Food additives, food and the concept of ‘food addiction’: Is stimulation of the brain reward circuit by food sufficient to trigger addiction?

Food additives, food and the concept of ‘food addiction’: Is stimulation of the brain reward circuit by food sufficient to trigger addiction?

Accepted Manuscript Title: Food additives, food and the concept of ‘food addiction’: Is stimulation of the brain reward circuit by food sufficient to ...

825KB Sizes 0 Downloads 40 Views

Accepted Manuscript Title: Food additives, food and the concept of ‘food addiction’: Is stimulation of the brain reward circuit by food sufficient to trigger addiction? Authors: A.Y. Onaolapo, O.J. Onaolapo PII: DOI: Reference:

S0928-4680(17)30175-X https://doi.org/10.1016/j.pathophys.2018.04.002 PATPHY 939

To appear in:

Pathophysiology

Received date: Revised date: Accepted date:

2-12-2017 26-2-2018 7-4-2018

Please cite this article as: Onaolapo AY, Onaolapo OJ, Food additives, food and the concept of ‘food addiction’: Is stimulation of the brain reward circuit by food sufficient to trigger addiction?, Pathophysiology (2010), https://doi.org/10.1016/j.pathophys.2018.04.002 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 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.

Food additives, food and the concept of ‘food addiction’: is stimulation of the brain reward circuit by food sufficient to trigger addiction?

1

IP T

Onaolapo AY1 and Onaolapo OJ2*

Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of

2

SC R

Technology, Ogbomoso, Oyo State, Nigeria. [email protected],

Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University

N

U

of Technology, Osogbo, Osun State, Nigeria. [email protected].

M

A

*Corresponding Author:

A

CC E

PT

ED

Graphical abstract

1

Abstract In the last few years, the concept of ‘food addiction’ has continued to gain popularity, with human and animal studies demonstrating the differential effects of foods that are high in fat,

IP T

sugar or protein on appetite, satiety, eating behaviour and the development of food addiction. However, a number of studies have disputed the occurrence of food addiction in humans.

SC R

Questions have also arisen regarding the possible impacts that food additives may have on the development of food addiction or eating disorders. Also, it is known that alterations in food

composition and the presence of food additives (flavour enhancers, sugars, sugar substitutes,

U

and non-nutritive sweeteners) are factors that generally influence the sensory perception of

N

food. Our understanding of the potential roles of central neurotransmitters (such as

A

dopamine) and certain neuropeptides in the evolution of food addiction is also evolving; but

M

presently, there isn’t sufficient scientific evidence to consider any food ingredient, micronutrient or standard food-additive as addictive. In this review, the relevant literatures

ED

dealing with the concept of ‘food addiction’ are examined, and the factors which may

sugars,

sugar

PT

predispose to food addiction are discussed. The possible influences that flavour-enhancers, substitutes

and

non-nutritive

sweeteners

may

exert

on

central

CC E

neurotransmission, neurotransmitter/receptor interactions, appetite, satiety, conditionedpreferences and the brain reward system are also highlighted.

A

Keywords: Brain reward system; Conditioned preference; Food addiction; Flavour enhancer; Food sweeteners; Overeating

2

1.0

Introduction



‘Food addiction’ is a behavioural addiction that is characterised by compulsive

consumption of palatable foods, which is associated with activation of the brain reward system in humans and animals (Olsen, 2011; Hebebrand et al., 2014). It has also been defined as a clinically-significant physical and/or psychological dependence on high sugar, high fat,

IP T

and highly-palatable foods (Brownell and Gold, 2012, 2013). Over the last few years, the concept of food addiction has become increasingly popular and this is reflected not only in

SC R

the volume of media reports and health blogs (Marlow, 2013; Avena and Talbott, 2014; Tarman and Werdell, 2014) but also in a substantial increase in scientific literature (Corwin

U

and, Grigson, 2009; Gearhardt et al., 2011a; Krashes and Kravitz, 2014; Muele, 2015; Schulte

N

et al., 2016) dedicated to the topic. Addiction to food has been implicated in foodcraving/bingeing and is also considered an important predisposing factor to obesity (Davis et

M

A

al., 2011; Gearhardt et al., 2011a; Schulte et al., 2016). An ‘addictive’ food, nutrient, or food ingredient is believed to possess an inherent ability to induce dependence (Hebebrand et al.,

ED

2014). Hence, the concept of food addiction is based on the idea that certain foods (or their additives) may have addictive potentials (Muele, 2015). Human and animal studies have

PT

continued to report the differential effects of high-fat, high-sugar or high-protein diet on eating behaviour (Avena et al., 2008a, 2008b; Avena et al., 2012a; Schulte et al., 2015;

CC E

Tulloch et al., 2015; Schulte et al., 2016) and brain neurotransmitter chemistry (Bocarsly et al., 2010). A number of rodent studies have also demonstrated the effects of exposure to

A

sugars, fats and highly-palatable diets on food-preferences, brain-reward functioning and the risk for obesity (Avena et al., 2008a, 2008b, 2012a; Johnson and Kenny, 2010; Borengasser et al., 2014). Genetic traits that may predispose to food addiction have also been studied (Velázquez-Sánchez et al., 2014).

3

Food additives are substances that are added to foods or drinks to maintain/enhance safety, freshness, texture, appearance, taste or flavour (WHO, 2017). For centuries, humans have used salt to preserve meats and fish, herbs and spices to enhance food flavour and sugars to preserve fruits. However, in recent times, the list of indications for food additives is expanding and they are increasingly being used to make food safe, stable, nutritious,

IP T

convenient, colourful, flavourful and affordable. Hence, different types of food additives have been (and are still being) developed to meet the growing demands of the food industry.

SC R

Food additives can be synthetic or derived from plants/animals, and they have been grouped

by the World Health Organisation and the Food and Agricultural Organisation into three

U

broad categories (Flavour enhancers, Enzymes and Others), which are based on their function

N

(WHO, 2017). Flavour enhancers constitute the majority of additives used in foods and they

A

include salt and monosodium glutamate. Enzyme preparations include additives which may

M

be naturally-occurring proteins or synthetic compounds which assist in breaking down larger molecules into their smaller building blocks, such as yeasts used in improving the dough or

ED

for alcohol fermentation. Other additives include nutritive (sugars) or non-nutritive (aspartame and saccharin) sweeteners and food preservation/colouring agents.

PT

Recently, interests in the potential associations of food additives, processed foods and food addiction have increased (Morris et al., 2008; Cocores and Gold, 2009; Blaylock, 2014), with

CC E

suggestions that consumption of highly-processed foods promotes food addiction (Johnson and Kenny, 2010; Gearhardt et al., 2011b). These suggestions are related to the fact that

A

while many food ingredients lack intoxication capacity, studies have shown that certain foods or their additives (e.g saccharin) appear to induce reinforcement behaviours that are similar to or possibly exceed those associated with drugs of abuse (Lenoir et al., 2007; Gearhardt et al., 2011b). Also, in similarity to drugs of abuse, they may trigger addiction-like neuroadaptive responses in the brain reward circuits (Johnson and Kenny, 2010). Again, highly-processed

4

foods may have been altered in ways similar to addictive drugs (Gearhardt et al., 2011b) and certain formulations of processed foods have been designed to maximise palatability and/or reward (Spence, 2012). However, a number of researchers are of the opinion that while there are speculations regarding the factors that are present in foods which may stimulate addiction, there isn’t sufficient scientific evidence yet to consider any food ingredient, micronutrient or

IP T

standard food-additive as addictive (Hebebrand et al., 2014). In this review, we highlight the results of studies on the possible addictive potential of selected food additives (culinary

SC R

flavour enhancers and food sweeteners) by examining the relevant scientific literature on the associations that exist among factors such as food, food additives, the brain reward system

1.1

U

and food addiction. Food and food addiction

A

N

The construct “food addiction” was first introduced by T.G. Randolph in 1956,

M

around a period when addictive-like consumption of wheat, corn, potatoes, chocolate, coffee, milk and eggs were reported (Randolph, 1956). However, in recent years, this construct has

ED

become popular because it helped to understand the influences that psychological factors could exert on weight gain (Brownell and Gold, 2013), especially following an increase in the

PT

prevalence of obesity in the United States of America (and world-wide) in the last few decades (Finucane et al., 2011; Ogden et al., 2012). Food addiction is not currently

CC E

recognised as a disorder in the Diagnostic and Statistical Manual of Mental Disorders (DSM5). However, studies evaluating the occurrence of food addiction in humans (Flint et al.,

A

2014; Pursey et al., 2015) have utilised the Yale Food Addiction Scale (YFAS), a validated and reliable instrument based primarily on the diagnostic criteria for substance dependence in the DSM-IV-TR (APA. 2000) which measures behavioural indicators of eating disorders (Gearhardt et al., 2009). Despite the fact there is a paucity of population-based prevalence rates for food addiction, studies in subjects not diagnosed with an eating disorder had

5

reported prevalence rates of 5 to 26% (Ziauddeen and Fletcher, 2013); while in studies that have included subjects with a diagnosed eating disorder, higher rates of food addiction have been reported (Pursey et al., 2014). There are growing interests in defining the roles played by the food reward system in regulating food intake, as well as the possible links that may exists between this system and

IP T

the development of food addiction in humans. In recent times, questions have also arisen

about the roles played by food additives like sugars, non-nutritive sweeteners and culinary

SC R

flavour enhancers in modulating the food reward towards food addiction (figure 1). Studies in laboratory animals have suggested the presence of homeostatic factors that control energy

U

intake and energy demand, in effect precisely modulating body weight over prolonged

N

periods of time (Woods, 2009). A few other studies have also reported that the hedonic

A

properties of food may influence feeding beyond the daily energy requirements, in essence

M

possibly contributing to weight gain and obesity (Kenny, 2011). The regulation of food intake (figure 2) has been demonstrated to involve strong associations

ED

that exist between homeostatic and non-homeostatic factors, the balance of which ensures the maintenance of body weight (Hagan and Niswender, 2012). The homeostatic control

PT

(regulated by energy stores and energy demands) which mediates how much food an individual consumes is usually subject to non-homeostatic or hedonic (consumption

CC E

motivated by reward systems) influences. It has been observed that reward-related signals can override homeostatic signals, essentially contributing to the consumption of more foods

A

above the body’s immediate energy requirement (Begg and Woods, 2013; Alonso-Alonso et al., 2015). In last few decades, the consumer response to the availability of high-energy and/or highly-palatable foods may be a reflection of the influences that such reward-related signals can exert.

6

The additive potential of some foods and food substances may be comparable to that of common drugs of abuse (like cocaine, marijuana and methamphetamine) whose use is characterised by episodes of negative consequences (abuse) and physiological dependence (Alonso-Alonso et al., 2015). These drugs are known to enhance

dopamine release (from

dopaminergic neurons in the ventral tegmental area) to brain regions that are associated with

IP T

the reward system, such as the prefrontal cortex, amygdala and the nucleus accumbens. In the

brain reward system, the patterns of neural responses associated with palatable foods have

SC R

been shown to be similar to those associated with drugs of abuse (Ahmed et al., 2002; Kalivas and O'Brien, 2008; Janes et al., 2010). Similarly, palatable or highly-processed foods

U

have reinforcing effects which may be mediated by dopamine’s influences on the brain

N

reward system (Alonso-Alonso et al., 2015). Also, palatable foods have been observed to

A

activate reward-learning regions of the brain, as well as dopamine signalling (Small et al., 2003). Repeated intake of foods that are high in sugar is associated with increased D1

M

receptor binding (in the shell and core of the NA) and increased μ-opioid receptor binding in

ED

the cingulate cortex, hippocampus, accumbens shell and locus coeruleus; changes which are similar to those associated with a number of drugs of abuse (Colantuoni et al., 2001). A

PT

down-regulation of post-synaptic D2 receptors in the nucleus accumbens shell and dorsal striatum (as seen in drug addicts) had also been reported in animals that were fed high-

CC E

fat/sugar foods (Johnson and Kenny, 2010; Bello et al., 2002; Stice et al., 2010a). The exposure of obese subjects to palatable food cues has also been associated with hyper-

A

responsiveness in the reward-related brain regions (Stoeckel et al., 2008; Stice et al., 2010b). From the aforegoing, dopamine is linked to the reinforcing effects of food, and studies have demonstrated that food consumption results in striatal (dorsal) dopamine release with the degree of pleasure derived from eating correlating positively with the quantity of dopamine released (Small et al., 2003). Administration of dopamine antagonists has also been

7

associated with increased food intake and weight gain, while dopamine agonists cause a reduction in food intake and an ensuing weight loss (Epstein et al., 2007). Neuropeptides (orexigenic and anorexigenic) which are involved in the regulation of food consumption and metabolism have also been evaluated for their roles in the neurobiological responses of drug and alcohol-dependence (Thiele et al., 2003; Barson and Leibowitz, 2016).

IP T

The brain expression of these neuropeptides has been reported to be altered following bingelike consumption of palatable foods (Alcaraz-Iborra et al., 2014) or drugs (Navarro et al.,

SC R

2008; Carvajal et al., 2015). Central administration of Agouti-related peptide (a melanocortin

receptor antagonist) activates midbrain dopamine neurons and induces consumption of fat

U

and sugar-enriched foods (Davis et al., 2011). Ghrelin, leptin and insulin are homeostatic

N

regulators of food intake and via their influences on the dopaminergic systems, they mediate

A

between the homeostatic and hedonic mechanisms of food intake (Kenny, 2011; Egecioglu et

M

al., 2011; Hebebrand et al., 2014).With the discovery of leptin receptors expression on midbrain dopamine neurons (Elmquist et al., 1998), there have been suggestions that

ED

mesolimbic dopamine pathways could also mediate the effect of leptin on food intake (Lerma-Cabrera, et al., 2016).

Anatomy of the brain reward system

PT

1.2

The brain reward system is made up of a group of neurons, nerve cells and brain

CC E

nuclei that are involved in the modulation and control of associative learning, pleasure and incentive salience (Berridge, 2012; Berridge and Kringelbach, 2015; Schultz, 2015). Reward

A

is the ability of an object to stimulate appetitive or approach behaviour, choices and emotional behaviour (Schultz, 2015). The association between a feeling of reward and substance use has been linked to the reason why humans use these substances, or why laboratory animals self-administer them (Wise, 2009). The rewarding effects of substances have been linked to their ability to increase dopamine (DA) secretion in the brain regions that

8

mediate reward (especially in the nucleus accumbens) (Di Chiara and Imperato, 1988; Nestler et al., 2004); although studies have also demonstrated that DA’s role in reward does not usually equate with hedonic responses which are mediated partly by endogenous opioids and cannabinoids. The neural circuitry tasked with the mediation of reward and/or addiction involves a network of brain regions that continues to evolve in number and/or complexity

IP T

(Wise and Koob, 2014).

Brain regions that constitute the reward system are located primarily within a cortico-basal

SC R

ganglia-thalamo-cortical loop (Yager et al., 2015), which modulates the development and

maintenance of addiction (Nestler, 2013, van Huijstee and Mansvelder, 2014). DA neurons in

U

the ventral tegmental area (VTA) of the midbrain are very important (Lammel et al. 2012),

N

because they form strong reciprocal connections with several other brain regions like the

A

nucleus accumbens, lateral hypothalamus, and prefrontal cortex (Calabresi et al. 2007; Luo et

M

al., 2015). The striatum (dorsal striatum and nucleus accumbens) is also very important in the reward pathway, as it serves as the main integration site for the cortico-basal ganglia-thalamic

ED

circuitry by receiving a variety of inputs. It receives GABAergic inputs from striatal interneurons (Dautan et al., 2014), cholinergic inputs from the interneurons of the striatum

PT

and brainstem (Yager et al., 2015), as well as dopaminergic inputs from the VTA (Swanson, 1982) and the substantia nigra (Yager et al., 2015). It also receives glutamatergic inputs from

CC E

the prefrontal cortex, amygdala, hippocampus and thalamus (Finch, 1996, Britt et al., 2012). For a detailed discussion on the brain reward system, see Berridge and Kringelbach (2015)

A

and Schultz (2015). 1.3

The food reward system Intrinsically, eating has rewarding and reinforcing effects, and studies have

demonstrated that food consumption activates the regions associated with reward in the brain (Hebebrand et al., 2014). However, food consumption is regulated by a neural circuitry that

9

includes the dorsolateral prefrontal cortex, striatum, amygdala, midbrain and the hypothalamus (Berthoud, 2011). Together, these systems regulate all aspects of feeding behaviour, including both homeostatic and hedonic food consumption (Hebebrand et al., 2014). The food reward system consists of at least of two parts, a sensory and/or a postingestive component (Avena et al., 2008a, 2008b). Gustatory information (usually

IP T

perceived by the taste receptors) send signals that ascend through the thalamus, terminating in the orbitofrontal cortex and anterior insula/frontal operculum (Kobayakawa et al.,1999;

SC R

Small, 2006); while the amygdala makes reciprocal connections at all levels of the gustatory

pathway (Yang, 2010). Mesolimbic DA system also aids the hedonic recognition of the

U

stimulus and satiation that follow consumption of palatable foods (Goldstein and Volkow,

N

2002; Haltia et al., 2007; Stice et al., 2008).

A

Imaging studies have revealed an increased activation in the lateral orbitofrontal cortex,

M

insula, amygdala, frontal operculum and striatum in anticipation of palatable food (Stice et al., 2008) and in response to pictures of palatable foods (Rothmund et al., 2007; Stoeckel et

ED

al., 2008) in obese compared to control subjects. In humans (unlike laboratory animals), the influence of cognition and executive functioning influences behavioural drives for palatable

PT

food. This executive functioning support self-regulation of eating and have been mapped to brain regions which include the dorsolateral prefrontal cortex, the dorsal anterior cingulate,

CC E

and the parietal cortex (Alonso-Alonso et al., 2015). 1.4

Food, food constituents and food consumption

A

Foods and/or food constituents have been shown to have varying abilities for

induction of satiation. Studies have reported that foods that are high in dietary protein tend to increase the perception of satiety (Labayen et al., 2004; Schoeller and Buchholz, 2005; Luhovyy et al., 2008; Thomas and Chapman, 2008). Several studies have also reported that foods that are high in protein are less liked than low-protein foods (Johnson and Vickers,

10

1992; Vanderwater and Vickers, 1994). High-protein foods have also been shown to produce an increase in sensory-specific satiety, compared to low-protein foods (Johnson and Vickers, 1992; Vanderwater and Vickers, 1994; Sivertsen et al., 2010). Sugar- and fat-rich foods are highly palatable, and studies have shown that foods rich in sugar and/or fat can promote overeating (Berthoud, 2011; Egecioglu et al., 2011; Ryan et al., 2012).

IP T

The ability of sweet taste and fatty flavour to activate the brain reward systems and promote

food consumption has been demonstrated by a number of animal studies (Hajnal and

SC R

Norgren, 2008; Liang et al., 2006). Also, the ability of these foods to cause obesity in rodents

has been observed (Sclafani, 1993; Speakman et al., 2008). It is now generally accepted that

U

food can be made more palatable by: the addition of biopolymers like transglutaminase

N

(which improves food texture), adding soy protein sauce or carrageenan mix, varying the fat

A

content (Muguruma et al., 2003; Pietrasik and Duda, 2000), addition of flavour enhancers

M

{like monosodium glutamate, inosine-5-monophosphate (IMP-5) (Bellisle, 1999; Yamaguchi and Ninomiya, 2000) and sodium guanoate}, or food sweeteners like non-nutritive

1.4.1

ED

sweeteners.

How do high-fat/high-sugar diets promote overeating and/or food addiction?

PT

There have been reports showing that foods that are rich in sugar and/or fats can stimulate segments of the reward system (including taste receptors, postingestive signals and

CC E

neuropeptide systems) enough to result in overeating or binge eating. A number of rodent studies have also demonstrated that foods that are rich in both sugar and fat stimulate

A

excessive eating. Consumption of foods that are high in sugar has been associated with increased secretion of DA and acetylcholine (ACH) in the nucleus accumbens (Avena et al., 2012b). Increasing palatability of food or food constituents (as it occurs with high-sugar or high-fat diets) has also been associated with a reduction in the effectiveness with which intestinal and post-absorptive satiation signals suppress food intake (Sclafani, 2013). Johnson

11

and Kelly (2010) also demonstrated that excessive intake of highly-palatable food results in a down-regulation of the expression of striatal D2 receptors. The body controls food intake and energy expenditure by a series of impulses which include satiation. Satiation represents a cumulation of the inhibitory signals that are induced by the ingestion of food substances. These signals may be cognitive, digestive, sensory or hormonal.

IP T

Satiation results in a feeling of being full and ultimately signals the end of a meal (Woods and D'Alessio, 2008). The duration or intensity of satiety that follows a meal and persists

SC R

until hunger pangs begin is also modulated by various signals which form part of the satiety cascade (Blundell et al., 1993). Studies have shown that sensory and cognitive processes

U

interact with post ingestive and post absorptive peripheral and central mechanisms that help

N

to maintain satiety. Satiation signals generally reflect the nutrient and caloric content of a

A

meal. The gut peptides involved in the induction of satiation include cholecystokinin,

M

enterostatin, amylin, glucagon-like peptide, peptide tyrosine-tyrosine, apolipoprotein A-IV, glucagon, oxyntomodulin and members of the bombesin family of peptides (Moran, 2004;

ED

Woods and D’Alessio, 2008).

In addition to satiation signals, there are also adiposity signals which in conjunction with

PT

satiation signals modulate food intake and metabolism. Adiposity signals reflect the amount of stored body fat, insulin which is secreted by the pancreatic β-cells and leptin secreted from

CC E

adipocytes; with both insulin and leptin correlating with the body’s total adiposity and energy store (Polonsky et al., 1988; Frederich et al., 1995; Schwartz et al., 1996). Increased flux of

A

insulin or leptin stimulates receptors in the hypothalamus and other brain regions to reduce food intake (Woods et al., 1979; Brief and Davis, 1984; Seeley et al., 1996); while a reduction of adiposity-signalling in the brain (in experimental animals) has been associated with hyperphagia (Cohen et al., 2001; Obici et al., 2002). Adiposity signals (unlike satiation signals) exert long-lasting effects spanning several meals or days, and stabilising adipose

12

stores (Ryan et al., 2012). Studies have shown that chronic exposure to foods that are high in fat raises the body’s adiposity and satiation threshold, such that higher levels of signals are required to suppress feeding (Ryan et al., 2012). Apart from the direct effects of palatable flavours of high-sugar and high-fat foods in preventing satiation, studies have also revealed that sugar and fat can induce post-oral

IP T

processes which stimulate food intake and food-preference (Sclafani and Ackroff, 2012). In

the central nervous system, molecular lipid sensors have been discovered, which link high fat

SC R

diets to over-eating and/or food addiction. The peroxisome proliferator activated receptors

(PPARs) are nuclear receptors which are usually activated by intracellular lipids and regulate

U

gene expression. An isoform of this molecule (PPARγ) which has been located in key regions

N

of the hypothalamus that are associated with the regulation of energy balance (Sarruf et al.,

A

2009) is also expressed in adipocytes, where it acts as a master regulator of adipogenesis

M

(Tontonoz et al.,1994; Patel et al., 2003) and lipogenesis (Patel et al., 2003; Lehrke and Lazar, 2005). From the results of some studies (Lu et al., 2011; Ryan et al., 2011),

ED

researchers have deduced that central activation of PPARγ (most likely hypothalamic) result in increased food intake and the accumulation of body fat, whilst the effects of local

PT

administration of antagonist (Ryan et al., 2011) or neuron- specific gene deletion (Lu et al., 2011) resulted in a decrease in the consumption of diets that are high in fat. So from the fore-

CC E

going, for any food or food constituent to stimulate or induce food addiction, it must influence at least one or more of the different peripheral or central signals that the body uses

A

to maintain food and energy balance. 1.5

Flavour, taste and food preference The flavour of food which is perceived by smell, texture and/or taste is a sensation

that provides individuals with information relating to the nutritional quality of food. This sensation may also stimulate cephalic phase digestive responses which facilitate food

13

utilisation, as well as induce a hedonic response that either stimulate likeness or distaste for the food, regardless of the presence (or not) of a homeostatic need for food (Smeets et al., 2010; Berthoud et al., 2011). Studies have revealed that taste stimulation acts directly on the brain reward system to drive eating behaviours, while in addition to taste cues, smell and/or texture stimuli provide conditioned reward signals (Hajnal and Norgren, 2008; Yamamoto

IP T

and Shimura, 2008). Three basic tastes have been described with regards to food preferences;

the sweet taste of sugar, the taste for fat or lipids in some species (Khan and Besnard, 2009),

SC R

and umami, which is the taste of glutamate and certain nucleotides that add a savoury flavour

to food (Yamaguchi and Ninomiya, 2000). While there may exist an innate preference for

U

sweet, fatty and umami tastes, generally, most flavour preferences are learned (Yamamoto

N

and Ueji, 2011; Sclafani and Ackroff, 2012) and can be enhanced by the postoral actions of

A

sugars (Myers and Sclafani, 2001) or other nutrients. The hedonic response to the

M

consumption of fat derives from an innate preference modulated by orosensory factors and learned preferences that can be modulated by postoral factors (Ackroff and Sclafani, 2010).

ED

Studies have revealed that in humans (Drewnowski et al., 1992) and rodents (Takeda et al., 2000; Yoneda et al., 2007a. Yoneda et al., 2007b), a conditioned flavour, taste or place-

PT

preference for fat exists. This preference also increases with higher concentrations of fat (Figlewicz and Benoit, 2009; Manabe et al., 2010). Some fats have also demonstrated the

CC E

presence of a concentration-dependent conditioned place-preference (Figlewicz, 2004; Imaizumi et al., 2000) and reinforcing effect in an operant task in rodents.

A

1.6

Is food addiction a disease, or the consequence of disease? Several scientists and researchers have questioned the validity of the term ‘food

addiction’, with scientific literature for and against the validity of the construct, as well as a large body of work examining the clinical presentations and neuroscience of food addiction (Randolph, 1956; Gearhardt et al., 2009, 2011a, 2011b; Davis et al., 2011; Ziauddeen and

14

Fletcher, 2013; Brownell and Gold, 2013; Hebebrand et al., 2014; Pursey et al., 2015; LermaCabrera et al., 2016). While evidences for its existence and implications in humans are still evolving, the possibility of food addiction being a cause, co-morbidity or possibly a consequence of obesity is still being debated. Its relationship to the development of compulsive eating and/or obesity has also been queried (Ziauddeen and Fletcher, 2013). A

IP T

number of researchers have suggested that while arguments and evidences showing that

obesity may indeed result from an addictive tendency to consume food (Davis and Carter,

SC R

2009; Gearhardt et al., 2009), caution against solely attributing the development and

neurobiology of obesity to food addiction should be exercised (Volkow et al., 2012). There

U

have also been suggestions that food addiction may correlate more with binge eating-

N

disorder (Davis et al., 2011; Avena et al., 2012a; Gearhardt et al., 2012) which is

A

characterised by excessive consumption of highly palatable food within short periods of time,

M

followed by loss of control over eating (Velázquez-Sánchez et al., 2015). Rats exposed to an operant limited access to a sucrose diet have an escalation in their response to the diet and

ED

show excessive food intake within a short period of time (Blasio et al., 2013; Cottone et al., 2012; Velazquez-Sanchez et al., 2014).

PT

The conditioned place or flavour-preference paradigm has been applied widely to study the reward value of a number of drugs of abuse (Tzschentke 1998, 2007). One of the advantages

CC E

of the paradigm is that it allows the assessment of the reward value of substances over a relatively brief exposure period, thereby rapidly offering insights into their additive

A

potentials. The conditioned place preference (CPP) utilises the pairing of a conditioned stimulus (chamber) with an unconditioned stimulus (administered drug or flavour) to unmask the behavioural response to stimulation (or otherwise) of the brain reward system (Watanabe, 2013). This paradigm usually involves a two-choice test that ascertains the flavour preference for two flavours; one (the conditioned stimulus) which is paired with a nutrient that is orally

15

consumed or infused via a post-oral route such as the intragastric infusion and a second flavour (the unconditioned stimulus) which is paired with a nonnutritive source ((Sclafani and Ackroff, 2012). In a rat model of binge-eating, an increased conditioned place preference, heightening of food-seeking behaviour and inflexible response to palatable food were observed (Velázquez-Sánchez et al., 2015).Presently, there is no sufficient scientific evidence

IP T

to support the classification of food addiction as a disease entity; neither can a conclusion be reached on its precise relationship with other eating disorders. However, research into this

2.0

SC R

area continues to unfold.

Food additives and the stimulation of the brain reward system

U

The development of the food industry in the 21st century has allowed the creation and

N

modification of food and food additives, resulting in the production of large quantities of

A

highly-palatable foods that may have the ability to stimulate the reward centres of the brain.

M

An obvious up-side to the development and use of food additives is their inherent ability to enhance food-palatability and increase the consumption of low calorie foods that on their

ED

own are not palatable. This has become important to food scientists and researchers who have suggested that it may be possible (through food-reformulation) to produce foods that not only

PT

suppress appetite, but are also desirable to eat; in a bid to influence behavioural change and facilitate the consumption of healthier low-energy foods. The possible down-side to the use

CC E

of food additives may have been reiterated by food critics and a number of other scientists (Yang, 2010; Cargil, 2017) who are of the opinion that the inclusion of food additives (which

A

have the tendency to stimulate the reward centres and/or increase DA secretion, especially in brain regions that modulate the reward system) in foods would result in overeating and/or compulsive eating (which occur with food addiction), ultimately predisposing to obesity or binge eating disorder. However, there is a dearth of scientific literature on the possible addiction-like potential of the commonly-available food additives (when administered alone

16

or especially when added to foods) either in humans or animals. Hence, the debates for and against food additives continue. In the remaining sections of this review, we examine available scientific literature for and against the possible addictive potential of some common food additives (flavour enhancers and food sweeteners), and the implications (if any) for human health and well-being. Flavour enhancers

IP T

2.1

Flavour is the sensory impression or biological perception of food or food substances,

SC R

which is determined primarily by perceptions from the chemical senses of taste. The

perception of flavours depends on structure, texture and oral manipulation of food. The

U

flavour of the food can be modified with the use of flavours or flavour enhancers. Flavourants

N

or flavouring agents include all types of natural or synthetic mixtures used for impacting a

agents

are

now

available

to

the

modern

food

industry,

M

flavouring

A

defined flavour or enhancing an inherent flavour. A wide range of natural and synthetic

with monosodium glutamate and inosine monophosphate being common examples of

ED

culinary flavour enhancers. 2.1.1 Monosodium glutamate

PT

Monosodium glutamate (MSG), a sodium salt of glutamate, is found in various concentrations in numerous food products. Glutamate (either in the non-protein or protein-

CC E

bound forms) has been a part of culinary flavour component of human diets for many years. MSG’s effect in humans (Geha et al., 2000; He et al., 2008) and rodents have been evaluated

A

by a number of studies (Falalieieva et al., 2010; Leshchenko et al., 2012; Onaolapo OJ et al., 2011, 2015, 2016a, 2016b; Onaolapo AY et al., 2013). MSG is a widely-available culinary flavour enhancer approved by food-regulating agencies all over the world for use in improving the palatability of foods and food substances. In recent times, there have been suggestions of a possible addictive potential associated with its use; and this has continued to

17

be the focus of health and nutrition debates, and the central theme in a number of nutrition and health-related blogs (Moskin, 2008; Blaylock, 2014). MSG is the prototype of the umami taste sensation (Hayakawa and Kawai, 2003), the fifth basic taste, which evokes the taste of glutamate and similar nucleotides present in animal protein, vegetables and cured foods like cheese. Its savoury flavour (McCabe and Rolls,

IP T

2007) has been used to improve the palatability and acceptability of a wide variety of foods

and food substances (Yamaguchi and Kumiko, 2000; Prescot, 2004) worldwide. The

SC R

consumption of MSG has been reported to be in excess of 2.5 million tons per year, with

suggestions that this widespread use may be due to its role as a flavour enhancer as well as its

U

possible physiological and nutritional role in metabolism (Torii, 2013). The flavourenhancing property (Mathey et al., 2001) and possible low satiety value (Rogers and

A

N

Blundell, 1990) have been suggested as important factors in MSG’s potential for sustaining

M

higher energy intake (Masic and Yeoman, 2013). There have also been reports suggesting that MSG, possibly through its effects at the umami taste receptors may cause the detection of

ED

certain nutrients and therefore enhance their consumption (Bachmanov et al., 2009). Studies have however revealed that diets supplemented with MSG are associated with a long-term

PT

maintenance of stable energy intake (Schiffman et al., 1994; Essed et al., 2007), and that MSG, by its interaction with macronutrients like proteins has the potential to enhance satiety

CC E

over time (Viarouge et al., 1992). MSG has been reported to increase hedonic stimulation, hence, it may have the potential to stimulate appetite and/or food intake, and it may also help

A

to maintain stable energy levels (Yeomans, 1996; Masic and Yeomans, 2013). Debates regarding the ability of MSG to cause overeating or induce compulsive eating in predisposed individuals have continued till date, with a consensus that individuals differ in their perception of the umami or glutamate taste (Lugaz et al., 2002; Raliou et al., 2009). These variations, which may be genetic (Chen et al., 2009; Shigemura et al., 2009; Raliou et al.,

18

2009) has also been demonstrated in rodents (Ninomiya et al., 1992). However, the possible effects MSG on induction of satiety and satiation are yet to be determined (Masic and Yeomans, 2013). It is also not known if the variations in taste perception of glutamate induces or modulates glutamate preference or intake (Bachmanov et al., 2009). Also, there have been suggestions that variations in umami taste responsiveness may be associated with

IP T

postingestive effects (Bachmanov et al., 2009) that may be mediated by different

mechanisms. Such mechanisms include nutritive value (which is dependent on the energy

SC R

generated from its catabolism), stimulation of vagus activity, the release of hormones and

changes in gastrointestinal secretion of peptides (Niijima, 1991; Wu, 1998; Brosnan, 2000;

U

Kondoh et al., 2000).

N

While examining the influence of MSG on appetite (by studying the effects of consuming

A

soups of varying concentrations of MSG and macronutrient content) in 24 non-obese males,

M

Masic and Yeomans (2013) reported that: a) food-added MSG significantly increased palatability which was associated with a smaller decrease in hunger immediately after

ED

ingestion of soup b) there was a reduction in the feeling of fullness immediately after ingestion of the high-protein soup c) the MSG added to protein soup resulted in a lower

PT

increase in satiety compared to MSG added to carbohydrate, and no MSG added soups conditions d) the increase in hunger and the decrease in the feeling of satiety that were

CC E

observed in the subsequent 2 hour period were significantly lower in the individuals that had MSG conditioned with the protein than the groups with no MSG added to protein soup within

A

30 and 60 min post-ingestion. They concluded that MSG may have a bi-phasic effect on appetite, a hedonic satisfaction (Yeomans, 1996, Masic and Yeomans, 2013) that reduces satiation and stimulates appetite and intake, and a potential for enhancing post-ingestive satiety, particularly in relation to protein ingestion (Masic and Yeomans, 2013).

19

MSG’s role as a flavour-enhancer ordinarily suggests that its presence in foods will enhance consumption, which may result in weight gain. However, inspite of MSG’s ability to enhance preference for itself and other flavours related to it (or elicit a conditioned preference), the reports of its effects on body weight remain conflicting. There have been studies suggesting an increase (Oleksandra et al., 2014), a decrease (Kondoh et al., 2008; Onaolapo OJ et al,

IP T

2015, 2016a), or no effect (Onaolapo AY et al., 2012; Tordoff, et al., 2012). Studies in which MSG was administered to neonatal rats reported weight gain (Oleksandra et al., 2014). In

SC R

studies using adult rodents, no change in body weight was observed when MSG is added to

food or water (Tordoff, et al., 2012). However, when administered by gavage, a dose-related

U

weight loss has been observed (Onaolapo OJ et al, 2015, 2016a).

N

Other issues that have been raised include the possibility of MSG inducing reward or

A

addiction (Onaolapo et al., 2017), and till date, studies continue to examine the possible

M

rewarding or addiction-like effects of oral MSG in rodents (Bachmanov et al. 2000, 2009, Ackroff et al. 2012; Ackroff and Sclafani, 2013, 2016; Onaolapo OJ et al., 2017). It has been

ED

shown that MSG has a post-oral reward component, evidenced by low MSG intakes and the lack of expression of preference for MSG, relative to water (in MSG-naïve C57BL/6J mice)

PT

on exposure to ascending concentrations of MSG. An increase in intake and preference across a range of MSG concentrations was also observed in MSG-conditioned C57BL/6J mice

CC E

(Bachmanov et al. 2000; Ackroff et al. 2012). The enhancement of MSG preference (with experience) has led to suggestions that a flavour-conditioning process may be involved,

A

which could be related to the post-oral effects of MSG, which increases flavour preference due to its association with rewarding effects that are detected in the gut (Ackroff et al. 2012; Sclafani and Ackroff 2012). There are also studies that have revealed that MSG reinforces (through conditioning) preferences for its own flavour and flavours related to it (Ackroff and Sclafani, 2013). MSG (at increasing concentrations) was associated with the induction of

20

conditioned taste preference for either MSG itself or MSG-paired flavours (Bachmanov et al. 2000, 2009, Ackroff et al. 2012; Ackroff and Sclafani, 2013, 2016). Also observed in a number of these studies were the strain differences in MSG intake, preference and flavourconditioning (Ackroff and Sclafani, 2013, 2016). In a recent study, Onaolapo et al. (2017) examined the effects of oral MSG in MSG-naïve (no prior MSG exposure) and MSG-

IP T

pretreated (administered increasing doses of MSG daily for 21 days via an oral cannula, prior

to conditioning) mice exposed to the conditioned place preference paradigm (CPP paradigm).

SC R

The results obtained suggested that the administration of MSG was rewarding, and an

association was learnt, as both MSG-naïve and MSG-pretreated mice showed ‘MSG-paired’

U

chamber preference. However, pre-treatment with MSG potentiated MSG’s ability to induce

A

2.1.1.i Glutamate and the brain reward system

N

CPP (Onaolapo OJ et al., 2017).

M

There have been reports (Tzschentke and Schmidt, 2003; Kawasaki et al., 2005; He et al., 2014) that suggest that endogenous brain glutamate plays a role in the development and

ED

maintenance of drug addiction. This could possibly be related to its effects on processes such as reinforcement, sensitisation, reinforcement learning and habit learning (Tzschentke and

PT

Schmidt, 2003). The dopaminergic projections that link the different brain regions (VTA to the NAc, and the VTA to the PFC) are crucial pathways of the reward system, through which

CC E

many drugs of addiction act. The glutamate system is believed to have a modulatory role on the activities of the dopaminergic system in the evolution and sustenance of drug addiction

A

(Tzschentke and Schmidt, 2000). However, this does not mean that exogenous glutamate would have similar effects on the brain reward system. Researchers examining the mechanisms involved in glutamate consumption or its effect on appetite have suggested that it may be related to the chemosensory perception of its taste and/or its postingestive effects (Bachmanov et al., 2009). Consumption of MSG has been linked to a direct or indirect

21

increase in brain stimulation. Kondoh et al. (2009) demonstrated that a luminal presence of glutamate results in the stimulation of gut L-glutamate receptors that activates vagal afferent nerve fibres, which via vagal inputs then go on to stimulate brain regions (like the habenular nucleus) which exerts an influence on other regions of the brain. Functional magnetic resonance imaging and blood oxygen level dependent (BOLD) signals have shown that an

IP T

intragastric load of MSG results in the activation of brain regions such as amygdala, insular cortex, lateral hypothalamus, medial preoptic area and the dorsomedial hypothalamus

SC R

(Tsurugizawa et al., 2009; Uematsu et al., 2009; 2010; Tsurugizawa and Torii, 2010).

Hence, the luminal presence of glutamate is linked to its ability to influence the brain regions

U

that modulate reward. However, despite the fact that some studies may have demonstrated

N

that MSG has the potential to stimulate the brain reward system, its ability to ultimately

A

induce addiction (or potentiate the addictive potential of other food macronutrients) still

M

needs further evaluation.

2.1.2 Purine nucleotides: inosine monophosphate

ED

Purine nucleotides like inosine monophosphate and guanosine monophosphate have both health and economic benefits in the food and biotechnology industries, where they have

PT

found value as food flavouring agents that also have nutritional and pharmaceutical properties (Ledesma-Amaro et al., 2013). The umami taste sensation is also the taste quality of inosine

CC E

monophosphate (IMP), guanosine monophosphate (GMP) and a few other L-amino acids. IMP and GMP have flavour enhancing capabilities via exerting synergistic effects in

A

enhancing the umami flavour when combined with MSG (Jinap and Hajeb, 2010). Studies have also revealed that these purine nucleotides (inosine and guanosine) have health benefits that are related to their antioxidant, cardiotonic, immunomodulatory and neuroprotective properties (Aviado, 1983; Hasko et al., 2004; Shen et al., 2005; Tsuda, 2005).

22

IMP was discovered in 1913 when it was found that the active ingredient in dried bonito (which is traditionally used amongst the Japanese to make dashi, a soup stock) was 5′inosinate (salt of 5′-inosinic acid) (Kodama, 1913; Kurihara, 2015). IMP and GMP are synthesised industrially by microbial fermentation, using either RNA extraction with subsequent breakdown into free nucleotides; or by improved metabolic biosynthesis with

IP T

excretion of the nucleosides into the culture medium, and subsequent enzymatic or chemical

phosphorylation (Ledesma-Amaro et al., 2013; 2015). In vivo, IMP is the first nucleotide to

SC R

be formed during purine synthesis and is present in nucleic acids and adenosine triphosphate, which are the body’s chemical energy store in muscles and other tissues (Watterson et al.

U

2007).

N

IMP is a colourless, odourless, white crystalline compound that is usually marketed alongside

A

MSG. IMP (like MSG) elicits the umami taste sensation in humans and rodents, via its ability

M

to stimulate gustatory cells in taste buds and interact with taste receptors like T1R1 and T1R3 heterodimers (Ninomiya et al., 2000; Nelson et al., 2002). IMP and MSG are marketed

ED

together because studies have shown that compounds that elicit umami display synergism (Kumazawa and Kurihara, 1990; Yamaguchi and Ninomiya, 2000). There is scientific

PT

evidence demonstrating that the addition of 0.5 mM IMP to MSG resulted in an increase in preference scores without significantly affecting detection thresholds in food (Schiffman et

CC E

al., 1994). Zhang et al. (2008) reported that the combination of MSG and IMP augmented the umami taste sensation via stimulation of the T1R1 and T1R3 receptors in the oral cavity. de

A

Araujo et al. (2003) showed that both compounds are recognised in the brain as the same taste. Tsurugizawa et al. (2011) reported that the gastrointestinal stimulation resulting from an intragastric load of MSG and IMP induced BOLD signals in the same brain regions, but with differences in the time course of the BOLD response. They also reported that while MSG and IMP induced BOLD signals in the lateral hypothalamus, nucleus of the solitary

23

tract and insular cortex, only MSG increased the BOLD signals in the amygdala. This observation led to the suggestion that the mechanism of perception of MSG and IMP in the gastrointestinal tract differed from that of the taste-sensing system of the oral cavity (Tsurugizawa et al., 2011). Questions regarding the possible effects of the MSG/IMP synergism on appetite and satiety

IP T

have been raised, with only a few studies examining this effect. Masic and Yeomans (2014)

concluded that in addition to increasing the flavour pleasantness of foods, a combination of

SC R

MSG/IMP (when added to a low-energy preload like a high protein diet) elicited a biphasic

effect. First, by stimulating appetite during ingestion (an appetiser effect), and secondly, by

U

enhancing postingestive satiety.

N

There is a dearth of scientific literature examining the possible rewarding or addiction-like

A

effects of IMP or MSG/IMP combinations, either in humans or rodents. However, a number

M

of studies have examined the effects of dried bonito dashi (a broth that is made from dried bonito tuna, which is frequently used in Japanese cuisine) that contains IMP amongst other

ED

amino acids. The taste component of dashi per 100g stock includes about 108 mg inosinate and glutamate (Honda et al., 2006). There are suggestions that the taste preference for dashi

PT

solution in rats is not solely linked its umami-salty taste (Kawasaki et al. 2008). The results of a conditioned taste aversion experiment in mice also revealed that the dashi flavour includes

CC E

the five recognised basic taste qualities (Delay and Kondoh 2013). There is also evidence suggesting that both olfactory and taste stimuli are needed, but not exclusive to the induction

A

of the dashi preference (Kawasaki et al. 2008) in rodents, and that post-oral nutritive feedback enhances dashi preference (Kondoh et al. 2012a, 2012b). Ackroff et al (2014) examined the effect of dashi on reward, using a series of three experiments. The first experiment showed no evidence for postoral dashi preference following an intragastric selfinfusion, while the second experiment showed an inability of the preferred dashi solution to

24

condition a preference when orally consumed. In the third experiment using taste-impaired genetically modified mice, it was observed that dashi preference in mice depended upon the Trpm5 signalling system which mediates the taste cell response to umami, sweet and bitter tastes. Although C57BL/6J mice did not acquire preferences for dashi-paired flavours, oral exposure to 0.1% inosinate enhanced subsequent dashi preference. However, the

IP T

concentration of IMP and other umami components in dashi may not be sufficient to induce post-oral flavour conditioning (Ackroff et al., 2014). Studies using the CPP have

SC R

demonstrated that dashi is able to mediate reward only in combination with a nutritive

sweetener like dextrin (Kawasaki et al. 2011). This suggests that dashi may be an orally-

U

attractive substance without an inherent post-oral reward component. It could be deduced

N

from the foregoing that while IMP and/or MSG may be able to condition flavour or taste-

A

preference, in food, they are usually present in concentrations that are insufficient to

2.2

M

stimulate the brain reward system. However, more studies are needed to validate this theory. Food sweeteners

ED

Sweeteners are food additives which are used to impart a sweet taste in food and food substances. They elicit pleasurable sensations, in addition to providing nutritive or non-

PT

nutritive energy. Concerning their regulation, nutritive sweeteners like sucrose or sucrose substitutes (monosaccharide, disaccharide and polysaccharide polyols) are generally

CC E

recognised as safe (GRAS), or have petitions filed for GRAS. Sugar is a term that refers to sweet, soluble carbohydrates, which are food additives used to

A

sweeten food and food substances. There are different forms of sugars, with simple sugars (also known as monosaccharides) including glucose, fructose and galactose. Another type of sugar is the disaccharides, including sucrose (which is commonly available as either granulated sugar or table sugar), maltose and lactose. Oligosaccharides or polysaccharides are sugars with longer chains. Sucrose is present in all green plants, where it is an early product

25

of photosynthesis and the main agent for the translocation of carbon to the rest of the plant. In sugarcane and sugar beet, high concentrations of sugars are found which is sufficient for commercial extraction. In the last few decades, there are questions regarding the health implications of diets that are high in sugars, more especially refined sugars. Although studies examining the health impacts remain inconclusive, excessive use of sugar has been linked to

IP T

an increased risk of cardiovascular disease (Brown et al., 2008; Stanley et al., 2009), diabetes

mellitus (Apovian, 2004; Gross et al., 2004), obesity (de Ruyter, et al., 2012), cognitive

SC R

decline (Lakhan and Kirchgessner, 2013; Chiavaroli et al., 2014) and tooth decay (Zero et al., 2009; Jacob et al., 2016). More recently, diets that are high in sugars have been implicated in

U

the development of food addiction or specifically sugar addiction, and obesity.

N

Polyols or sugar alcohols are nutritive, low calorie, digestible carbohydrates that have been

A

associated with the addition of sweetness to foods without significantly increasing energy.

M

Polyols are obtained from the substitution of an aldehyde group with a hydroxyl (Shankar et al., 2013). Examples of polyols include hydrogenated monosaccharides (mannitol, sorbitol),

ED

hydrogenated disaccharides (isomalt, lacitol, maltitol) and hydrogenated starch hydrolysates (Shankar et al., 2013; Grembecka, 2015). There are also mixed compounds like polydextrose

PT

which is a combination of corn sugar and sorbitol. Polyols tend to occur in small quantities in animals and plants (Mäkinen, 1984). Acceptable daily intake (ADI) levels are yet to be

CC E

specified for these polyols. However, when consumed in large quantities, they have been associated with potent laxative effects and gastrointestinal symptoms like abdominal

A

discomfort, bloating and flatulence (Grabitske and Slavin, 2008; EFSA, 2011). In addition to their ability to impart a sweet taste, polyols have been shown to improve the texture, holding moisture and cooling sensation of food products in the mouth, as well as serve as preservatives (Health Canada, 2005). Polyols also have prebiotic and anti-cariogenic effects, and help with the normalisation of intestinal function (Grabitske and Slavin, 2008; EFSA,

26

2011). They have also been associated with weight loss and reduction in energy intake, due to their lower caloric value (Grabitske and Slavin, 2008). They are recommended for use in diabetics because they induce only a small increase in blood glucose or insulin secretion (Nakamura, 2005; Grabitske and Slavin, 2008; Wheeler and Pi-Sunyer, 2008; EFSA, 2011).

from about 25 to 100 %, compared with sucrose (Grembecka, 2015).

IP T

Erythritol and xylitol are widely used as bulk sweeteners. The sweetness of polyols varies

Six non-nutritive sweeteners (acesulfame-K, aspartame, neotame, saccharin, sucralose and

SC R

stevia) with intense sweetening potential are approved and classified as generally recognised as safe (GRAS) for use in humans by the United states Food and Drug Administration (FDA)

U

and the European Food Safety Authority (FDA, 2015; EFSA, 2015). Siraitia Grosvenorii

N

Swingle Fruit Extract (SGFE) known as Luo Han Guo in Chinese, is a traditional Chinese

A

herbal-based sweetener which was also granted GRAS status by the FDA (FDA, 2014). Two

M

others which have been approved for use in other regions of the world, but not in the United States of America are alitame (approved for use in Australia, China, Mexico and New

ED

Zealand) (Ellis, 1995; Australia New Zealand Food Authority, 2002) and cyclamate ( used in over fifty countries). Non-nutritive sweeteners (NNS) are usually about 30–13000 times

PT

sweeter than sucrose (Shankar et al., 2013), and have established acceptable daily intake limit currently available for each of them. However, there are ongoing debates on the safety of

CC E

(NNS), with a number of studies examining the possible effects of the different NNS in humans or rodents (Weihrauch and Diehl, 2004; Akihisa et al., 2007; Magnuson et al., 2007;

A

Lin and Curhan, 2011; Gardner et al., 2012; Pepino et al., 2013; Liu et al., 2013; Suez et al., 2014). Aspartame is made up of two amino acid isolates (phenylalanine, aspartic acid) and methanol in a ratio of 5:4:1. It also breaks down into these components on ingestion. The central effects of aspartame have been credited to alterations in the concentrations of catecholamines

27

such as epinephrine, norepinephrine and dopamine (Coulombe and Sharma, 1986). In recent studies examining the effects of increasing doses of aspartame in mice, dose-related behavioural, biochemical and morphologic changes involving different regions of the brain (Onaolapo AY et al., 2016, 2017a, 2017b) were observed. Acesulfame-potassium (acesulfame) is a white crystalline powder and a potassium salt of 6-methyl-123-axathiazine-

IP T

4(3H)-one 2, 2-dioxide. It has a bitter after-taste when used alone as a sweetener (Horne et al.

2002), so it is usually combined other sweeteners (sucralose or aspartame) allowing the

SC R

sweeteners mask each other’s after-taste, with an increased sweetness derived. Acesulfame is

excreted unmetabolised in humans; therefore, in addition to being a non-caloric sweetener, it

U

does not alter potassium metabolism (ADA, 2004). Sucralose is derived from a process that

N

substitutes 3 chloride atoms for 3 hydroxyl groups on the sucrose molecule. The bulk of

A

sucralose is not metabolised in humans and as it is excreted in the faeces unchanged, and

M

while 14–20% of it is absorbed from the gastrointestinal tract, this is also excreted in the urine within 5 days (Roberts et al., 2000; John et al., 2000).

ED

2.2.1 Nutritive Sweeteners (Sugars and modified-sugars) and food addiction Food sweeteners have the ability to increase palatability and consumption of nutrient-

PT

dense foods or beverages, thereby promoting a healthy diet. However, questions continue to be raised over the possible effects of food sweeteners on appetite, satiety, brain reward

CC E

system and food addiction. Studies investigating the construct ‘sugar addiction’ have been conflicting in their reports, with some researchers and food scientists asserting that

A

consumption of high sugar diets or sugar stimulates the brain reward system in ways that are similar to drugs of addiction (Avena et al., 2008b).Whereas, others have suggested that sugar addiction does not occur in humans (Hedebrand et al., 2014; Westwater et al., 2015). Studies have shown that sugar is easily detected by the sweet taste receptors in the oral cavity, and it elicits a strong taste or flavour preference in many animals (Bachmanov et al., 2011). Also,

28

sugars can be detected by gut chemosensors, as well as chemosensors in post-absorptive sites (like liver, pancreas and brain), providing a possible source of the positive feedback signals of sugar-conditioned flavour preferences. In humans and rodents, the sweet taste sensation of sugar has been linked to stimulation of the heterodimeric T1R2+T1R3 sweet receptor which are located in taste buds on the tongue and palate (Bachmanov et al., 2011; Treesukosol et al.,

IP T

2011). This activates an intracellular signalling mechanism that is coupled to the inositol 1,

4, 5-trisphosphate receptor 3, G protein α-gustducin phospholipase Cβ2 and the transient

SC R

receptor potential cation channel Trpm5 (Chaudhari and Roper, 2010). Intragastric or intraduodenal infusion of sugars has been associated with a post-oral enhancement of the

U

conditioned flavour-preference in rodents (Sclafani and Glendinning, 2005) and humans

N

(Yeomans et al., 2008). Once developed, conditioned flavour-preference for sugars has been

A

observed to persist for days, and is also resistant to extinction (Myers, 2007). This suggests

M

that conditioning to sugar produces a long-lasting increase in the reward value (Sclafani and Ackroff, 2012). The mechanism by which sugars may cause or potentiate food addiction has

ED

been discussed in section 1.4.1.

In the light of studies that have demonstrated the possible deleterious metabolic effects of

PT

excessive consumption of sugar and its ability to increase the risk of addiction to foods; there has been an increase in the advocacy for the substitution of sugar with either nutritive low

CC E

calorie sweeteners like polyols, or non nutritive sweeteners like aspartame. However, there are still debates on the possible effects that these sugar substitutes may exert on appetite and

A

satiety (or their ability to influence the brain reward system), since they also stimulate the same sweet taste receptors as sugars, and may therefore elicit the same postingestive and/or post-absorptive effects. While there is a dearth of scientific literature examining the ability of sugar alcohols to condition preferences or activate the brain reward circuit, studies have evaluated the effects

29

of polyols (xylitol, lacitol) and glucose polymers like polydextrose (PDX) on appetite and satiety (Gee and Johnson, 2005; King et al., 2005; Olli et al., 2011; Kumar et al., 2012; Ibarra et al., 2016). PDX has been shown to increase satiety when administered as a supplement about 60-90 minute before an ad libitum lunch (exerting no such effect when administered with breakfast) and it also reduces energy intake (Hull et al., 2012; Astbury et al., 2013;

IP T

Ranawana et al., 2013). It has also been associated with enhanced satiety and satiation when

it replaced 30% of the other available carbohydrates in a diet (Konings et al., 2014).

SC R

However, effects of supplementation of lower concentrations of PDX on appetite and satiety

were inconsistent (King et al., 2005; Schwab et al., 2006; Astbury et al., 2013). PDX is high

U

in fibre and this is believed to be partly responsible for its effects on appetite and satiety,

N

although other factors have also been considered. A recent study examining the effects of a

A

PDX-supplemented meal on appetite regulation in obese participants, reported that PDX-

M

supplemented diet enhanced satiety and reduced hunger after a high-fat diet (Olli et al., 2011).These effects were associated with stimulation of the postprandial secretion of

ED

glucagon-like peptide 1, a satiety hormone (Olli et al., 2011). Other studies have also associated PDX with increase in the secretion of glucagon-like peptide 1 and peptide YY, and

PT

lower ghrelin levels in lean men (Astbury et al., 2014). The impact of polyols on satiety and satiation has also been reported (Kumar et al., 2012). Polyols have been observed to enhance

CC E

satiety, stimulate the secretion of glucagon-like peptide1, and increase acute postprandial concentrations of plasma peptide YY (Massimino et al., 1998; Gee and Johnson, 2005;

A

Lesniewska et al., 2006). Polyols also delay gastric emptying, which has been suggested to be partly responsible for their effects on satiety. 2.2.2 Non-Nutritive Sweeteners and food addiction There is increasing evidence to suggest that artificial sweeteners (like aspartame and saccharin) activate the food reward circuit, using pathways that differ from those used by

30

sugar and other natural sweeteners. NNS are low calorie sweeteners, so this could possibly eliminate the postingestive component (Yang, 2010) that is dependent on metabolic products of the food (Sclafani and Ackroff, 2004). Therefore, NNS activate the gustatory component of the reward pathway differently from caloric sweeteners. The ingestion of sucrose (compared to saccharin) was associated with an increased activation of higher gustatory areas

IP T

like the orbitofrontal cortex, insula and amygdala (Haase et al., 2009). Imaging studies comparing the effects of sucrose and sucralose on central taste pathways demonstrated that

SC R

although both sucrose and sucralose activated primary taste pathways, sucrose elicits a

stronger brain response in the anterior insula, frontal operculum, striatum and anterior

U

cingulate, compared to sucralose (Frank et al., 2008). Also, only sucrose stimulates

N

dopaminergic midbrain areas, suggesting that the brain probably distinguishes caloric from

A

non-caloric sweeteners (Frank et al., 2008). Functional imaging studies (in male subjects with

M

normal weight) following sucralose ingestion also revealed an absence of the prolonged hypothalamic signal depression that was observed with glucose ingestion (Smeets et al.,

ED

2005). The results obtained from a human study also suggested that habitual use of NNS was associated with alterations in reward-processing of sweet taste and modulation of

PT

consumption (Green and Murphy, 2012). Rudenga and Small (2012) also tested the hypothesis that sweet taste may produce a differential response in brain regions that signal the

CC E

post-ingestive effects of flavours. They observed an inverse relationship between amygdala response to sucrose and artificial sweetener use, and a trend towards a similar effect in the

A

insula. These findings also support suggestions that experiencing sweet taste without calories degrades the predictive capacity of sweet sensation in the amygdala and insula (Davidson and Swithers, 2004). Studies have evaluated the effect of NNS on appetite, satiety and weight, with conflicting reports. There have been suggestions that because NNS are sweet, they could potentiate

31

sugar-craving and sugar dependence (Yang, 2010) leading to overeating and weight gain. A number of studies have suggested that acute administration of NNS in vehicles that are energy deficient (water or chewing gum) result in an increase in hunger compared to vehicle alone (Black et al., 1993; Blundell and Hill, 1986; Rogers et al., 1988; Tordoff et al., 1990). However, when administered with high energy vehicle, no alteration of hunger, relative to

IP T

vehicle alone or vehicle sweetened with sucrose (Rolls et al., 1989; Maone et al., 1990; Drewnowski et al., 1994) was observed. Some studies have however suggested that low

SC R

calorie NNS neither promote nor suppress appetite (Bellisle and Drewnowski 2007; Renwick and Molinary, 2010). A study evaluating the effects of NNS-containing beverages (compared

U

to sugar-sweetened beverages) on satiety, reported no difference in satiety ratings in children

N

over an 18-month intervention trial (de Ruyter et al. 2013); similar effects were observed in

A

adults in another study (Anton et al., 2010). Studies in rodents have also demonstrated

M

preference for non nutritive sweeteners like stevia compared to water (Martínez et al., 2016), and conditioned flavour preference; although preference for nutritive sweeteners exceeded

ED

that observed for NNS (Sclafani and Ackroff, 2017). From the foregoing it can be said that NNS do not elicit the same effects on appetite, satiety, conditioned preferences or the brain

PT

reward circuit as observed with sugars. Also, they are not likely to potentiate the effects of

CC E

other foods or food substances. Conclusion

In conclusion, while the validity of the concept of food addiction continues to be debated,

A

studies have continued to show that deliberate alterations in the composition of foods can change the behavioural response to their consumption, due to differences in sensory perception. However, the scientific evidences in support of food addiction are not yet sufficient to categorically state that any particular food ingredient, micronutrient or standard food-additive is addictive.

32

Therefore, research needs to focus more on understanding the mechanisms by which food constituents may modulate the brain response to their consumption. Also, the probable longterm changes (in brain chemistry and structure) that may accompany the consumption of

IP T

certain foods or food-additives need to be further investigated.

Acknowledgement

SC R

This research did not receive any specific grant from agencies in the public, commercial, or not-for-profit sectors.

U

Conflict of interest:

Onaolapo AY declares no conflict of interest related to the content of this manuscript.

A

N

Onaolapo OJ declares no conflict of interest related to the content of this manuscript.

M

Ethical statement

This article does not contain any studies with human participants performed by any of the authors. All studies

ED

referenced in this article performed by the authors using animal were in compliance with international, national,

A

CC E

PT

and/or institutional guidelines for the care and use of animals.

33

References 1.

Ackroff K, Kondoh T. Sclafani A (2014) Dried Bonito Dashi: a preferred fish broth without postoral reward actions in mice. Chem Senses, 39, 159–166. Ackroff K, Sclafani (2016) A Flavor Preferences Conditioned by Dietary Glutamate. Adv

IP T

2.

Nutr.7: 845S-852S.

Ackroff K, Sclafani A (2010) Oral and postoral determinants of dietary fat appetite. In:

SC R

3.

JP M, J l.C., editors. Fat Detection: Taste, Texture, and Post Ingestive Effects. CRC

Ackroff K, Sclafani A (2013) Flavour preferences conditioned by oral monosodium

N

4.

U

Press; Boca Raton: Chapter 12.

Ackroff K, Weintraub R, Sclafani A (2012) MSG intake and preference in mice are

M

5.

A

glutamate in mice. Chem. Senses 38: 745–758. doi:10.1093/chemse/bjt049.

influenced by prior testing experience. Physiol Behav. 107:207–217. ADA: American Dietetic Association (2004)

Position of the American Dietetic

ED

6.

Association: use of nutritive and nonnutritive sweeteners. J Am Diet Assoc. 104:255-75. Ahmed SH, Kenny PJ, Koob GF, Markou A(2002) Neurobiological evidence for hedonic

PT

7.

allostasis associated with escalating cocaine use. Nat Neurosci. 5:625-6. Akihisa T, Hayakawa Y, Tokuda H, Banno N, Shimizu N, Suzuki T, Kimura Y (2007)

CC E

8.

Cucurbitane glycosides from the fruits of Siraitia gros venorii and their inhibitory effects

A

on Epstein-Barr virus activation. J Nat Prod. 70:783-8.

9.

Alcaraz-Iborra M, Carvajal F, Lerma-Cabrera JM, Valor LM, Cubero I (2014) Binge-like consumption of caloric and non-caloric palatable substances in ad libitum-fed C57BL/6J mice: pharmacological and molecular evidence of orexin involvement. Behav Brain Res. 2014 Oct 1; 272():93-9.

34

10. Alonso-Alonso M, Woods SC, Pelchat M, Grigson PS, Stice E, Farooqi S, et al (2015) Food reward system: current perspectives and future research needs. Nutr Rev. 73: 296– 307. doi: 10.1093/nutrit/nuv002. 11. Anton SD, Martin CK, Han Het al. (2010) Effects of stevia, aspartame, and sucrose on food intake, satiety, and postprandial glucose and insulin levels. Appetite 55: 37–43.

IP T

12. APA: American Psychiatric Association (2000) Diagnostic and Statistical Manual of Mental Disorders (fourth ed), American Psychiatric Association, Washington, DC.

SC R

13. Apovian CM (2004). Sugar-sweetened soft drinks, obesity, and type 2 diabetes. JAMA: The J.Am. Med. Association. 292: 978. doi:10.1001/jama.292.8.978.

U

14. Arias-Carrión O, Stamelou M, Murillo-Rodríguez E, Menéndez-González M, Pöppel E

N

(2010) Dopaminergic reward system: a short integrative review. International Archives

A

of Medicine 3:24 doi:10.1186/1755-7682-3-24

M

15. Astbury NM, Taylor MA, French SJ, Macdonald IA(2014) Snacks containing whey protein and polydextrose induce a sustained reduction in daily energy intake over 2 wk

ED

under free-living conditions. Am J Clin Nutr. 99:1131-40. 16. Astbury NM, Taylor MA, Macdonald IA (2013) Polydextrose results in a dose-

110934-42.

PT

dependent reduction in ad libitum energy intake at a subsequent test meal. Br J Nutr.

CC E

17. Australia New Zealand Food Authority (2002) Australia New Zealand Food Standards Code, Standard 1.3.1 Food Additives, Canberra (available at http://www.anzfa.gov.au)

A

18. Avena NM, Bocarsly ME, Rada P, Kim A, Hoebel BG (2008a) After daily bingeing on a sucrose

solution,

food

deprivation

induces

anxiety

dopamine/acetylcholine imbalance. Physiol Behav. 94:309–315.

35

and

accumbens

19. Avena NM, Rada P, Hoebel BG (2008b) Evidence for sugar addiction: Behavioral and neurochemical effects of intermittent, excessive sugar intake. Neurosci Biobehav Rev. 32: 20-39. 20. Avena NM, Gold JA, Kroll C, Gold MS (2012a) Further developments in the neurobiology of food and addiction: Update on the state of the science. Nutrition.

IP T

28:341–343.

21. Avena NM, Bocarsly ME, Hoebel BG (2012b) Animal models of sugar and fat bingeing:

SC R

relationship to food addiction and increased body weight. Methods Mol Biol. 829:35165.

U

22. 23. Avena NM, Talbott JR (2014) Why diets ail (Because you’re addicted to sugar):Science

N

explains how to end cravings, lose weight and get health. ISBN 978-1-60774-486-3

A

24. Aviado DM (1983) Inosine: a naturally occurring cardiotonic agent. J Pharmacol.

M

14:24

ED

25. Bachmanov AA, Bosak NP, Floriano WB, Inoue M, Li X, Lin C, Murovets VO, Reed DR, Zolotarev VA, Beauchamp GK (2011) Genetics of sweet taste preferences. Flavour

PT

Fragr J. 26:286-294.

26. Bachmanov AA, Inoue M, Ji H, Murata Y, Tordoff MG, Beauchamp GK (2009)

CC E

Glutamate taste and appetite in laboratory mice: physiologic and genetic analyses Am J Clin Nutr 90: 756S–763S.

27. Bachmanov AA, Tordoff MG, Beauchamp GK (2000) Intake of umami-tasting solutions

A

by mice: a genetic analysis. J Nutr. 130:935S–941S.

28. Barson JR, Leibowitz SF (2016) Hypothalamic neuropeptide signaling in alcohol addiction. Prog Neuropsychopharmacol Biol Psychiatry. 65:321-9. 29. Begg DP, Woods SC (2013) The endocrinology of food intake. Nat. Rev Endocrinol. 9:584-97. 36

30. Bellisle F (1999) Glutamate and the umami taste: sensory, metabolic, nutritional and behavioral considerations. A review of the literature published in the last 10 years. Neurosci Biobehav Rev. 23:423–38. 31. Bellisle F and Drewnowski A (2007) Intense sweeteners, energy intake and the control of body weight. European J. Clinical Nutr. 61: 691–700.

IP T

32. Bello NT, Lucas LR, Hajnal A (2002) Repeated sucrose access influences dopamine D2 receptor density in the striatum. Neuroreport. 13:1575-8.

SC R

33. Berridge KC (2012) From prediction error to incentive salience: mesolimbic computation of reward motivation. 35:1124–1143.

U

34. Berridge KC, Kringelbach ML (2015). Pleasure systems in the brain. Neuron. 86: 646–

N

664.

A

35. Berthoud HR (2011) Metabolic and hedonic drives in the neural control of appetite: who

M

is the boss? Curr. Opin. Neurobiol., 21:888-89.

36. Berthoud HR, Lenard NR, Shin AC (2011) Food reward, hyperphagia, and obesity. Am J

ED

Physiol Regul Integr Comp Physiol300:R1266-77. 37. Black RM, Leiter LA, Anderson GH (1993) Consuming aspartame with and without

PT

taste: differential effects on appetite and food intake of young adult males. Physiol Behav. 53:459–66.

CC E

38. Blasio A, Steardo L, Sabino V, Cottone P (2014) Opioid system in the medial prefrontal cortex mediates binge-like eating. Addict Biol. 19:652-62.

A

39. Blaylock MD (2014) MSG: The hidden addiction. Newsmax health, Newsmax.com, Assessed November, 2017.

40. Blundell JE, Hill AJ (1986) Paradoxical effects of an intense sweetener (aspartame) on appetite. Lancet 1:1092–3.

37

41. Blundell JE, Lawton CL, Hill AJ (1993) Mechanisms of appetite control and their abnormalities in obese patients. Horm Res. 39 S3:72–6. 42. Bocarsly ME, Powell ES, Avena NM, Hoebel BG (2010) High fructose corn syrup causes characteristics of obesity in rats: Increased body weight, body fat, and triglyceride levels. Pharmacol Biochem Behav 97:101–106. doi: 10.1016/j.pbb.2010.02.012.

IP T

43. Borengasser SJ, Kang P, Faske J, Gomez-Acevedo H, Blackburn ML, Badger TM. (2014). High fat diet and in utero exposure to maternal obesity disrupts circadian rhythm

SC R

and leads to metabolic programming of liver in rat offspring. PLoS one. 9:e8420-9.

44. Brief DJ, Davis JD (1984) Reduction of food intake and body weight by chronic

U

intraventricular insulin infusion. Brain Res Bull. 12:571-5.

N

45. Britt JP, Benaliouad F, McDevitt RA, S (2012) Synaptic and behavioral profile of

A

multiple glutamatergic inputs to the nucleus accumbens. Neuron. 76:790-803.

M

46. Brosnan JT (2000) Glutamate, at the interface between amino acid and carbohydrate metabolism. J Nutr 130:S988–90.

ED

47. Brown CM, Dulloo A G, Montani J-P (2008). Sugary drinks in the pathogenesis of obesity and cardiovascular diseases. Int. J. Obesity. 32: S28.

PT

48. Brownell KD, Gold MS (2012) Food and Addiction: A Comprehensive Handbook Oxford University Press, Oxford, UK.

CC E

49. Brownell KD, Gold MS (2013) Food and addiction: scientific, social, legal, and legislative implications. K.D. Brownell, M.S. Gold (Eds.), Food and Addiction, Oxford

A

University Press, Oxford, pp. 439-446.

50. Calabresi P, Picconi B, Tozzi A, Di Filippo M (2007) Dopamine-mediated regulation of corticostriatal synaptic plasticity. Trends Neurosci. 30:211-9. 51. Cargil K. (2017) Stop calling food addictive. Food science Opinion. The Guardian, Guardian News and Media Limited USA.

38

52. Carvajal F, Alcaraz-Iborra M, Lerma-Cabrera JM, Valor LM, de la Fuente L, SanchezAmate Mdel C, Cubero I(2015) Orexin receptor 1 signaling contributes to ethanol bingelike drinking: Pharmacological and molecular evidence. Behav Brain Res. 2015; 287():230-7. 53. Chaudhari N, Roper SD (2010) The cell biology of taste. J Cell Biol 190: 285– 296.

IP T

54. Chen Q-Y, Alarcon S, Tharp A, et al (2009) Perceptual variation in umami taste and polymorphisms in TAS1R taste receptor genes. Am J Clin Nutr 90: S770–9.

SC R

55. Chiavaroli L, HaV, de Souza RJM, Kendall CWC, Sievenpiper JL (2014) Fructose in

obesity and cognitive decline: is it the fructose or the excess energy? Nutrition Journal.

U

13: 27.

N

56. Cocores JA, Gold MS (2009) The Salted Food Addiction Hypothesis may explain

A

overeating and the obesity epidemic. Medical Hypotheses. 73:892–899.

M

57. Cohen P, Zhao C, Cai X, Montez JM, Rohani SC, Feinstein P, et al (2001) Selective deletion of leptin receptor in neurons leads to obesity. J Clin Invest. 108:1113-21.

ED

58. Colantuoni C, Schwenker J, McCarthy J, Rada P, Ladenheim B, Cadet JL, et al (2001) Excessive sugar intake alters binding to dopamine and mu-opioid receptors in the brain.

PT

Neuroreport. 12:3549-52.

59. Corwin RL, Grigson PS (2009) Symposium Overview—Food Addiction: Fact or Fiction?

CC E

J Nutr 139:617–619. doi: 10.3945/jn.108.097691.

60. Cottone P, Wang X, Park JW, Valenza M, Blasio A, Kwak J, et al (2012) Antagonism of

A

sigma-1 receptors blocks compulsive-like eating. Neuropsychopharmacol. 37:2593-604.

61. Dautan D, Huerta-Ocampo I, Witten IB, Deisseroth K, Bolam JP, Gerdjikov T, MenaSegovia J (2014) A major external source of cholinergic innervation of the striatum and nucleus accumbens originates in the brainstem. J Neurosci. 34:4509-18.

39

62. Davidson TL, Swithers SE (2004) A Pavlovian approach to the problem of obesity. Int J Obes Relat Metab Disord. 28:933-5. 63. Davis C, Carter JC (2009) Compulsive overeating as an addiction disorder. A review of theory and evidence. Appetite. 53:1–8.

‘food addiction’ is a valid phenotype of obesity. Appetite 57:711–717.

IP T

64. Davis C, Curtis C, Levitan RD, Carter JC, Kaplan AS, Kennedy JL (2011) Evidence that

taste in the human brain. J Neurophysiol. 90:313–319.

SC R

65. de Araujo IE, Kringelbach ML, Rolls ET, Hobden P (2003) Representation of umami

66. de Ruyter JC, Katan MB, Kuijper LD et al (2013) The effect of sugar-free versus sugar-

U

sweetened beverages on satiety, liking and wanting: an 18 month randomized double-

N

blind trial in children. PLoS ONE8: e78039.

A

67. de Ruyter JC, Margreet RO, Jacob CS, Martijn BK (2012). A trial of sugar-free or

M

sugar-sweetened beverages and body weight in children New England J. Med. 367: 1397–1406.

ED

68. Delay ER, Kondoh T (2015) Dried-bonito dashi: taste qualities evaluated using conditioned taste aversion methods in wild-type and T1R1 knockout mice. Chem Senses

PT

40:125–40.

69. Di Chiara G, Imperato A (1988) Drugs abused by humans preferentially increase

CC E

synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc Natl Acad Sci U S A. 85:5274-8.

A

70. Drewnowski A, Kurth C, Holden-Wiltse J, Saari J (1992) Food preferences in human obesity: carbohydrates versus fats. Appetite. 18:207-21.

71. Drewnowski A, Massien C, Louis-Sylvestre J, Fricker J, Chapelot D, Apfelbaum M. (1994) Comparing the effects of aspartame and sucrose on motivational ratings, taste preferences, and energy intakes in humans. Am J Clin Nutr 59:338–45.

40

72. EFSA (2011) Scientific opinion on the substantiation of health claims related to the sugar replacers

xylitol,

D-tagatose,

xylitol,

sorbitol,

mannitol,

maltitol,

lactitol,

isomalt,erythritol, D-tagatose, isomaltulose, sucralose and polydextrose and maintenance of tooth mineralisation by decreasing tooth demineralisation. EFSA Journal 9: 2076. 73. EFSA

European

Food

Safety

Authority

(2015)

IP T

www.efsa.europa.eu/en/topics/sweeteners.

Sweeteners.

74. Egecioglu E, Skibicka KP, Hansson C, Alvarez-Crespo M, Friberg PA, Jerlhag E, et al

SC R

(2011) Hedonic and incentive signals for body weight control. Rev. Endocr. Metab. Disord., 12:141-151

U

75. Ellis, J. W. (1995). Overview of Sweeteners. J. Chem. Edu. 72:671–675.

N

76. Elmquist JK, Bjorbaek C, Ahima RS, Flier JS, Saper CB (1998) Distributions of leptin

A

receptor mRNA isoforms in the rat brain. J Comp Neurol. 395(:535–547.

M

77. Epstein LH, Leddy JJ, Temple JL, Faith MS (2007) Food reinforcement and eating: a multilevel analysis. Psychol Bull133:884-906.

ED

78. Essed NH, van Staveren WA, Kok FJ, de Graaf C (2007) No effect of 16 weeks flavor enhancement on dietary intake and nutritional status of nursing home elderly. Appetite.

PT

48:29–36.

79. Falalieieva TM, Kukhars'kyĭ VM, Berehova TV (2010) Effect of long-term monosodium

CC E

glutamate administration on structure and functional state of the stomach and body weight in rats]. Fiziol Zh 56:102-10.

A

80. FDA (2014) Letter Notifying FDA for GRAS Status. 81. Figlewicz DP, Bennett J, Evans SB, Kaiyala K, Sipols AJ, Benoit SC (2004) Intraventricular insulin and leptin reverse place preference conditioned with high-fat diet in rats. Behav Neurosci. 118:479-87.

41

82. Figlewicz DP, Benoit SC (2009) Insulin, leptin, and food reward: update 2008. Am J Physiol Regul Integr Comp Physiol296:R9-19. 83. Finch DM (1996) Neurophysiology of converging synaptic inputs from the rat prefrontal cortex, amygdala, midline thalamus, and hippocampal formation onto single neurons of the caudate/putamen and nucleus accumbens. Hippocampus. 6:495-512.

IP T

84. Finucane MM, Stevens GA, Cowan MJ, Danaei G, Lin JK, Paciorek CJ, et al (2011)

National, regional, and global trends in body-mass index since 1980: systematic analysis

SC R

of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet. 377:557-567.

U

85. Flint AJ, Gearhardt AN, Corbin WR, Brownell KD, Field AE, Rimm EB (2014) Food-

N

addiction scale measurement in 2 cohorts of middle-aged and older women. American J.

A

Clin. Nutr. 99:578-586.

Sweeteners

Permitted

for

M

86. Food and Drug Administration 2015. Additional Information about High-Intensity use

in

Food

in

the

United

States.

ED

www.fda.gov/food/foodadditivesingredients. 87. Frank GK, Oberndorfer TA, Simmons AN, Paulus MP, Fudge JL, Yang TT, Kaye WH

PT

(2008) Sucrose activates human taste pathways differently from artificial sweetener. Neuroimage. 39:1559-69.

CC E

88. Frederich RC, Hamann A, Anderson S, Löllmann B, Lowell BB, Flier JS (1995) Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin

A

action. Nat Med. 1:1311-4.

89. Gardner C, Wylie-Rosett J, Gidding SS, Steffen LM, Johnson RK, Reader D, Lichtenstein AH (2012) Nonnutritive Sweeteners: Current Use and Health Perspectives A Scientific Statement from the American Heart Association and the American Diabetes Association. Diabetes Care 35: 1798–1808.

42

90. Gearhardt AN, Corbin WR, Brownell KD (2009) Preliminary validation of the Yale Food Addiction Scale. Appetite. 52:430-436. 91. Gearhardt AN, Davis C, Kuschner R, Brownell KD (2011b) The addiction potential of hyperpalatable foods. Current Drug Abuse Reviews 140-145:1874-4737/11 92. Gearhardt AN, Grilo CM, DiLeone RJ, Brownell KD, Potenza MN (2011a) Can food be

IP T

addictive? Public health and policy implications. Addiction 106:1208–1212

93. Gearhardt AN, White MA, Masheb RM, Morgan PT, Crosby RD, Grilo CM (2012) An

SC R

examination of the food addiction construct in obese patients with binge eating disorder. Int J Eat Disord. 45:657–663.

U

94. Gee JM, Johnson IT (2005) Dietary lactitol fermentation increases circulating peptide

N

YY and glucagon-like peptide-1 in rats and humans Nutrition.21:1036-43.

A

95. Geha RS, Beiser A, Ren C, Patterson R, Greenberger PA, Grammer LC, et al (2000) A.

M

review of alleged reaction to monosodium glutamate and outcome of a multicenter double-blind placebo-controlled study, J. Nutr. 130:1058S-62S.

ED

96. Goldstein RZ, Volkow ND (2002) Drug addiction and its underlying neurobiological basis: neuroimaging evidence for the involvement of the frontal cortex. Am J Psychiatry.

PT

159:1642–1652.

97. Grabitske HA, Slavin JL (2008) Perspectives in practice low-digestible carbohydrates in

CC E

practice. J Am Diet Assoc 108:1677–1681

98. Green E, Murphy C (2012) Altered processing of sweet taste in the brain of diet soda

A

drinkers. Physiol Behav. 107:560-7.

99. Grembecka M (2015) Sugar alcohols—their role in the modern world of sweeteners: a review European Food Res. Technol. 24:1–14

43

100.Gross LS, Li L, Earl SF, Simin L (2004). "Increased consumption of refined carbohydrates and the epidemic of type 2 diabetes in the United States: an ecologic assessment". American J. Clinical Nutr. 79:774–779. 101.Haase L, Cerf-Ducastel B, Murphy C (2009) Cortical activation in response to pure taste stimuli during the physiological states of hunger and satiety. Neuroimage. 44:1008-21.

IP T

102.Hagan S, Niswender KD (2012) Neuroendocrine regulation of food intake. Pediatr Blood Cancer. 58:149-53.

SC R

103.Hajnal A, Norgren R. Dopamine release by sucrose. In: The Senses: A Comprehensive Reference, edited by Basbaum AI, Kaneko A, Shepherd GM, Westheimer G, Albright

U

TD, Masland RH, Dallos P, Oertel D, Firestein S, Beauchamp GK, Bushnell MC, Kaas

N

JH, Gardner E, editors. New York: Academic, pg. 459–468

A

104.Haltia LT, Rinne JO, Merisaari H, Maguire RP, Savontaus E, Helin S. et al (2007)

Synapse. 61:748–756.

M

Effects of intravenous glucose on dopaminergic function in the human brain in vivo.

ED

105.Hasko G, Sitkovsky MV, Szabo (2004) Immunomodulatory and neuroprotective effects of inosine. Trends Pharmacol Sci. 25:152–7.

PT

106. Hayakawa Y, Kawai M (2003) Taste properties of L-amino acid solutions at suprathreshold concentration. Jap J Taste Smell Res 10:463–6

CC E

107.He K, Zhao L, Daviglus ML, Dyer AR, Van Horn L, Garside D, et al (2008). Association of monosodium glutamate intake with overweight in Chinese adults: the INTERMAP

A

Study. Obesity, 16:1875–80.

108.He Z, Chen Y, Dong H, Su R, Gong Z, Yan L (2014) Inhibition of vesicular glutamate transporters contributes to attenuate methamphetamine-induced conditioned place preference in rats. Behav Brain Res. 267:1-5. doi: 10.1016/j.bbr.2014.02.047.

44

109.Health

Canada

(2005)

http://www.hc-sc.gc.ca/fn-an/securit/addit/sweeten-

edulcor/polyols_polydextose_factsheet-polyols_polydextose_fiche-eng.php 110.Hebebrand J, Albayrak Ö, Adan R, Antel J, Dieguez C, de Jong J, Leng G, Menzies J, Mercer JG, Murphy M, van der Plasse G, Dickson SL (2014). Eating addiction, rather than food addiction, better captures addictive-like eating behavior. Neurosci Biobehav

IP T

Rev 47: 295–306. doi:10.1016/j.neubiorev.2014.08.016

111.Honda M, Ishizaki T, Kuroda M (2006) The effect of dried skipjack soup stock on visual

112.Horne J, Lawless HT, Speirs W, Sposato D

SC R

fatigue. J Jpn Soc Food Sci Tech . 53:443–446. (in Japanese with English abstract)

(2002) Bitter taste of saccharin and

U

acesulfame-K. Chem Senses. 27:31-8.

N

113.Hull S., Re R., Tiihonen K., Viscione L., Wickham M(2012) Consuming polydextrose in

A

a mid-morning snack increases acute satiety measurements and reduces subsequent

M

energy intake at lunch in healthy human subjectsAppetite, 59:706-712, 114.Ibarra A, Astbury NM, Olli K, Alhoniemi E, Tiihonen K (2016) Effect of Polydextrose

ED

on Subjective Feelings of Appetite during the Satiation and Satiety Periods: A systematic review and meta-analysis. Nutrients. 8: E45. doi: 10.3390/nu8010045.

PT

115.Imaizumi M, Takeda M, Fushiki T (2000) Effects of oil intake in the conditioned place preference test in mice. Brain Res. 870:150-6.

CC E

116.Jacob M, Tripathi AM, Yadav G, Saha S (2016) Nutritive and Non-Nutritive Sweeteners: A Review. Int J Oral Health Med Res. 2:149-153.

A

117.Janes AC, Pizzagalli DA, Richardt S, deB Frederick B, Chuzi S, Pachas G, Cetal (2010) Brain reactivity to smoking cues prior to smoking cessation predicts ability to maintain tobacco abstinence. Biol Psychiatry. 67:722-9. 118.Jinap S, Hajeb P (2010) Glutamate. Its applications in food and contribution to health. Appetite. 55:1–10.

45

119.John BA, Wood SG, Hawkins DR (2000) The pharmacokinetics and metabolism of sucralose in the mouse. Food Chem Toxicol. 38:S107-10.Coulombe RA, Sharma RS (1986). Neurobiochemical alterations induced by the artificial sweetener aspartame. (Nutrasweet). Toxicol Appl Pharmacol 83, 79–85. | 120.Johnson J, Vickers Z (1992) Factors influencing sensory-specific satiety. Appetite.

IP T

19:15–31. Labayen I, Díez N, Parra D, González A, Martínez JA (2004) Total and

endogenous lipid oxidation in obese women during a 10 weeks weight loss program

SC R

based on a moderately high protein energy-restricted diet. Nutr Res. 24:7–18.

121.Johnson PM, Kenny PJ (2010) Dopamine D2 receptors in addiction-like reward

U

dysfunction and compulsive eating in obese rats. Nat Neurosci 13: 635-41.

N

122.Kalivas PW, O'Brien C (2008) Drug addiction as a pathology of staged neuroplasticity.

A

Neuropsychopharmacol. 33:166-80.

M

123.Kawasaki H, Yamada A, Fuse R, Fushiki T (2008) Preference for dried bonito broth in olfactory-blocked or taste nerve-sectioned mice in the two-bottle choice test. Biosci

ED

Biotechnol Biochem. 72:2840–2846.

124.Kawasaki H, Yamada A, Fuse R, Fushiki T (2011) Intake of dried bonito broth flavored

PT

with dextrin solution induced conditioned place preference in mice. Biosci Biotechnol Biochem. 75:2288-92.

CC E

125.Kawasaki Y, Jin C, Suemaru K, Kawasaki H, Shibata K, Choshi T, Hibino S, Gomita Y, Araki H (2005) Effect of glutamate receptor antagonists on place aversion induced by

A

naloxone in single-dose morphine-treated rats. Br J Pharmacol. 145: 751–757.

126.Kenny PJ (2011) Reward mechanisms in obesity: new insights and future directions. Neuron. 69:664-79. 127.Khan NA, Besnard P (2009) Oro-sensory perception of dietary lipids: new insights into the fat taste transduction. Biochim Biophys Acta. 1791:149-55.

46

128.King NA, Craig SA, Pepper T, Blundell JE (2005) Evaluation of the independent and combined effects of xylitol and polydextrose consumed as a snack on hunger and energy intake over 10 d. Br J Nutr. 93:911–5. 129.Kobayakawa T, Ogawa H, Kaneda H, Ayabe-Kanamura S, Endo H, Saito S (1999) Spatio-temporal analysis of cortical activity evoked by gustatory stimulation in humans.

IP T

Chem Senses. 24:201–209.

130.Kodama S (1913) Separation methods of inosinic acid,” J. Chem Soc. Tokyo, 34:751–

SC R

757.

131.Kondoh T, Mallick HN, Torii K (2009) Activation of the gut-brain axis by dietary

U

glutamate and physiologic significance in energy homeostasis. American J Clin Nutr. 90:

Ueta Y, Hori E, Nishijo H (2012a) Experience-based

A

132.Kondoh T, Matsunaga T,

N

832S–7S.

M

enhancement of preference for dried-bonito dashi (a traditional japanese fish stock) in various rodent strains. Chem Senses. 38:275.

ED

133. Kondoh T, Matsunaga T, Yamazaki H, Kambe T, Nagao M (2012b) Enhancement of preference for dried-bonito dashi (a traditional Japanese fish stock) by prior food

PT

experience. tockholm (Sweden): International Symposium on Olfaction and Taste http://www.isotxvi.com/ISOT_Abstracts_final.pdf.

CC E

134.Kondoh T, Mori M, Ono T, Torii K (2000) Mechanisms of umami taste preference and aversion in rats. J Nutr 130: S966–70.

A

135.Kondoh, T. and Torii, K. MSG intake suppresses weight gain, fat deposition and plasma leptin levels in male Sprague–Dawley rats. Physiol Behav, (2008), 95: 135–144.

136.Konings E, Schoffelen PF, Stegen J, Blaak EE (2014) Effect of polydextrose and soluble maize fibre on energy metabolism, metabolic profile and appetite control in overweight men and women. Br J Nutr. 11:111-21.

47

137.Krashes MJ, Kravitz AV (2014) Optogenetic and chemogenetic insights into the food addiction hypothesis. Front Behav Neurosci 8:1–9. 138.Kumar V, Sinha AK, Makkar HP, de Boeck G, Becker K (2012) Dietary roles of nonstarch polysaccharides in human nutrition: a review. Crit Rev Food Sci Nutr. 52:899935.

IP T

139.Kumazawa T, Kurihara K (1990) Large synergism between monosodium glutamate and

5′-nucleotides in canine taste nerve responses, Am J. Physiol—Regul. Integ. Comp

SC R

Physiol, 259:R420–R426.

140.Kurihara K (2015) Umami the Fifth Basic Taste: History of Studies on Receptor

U

Mechanisms and Role as a Food Flavor. 189402, http://dx.doi.org/10.1155/2015/189402

N

141.Lakhan SE, Kirchgessner A (2013) The emerging role of dietary fructose in obesity and

A

cognitive decline. Nutr J. 12:114. doi: 10.1186/1475-2891-12-114.

M

142.Lammel S, Lim BK, Ran C, Huang KW, Betley MJ, Tye KM, et al (2012) Input-specific control of reward and aversion in the ventral tegmental area. Nature. 491:212-7

ED

143.Ledesma-Amaro R, Buey RM, Revuelta JL (2015) Increased production of inosine and guanosine by means of metabolic engineering of the purine pathway in Ashbya gossypii.

PT

Microbial Cell Factories. 14:58.

144.Ledesma-Amaro R, Jimenez A, Santos MA, Revuelta JL (2013)Biotechnological

CC E

production of feed nucleotides by microbial strain improvement. Process Biochem. 48:1263–70.

A

145.Lehrke M, Lazar MA (2005) The many faces of PPARgamma. Cell. 123:993-9. 146.Lenoir M, Serre F, Cantin L,Ahmed SH (2007). Intense sweetness surpasses cocaine reward. PLoS one 2:698-708. 147.Lerma-Cabrera JM, Carvajal F Lopez-Legarrea F (2016) Food addiction as a new piece of the obesity framework, Nutr J. 15:5.

48

148.Leshchenko IV, Shevchuk VH, Falalieieva TM, Beregova TV (2012)The influence of long-term monosodium glutamate feeding on the structure of rats pancreas. Fiziol Zh. 58:59-65. 149.Lesniewska V, Rowland I, Cani PD, Neyrinck AM, Delzenne NM, Naughton PJ (2006) Effect on components of the intestinal microflora and plasma neuropeptide levels of

IP T

feeding Lactobacillus delbrueckii, Bifidobacterium lactis, and inulin to adult and elderly rats. Appl Environ Microbiol. 72:6533-8.

SC R

150.Liang NC, Hajnal A, Norgren R (2006) Sham feeding corn oil increases accumbens dopamine in the rat. Am J Physiol Regul Integr Comp Physiol. 291:R1236-9.

U

151.Lin J, Curhan GC (2-11) Associations of sugar and artificially sweetened soda with

N

albuminuria and kidney function decline in women. Clin J Am Soc Nephrol6:160-6.

A

152. Liu D-D, Ji X-W, Li R-W (2013) Effects of Siraitia grosvenorii Fruits Extracts on

M

Physical Fatigue in Mice Iran J Pharm Res 12: 115–121. 153.Lu M, Sarruf DA, Talukdar S, Sharma S, Li P, Bandyopadhyay G, et al (2011) Brain

ED

PPAR-γ promotes obesity and is required for the insulin-sensitizing effect of thiazolidinediones. Nat Med. 17:618-22.

PT

154.Lugaz O, Pillias AM, Faurion A (2002) A new specific ageusia: some humans cannot taste L-glutamate. Chem Senses. 27:105–15.

CC E

155.Luhovyy BL, Akhavan T, Anderson GH (2007) Whey proteins in the regulation of food intake and satiety. J Am Coll Nutr. 26:704S–712.

A

156.Luo M, Zhou J, Liu Z (2015) Reward processing by the dorsal raphe nucleus: 5-HT and beyond. Learn Mem. 22: 452–460. doi: 10.1101/lm.037317.114

157.Magnuson BA, Burdock GA, Doull J, et al. (2007) Aspartame: a safety evaluation based on current use levels, regulations, and toxicological and epidemiological studies. Crit Rev Toxicol 37:629–727.

49

158.Mäkinen KK (1084) Effect of long-term, peroral administration of sugar alcohols on man. Swed Dent J. 8:113-24. 159.Manabe Y, Shigenobu M, Fushiki T (2010) Preference for high fat foods in animals. In: Montmayeur JP l.C.J., editor. Fat Detection: Taste, Texture, and Post Ingestive Effects. CRC Press; Boca Raton: Chapter 10.

IP T

160.Maone TR, Mattes RD, Bernbaum JC, Beauchamp GK (1990) A new method for delivering a taste without fluids to preterm and term infants. Dev Psychobiol 23:179–91.

SC R

161.Marlow S (2013) Food addiction: Could overeating be compulsive? BBC News London, United Kingdom. Assessed November, 2016.

U

162.Martínez PN, Luis JA, Méndez CP (2016) Stevia preferences in Wistar rats Psicothema.

N

28:442-447

A

163.Masic U, Yeomans MR (2013) Does monosodium glutamate interact with macronutrient

M

composition to influence subsequent appetite? Physiol. Behav. 116–117: 23–29. 164.Masic U, Yeomans MR (2014) Umami flavor enhances appetite but also increases

ED

satiety. Am J Clin Nutr. 100:532-538.

165.Massimino SP, McBurney MI, Field CJ, Thomson AB, Keelan M, Hayek MG, Sunvold

PT

GD (1998) Fermentable dietary fiber increases GLP-1 secretion and improves glucose homeostasis despite increased intestinal glucose transport capacity in healthy dogs. J

CC E

Nutr. 128 1786-93.

166.Mathey M, Siebelink E, de Graaf C, Van Staveren WA (2001) Flavor enhancement of

A

food improves dietary intake and nutritional status of elderly nursing home residents. J Gerontol A Biol Sci Med Sci. 56:M200–5.

167.McCabe C, Rolls ET (2007) Umami: A delicious flavour formed by convergence of taste and olfactory pathways in the human brain. European J Neurosci 25: 1855–64.

50

168.Meule A (2015) Back by Popular Demand: A narrative review on the history of food addiction research. Yale J Biol Med. 88:295–302. 169.Moran TH (2004) Gut peptides in the control of food intake: 30 years of ideas. Physiol Behav. 82:175-80. 170.Morris MJ, Na ES, Johnson AK (2008) Salt craving: the psychobiology of pathogenic

IP T

sodium intake. Physiol Behav 94:70.

171.Moskin J (2008) Yes, MSG, the secret behind the savour. The New York Times nytimes

SC R

.com. Assessed November, 2016

172.Muguruma M, Tsuruoka K, Katayama K, Erwanto Y, Kawahara S, Yamaguchi K, et al.

U

(2003) Soybean and milk proteins modified by transglutaminase improves chicken

N

sausage texture even at reduced levels of phosphate. Meat Sci. 63:191–7.

A

173.Myers KP (2007) Robust preference for a flavor paired with intragastric glucose acquired

M

in a single trial. Appetite. 2007 Jan; 48(1):123-7.

174.Nakamura S (2005) Bioavailability of cellobiose and other non-digestible and/or

ED

nonabsorbable sugar substitutes and related topics. Nutrition 21:1158–1159. 175.Navarro M, Cubero I, Knapp DJ, Breese GR, Thiele TE

(2008) Decreased

PT

immunoreactivity of the melanocortin neuropeptide alpha-melanocyte-stimulating hormone (alpha-MSH) after chronic ethanol exposure in Sprague-Dawley rats. Alcohol

CC E

Clin Exp Res. 32:266-76.

176.Nelson G, Chandrashekar J, Hoon MA, Feng L, Zhao G, Ryba NJ, Zuker CS (2002) An

A

amino-acid taste receptor. Nature. 416:199–202.

177.Nestler EJ (2004) Historical review: Molecular and cellular mechanisms of opiate and cocaine addiction. Trends Pharmacol Sci. 2:210-8. 178.Nestler EJ (2013) Cellular basis of memory for addiction. Dialogues Clin Neurosci. 15:431-43.

51

179.Niijima A(1991) Effects of oral and intestinal stimulation with umami substance on gastric vagus activity. Physiol Behav 49:1025–8. 180.Ninomiya Y, Kurenuma S, Nomura T, Uebayashi H, Kawamura H (1992)

Taste

synergism between monosodium glutamate and 5′-ribonucleotide in mice. Comp Biochem Physiol A Comp Physiol 101:97–102.

IP T

181.Ninomiya Y, Nakashima K, Fukuda A, Nishino H, Sugimura T, Hino A, Danilova V, Hellekant G(2000) Responses to umami substances in taste bud cells innervated by the

SC R

chorda tympani and glossopharyngeal nerves. J Nutr. 130:950S–953S.

182.Obici S, Feng Z, Karkanias G, Baskin DG, Rossetti L (2002) Decreasing hypothalamic

U

insulin receptors causes hyperphagia and insulin resistance in rats. Nat Neurosci. 5:566-

N

72.

A

183.Ogden CL, Carroll MD, Kit BK, Flegal KM (2012) Prevalence of obesity in the United

M

States, 2009–2010 NCHS Data Brief. 82:1-8.

184.OJ Onaolapo, OS Aremu, AY Onaolapo (2017). Monosodium glutamate-associated

ED

alterations in open-field,anxiety-related and conditioned place-preference behaviours in mice. Naunyn-Schmiedeberg's Arch. Pharmacol. 390(7):677-689., doi: 10.1007/s00210-

PT

017-1371-6

185.Oleksandra AS, Oleksandr VV, Falalyeyeva T.M, Beregova TV, Babenko LP, et al

CC E

(2014) The efficacy of probiotics for monosodium glutamate-induced obesity: Dietology concerns and opportunities for prevention EPMA J. 5: 2.

A

186.Olli K, Salli K, Alhoniemi E, Saarinen M, Ibarra A, Vasankari T, Rautonen N, Tiihonen K. (2015) Postprandial effects of polydextrose on satiety hormone responses and subjective feelings of appetite in obese participants. Nutr J. 14:2. 187.Olsen CM (2011) "Natural rewards, neuroplasticity, and non-drug addictions. Neuropharmacol. 61:1109–1122.

52

188.Onaolapo A.Y, Onaolapo O.J, Mosaku T.J, Onigbinde O.A and Oyeleke A (2013) A histological study of the hepatic and renal effects of subchronic low dose oral monosodium glutamate in Swiss albino mice. BJMMR 3:294-306. 189.Onaolapo AY, Abdusalam SZ, Onaolapo OJ, (2017b) Silymarin attenuates aspartameinduced variation in mouse behaviour, cerebrocortical morphology and oxidative stress

IP T

markers Pathophysiology 24 (2017) 51–62

190.Onaolapo AY, Onaolapo OJ, Nwoha PU (2017a) Aspartame and the hippocampus:

SC R

Revealing a bi-directional, dose/time-dependent behavioural and morphological shift in mice. Neurobiol Learn Memory 139:76–88.

U

191.Onaolapo AY, Onaolapo OJ, Nwoha PU, (2016a) Alterations in behaviour, cerebral

N

cortical morphology and cerebral oxidative stress markers following aspartame ingestion.

A

J Chem Neuroanatomy 78:42–56.

M

192.Onaolapo OJ, Onaolapo AY (2011) Acute low dose monosodium glutamate retards novelty induced behaviours in male Swiss albino mice. J Neuro Brain Health 3:51–56

ED

193.Onaolapo OJ, Onaolapo AY, Akanmu MA, Olayiwola G (2015) Foraging enrichment modulates open field response to monosodium glutamate in mice. Ann Neurosci 22:162–

PT

170.

194.Onaolapo OJ, Onaolapo AY, Akanmu MA, Olayiwola G (2016a) Evidence of alterations

CC E

in brain structure and antioxidant status following ‘low-dose’ monosodium glutamate ingestion. Pathophysiology 23:147–156

A

195.Onaolapo OJ, Onaolapo AY, Akanmu MA, Olayiwola G (2016b) Changes in spontaneous working-memory, memory-recall and approach-avoidance following Blow dose^ monosodium glutamate in mice. AIMS Neurosci 3:317–337.

53

196.Patel NG, Holder JC, Smith SA, Kumar S, Eggo MC (2003) Differential regulation of lipogenesis and leptin production by independent signaling pathways and rosiglitazone during human adipocyte differentiation. Diabetes. 52:43-50. 197.Pepino MY, Tiemann CD, Patterson BW, Wice BM, Klein S (2013) Sucralose affects glycemic and hormonal responses to an oral glucose load Diabetes Care. 36:2530-5.

IP T

198.Pietrasik Z, Duda Z (2000) Effect of fat content and soy protein/carrageenan mix on the quality characteristics of comminuted, scalded sausages. Meat Sci. 56:181–8

SC R

199.Polonsky KS, Given BD, Hirsch L, Shapiro ET, Tillil H, Beebe C, et al (1988)

Quantitative study of insulin secretion and clearance in normal and obese subjects. J Clin

U

Invest. 81(2):435-41.

N

200.Prescott J (2004) Effects of added glutamate on liking for novel food flavours. Appetite

A

42:143–50.

M

201.Pursey KM, Collins CE, Stanwell P, Burrows TL. (2015) The stability of 'food addiction' as assessed by the Yale Food Addiction Scale in a non-clinical population over 18-

ED

months. Appetite. 96:533-538.

202.Pursey, K. M., Stanwell, P., Gearhardt, A. N., Collins, C. E., & Burrows, T. L. (2014).

PT

The prevalence of food addiction as assessed by the Yale Food Addiction Scale: A systematic review. Nutrients. 6:4552-4590.

CC E

203.Raliou M, Wiencis A, Pillias A-M, et al (2009) Non-synonymous single nucleotide polymorphisms in human tas1r1, tas1r3, and mGluR1 and individual taste sensitivity to

A

glutamate. Am J Clin Nutr 90:789S–99S.

204.Ranawana V, Muller A, Henry CJ (2013) Polydextrose. Its impact on short-term food intake and subjective feelings of satiety in males-a randomized controlled cross-over studyEuropean J. Nutrition, 52:885-893,

54

205.Randolph TG (1956) The descriptive features of food addiction; addictive eating and drinking. QJ Stud. Alcohol, 17:198-224 206.Renwick AG, Molinary SV (2010) Sweet-taste receptors, low- energy sweeteners, glucose absorption and insulin release.The British J. Nutri. 104: 1415–20.

pharmacokinetics in man. Food Chem Toxicol. 38:S31-41.

IP T

207.Roberts A, Renwick AG, Sims J, Snodin DJ (2000) Sucralose metabolism and

208.Rogers PJ, Blundell JE (1990) Umami and appetite — effects of monosodium glutamate

SC R

on hunger and food-intake in human subjects. Physiol Behav 48:801–4.

209. Rogers PJ, Carlyle JA, Hill AJ, Blundell JE.(1988) Uncoupling sweet taste and calories:

U

comparison of the effects of glucose and three intense sweeteners on hunger and food

N

intake. Physiol Behav 43:547–52.

A

210.Rolls BJ, Laster LJ, Summerfelt A (1989) Hunger and food intake following

M

consumption of low-calorie foods. Appetite 13:115–27. 211. Rothemund Y, Preuschhof C, Bohner G, Bauknecht H, Klingebiel R, Flor H, etal (2007)

ED

Differential activation of the dorsal striatum by high-calorie visual food stimuli in obese individuals. Neuroimage. 37:410–421.

PT

212.Rudenga KJ, Small DM (2012) Amygdala response to sucrose consumption is inversely related to artificial sweetener use. Appetite. 58:504-7.

CC E

213.Ryan KK, Li B, Grayson BE, Matter EK, Woods SC, Seeley RJ(2011) A role for central nervous system PPAR-γ in the regulation of energy balance. Nat Med. 17:623-6.

A

214.Ryan KK, Woods SC, Seeley RJ (2012) Central nervous system mechanisms linking the consumption of palatable high-fat diets to the defense of greater adiposity. Cell Metab. 15:137-49.

55

215.Sarruf DA, Yu F, Nguyen HT, Williams DL, Printz RL, Niswender KD, et al (2009) Expression of peroxisome proliferator-activated receptor-gamma in key neuronal subsets regulating glucose metabolism and energy homeostasis. Endocrinology. 150:707-12. 216.Schiffman SS, Sattely-Miller EA, Zimmerman IA, Graham BG, Erickson RP (1994) Taste perception of monosodium glutamate (MSG) in foods in young and elderly

IP T

subjects. Physiol Behav. 56:265–75.

217.Schoeller DA, Buchholz A (2005) Energetics of obesity and weight control: does diet

SC R

composition matter? J Am Diet Assoc105:S24–S28.

218.Schulte EM, Avena NM, Gearhardt AN (2016) Which Foods May Be Addictive? The

U

roles of processing, fat content, and glycemic Load. PLoS One 10:e0117959. doi:

N

10.1371/journal.pone.0117959.

A

219.Schulte EM, Tuttle HM, Gearhardt AN (2015) Belief in food addiction and obesity

M

related policy support. PLoS One. 11: e0147557. 10.1371/journal.pone.0147557 220.Schultz W (2015). Neuronal reward and decision signals: from theories to data.

ED

Physiological Reviews. 95: 853–951.

221.Schwab U, Louheranta A, Törrönen A, Uusitupa M (2006) Impact of sugar beet pectin

PT

and polydextrose on fasting and postprandial glycemia and fasting concentrations of serum total and lipoprotein lipids in middle-aged subjects with abnormal glucose

CC E

metabolism.Eur J Clin Nutr. 60:1073-80.

222.Schwartz MW, Peskind E, Raskind M, Boyko EJ, Porte D Jr (1996) Cerebrospinal fluid

A

leptin levels: relationship to plasma levels and to adiposity in humans. Nat Med. 2:58993.

223.Sclafani (2013) A Gut-Brain Nutrient Signaling: Appetition vs. Satiation. Appetite. 71:454-8.

56

224.Sclafani A (1993) Dietary obesity. In: Stunkard AJ, Wadden TA, editors. Obesity: Theory and Therapy. New York: Raven Press, pp. 125–136 225.Sclafani A, Ackroff K (2012) Role of gut nutrient sensing in stimulating appetite and conditioning food preferences. Am J Physiol Regul Integr Comp Physiol. 302:R1119-33.

nutritive sweeteners in mice. Physiol Behav 1;173:188-199.

IP T

226.Sclafani A, Ackroff K (2017) Flavor preferences conditioned by nutritive and non-

227.Sclafani A, Ackroff K(2004) The relationship between food reward and satiation

SC R

revisited. Physiol Behav. 82:89-95.

228.Sclafani A, Glendinning JI (2005) Sugar and fat conditioned flavor preferences in

U

C57BL/6J and 129 mice: oral and postoral interactions. Am J Physiol Regul Integr Comp

N

Physiol. 289:R712-20.

A

229.Seeley RJ, van Dijk G, Campfield LA, Smith FJ, Burn P, Nelligan JA, et al (1996)

M

Intraventricular leptin reduces food intake and body weight of lean rats but not obese Zucker rats. Horm Metab Res. 28:664-8.

ED

230.Shankar P, Ahuja S, Sriram K (2013) Non-nutritive sweeteners: review and update. Nutrition 29:1293–1299.

PT

231.Shen H, Chen G-J, Harvey BK, Bickford PL, Wang Y (2005) Inosine reduces ischemic brain injury in rats. Stroke. 36: 654–659.

CC E

232.Shigemura N, Shirosaki S, Ohkuri T, et al (2009)Variation in umami perception and in candidate genes for the umami receptor in mice and humans. Am J Clin Nutr 90:S764–9.

A

233.Sivertsen HK, Ueland Ø, Westad F Ø (2010) Development of satiating and palatable high-protein meat products by using experimental design in food technology Food Nutr Res 54, doi: 10.3402/fnr.v54i0.5114 234.Small DM (2006) Central gustatory processing in humans. Adv Otorhinolarygol. 63:191–220.

57

235.Small DM, Jones-Gotman M, Dagher A (2003) Feeding-induced dopamine release in dorsal striatum correlates with meal pleasantness ratings in healthy human volunteers. Neuroimage. 19:1709-15. 236.Smeets PA, Erkner A, de Graaf C (2010) Cephalic phase responses and appetite. Nutr Rev. 68:643-55.

IP T

237.Smeets PAM, de Graaf C, Stafleu A, van Osch MJP, van der Grond J (2005) Functional magnetic resonance imaging of human hypothalamic responses to sweet taste and

SC R

calories. Am J Clin Nutr. 82:1011–1016

238.Speakman J, Hambly C, Mitchell S, Król E (2008) The contribution of animal models to

U

the study of obesity. Lab Anim. 42:413-32.

N

239.Spence C (2012) Auditory contributions to flavour perception and feeding behaviour.

A

Physiol. Behav. 107:505–515.

M

240.Stanley WC, Shah KB, Essop MF (2009). Does Junk Food Lead to Heart Failure? Importance of Dietary Macronutrient Composition in Hypertension" Hypertension. 54:

ED

1209–10.

241.Stice E, Spoor S, Bohon C, Veldhuizen MG, Small DM (2008) Relation of reward from

PT

food intake and anticipated food intake to obesity: a functional magnetic resonance imaging study. J Abnorm Psychol. 117:924-35.

CC E

242.Stice E, Yokum S, Blum K, Bohon C (2010b) Weight gain is associated with reduced striatal response to palatable food. J Neurosci. 30:13105-9.

A

243.Stice E, Yokum S, Bohon C, Marti N, Smolen A (2010) Reward circuitry responsivity to food predicts future increases in body mass: moderating effects of DRD2 and DRD4. Neuroimage. 50:1618-25.

58

244.Stoeckel LE, Weller RE, Cook EW, Twieg DB, Knowlton RC, Cox JF. Widespread reward-system activation in obese women in response to pictures of high-calorie foods. Neuroimage. 2008;41(2):636–647. 245.Suez J, Korem T, Zeevi D, Zilberman-Schapira G, Thaiss CA, Maza O, et al (2014) Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature.

IP T

514:181-6.

246.Swanson LW (1982) The projections of the ventral tegmental area and adjacent regions:

SC R

a combined fluorescent retrograde tracer and immunofluorescence study in the rat. Brain Res Bull. 1982 Jul-Dec; 9(1-6):321-53.

U

247.Takeda M, Imaizumi M, Fushiki T (2000) Preference for vegetable oils in the two-bottle

N

choice test in mice. Life Sci. 67:197-204.

A

248.Tarman V, Werdell P (2014) Food Junkies: The truth about food addiction. Toronto,

M

Canada: Dundurn.

249.Thiele TE, Navarro M, Sparta DR, Fee JR, Knapp DJ, Cubero I (2003) Alcoholism and

ED

obesity: overlapping neuropeptide pathways? Neuropeptides. 37:321-37. 250.Thomas S, Chapman S. Chipping Campden, UK (2008) A review of ingredients,

PT

physiological mechanisms and measurements involved in the enhancement of satiation. Review No. 60.

CC E

251.Tontonoz P, Hu E, Spiegelman BM (1994) Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor.Cell. 79:1147-56.

A

252.Tordoff MG, Aleman TR, Murphy MC(2012) No effects of monosodium glutamate consumption on the body weight or composition of adult rats and mice. Physiol Behav. 107: 338-345. 253.Tordoff MG, Alleva AM (1990) Oral stimulation with aspartame increases hunger. Physiol Behav 47:555–9.

59

254.Torii K, Uneyama H, Nakamura E (2013) Physiological roles of dietary glutamate signaling via gut-brain axis due to efficient digestion and absorption. J Gastroenterol. 48:442-51. 255.Treesukosol Y, Smith KR, Spector AC (2011) The functional role of the T1R family of receptors in sweet taste and feeding. Physiol Behav 105: 14– 26.

IP T

256.Tsuda K (2005) Inosine, calcium channels, and neuroprotection against ischemic brain injury. Stroke. 36:1823.

SC R

257.Tsurugizawa T, Torii K (2010) Physiological roles of glutamate signaling in gut and brain function. Biol Pharm. Bull. 33: 1796-1799.

U

258.Tsurugizawa T, Uematsu A, Uneyama H, Torii K (2011) Different BOLD responses to

N

intragastric load of L-glutamate and inosine monophosphate in conscious rats. Chem

A

Senses. 36:169-76.

M

259.Tsurugizawa T, Uematsu A, Nakamura E, Hasumura M, Hirota M, Kondoh T, Uneyama H, Torii K (2009) Mechanisms of neural response to gastrointestinal nutritive

ED

stimuli: The gut-brain axis, Gastroenterol. 137: 262-273. 260.Tulloch AJ, Murray S, Vaicekonyte R, Avena NM (2015) Neural responses to

1218.

PT

macronutrients: hedonic and homeostatic mechanisms. Gastroenterology 148:1205–

CC E

261.Tzschentke TM (1998) Measuring reward with the conditioned place preference paradigm: a comprehensive review of drug effects. Recent progress and new issues.

A

Prog. Neurobiol. 56: 613–72.

262.Tzschentke TM (2007) Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol. 12:227-462.

60

263.Tzschentke TM, Schmidt WJ (2000) Functional relationship among medial prefrontal cortex, nucleus accumbens, and ventral tegmental area in locomotion and reward. Crit Rev Neurobiol 14:131–142 264.Tzschentke TM, Schmidt WJ (2003) Glutamatergic mechanisms in addiction. Mol Psychiatry 8:373-82.

IP T

265.Uematsu A, Tsurugizawa T, Kondoh T, Torii K (2009) Conditioned flavour preference

learning by intragastric administration of L-glutamate in rats. Neurosci Lett. 451:190–

SC R

193.

266.Uematsu A, Tsurugizawa T, Uneyama H, Torii K (2010) Brain-gut communication via

(2014) Glutamatergic synaptic plasticity in the

N

267.van Huijstee AN, Mansvelder HD

U

vagus nerve modulate conditioned flavor preference. Eur J Neurosci. 31:1136–1143.

A

mesocorticolimbic system in addiction. Front Cell Neurosci. 8:466.

M

268.Vanderwater K, Vickers Z (1994) High-protein foods produce greater sensory-specific satiety. Physiol Behav. 59:579–83.

High

trait

ED

269.Velázquez-Sánchez C, Ferragud A, Moore CF, Everitt BJ, Sabino V, Cottone P (2014) impulsivity

predicts

food

addiction-like

behavior

in

the

rat.

PT

Neuropsychopharmacology. 39:2463–2472. 270.Velázquez-Sánchez C, Jeffrey W. Santos JW, Smith KL, Ferragud A, Sabino V, Cottone

CC E

P (2015) Seeking behavior, place conditioning and resistance to conditioned suppression of feeding in rats intermittently exposed to palatable food. Behav Neurosci. 129: 219–

A

224.

271.Viarouge C, Caulliez R, Nicolaidis S (1992) Umami taste of monosodium glutamate enhances the thermic effect of food and affects the respiratory quotient in the rat. Physiol Behav 52:879–84.

61

272.Volkow ND, Wang GJ, Tomasi D, Baler RD (2012) Obesity and addiction: neurobiological overlaps. Obes Rev 14(1):2-18. 273.Watanabe S (2013) Possible false negative results in conditioned place preference induced by low-dose morphine in mice. J. Addiction Res. Therapy. S4:013. doi:10.4172/2155-6105.S4-013.

IP T

274.Watterson SH, Chen P, Zhao Y, Gu HH, Dhar TG, Xiao Z, Ballentine SK, Shen Z,

Fleener CA, Rouleau KA, et al (2007) Acridone-based inhibitors of inosine 5’-

(6-(4-ethylpiperazin-1-yl)pyridin-3-yl)propan-2-yl)-2-

SC R

monophosphate dehydrogenase: discovery and SAR leading to the identification of N-(2-

fluoro-9-oxo-9,10-

U

dihydroacridine-3-carboxamide (BMS-566419). J Med Chem. 50:3730–3742

N

275.Weihrauch MR, Diehl V (2004) Artificial sweeteners--do they bear a carcinogenic risk?

A

Ann Oncol. 15:1460-5.

M

276.Westwater ML, Fletcher PC, Ziauddeen H (2016) Sugar addiction: the state of the science European Journal of Nutrition. 55: 55–69.

Assoc 108:S34–S39.

ED

277.Wheeler ML, Pi-Sunyer X (2008) Carbohydrate issues. Type and amount. J Am Diet

PT

278.WHO -World health Organisation (2017) Food additives Fact sheet. 279.Wise RA (2009) Roles for nigrostriatal--not just mesocorticolimbic--dopamine in reward

CC E

and addiction. Trends Neurosci. 32:517-24.

280.Wise RA, Koob GF (2014) The development and maintenance of drug addiction.

A

Neuropsychopharmacology39:254-62.

281.Woods SC (2009) The control of food intake: behavioral versus molecular perspectives. Cell Metab 9:489-98. 282.Woods SC, D'Alessio DA (2008) Central control of body weight and appetite. J Clin Endocrinol Metab. 93:S37-50.

62

283.Woods SC, Lotter EC, McKay LD, Porte D Jr (1979) Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature. 282:503-5. 284.Wu G (1998) Intestinal mucosal amino acid catabolism. J Nutr. 128:1249–52 285.Yager LM, Garcia AF, Wunsch AM, Ferguson SM (2015). "The ins and outs of the striatum: Role in drug addiction". Neuroscience. 301: 529–541.

IP T

286.Yamaguchi S, Ninomiya K (2000) Umami and food palatability. Am Soc Nutr Sci. 921S–926S.

SC R

287.Yamamoto T, Shimura T. Roles of taste in feeding and reward. In: The Senses: A

Comprehensive Reference, edited by Basbaum AI, Kaneko A, Shepherd GM,

U

Westheimer G, Albright TD, Masland RH, Dallos P, Oertel D, Firestein S, Beauchamp

N

GK, Bushnell MC, Kaas JH, Gardner E, editors. New York: Academic, pg. 437–458 Front Syst

A

288.Yamamoto T, Ueji K (2011) Brain mechanisms of flavor learning. Neurosci5:76.

M

289.Yang Q (2010) Gain weight by “going diet?” Artificial sweeteners and the neurobiology

ED

of sugar cravings Yale J Biol Med. 83:101–108. 290.Yeomans MR (1996) Palatability and the micro-structure of feeding in humans: the

PT

appetizer effect. Appetite 27:119–33. 291.Yeomans MR, Leitch M, Gould NJ, Mobini S (2008)Differential hedonic, sensory and

CC E

behavioral changes associated with flavor-nutrient and flavor-flavor learning Physiol Behav. 93:798-806.

A

292.Yoneda T, Saitou K, Mizushige T, Matsumura S, Manabe Y, Tsuzuki S, et al (2007a) The palatability of corn oil and linoleic acid to mice as measured by short-term twobottle choice and licking tests. Physiol Behav. 91:304-9.

63

293.Yoneda T, Taka Y, Okamura M, Mizushige T, Matsumura S, Manabe Y, etal (2007b) Reinforcing effect for corn oil stimulus was concentration dependent in an operant task in mice. Life Sci. 81:1585-92. 294.Zero DT, Fontana M, Martínez-Mier EA, Ferreira-Zandoná A, Ando M, GonzálezCabezas C, Bayne S. (2009). "The Biology, Prevention, Diagnosis and Treatment of

IP T

Dental Caries: Scientific Advances in the United States" Journal of the American Dental Association 140: 25S–34S.

SC R

295.Zhang F, Klebansky B, Fine RM, Xu H, Pronin A, Liu H, Tachdjian C, Li X. (2008)

Molecular mechanism for the umami taste synergism. Proc Natl Acad Sci U S A.

U

105:20930–20934.

N

296.Ziauddeen H, Fletcher PC (2013) Is food addiction a valid and useful concept? Obesity

A

CC E

PT

ED

M

A

Reviews. 14:19-28.

64

297. Figure legends Figure 1: Can food additives in food stimulate the brain reward circuit resulting in food addiction? Does eating excessively potentiate the reward system that includes the substantia

ED

M

A

N

U

SC R

IP T

nigra (SNc) and the ventral tegmental area (VTA) enough to perpetuate food addiction?

Figure 2: The regulation of food intake involves a balance between homeostatic factors

PT

(regulated by energy stores and energy demands) and hedonic influences (consumption

A

CC E

motivated by reward systems).

65

66

A ED

PT

CC E

IP T

SC R

U

N

A

M