tastant to a sweet taste receptor, a G-protein coupled receptor with two 7-transmembrane subunits T1R2/T1R3 [18]. Information from activated sweet taste receptor cells is conveyed to the brain via presynaptic cells which stimulate afferent cranial nerve fibers [15]. As with other signaling networks, this system can become saturated. Prolonged or repeated exposure to a taste stimulus can lead to an acute adaptation or a reduction in responsiveness and sensitivity to the stimulus. Adaptation in the taste system has been extensively documented [1924]. This adaptation is mediated, at least in part, by physiological changes at the level of the taste receptor cell via receptor desensitization [25,26]. Repeated oral stimulation with natural, caloric sweeteners such as glucose and sucrose results in reduced responsiveness to, and perceived intensity of, both naturally and artificially sweet stimuli [20,25]. NNSs elicit the perception of sweet taste at low concentrations by binding with high affinity to one or more sites on the T1R2/T1R3 heterodimer [2729]. This raises the possibility that prolonged or repeated receptor stimulation by high affinity NNSs may augment sensory adaptation and reduce sweet taste sensitivity, which may in turn influence the acceptability of caloric and non-caloric sweeteners. However, there are many outstanding questions. First, while cross-adaptation has been found to occur consistently when the adapting solution is a sugar, studies have reported that NNSs, unlike their caloric counterparts, do not reliably produce cross-adaptation, and in some cases enhance the intensity of a sweet test stimulus, especially when the stimulus is caloric in nature [20,30]. Interpretation of such studies is complicated by the presence of a salient bitter component in many NNSs. In addition, previous studies have demonstrated that adaptation to certain compounds can induce particular taste qualities in water. Notably, Mcburney and Shick [31] and Bartoshuk [32] demonstrated that water acquires a sweet taste following adaptation to a bitter stimulus. This phenomenon may account for the sweetness enhancement effects following adaptation to NNSs. Particularly, adaptation to NNSs may produce a reduction in the sweetness of the test solution through cross-adaptation concomitant to an enhancement of sweet water taste which adds to the overall sweetness of the test solution [20,33]. It is also worth noting that NNSs are commonly consumed not in isolation but in mixtures with other sweetener types and/or with sour tastants such as citric acid. Nevertheless, NNS application to sweet taste receptor-expressing cells in vitro produces greater down-regulation of taste receptor subunits, suggesting that the magnitude of adaptation produced by sweet tastants may relate to relative binding affinity or differences in receptor site interactions [34]. Therefore, future work to characterize the effects of taste interactions and adaptation on taste sensitivity across a range of concentrations and NNS compounds at the molecular, cellular, and perceptual level is called for.

Second, the relationship between peripheral taste sensitivity, appetite, and intake is not fully understood (for review, see [35,36]), though some evidence suggests that prandial orosensory stimulation may contribute to the regulation of food intake. Studies have demonstrated that increased oral processing time (i.e., the length of time the food stimulus remains in the mouth) and increased orosensory exposure per unit of liquid or semisolid food consumption (i.e., sip or bite size) promotes satiation and decreases total intake [3739]. Accordingly, Lavin et al. [40] reported differences in intake of a test lunch after chewing sucrose-containing pastilles over 10 min compared to consumption of the same sucrose amount consumed in liquid form over 2 min. However, these studies were not restricted to oral exposure and contributions of post-ingestive mechanisms cannot be ruled out. Indeed, studies comparing oral ingestion of nutrients to gastric infusion have demonstrated that oral administration generates a slower rate of gastric emptying, which may mediate reductions in appetite and intake [41,42]. In an effort to disentangle the effects of oral and post-ingestive mechanisms on intake, a recent study by Wijlens et al. [43] simultaneously but independently manipulated oral and gastric stimulation using a modified sham feeding (MSF) procedure. Compared to a no-stimulation control condition, oral exposure via MSF over 8 min – but not 1 min – paired with simultaneous gastric loads of 100 or 800 mL (infused at a constant rate of 100 mL/min) produced comparable reductions in ad libitum energy intake. Importantly, duration of oral exposure influenced the magnitude of suppression of energy intake, whereas gastric volume load did not. These findings suggest that orosensory stimulation may be at least as effective in suppressing intake as gastric volume. Nevertheless, whether the satiating effects of longer orosensory stimulation occur in the absence of post-ingestive stimulation remains unclear.

Studies employing modified sham feeding of sweetened solutions in the absence of post-ingestive stimulation have reported mixed effects on intake. Klein et al. found that sham intake of unsweetened and sucrose-sweetened flavored solutions increased as a function of sucrose concentration, suggesting that orosensory stimulation in the absence of post-ingestive feedback may enhance intake [44]. Similar effects were reported for sham intake of solutions sweetened with the NNS aspartame, with exaggerated effects observed in women with bulimia nervosa [45]. A tempting interpretation of these findings is that orosensory stimulation with sweeteners – independent of inhibitory post-ingestive nutrient stimulation – provokes greater intake. However, these studies are limited by small sample sizes and exclusion of male participants. Moreover, only sham intake of the solutions used for orosensory stimulation was measured in these studies. As such, extrapolation of these findings to real ingestion and to intake of other sweet tastants or nutrient classes should be performed with caution.

These studies highlight the need for further investigations of oral determinants of periprandial appetite and energy regulation as well as on the possible role of NNS consumption on altering these processes. In particular, more rigorous attempts to isolate contributions of orosensory mechanisms related to taste hedonics, gustatory sensitivity, and cephalic phase responses using both nutritive and non-nutritive sweet stimuli are called for.

2.2. Persistent alterations in taste perception

Many studies have interrogated the association between gustatory perception and obesity in humans with mixed results [4652]. Several studies have reported a relationship between body mass index (BMI) and reduced suprathreshold sweet taste intensity perception [48,53], while others have found no such relationship [51,54]. Studies examining the relationship between sweet taste thresholds and body weight have reported lower thresholds in obese adolescents [49] but not in adult populations [46,51,55]. Interpretation of these conflicting findings is hindered by variations in the psychophysical approaches used to assess taste sensitivity, which limits inter-study comparisons [53]. Studies in rodents have demonstrated that high fat diet-induced obese (DIO) rats display higher sucrose preference and reduced lingual expression of the T1R3 subunit [56]. However, Chen et al. observed paradoxical effects, finding that reduced T1R3 expression in DIO rats was associated with a marked reduction in sweetener consumption and preference [57]. Notably, the latter study used the NNS saccharin, rather than caloric sucrose, to assess sweet taste preferences and consumption. Previous studies have demonstrated that rodents develop preferences for caloric sweeteners but not NNSs in the absence of oral taste signaling [5860]. Thus, if high fat diet-induced reductions in lingual T1R3 expression lead to impaired taste sensitivity, preferences for caloric and non-caloric sweeteners might be impacted differently, as NNS preferences appear to depend upon oral taste signaling whereas caloric sweetener preferences are maintained in the absence of orosensory stimulation. However, taste sensitivity was not directly assessed in these studies, so it is unclear whether the observed changes in preference reflect an altered ability to detect sweeteners.

In contrast, Roux-en-Y gastric bypass (RYGB) reduces sucrose preference and intake in rodents and decreases sucrose taste detection thresholds in bypass patients [61]. The impact of changes in taste sensitivity on weight loss or food intake following RYGB remains to be empirically determined [62], but it is possible that these alterations in taste perception may contribute to the procedure’s clinical efficacy. However, there was no difference in hedonic ratings of sucrose solutions by gastric bypass patients pre- vs. post- surgery. This may be attributable to a lack of correspondence between sucrose detection thresholds and suprathreshold taste sensitivity. Alternatively, it is possible that the scaling procedure used to measure hedonic valuation of taste stimuli lacked the sensitivity to detect individual changes in sweetener acceptability [53].

With respect to NNSs, there is some evidence that consumption might produce persistent taste alterations. Mice pups exposed to the NNS acesulfame potassium (AceK) via maternal milk demonstrated increased preference thresholds for saccharin and sucrose in adulthood [63]. These behavioral changes were accompanied by alterations in the expression of proteins involved in taste signal transduction, including increased expression of T1R2 in the tongue and reduced expression of Gα-gustducin and leptin receptor Ob-Rb in the soft palate. These findings raise the possibility that excessive exposure to NNSs may produce alterations in taste transduction physiology. In vitro evidence supports this notion, though there is some discrepancy with regards to the directionality of effects. Ren et al. found that exposure of mouse hypothalamic cells to a hypoglycemic medium resulted in increased expression of the Tas1r2 gene which encodes the T1R2 subunit of the sweet taste receptor, whereas exposure to higher concentrations of glucose produced the opposite effect [34]. The addition of sucralose into the medium produced an even more robust reduction in Tas1R2 expression, with levels decreasing approximately 300% relative to baseline, raising the possibility that effect magnitude might be related to receptor affinity. Taken together, NNS exposure appears to produce some effect on taste receptor physiology in rodents, but the direction of these effects and their impact on taste sensitivity and preference cannot be determined on the basis of the limited evidence available. It is also important to consider that findings from rodent models investigating the relationship between sweet taste and preference may not translate to humans, as rodents appear to differ in their attraction to, and preference for, certain caloric and non-caloric sweeteners [6466].

Whether NNS exposure alters taste perception in humans is unknown; however, there is some evidence to support this possibility. Neuroimaging studies have reported altered taste processing in heavy NNS users [67,68]. In particular, Small et al. reported an inverse association between NNS use and blood oxygen level dependent (BOLD) responses in the amygdala and insula in response to sucrose [68]. These findings have commonly been considered in the context of cue—reward associations (see below), but an alternative interpretation is plausible. BOLD responses in the amygdala and insula are also sensitive to taste intensity perception [69,70]. Thus, it is possible that altered activity in these regions in heavy NNS consumers reflects decreased afferent signaling and perceived intensity of sweet tastants.

3. Extra-oral Mechanisms

3.1. Extra-oral sweet taste receptors

Functional sweet taste receptors have also been identified in a variety of extra-oral tissues, including, but not limited to, the brain, pancreas, and gut. These receptors have been implicated in metabolic processes such as glucose sensing, secretion of satiety hormones, and glycemic control (for review see [71]). Intestinal enteroendocrine cells express a number of taste transduction molecules found in oral taste cells, including T1R2, T1R3 and Gα-gustducin [72]. Studies have shown that application of caloric sweeteners and NNSs to enteroendocrine cells in vitro can elicit secretion of incretin hormones glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) from these cells in vitro via a T1R3-dependent mechanism [72,73]. Additionally, both natural sweeteners and NNSs increase expression and membrane trafficking of glucose transporters SGLT-1 and GLUT2 through their actions on intestinal sweet taste receptors [73,74].

However, there are conflicting data regarding the effects of NNSs on glucoregulation and gut hormone secretion in vivo. Several studies have shown that administration of NNSs increases SGLT-1- and GLUT2-dependent glucose absorption in mice (Margolskee et al., 2007b; Mace et al., 2007). In contrast, Fujita et al. failed to observe effects of acute NNS consumption in naïve rats on hormone secretion, reporting no effect of oral gavage of NNSs on incretin release or glycemic excursion in response to an intra-peritoneal glucose tolerance test (IPOGTT) in rats [75].

Investigations in humans have similarly yielded contradictory findings on this subject. Several studies have reported that oral consumption of an NNS-sweetened preload beverage increases GLP-1 in response to an OGTT in healthy subjects [7678]. However, these studies reported discrepant effects on blood glucose. In a similar study in obese subjects, Pepino et al. found no effects of a sucralose preload on GLP-1 response to OGTT; however, increases in glucose, c-peptide, and insulin release as well as decreases in insulin clearance were observed [79]. Thus, there is a lack of consensus regarding glycemic effects of oral NNS consumption in humans. In contrast, studies employing intragastric or intraduodenal administration of NNSs have been remarkably unanimous in their findings, demonstrating no effect of NNSs on gut hormone secretion [8082]. In an attempt to parse the contributions of oral and post-ingestive mechanisms, Ford et al. measured hormonal responses to oral or modified sham ingestion of a NNS preload and found that neither method of delivery produced changes in gut hormone secretion [83].

In summary, though data from in vitro studies strongly suggest that NNSs are physiologically active in the gut, in vivo studies in rodents and humans present conflicting evidence regarding the ability of NNSs to evoke post-ingestive responses. Unfortunately, the interpretation of these in vivo studies is hindered by marked methodological variations, including type of NNS employed, route of administration, duration of exposure, body weight status, and prior exposure to NNSs.

Sweet taste receptors have also been identified in several regions of the brain, including the hypothalamus, where they may be directly involved in glucose homeostasis. As mentioned above, Ren et al. [34] reported that sweet taste receptor expression patterns in murine hypothalamic cells varied according to the glycemic index of the medium to which they were exposed, and sucralose exposure reduced the expression of Tas1R2. Recent evidence suggests that some NNSs may cross the blood brain barrier and interact with neurally-expressed sweet taste receptors in vivo [84]. It has been proposed that activation of these receptors by NNSs in nutrient-sensing brain regions may provide inaccurate feedback about extracellular glucose levels, which could in turn alter glucose homeostasis and intake [85].

3.2. Gut microbiota

The microbiome has been linked to multiple physiological roles, and growing evidence implicates gut microbiota in obesity and metabolic abnormalities [86]. The composition and function of the gut microbiota fluctuate between and within individuals and appear to be influenced by a variety of environmental factors, including diet [8789]. Alterations in gut microbiota have been linked to obesity and type II diabetes [9094]. These findings have fueled the proposition that interactions between diet and gut microbiota may promote a vulnerability to obesity and related metabolic disturbances. Studies dating back to 1980 have reported associations between NNS exposure and alterations in microbiomes or bacteria in culture [9598], raising the possibility that NNSs might exert effects on human health via interactions with gut microbiota.

In support of this notion, a recent study by Suez et al. demonstrated that consumption of NNSs produces glucose intolerance via alterations to the gut microbiota [99]. In this study, commercial saccharin added to the drinking water of mice induced glucose intolerance in both lean and high-fat diet-fed (HFD) obese mice, and antibiotic treatment reversed these metabolic derangements. Transference of intestinal microbiota from NNS-consuming mice to controls replicated the glucose intolerance phenotype. Analysis of fecal microbiota composition revealed marked differences in microbial composition and function between saccharin-exposed mice and controls. In particular, saccharin consumption produced compositional alterations in bacterial taxa that have previously been linked to type II diabetes in humans, including Bacteroides and Clostridiales. Additionally, saccharin consumption was associated with enriched microbial metabolic pathways characteristic of enhanced energy harvest, a pattern that has previously been associated with obesity in mice. These findings were also recapitulated in humans. In a large sample of subjects, NNS consumption was correlated with clinical parameters of metabolic syndrome including body weight, fasting blood glucose, and impaired glucose tolerance. Gut bacterial populations in NNS consumers were distinct from non-consumers, and this was not accounted for by differences in BMI. Moreover, when placed on a regimen of controlled high saccharin intake, normal non-consumers of NNSs exhibited elevated blood glucose levels and altered gut microbiota composition after just 5–7 days. Transference of microbiota from these saccharin-exposed human subjects to lean NNS-naïve mice induced significant glucose intolerance, suggesting a causal role for saccharin-induced microbiota alterations. It should be noted, however, that these effects were observed in just four of seven participants. Moreover, the NNS regimen administered in both the rodent and human interventions represents the FDA’s maximal acceptable daily intake of commercial saccharin – an amount that exceeds the average intake of sugar substitutes for even the heaviest users. As such, these findings should be interpreted with caution until they can be replicated with a more ecologically relevant dose in a larger-scale, randomized controlled trial. Nevertheless, these findings introduce an unexpected and heretofore unexplored mechanism by which NNSs may produce detrimental metabolic consequences.

3.3. Uncoupling taste and post-ingestive consequences

An inability of NNSs to reliably elicit post-ingestive responses may not equate to physiological inertia. An alternative interpretation invoking the principles of classical Pavlovian conditioning postulates that repeated consumption of NNSs might disrupt energy regulation by degrading conditioned associations between sweet taste and its post-oral consequences [100]. Gustatory signals originating from foods are linked with post-ingestive metabolic consequences, thereby forging a conditioned association between orosensory cue and biological outcome. As a result of these learned associations, orosensory signals can guide ingestive behavior in an adaptive manner, promoting intake of foods associated with positive biological outcomes (e.g. energy absorption and utilization) and suppressing intake of foods associated with negative biological outcomes (e.g. malaise) [101]. These conditioned sensory cues can independently elicit a series of anticipatory pre-absorptive physiological responses – such as salivation and gastric acid secretion, secretion of metabolic hormones such as insulin, leptin, and ghrelin, and thermogenesis – referred to as cephalic phase responses (CPRs) [102]. CPRs serve to facilitate digestion, absorption, and metabolism [103], and may also dynamically modulate appetite and satiety in a manner that serves to protect homeostasis [104]. In natural settings, sweet taste reliably predicts the presence of carbohydrates that serve as an energy source. By virtue of this association, sweet tastes in the mouth would be expected to elicit CPRs that signal and prepare for the impending arrival of carbohydrates in the gut, and the available body of evidence strongly supports this suggestion (for review, see [103]). When a conditioned stimulus (CS) is repeatedly presented in the absence of the unconditioned stimulus (US), it loses its predictive value and its ability to elicit the conditioned response. Based on this principle, it has been suggested that repeated experience with NNSs, which provide the conditioned orosensory stimulus of sweet taste in the absence of post-ingestive nutritive consequences, might lead to a suppression of conditioned CPRs [105]. Further, this suppression may persist even when sweet taste is once again accompanied by caloric content due to a devaluation of the CS [106]. This CS-US decoupling could impair the ability of sweet taste to predict energy availability and appropriately guide intake.

Supporting evidence for this hypothesis was obtained in rodents exposed to inconsistent pairings of sweet taste and calories. Rats with a history of exposure to NNS-sweetened foods and liquids showed increased weight gain, energy intake, and adiposity compared to control rats exposed to similar diets sweetened with glucose, for whom sweet taste consistently matched caloric content [107,108]. Moreover, NNS-exposed rats displayed an impaired ability to compensate for additional calories consumed in a novel, calorically-sweetened pre-meal by reducing intake at subsequent feeding opportunities, and also demonstrated blunted thermic responses to caloric sweet meals [107]. A similar study corroborated the effects of NNS exposure on body weight and energy intake, and further demonstrated that, compared to glucose-exposed controls, NNS-exposed rats displayed increased blood glucose and decreased GLP-1 in response to an OGTT [109]. Consistent with the notion that NNS experience interferes with ability of sweet orosensory cues to elicit CPRs, these glycemic impairments were not observed when glucose was administered by intragastric gavage, bypassing the oral cavity. Taken together, these data support the notion that NNS consumption may disrupt energy homeostasis by interfering with the predictive relationship between sweet taste and post-ingestive outcomes. It is important to note that these findings contradict earlier work by Berthoud et al. [110], who found that ingestion of a saccharin solution reliably elicits a rapid cephalic phase insulin response (CPIR) in rats which was not easily extinguished, suggesting that this response may possess an unconditioned component. However, CPIR was measured over only 10 trials in this study, which may be insufficient to produce extinction.

3.4. Uncoupling taste and reward

Whether prolonged NNS exposure alters physiological responses to caloric sweeteners in humans remains unclear. However, neuroimaging studies have provided some evidence that NNS consumption may alter the relationship between sweet taste and reward. Frank et al. found that tasting sucrose and sucralose activated common taste pathways, but absolute brain response after sucrose was stronger than for sucralose [111]. Furthermore, sucrose – but not sucralose – recruited strong connectivity between primary taste pathways and midbrain reward circuits in relation to behavioral pleasantness ratings. These findings were confirmed by Smeets et al., who reported that striatal activation was greater in response to a naturally sweetened solution, whereas NNSs produced greater amygdala activation [112]. These data suggest that the brain is capable of distinguishing between caloric sweeteners and NNSs even though both are perceived as similarly sweet. This might be related to post-ingestive responses associated with caloric vs. non-caloric sweeteners. Alternatively, it may reflect differences in taste profile between NNSs and natural sweeteners. Though these studies controlled the perceived sweetness intensity of the two stimuli, they did not assess whether subjects were able to discriminate between the two sweetener types on the basis of other taste characteristics. Thus, this possibility cannot be excluded.

Recent studies have reported altered processing of caloric and non-caloric sweet taste stimuli in habitual NNS consumers. A study by Green and Murphy revealed that sweet taste elicited greater activation of reward-related brain regions in self-reported diet soda drinkers compared to non-diet soda drinkers, and that habitual diet soda drinkers did not demonstrate differential brain responses to nutritive and NNSs [67]. These results suggest that regular NNS consumption may be associated with changes in the reward experienced from caloric and non-caloric sweeteners. Further, Rudenga and Small reported that frequency of NNS use is negatively associated with brain response to sucrose in the amygdala and insula [68]. The amygdala is critically involved in flavor-nutrient conditioning in rodents [113115], and in the central representation of the reward value of sensory-predictive cues [116]. The amygdala is also activated to a greater extent by sensory cues that predict the immediate arrival of caloric vs. non-caloric solutions [117]. The insula has also been implicated in integrating orosensory and homeostatic signals [118], and was found to interact more strongly with feeding-related regions such as the hypothalamus and striatum in response to nutritive as opposed to non-nutritive taste stimuli [119]. Based on these observations, it might be speculated that blunted amygdala and insula response to sucrose in habitual NNS consumers may reflect a reduction in the predictive value of sucrose and a decoupling of sensory cue from reward, though future work in which NNS exposure is manipulated experimentally is needed to bolster this interpretation.

Contrary to these findings, a recent clinical trial reported that repeated consumption of NNS- and sucrose-sweetened versions of a drink did not alter the reward value of either version [120]. In this study, subjects consumed fixed portions of sucrose- and NNS-sweetened versions of a beverage that were distinguishable by means of colored labels. Each version was offered 10 times in semi-random order over a 20-day conditioning period. Before and after this conditioning phase, the reward value of each drink was assessed using behavioral tasks measuring implicit and explicit wanting, liking, and expected satiety. Additionally, BOLD response to NNS- and sucrose-sweetened liquids was measured before and after conditioning. Outcomes of both behavioral tasks and fMRI data indicated that conditioning with repeated exposures did not affect the reward value of either version of the drink. These findings suggest that the learned relationships between sweet taste and reward might be relatively resistant to extinction, though it is possible that the limited exposure to NNS in this study was insufficient to degrade conditioned associations that developed over a lifetime.

3.5. Cognitive influences

In humans, beliefs and expectations about the caloric content of a food or beverage may influence brain function and metabolism. For example, consuming the same milkshake produces greater decreases in circulating ghrelin when participants believe that it is “indulgent” as opposed to “sensible” [121]. Likewise, a flavored beverage produces greater hypothalamic response when preceded by the label “treat” compared to the label “healthful”, with the overall pattern of brain activation associated with the treat label more closely resembling response to a prototypical treat (milkshake) [122]. There is also evidence that consuming food believed to be low in calorie content can produce reduced satiety and lead to “rebound” eating [123,124], though not all studies support such an effect [125,126]. These findings raise the possibility that NNSs might influence intake and metabolism simply by creating the impression that a food or beverage is less caloric than its actual energy content.

A second way in which NNS may impact cognition is by passing through the blood brain barrier to produce deleterious effects on brain tissue. In a recent study, Cong et al. [84] demonstrated that orally ingested AceK is able to cross the mouse blood brain barrier and accumulate in brain tissue. Chronic consumption (40 weeks) of this NNS produced neurosynaptic- and metabolism-related genomic and proteomic abnormalities in the hippocampus, including reduced protein expression of the T1R3 subunit and the glucose transporter Glut1, functional ATP depletion, and dysregulation of proteins involved in cell growth and survival. They further demonstrated that the chronically exposed animals showed signs of impaired hippocampal-dependent learning, as assessed with the Morris Water Maze. AceK-treated T1R3 knockout mice failed to exhibit these cognitive impairments, suggesting an integral role for the T1R3 subunit. It should be noted that while the doses of AceK employed in this study were within the acceptable daily intake range for AceK set by the FDA, daily AceK intake by the experimental animals was likely higher than what most humans experience on average. Moreover, AceK may be unique among NNSs in its ability to cross the blood brain barrier, as there is no evidence that other NNSs accumulate in the blood or are absorbed from the intestine. Thus, the epidemiological relevance of these findings may be limited to suprathreshold levels of intake of AceK.

Nevertheless, the possibility that some NNSs might cross the blood brain barrier to produce hippocampal damage may be relevant for consideration of the role of NNSs in energy balance and metabolism. The hippocampus is sensitive to satiety signals in humans and animals [127,128], and there is a strong body of evidence demonstrating detrimental effects of high fat and high sugar diets on hippocampal function. Excess consumption of high fructose corn syrup, and to a lesser extent, sucrose, impaired hippocampal-dependent spatial learning and memory in rats [129]. Chronic exposure to a high-fat and refined sugar (HFS) diet has also been shown to reduce hippocampal levels of brain-derived neurotrophic factor, synaptic plasticity, and spatial learning performance in rodents [130]. In humans, self-reported (HFS) diet is associated with poorer performance on hippocampal-dependent memory tasks, as well as reduced accuracy in tracking prior food intake and diminished sensitivity to interoceptive hunger and satiety signals [131].

There is also evidence that hippocampal dysfunction may promote weight gain. Rats with neurotoxic lesions of the hippocampus exhibit excessive ad libitum feeding and weight gain [132]. Additionally, hippocampal lesions impaired the ability of rats to use interoceptive cues arising from 0 and 24 h food deprivation as discriminative signals [133]. In particular, lesioned rats exhibited increased appetitive responding in the presence of energy state cues that signaled non-reinforcement, whereas responding to reinforced cues remained intact. These data suggest that impaired discriminative responding following hippocampal lesions results from an inability to inhibit activation of the memory of food reinforcement. As a consequence, satiety cues may be less able to suppress the ability of food-related cues to evoke the memory of food reinforcement and excite appetitive and consummatory behavior [133135]. Taken together, these data suggest that dietary factors such as excess consumption of fats, refined sugars, and perhaps also of certain NNSs that cross the blood brain barrier and disrupt hippocampal function may impair sensitivity to interoceptive signals, dysregulate appetitive behavior, and thereby promote food intake.

4. Conclusions

The addition of NNSs to foods and beverages has become increasingly pervasive in the modern food environment. Although the existing literature on the biological consequences of NNSs, particularly in humans, remains highly controversial, amassing evidence suggests that NNSs are not physiologically inert, and may influence feeding and metabolism through a variety of peripheral and central mechanisms. The determinants of energy homeostasis and ingestive behavior are exceptionally diverse and complex. Contributions of oral, gastrointestinal, endocrine, and neural mechanisms, and the manner in which these systems interact to regulate energy balance, remain insufficiently understood. Conclusions about the impact of NNSs on human health are made within the context of the level of current understanding. As understanding advances so too should the consideration of the impact of NNS use on human health.

Highlights.

  • The health consequences of non-nutritive sweetener (NNS) use are controversial.

  • Evidence suggests NNSs may impact energy balance and metabolic function.

  • We review the evidence for central and peripheral physiological effects of NNSs.

  • Limitations of the current literature base and health implications are discussed.

Acknowledgments

The authors wish to acknowledge support from the National Institutes of Health (NIDCD R01 DC006706, NIDDK R01 DK095579, NCI R01 CA180030), NIH Training Grant T32 NS41228, and the NIH Predoctoral Training Program in the Neurosciences.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Huth PJ, Fulgoni VL, Keast DR, Park K, Auestad N. Major food sources of calories, added sugars, and saturated fat and their contribution to essential nutrient intakes in the U.S. diet: data from the national health and nutrition examination survey (2003–2006) Nutr J. 2013;12:116. doi: 10.1186/1475-2891-12-116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Malik VS, Schulze MB, Hu FB. Intake of sugar-sweetened beverages and weight gain: a systematic review. [accessed May 4, 2014];Am J Clin Nutr. 2006 84:274–88. doi: 10.1093/ajcn/84.1.274. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3210834&tool=pmcentrez&rendertype=abstract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Malik VS, Pan A, Willett WC, Hu FB. Sugar-sweetened beverages and weight gain in children and adults: a systematic review and meta-analysis. Am J Clin Nutr. 2013;98:1084–102. doi: 10.3945/ajcn.113.058362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Vartanian LR, Schwartz MB, Brownell KD. Effects of soft drink consumption on nutrition and health: a systematic review and meta-analysis. Am J Public Health. 2007;97:667–75. doi: 10.2105/AJPH.2005.083782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hu FB, Malik VS. Sugar-sweetened beverages and risk of obesity and type 2 diabetes: epidemiologic evidence. Physiol Behav. 2010;100:47–54. doi: 10.1016/j.physbeh.2010.01.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Malik VS, Popkin BM, Bray GA, Després JP, Willett WC, Hu FB. Sugar-sweetened beverages and risk of metabolic syndrome and type 2 diabetes: a meta-analysis. Diabetes Care. 2010;33:2477–83. doi: 10.2337/dc10-1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Johnson RK, Appel LJ, Brands M, Howard BV, Lefevre M, Lustig RH, et al. Dietary sugars intake and cardiovascular health: a scientific statement from the American Heart Association. Circulation. 2009;120:1011–20. doi: 10.1161/CIRCULATIONAHA.109.192627. [DOI] [PubMed] [Google Scholar]
  • 8.Fernstrom JD. Non-Nutritive Sweeteners and Obesity. Annu Rev Food Sci Technol. 2014 doi: 10.1146/annurev-food-022814-015635. [DOI] [PubMed] [Google Scholar]
  • 9.Rolls BJ. Effects of intense sweeteners on hunger, food intake, and body weight: a review. Am J Clin Nutr. 1991;53:872–878. doi: 10.1093/ajcn/53.4.872. [DOI] [PubMed] [Google Scholar]
  • 10.Mattes RD, Popkin BM. Nonnutritive sweetener consumption in humans: effects on appetite and food intake and their putative mechanisms. Am J Clin Nutr. 2009;89:1–14. doi: 10.3945/ajcn.2008.26792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Swithers SE. Artificial sweeteners produce the counterintuitive effect of inducing metabolic derangements. Trends Endocrinol Metab. 2013;24:431–41. doi: 10.1016/j.tem.2013.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Fowler SP, Williams K, Resendez RG, Hunt KJ, Hazuda HP, Stern MP. Fueling the obesity epidemic? Artificially sweetened beverage use and long-term weight gain. Obesity (Silver Spring) 2008;16:1894–1900. doi: 10.1038/oby.2008.284. [DOI] [PubMed] [Google Scholar]
  • 13.Renwick AG, Molinary SV. Sweet-taste receptors, low-energy sweeteners, glucose absorption and insulin release. Br J Nutr. 2010;104:1415–20. doi: 10.1017/S0007114510002540. [DOI] [PubMed] [Google Scholar]
  • 14.Benton D. Can artificial sweeteners help control body weight and prevent obesity? Nutr Res Rev. 2005;18:63–76. doi: 10.1079/NRR200494. [DOI] [PubMed] [Google Scholar]
  • 15.Yarmolinsky DA, Zuker CS, Ryba NJP. Common Sense about Taste: From Mammals to Insects. Cell. 2009;139:234–244. doi: 10.1016/j.cell.2009.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ventura AK, Mennella JA. Innate and learned preferences for sweet taste during childhood. Curr Opin Clin Nutr Metab Care. 2011;14:379–84. doi: 10.1097/MCO.0b013e328346df65. [DOI] [PubMed] [Google Scholar]
  • 17.Beauchamp GK, Mennella JA. Early flavor learning and its impact on later feeding behavior. J Pediatr Gastroenterol Nutr. 2009;48(Suppl 1):S25–30. doi: 10.1097/MPG.0b013e31819774a5. [DOI] [PubMed] [Google Scholar]
  • 18.Zhao GQ, Zhang Y, Hoon MA, Chandrashekar J, Erlenbach I, Ryba NJP, et al. The Receptors for Mammalian Sweet and Umami Taste. Cell. 2003;115:255–266. doi: 10.1016/S0092-8674(03)00844-4. [DOI] [PubMed] [Google Scholar]
  • 19.Schiffman SS, Pecore SD, Booth BJ, Losee ML, Carr BT, Sattely-Miller E, et al. Adaptation of sweeteners in water and in tannic acid solutions. Physiol Behav. 1994;55:547–559. doi: 10.1016/0031-9384(94)90116-3. [DOI] [PubMed] [Google Scholar]
  • 20.Schiffman S. Multiple receptor sites mediate sweetness: Evidence from cross adaptation. Pharmacol Biochem Behav. 1981;15:377–388. doi: 10.1016/0091-3057(81)90266-5. [DOI] [PubMed] [Google Scholar]
  • 21.Gent JF. An exponential model for adaptation in taste. [accessed February 6, 2015];Sens Processes. 1979 3:303–16. http://www.ncbi.nlm.nih.gov/pubmed/263646. [PubMed] [Google Scholar]
  • 22.Ganzevles PG, Kroeze JH. Effects of adaptation and cross-adaptation to common ions on sourness intensity. [accessed February 6, 2015];Physiol Behav. 1987 40:641–6. doi: 10.1016/0031-9384(87)90111-9. http://www.ncbi.nlm.nih.gov/pubmed/3671530. [DOI] [PubMed] [Google Scholar]
  • 23.DuBose CN, Meiselman HL, Hunt DA, Waterman D. Incomplete taste adaptation to different concentrations of salt and sugar solutions. Percept Psychophys. 1977;21:183–186. doi: 10.3758/BF03198723. [DOI] [Google Scholar]
  • 24.Alsiö J, Olszewski PK, Levine AS, Schiöth HB. Feed-forward mechanisms: Addiction-like behavioral and molecular adaptations in overeating. [accessed October 2, 2013];Front Neuroendocrinol. 2012 33:127–139. doi: 10.1016/j.yfrne.2012.01.002. http://www.sciencedirect.com/science/article/pii/S0091302212000039. [DOI] [PubMed] [Google Scholar]
  • 25.Tonosaki K, Funakoshi M. Cross-adapted sugar responses in the mouse taste cell. [accessed February 6, 2015];Comp Biochem Physiol A Comp Physiol. 1989 92:181–3. doi: 10.1016/0300-9629(89)90149-7. http://www.ncbi.nlm.nih.gov/pubmed/2566408. [DOI] [PubMed] [Google Scholar]
  • 26.Tonosaki K. Cross-adapted salt responses in the mouse taste cell. [accessed February 6, 2015];Brain Res. 1992 574:338–40. doi: 10.1016/0006-8993(92)90837-y. http://www.ncbi.nlm.nih.gov/pubmed/1638406. [DOI] [PubMed] [Google Scholar]
  • 27.Xu H, Staszewski L, Tang H, Adler E, Zoller M, Li X. Different functional roles of T1R subunits in the heteromeric taste receptors. Proc Natl Acad Sci U S A. 2004;101:14258–63. doi: 10.1073/pnas.0404384101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Nie Y, Vigues S, Hobbs JR, Conn GL, Munger SD. Distinct contributions of T1R2 and T1R3 taste receptor subunits to the detection of sweet stimuli. Curr Biol. 2005;15:1948–52. doi: 10.1016/j.cub.2005.09.037. [DOI] [PubMed] [Google Scholar]
  • 29.Farkas A, Híd J. The black agonist-receptor model of high potency sweeteners, and its implication to sweetness taste and sweetener design. J Food Sci. 2011;76:S465–8. doi: 10.1111/j.1750-3841.2011.02353.x. [DOI] [PubMed] [Google Scholar]
  • 30.Froloff N, Lloret E, Martinez JM, Faurion A. Cross-adaptation and molecular modeling study of receptor mechanisms common to four taste stimuli in humans. [accessed February 6, 2015];Chem Senses. 1998 23:197–206. doi: 10.1093/chemse/23.2.197. http://www.ncbi.nlm.nih.gov/pubmed/9589167. [DOI] [PubMed] [Google Scholar]
  • 31.McBurney DH, Shick TR. Taste and water taste of twenty-six compounds for man. Percept Psychophys. 1971;10:249–252. doi: 10.3758/BF03212815. [DOI] [Google Scholar]
  • 32.Bartoshuk LM. Water taste in man. Percept Psychophys. 1968;3:69–72. doi: 10.3758/BF03212715. [DOI] [Google Scholar]
  • 33.MCBURNEY D, BARTOSHUK L. Interactions between stimuli with different taste qualities☆. Physiol Behav. 1973;10:1101–1106. doi: 10.1016/0031-9384(73)90194-7. [DOI] [PubMed] [Google Scholar]
  • 34.Ren X, Zhou L, Terwilliger R, Newton SS, de Araujo IE. Sweet taste signaling functions as a hypothalamic glucose sensor. Front Integr Neurosci. 2009;3:12. doi: 10.3389/neuro.07.012.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Donaldson LF, Bennett L, Baic S, Melichar JK. Taste and weight: is there a link? Am J Clin Nutr. 2009;90:800S–803S. doi: 10.3945/ajcn.2009.27462Q. [DOI] [PubMed] [Google Scholar]
  • 36.Low YQ, Lacy K, Keast R. The role of sweet taste in satiation and satiety. Nutrients. 2014;6:3431–50. doi: 10.3390/nu6093431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Weijzen PLG, Smeets PAM, de Graaf C. Sip size of orangeade: effects on intake and sensory-specific satiation. Br J Nutr. 2009;102:1091–7. doi: 10.1017/S000711450932574X. [DOI] [PubMed] [Google Scholar]
  • 38.Bolhuis DP, Lakemond CMM, de Wijk RA, Luning PA, de Graaf C. Consumption with large sip sizes increases food intake and leads to underestimation of the amount consumed. PLoS One. 2013;8:e53288. doi: 10.1371/journal.pone.0053288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zijlstra N, de Wijk RA, Mars M, Stafleu A, de Graaf C. Effect of bite size and oral processing time of a semisolid food on satiation. Am J Clin Nutr. 2009;90:269–75. doi: 10.3945/ajcn.2009.27694. [DOI] [PubMed] [Google Scholar]
  • 40.Lavin JH, French SJ, Ruxton CHS, Read NW. An investigation of the role of oro-sensory stimulation in sugar satiety? Int J Obes Relat Metab Disord. 2002;26:384–8. doi: 10.1038/sj.ijo.0801829. [DOI] [PubMed] [Google Scholar]
  • 41.Cecil J, Francis J, Read N. Comparison of the Effects of a High-Fat and High-Carbohydrate Soup Delivered Orally and Intragastrically on Gastric Emptying, Appetite, and Eating Behaviour. Physiol Behav. 1999;67:299–306. doi: 10.1016/S0031-9384(99)00069-4. [DOI] [PubMed] [Google Scholar]
  • 42.Cecil JE, Francis J, Read NW. Relative contributions of intestinal, gastric, oro-sensory influences and information to changes in appetite induced by the same liquid meal. Appetite. 1998;31:377–90. doi: 10.1006/appe.1998.0177. [DOI] [PubMed] [Google Scholar]
  • 43.Wijlens AGM, Erkner A, Alexander E, Mars M, Smeets PAM, de Graaf C. Effects of oral and gastric stimulation on appetite and energy intake. Obesity (Silver Spring) 2012;20:2226–32. doi: 10.1038/oby.2012.131. [DOI] [PubMed] [Google Scholar]
  • 44.Klein DA, Schebendach JS, Devlin MJ, Smith GP, Walsh BT. Intake, sweetness and liking during modified sham feeding of sucrose solutions. Physiol Behav. 2006;87:602–6. doi: 10.1016/j.physbeh.2005.12.009. [DOI] [PubMed] [Google Scholar]
  • 45.Klein DA, Schebendach JE, Brown AJ, Smith GP, Walsh BT. Modified sham feeding of sweet solutions in women with and without bulimia nervosa. Physiol Behav. 2009;96:44–50. doi: 10.1016/j.physbeh.2008.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Skrandies W, Zschieschang R. Olfactory and gustatory functions and its relation to body weight. Physiol Behav. 2015;142C:1–4. doi: 10.1016/j.physbeh.2015.01.024. [DOI] [PubMed] [Google Scholar]
  • 47.Bartoshuk LM, Duffy VB, Hayes JE, Moskowitz HR, Snyder DJ. Psychophysics of sweet and fat perception in obesity: problems, solutions and new perspectives. Philos Trans R Soc Lond B Biol Sci. 2006;361:1137–48. doi: 10.1098/rstb.2006.1853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Sartor F, Donaldson LF, Markland DA, Loveday H, Jackson MJ, Kubis H-P. Taste perception and implicit attitude toward sweet related to body mass index and soft drink supplementation. [accessed January 9, 2014];Appetite. 2011 57:237–246. doi: 10.1016/j.appet.2011.05.107. http://www.sciencedirect.com/science/article/pii/S0195666311002649. [DOI] [PubMed] [Google Scholar]
  • 49.Overberg J, Hummel T, Krude H, Wiegand S. Differences in taste sensitivity between obese and non-obese children and adolescents. Arch Dis Child. 2012;97:1048–52. doi: 10.1136/archdischild-2011-301189. [DOI] [PubMed] [Google Scholar]
  • 50.Tucker RM, Edlinger C, Craig BA, Mattes RD. Associations between BMI and fat taste sensitivity in humans. Chem Senses. 2014;39:349–57. doi: 10.1093/chemse/bju006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Frijters JE, Rasmussen-Conrad EL. Sensory discrimination, intensity perception, and affective judgment of sucrose-sweetness in the overweight. J Gen Psychol. 1982;107:233–47. doi: 10.1080/00221309.1982.9709931. [DOI] [PubMed] [Google Scholar]
  • 52.Pepino MY, Finkbeiner S, Beauchamp GK, Mennella JA. Obese women have lower monosodium glutamate taste sensitivity and prefer higher concentrations than do normal-weight women. Obesity (Silver Spring) 2010;18:959–65. doi: 10.1038/oby.2009.493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bartoshuk LM, Duffy VB, Hayes JE, Moskowitz HR, Snyder DJ. Psychophysics of sweet and fat perception in obesity: problems, solutions and new perspectives. Philos Trans R Soc Lond B Biol Sci. 2006;361:1137–48. doi: 10.1098/rstb.2006.1853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Pepino MY, Mennella JA. Habituation to the pleasure elicited by sweetness in lean and obese women. [accessed January 9, 2014];Appetite. 2012 58:800–805. doi: 10.1016/j.appet.2012.01.026. http://www.sciencedirect.com/science/article/pii/S019566631200027X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Malcolm R, O’Neil PM, Hirsch AA, Currey HS, Moskowitz G. Taste hedonics and thresholds in obesity. [accessed May 6, 2015];Int J Obes. 1980 4:203–12. http://www.ncbi.nlm.nih.gov/pubmed/7419338. [PubMed] [Google Scholar]
  • 56.Zhang X, Wang Y, Long Y, Wang L, Li Y, Gao F, et al. Alteration of sweet taste in high-fat diet induced obese rats after 4 weeks treatment with exenatide. Peptides. 2013;47:115–23. doi: 10.1016/j.peptides.2013.07.015. [DOI] [PubMed] [Google Scholar]
  • 57.Chen K, Yan J, Suo Y, Li J, Wang Q, Lv B. Nutritional status alters saccharin intake and sweet receptor mRNA expression in rat taste buds. Brain Res. 2010;1325:53–62. doi: 10.1016/j.brainres.2010.02.026. [DOI] [PubMed] [Google Scholar]
  • 58.Ren X, Ferreira JG, Zhou L, Shammah-Lagnado SJ, Yeckel CW, de Araujo IE. Nutrient selection in the absence of taste receptor signaling. J Neurosci. 2010;30:8012–23. doi: 10.1523/JNEUROSCI.5749-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Damak S, Rong M, Yasumatsu K, Kokrashvili Z, Varadarajan V, Zou S, et al. Detection of sweet and umami taste in the absence of taste receptor T1r3. Science. 2003;301:850–3. doi: 10.1126/science.1087155. [DOI] [PubMed] [Google Scholar]
  • 60.de Araujo IE, Oliveira-Maia AJ, Sotnikova TD, Gainetdinov RR, Caron MG, Nicolelis MAL, et al. Food reward in the absence of taste receptor signaling. Neuron. 2008;57:930–41. doi: 10.1016/j.neuron.2008.01.032. [DOI] [PubMed] [Google Scholar]
  • 61.Bueter M, Miras AD, Chichger H, Fenske W, Ghatei MA, Bloom SR, et al. Alterations of sucrose preference after Roux-en-Y gastric bypass. Physiol Behav. 2011;104:709–21. doi: 10.1016/j.physbeh.2011.07.025. [DOI] [PubMed] [Google Scholar]
  • 62.Mathes CM, Spector AC. Food selection and taste changes in humans after Roux-en-Y gastric bypass surgery: a direct-measures approach. Physiol Behav. 2012;107:476–83. doi: 10.1016/j.physbeh.2012.02.013. [DOI] [PubMed] [Google Scholar]
  • 63.Li WL, Chen ML, Liu SS, Li GL, Gu TY, Liang P, et al. Sweet preference modified by early experience in mice and the related molecular modulations on the peripheral pathway. J Mol Neurosci. 2013;51:225–36. doi: 10.1007/s12031-013-0011-y. [DOI] [PubMed] [Google Scholar]
  • 64.Glendinning JI, Breinager L, Kyrillou E, Lacuna K, Rocha R, Sclafani A. Differential effects of sucrose and fructose on dietary obesity in four mouse strains. Physiol Behav. 2010;101:331–43. doi: 10.1016/j.physbeh.2010.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sclafani A, Abrams M. Rats show only a weak preference for the artificial sweetener aspartame. [accessed May 8, 2015];Physiol Behav. 1986 37:253–6. doi: 10.1016/0031-9384(86)90228-3. http://www.ncbi.nlm.nih.gov/pubmed/3737735. [DOI] [PubMed] [Google Scholar]
  • 66.Bachmanov AA, Tordoff MG, Beauchamp GK. Sweetener preference of C57BL/6ByJ and 129P3/J mice. [accessed May 8, 2015];Chem Senses. 2001 26:905–13. doi: 10.1093/chemse/26.7.905. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3718299&tool=pmcentrez&rendertype=abstract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Green E, Murphy C. Altered processing of sweet taste in the brain of diet soda drinkers. Physiol Behav. 2012;107:560–7. doi: 10.1016/j.physbeh.2012.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Rudenga KJ, Small DM. Amygdala response to sucrose consumption is inversely related to artificial sweetener use. Appetite. 2012;58:504–507. doi: 10.1016/j.appet.2011.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Small DM, Gregory MD, Mak YE, Gitelman D, Mesulam MM, Parrish T. Dissociation of neural representation of intensity and affective valuation in human gustation. Neuron. 2003;39:701–711. doi: 10.1016/S0896-6273(03)00467-7. [DOI] [PubMed] [Google Scholar]
  • 70.Spetter MS, Smeets PAM, de Graaf C, Viergever MA. Representation of sweet and salty taste intensity in the brain. Chem Senses. 2010;35:831–840. doi: 10.1093/chemse/bjq093. [DOI] [PubMed] [Google Scholar]
  • 71.Laffitte A, Neiers F, Briand L. Functional roles of the sweet taste receptor in oral and extraoral tissues. Curr Opin Clin Nutr Metab Care. 2014;17:379–85. doi: 10.1097/MCO.0000000000000058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jang HJ, Kokrashvili Z, Theodorakis MJ, Carlson OD, Kim BJ, Zhou J, et al. Gut-expressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proc Natl Acad Sci U S A. 2007;104:15069–74. doi: 10.1073/pnas.0706890104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Margolskee RF, Dyer J, Kokrashvili Z, Salmon KSH, Ilegems E, Daly K, et al. T1R3 and gustducin in gut sense sugars to regulate expression of Na+-glucose cotransporter 1. Proc Natl Acad Sci U S A. 2007;104:15075–80. doi: 10.1073/pnas.0706678104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Mace OJ, Affleck J, Patel N, Kellett GL. Sweet taste receptors in rat small intestine stimulate glucose absorption through apical GLUT2. J Physiol. 2007;582:379–92. doi: 10.1113/jphysiol.2007.130906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Fujita Y, Wideman RD, Speck M, Asadi A, King DS, Webber TD, et al. Incretin release from gut is acutely enhanced by sugar but not by sweeteners in vivo. Am J Physiol Endocrinol Metab. 2009;296:E473–9. doi: 10.1152/ajpendo.90636.2008. [DOI] [PubMed] [Google Scholar]
  • 76.Temizkan S, Deyneli O, Yasar M, Arpa M, Gunes M, Yazici D, et al. Sucralose enhances GLP-1 release and lowers blood glucose in the presence of carbohydrate in healthy subjects but not in patients with type 2 diabetes. Eur J Clin Nutr. 2014 doi: 10.1038/ejcn.2014.208. [DOI] [PubMed] [Google Scholar]
  • 77.Brown RJ, Rother KI. Non-nutritive sweeteners and their role in the gastrointestinal tract. J Clin Endocrinol Metab. 2012;97:2597–605. doi: 10.1210/jc.2012-1475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Brown RJ, Walter M, Rother KI. Ingestion of diet soda before a glucose load augments glucagon-like peptide-1 secretion. Diabetes Care. 2009;32:2184–6. doi: 10.2337/dc09-1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Pepino MY, Tiemann CD, Patterson BW, Wice BM, Klein S. Sucralose affects glycemic and hormonal responses to an oral glucose load. Diabetes Care. 2013;36:2530–5. doi: 10.2337/dc12-2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ma J, Chang J, Checklin HL, Young RL, Jones KL, Horowitz M, et al. Effect of the artificial sweetener, sucralose, on small intestinal glucose absorption in healthy human subjects. Br J Nutr. 2010;104:803–6. doi: 10.1017/S0007114510001327. [DOI] [PubMed] [Google Scholar]
  • 81.Ma J, Bellon M, Wishart JM, Young R, Blackshaw LA, Jones KL, et al. Effect of the artificial sweetener, sucralose, on gastric emptying and incretin hormone release in healthy subjects. Am J Physiol Gastrointest Liver Physiol. 2009;296:G735–9. doi: 10.1152/ajpgi.90708.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Steinert RE, Frey F, Töpfer A, Drewe J, Beglinger C. Effects of carbohydrate sugars and artificial sweeteners on appetite and the secretion of gastrointestinal satiety peptides. Br J Nutr. 2011;105:1320–8. doi: 10.1017/S000711451000512X. [DOI] [PubMed] [Google Scholar]
  • 83.Ford HE, Peters V, Martin NM, Sleeth ML, Ghatei MA, Frost GS, et al. Effects of oral ingestion of sucralose on gut hormone response and appetite in healthy normal-weight subjects. Eur J Clin Nutr. 2011;65:508–13. doi: 10.1038/ejcn.2010.291. [DOI] [PubMed] [Google Scholar]
  • 84.Cong W, Wang R, Cai H, Daimon CM, Scheibye-Knudsen M, Bohr VA, et al. Long-term artificial sweetener acesulfame potassium treatment alters neurometabolic functions in C57BL/6J mice. PLoS One. 2013;8:e70257. doi: 10.1371/journal.pone.0070257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Schiffman SS. Rationale for further medical and health research on high-potency sweeteners. Chem Senses. 2012;37:671–9. doi: 10.1093/chemse/bjs053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Janssen AWF, Kersten S. The role of the gut microbiota in metabolic health. FASEB J. 2015 doi: 10.1096/fj.14-269514. [DOI] [PubMed] [Google Scholar]
  • 87.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–63. doi: 10.1038/nature12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Muegge BD, Kuczynski J, Knights D, Clemente JC, González A, Fontana L, et al. Diet drives convergence in gut microbiome functions across mammalian phylogeny and within humans. Science. 2011;332:970–4. doi: 10.1126/science.1198719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Claesson MJ, Jeffery IB, Conde S, Power SE, O’Connor EM, Cusack S, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488:178–84. doi: 10.1038/nature11319. [DOI] [PubMed] [Google Scholar]
  • 90.Karlsson FH, Tremaroli V, Nookaew I, Bergström G, Behre CJ, Fagerberg B, et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013;498:99–103. doi: 10.1038/nature12198. [DOI] [PubMed] [Google Scholar]
  • 91.Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60. doi: 10.1038/nature11450. [DOI] [PubMed] [Google Scholar]
  • 92.Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–3. doi: 10.1038/4441022a. [DOI] [PubMed] [Google Scholar]
  • 93.Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341:1241214. doi: 10.1126/science.1241214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–31. doi: 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]
  • 95.Daly K, Darby AC, Hall N, Nau A, Bravo D, Shirazi-Beechey SP. Dietary supplementation with lactose or artificial sweetener enhances swine gut Lactobacillus population abundance. Br J Nutr. 2014;111(Suppl):S30–5. doi: 10.1017/S0007114513002274. [DOI] [PubMed] [Google Scholar]
  • 96.Abou-Donia MB, El-Masry EM, Abdel-Rahman AA, McLendon RE, Schiffman SS. Splenda alters gut microflora and increases intestinal p-glycoprotein and cytochrome p-450 in male rats. J Toxicol Environ Health A. 2008;71:1415–29. doi: 10.1080/15287390802328630. [DOI] [PubMed] [Google Scholar]
  • 97.Anderson RL, Kirkland JJ. The effect of sodium saccharin in the diet on caecal microflora. Food Cosmet Toxicol. 1980;18:353–355. doi: 10.1016/0015-6264(80)90188-1. [DOI] [PubMed] [Google Scholar]
  • 98.Palmnäs MSA, Cowan TE, Bomhof MR, Su J, Reimer RA, Vogel HJ, et al. Low-dose aspartame consumption differentially affects gut microbiota-host metabolic interactions in the diet-induced obese rat. PLoS One. 2014;9:e109841. doi: 10.1371/journal.pone.0109841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Suez J, Korem T, Zeevi D, Zilberman-Schapira G, Thaiss CA, Maza O, et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature. 2014;514:181–186. doi: 10.1038/nature13793. [DOI] [PubMed] [Google Scholar]
  • 100.Swithers SE, Martin AA, Davidson TL. High-intensity sweeteners and energy balance. Physiol Behav. 2010;100:55–62. doi: 10.1016/j.physbeh.2009.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sclafani A. Learned controls of ingestive behaviour. Appetite. 1997;29:153–8. doi: 10.1006/appe.1997.0120. [DOI] [PubMed] [Google Scholar]
  • 102.Power ML, Schulkin J. Anticipatory physiological regulation in feeding biology: cephalic phase responses. Appetite. 50:194–206. doi: 10.1016/j.appet.2007.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Mattes RD. Physiologic responses to sensory stimulation by food: nutritional implications. J Am Diet Assoc. 1997;97:406–13. doi: 10.1016/S0002-8223(97)00101-6. [DOI] [PubMed] [Google Scholar]
  • 104.Smeets PAM, Erkner A, de Graaf C. Cephalic phase responses and appetite. Nutr Rev. 2010;68:643–55. doi: 10.1111/j.1753-4887.2010.00334.x. [DOI] [PubMed] [Google Scholar]
  • 105.Davidson TL, Swithers SE. A Pavlovian approach to the problem of obesity. Int J Obes Relat Metab Disord. 2004;28:933–935. doi: 10.1038/sj.ijo.0802660. [DOI] [PubMed] [Google Scholar]
  • 106.Swithers SE, Martin AA, Davidson TL. High-intensity sweeteners and energy balance. Physiol Behav. 2010;100:55–62. doi: 10.1016/j.physbeh.2009.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Swithers SE, Davidson TL. A role for sweet taste: calorie predictive relations in energy regulation by rats. Behav Neurosci. 2008;122:161–173. doi: 10.1037/0735-7044.122.1.161. [DOI] [PubMed] [Google Scholar]
  • 108.Swithers SE, Baker CR, Davidson TL. General and persistent effects of high-intensity sweeteners on body weight gain and caloric compensation in rats. Behav Neurosci. 2009;123:772–780. doi: 10.1037/a0016139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Swithers SE, Laboy AF, Clark K, Cooper S, Davidson TL. Experience with the high-intensity sweetener saccharin impairs glucose homeostasis and GLP-1 release in rats. Behav Brain Res. 2012;233:1–14. doi: 10.1016/j.bbr.2012.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Berthoud HR, Bereiter DA, Trimble ER, Siegel EG, Jeanrenaud B. Cephalic phase, reflex insulin secretion. Neuroanatomical and physiological characterization. [accessed May 6, 2015];Diabetologia. 1981 20(Suppl):393–401. http://www.ncbi.nlm.nih.gov/pubmed/7014335. [PubMed] [Google Scholar]
  • 111.Frank GKW, Oberndorfer TA, Simmons AN, Paulus MP, Fudge JL, Yang TT, et al. Sucrose activates human taste pathways differently from artificial sweetener. Neuroimage. 2008;39:1559–1569. doi: 10.1016/j.neuroimage.2007.10.061. [DOI] [PubMed] [Google Scholar]
  • 112.Smeets PAM, Weijzen P, de Graaf C, Viergever MA. Consumption of caloric and non-caloric versions of a soft drink differentially affects brain activation during tasting. Neuroimage. 2011;54:1367–1374. doi: 10.1016/j.neuroimage.2010.08.054. [DOI] [PubMed] [Google Scholar]
  • 113.Johnson AW, Gallagher M, Holland PC. The basolateral amygdala is critical to the expression of pavlovian and instrumental outcome-specific reinforcer devaluation effects. J Neurosci. 2009;29:696–704. doi: 10.1523/JNEUROSCI.3758-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Touzani K, Bodnar RJ, Sclafani A. Dopamine D1-like receptor antagonism in amygdala impairs the acquisition of glucose-conditioned flavor preference in rats. Eur J Neurosci. 2009;30:289–98. doi: 10.1111/j.1460-9568.2009.06829.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Malkusz DC, Banakos T, Mohamed A, Vongwattanakit T, Malkusz G, Saeed S, et al. Dopamine signaling in the medial prefrontal cortex and amygdala is required for the acquisition of fructose-conditioned flavor preferences in rats. Behav Brain Res. 2012;233:500–7. doi: 10.1016/j.bbr.2012.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Gottfried JA, O’Doherty J, Dolan RJ. Encoding predictive reward value in human amygdala and orbitofrontal cortex. Science. 2003;301:1104–1107. doi: 10.1126/science.1087919. [DOI] [PubMed] [Google Scholar]
  • 117.Small DM, Veldhuizen MG, Felsted J, Mak YE, McGlone F. Separable substrates for anticipatory and consummatory food chemosensation. Neuron. 2008;57:786–797. doi: 10.1016/j.neuron.2008.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.de Araujo IE, Simon SA. The gustatory cortex and multisensory integration. Int J Obes (Lond) 2009;33(Suppl 2):S34–43. doi: 10.1038/ijo.2009.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Rudenga K, Green B, Nachtigal D, Small DM. Evidence for an integrated oral sensory module in the human anterior ventral insula. Chem Senses. 2010;35:693–703. doi: 10.1093/chemse/bjq068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Griffioen-Roose S, Smeets PAM, Weijzen PLG, van Rijn I, van den Bosch I, de Graaf C. Effect of replacing sugar with non-caloric sweeteners in beverages on the reward value after repeated exposure. PLoS One. 2013;8:e81924. doi: 10.1371/journal.pone.0081924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Crum AJ, Corbin WR, Brownell KD, Salovey P. Mind over milkshakes: mindsets, not just nutrients, determine ghrelin response. Health Psychol. 2011;30:424–9. doi: 10.1037/a0023467. discussion 430–1. [DOI] [PubMed] [Google Scholar]
  • 122.Veldhuizen MG, Nachtigal DJ, Flammer LJ, de Araujo IE, Small DM. Verbal descriptors influence hypothalamic response to low-calorie drinks. Mol Metab. 2013;2:270–80. doi: 10.1016/j.molmet.2013.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Cassady BA, Considine RV, Mattes RD. Beverage consumption, appetite, and energy intake: what did you expect? Am J Clin Nutr. 2012;95:587–93. doi: 10.3945/ajcn.111.025437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Faulkner GP, Pourshahidi LK, Wallace JMW, Kerr MA, McCaffrey TA, Livingstone MBE. Perceived “healthiness” of foods can influence consumers’ estimations of energy density and appropriate portion size. Int J Obes (Lond) 2014;38:106–12. doi: 10.1038/ijo.2013.69. [DOI] [PubMed] [Google Scholar]
  • 125.Gravel K, Doucet É, Herman CP, Pomerleau S, Bourlaud AS, Provencher V. “Healthy,” “diet,” or “hedonic”. How nutrition claims affect food-related perceptions and intake? Appetite. 2012;59:877–84. doi: 10.1016/j.appet.2012.08.028. [DOI] [PubMed] [Google Scholar]
  • 126.Vadiveloo M, Morwitz V, Chandon P. The interplay of health claims and taste importance on food consumption and self-reported satiety. Appetite. 2013;71:349–56. doi: 10.1016/j.appet.2013.09.005. [DOI] [PubMed] [Google Scholar]
  • 127.Small DM. Changes in brain activity related to eating chocolate: From pleasure to aversion. Brain. 2001;124:1720–1733. doi: 10.1093/brain/124.9.1720. [DOI] [PubMed] [Google Scholar]
  • 128.Min DK, Tuor UI, Chelikani PK. Gastric distention induced functional magnetic resonance signal changes in the rodent brain. Neuroscience. 2011;179:151–8. doi: 10.1016/j.neuroscience.2011.01.051. [DOI] [PubMed] [Google Scholar]
  • 129.Hsu TM, Konanur VR, Taing L, Usui R, Kayser BD, Goran MI, et al. Effects of sucrose and high fructose corn syrup consumption on spatial memory function and hippocampal neuroinflammation in adolescent rats. Hippocampus. 2015;25:227–39. doi: 10.1002/hipo.22368. [DOI] [PubMed] [Google Scholar]
  • 130.Molteni R, Barnard RJ, Ying Z, Roberts CK, Gómez-Pinilla F. A high-fat, refined sugar diet reduces hippocampal brain-derived neurotrophic factor, neuronal plasticity, and learning. [accessed March 27, 2015];Neuroscience. 2002 112:803–14. doi: 10.1016/s0306-4522(02)00123-9. http://www.ncbi.nlm.nih.gov/pubmed/12088740. [DOI] [PubMed] [Google Scholar]
  • 131.Francis HM, Stevenson RJ. Higher reported saturated fat and refined sugar intake is associated with reduced hippocampal-dependent memory and sensitivity to interoceptive signals. Behav Neurosci. 2011;125:943–55. doi: 10.1037/a0025998. [DOI] [PubMed] [Google Scholar]
  • 132.Davidson TL, Chan K, Jarrard LE, Kanoski SE, Clegg DJ, Benoit SC. Contributions of the hippocampus and medial prefrontal cortex to energy and body weight regulation. Hippocampus. 2009;19:235–52. doi: 10.1002/hipo.20499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Davidson TL, Kanoski SE, Chan K, Clegg DJ, Benoit SC, Jarrard LE. Hippocampal lesions impair retention of discriminative responding based on energy state cues. Behav Neurosci. 2010;124:97–105. doi: 10.1037/a0018402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Kanoski SE. Cognitive and neuronal systems underlying obesity. Physiol Behav. 2012;106:337–44. doi: 10.1016/j.physbeh.2012.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Kanoski SE, Davidson TL. Western diet consumption and cognitive impairment: links to hippocampal dysfunction and obesity. Physiol Behav. 2011;103:59–68. doi: 10.1016/j.physbeh.2010.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES