Next Article in Journal
Hepatic Steatosis in Patients with Celiac Disease: The Role of Packaged Gluten-Free Foods
Next Article in Special Issue
Supplementation with NAD+ and Its Precursors to Prevent Cognitive Decline across Disease Contexts
Previous Article in Journal
Magnesium Status and Ca/Mg Ratios in a Series of Children and Adolescents with Chronic Diseases
Previous Article in Special Issue
A Review of the Potential Consequences of Pearl Millet (Pennisetum glaucum) for Diabetes Mellitus and Other Biomedical Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A High-Sugar Diet Consumption, Metabolism and Health Impacts with a Focus on the Development of Substance Use Disorder: A Narrative Review

Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland
*
Author to whom correspondence should be addressed.
Nutrients 2022, 14(14), 2940; https://doi.org/10.3390/nu14142940
Submission received: 23 May 2022 / Revised: 13 July 2022 / Accepted: 15 July 2022 / Published: 18 July 2022
(This article belongs to the Special Issue Diet and Disease Development: Present and Future)

Abstract

:
Carbohydrates are important macronutrients in human and rodent diet patterns that play a key role in crucial metabolic pathways and provide the necessary energy for proper body functioning. Sugar homeostasis and intake require complex hormonal and nervous control to proper body energy balance. Added sugar in processed food results in metabolic, cardiovascular, and nervous disorders. Epidemiological reports have shown enhanced consumption of sweet products in children and adults, especially in reproductive age and in pregnant women, which can lead to the susceptibility of offspring’s health to diseases in early life or in adulthood and proneness to mental disorders. In this review, we discuss the impacts of high-sugar diet (HSD) or sugar intake during the perinatal and/or postnatal periods on neural and behavioural disturbances as well as on the development of substance use disorder (SUD). Since several emotional behavioural disturbances are recognized as predictors of SUD, we also present how HSD enhances impulsive behaviour, stress, anxiety and depression. Apart from the influence of HSD on these mood disturbances, added sugar can render food addiction. Both food and addictive substances change the sensitivity of the brain rewarding neurotransmission signalling. The results of the collected studies could be important in assessing sugar intake, especially via maternal dietary patterns, from the clinical perspective of SUD prevention or pre-existing emotional disorders. Methodology: This narrative review focuses on the roles of a high-sugar diet (HSD) and added sugar in foods and on the impacts of glucose and fructose on the development of substance use disorder (SUD) and on the behavioural predictors of drugs abuse. The literature was reviewed by two authors independently according to the topic of the review. We searched the PubMed and Scopus databases and Multidisciplinary Digital Publishing Institute open access scientific journals using the following keyword search strategy depending on the theme of the chapter: “high-sugar diet” OR “high-carbohydrate diet” OR “sugar” OR “glucose” OR “fructose” OR “added sugar” AND keywords. We excluded inaccessible or pay-walled articles, abstracts, conference papers, editorials, letters, commentary, and short notes. Reviews, experimental studies, and epidemiological data, published since 1990s, were searched and collected depending on the chapter structure. After the search, all duplicates are thrown out and full texts were read, and findings were rescreened. After the selection process, appropriate papers were included to present in this review.

Graphical Abstract

1. Introduction

Carbohydrates (=saccharides) are groups of chemical compounds that include sugars, starch, and cellulose [1]. Saccharides are divided into four chemical subgroups according to the degree of polymerization by indication of the length of the carbohydrate chain: monosaccharides (with only 1 sugar unit), disaccharides (with 2 sugar units), oligosaccharides (containing 3–9 sugar units), and polysaccharides (with ≥10 sugar units) (Table 1). The representatives of the monosaccharides are glucose, galactose, and fructose; examples of disaccharides include sucrose (glucose + fructose), maltose (2 glucose units) and lactose (glucose + galactose); and the polysaccharides are starches. Carbohydrates are crucial to nutrition and perform crucial roles in organisms. Monosaccharides are a source of energy for cells, and some biomolecules fulfil numerous roles in biosynthesis and build other subgroups of saccharides. Mono- and disaccharides act as sweeteners in many foods, as well as natural preservatives. Fructose is the sweetest of all food carbohydrates. Polysaccharides are used for energy storage and as structural components [2,3]. Processed, refined sugars (called added sugars), such as sucrose or high-fructose corn syrup (HFCS), provide little nutritional value to food and consist of empty calories. High values of added sugars appear in sugar-sweetened beverages (SSBs). Excess sugar bingeing is dangerous for the human body, contributing to the epidemic of obesity and overweight [4,5].

1.1. Daily Limits for Carbohydrate Intake

Dietary carbohydrates are crucial to nutrition and are found in a wide variety of natural and processed foods. The Institute of Medicine (IOM) established a limit for carbohydrates of 130 g per day for adults and children aged ≥1 year old. The IOM and the Dietary Guidelines for Americans recommend that carbohydrates should provide 45–65% of human daily caloric intake for all ages and genders [6,7]. Moreover, according to the Food and Drug Administration, the daily value for carbohydrates is 300 g per day in humans consuming a 2000-calorie diet [8]. In turn, the daily sugar limit for laboratory animals depends on the species, stage of development, strain, and individual preferences. In most cases, standard rodent diets are based on the safety margins reported by laboratories. To date, a specific need for carbohydrates has not been established, but rats are better at eating a diet rich in sugar, such as glucose, or its precursors, such as starch. Diets high in fat or sugar have contributed to the promotion of obesity and overweight. It should be mentioned that fructose and sucrose (as a source of fructose) can lead to anomalies compared with glucose [9,10]. The control laboratory rodent diet should contain between 55% and 65% of its energy requirement from carbohydrates, with additional fibre, at the required temperature and humidity conditions [10,11,12,13].

1.2. Added Sugar Consumption

In 2008 and 2012, adults in the United States consumed more than 28 kg of added sugar. The average sugar consumption in 2008 was 76.7 g (19 teaspoons), and in 2012, it was 77 g per day [14,15,16]. The average daily intake of added sugars to food in 2017–2018 was 17 teaspoons per day for children and adults in the USA. In particular, men consume 20% more teaspoons of added sugar than women [17]. In 2011–2014, American youth and adults consumed an average of 143 and 145 calories from SSB, respectively [18,19]. Currently, the World Health Organization (WHO) guideline recommends reducing free sugar (including monosaccharides and disaccharides) intake to less than 10% of total daily energy intake in adults and children, whereas the Institute of Medicine recommends that added sugar make up less than 25% of total calories [20,21]. Clinical trials and epidemiologic studies have shown that individuals who consume greater amounts of added sugar, especially sugar-sweetened beverages, tend to gain more weight and have a higher risk of obesity, type 2 diabetes mellitus, dyslipidaemia, hypertension, and cardiovascular disease [22].
Since 1990, the most important change in the eating patterns of United Kingdom consumers has been a reduction in high-fat milk consumption, especially among pre-schoolers, so children and adolescents have switched to sodas, fruit drinks, juices, and sweetened dairy products [23]. Additionally, in the UK, the total sales volume of SSB decreased, whereas volume sales of low- and zero-sugar drinks and water increased between 2015 and 2018 [24]. The consumption of fructose is high in Western diets, mainly due to the consumption of refined or processed sugars, such as HFCS [25]. In the United States, HFCS is found in nearly all processed foods that contain sweeteners. HFCS—due to its low cost of production—replaced sucrose as a sweetener [26].
Currently, little clinical research data about fructose and HFCS consumption are available [27]. Unfortunately, no clear dietary recommendations on fructose or HFCS consumption limits are available. Additional research is needed to determine the adverse effects of consuming added fructose-containing sugars. Several previous data have indicated that total daily caloric intake in the form of added sugars increased over the study period (1970–1999) [28], including fructose availability in grams per day [29] and total fructose intake [30,31]. Further research is needed to understand the impact of increased intake of fructose on human health. More than 10% of Americans daily calories come from fructose consumption [28]. When too much sugar or too many carbohydrates are eaten, all this energy must basically be stored inside the fat cells or fat droplets, and this fat can accumulate in internal organs, especially in the liver but also peripherally—subcutaneously. Many more calories from fructose are converted into abdominal fat, whereas less fat from glucose accumulates in the liver. HSDs, sucrose, and thus glucose and fructose cause dysregulation of lipid and carbohydrate metabolism in the body. Added sugar bingeing promotes an increased energy balance, weight gain, fat storage and consequently overweight and/or obesity [32,33,34,35]. An increase in high-sugar products and added sugar consumption has been observed since the 1980s (more visible in the USA) and suggests that sugar, not including fat, is the major factor contributing to and driving the current obesity epidemic and diabetes [36,37].
According to a WHO report, in 2016, more than 1.9 billion adults (18 years and older) were overweight, and over 650 million of them, mainly women were obese [38]. Glucose metabolism disturbances can occur in neurological diseases and have been reported in both clinical and preclinical studies. Alzheimer’s [39,40,41] and Parkinson’s diseases [42] lead to a decrease in glucose levels or degeneration of neurons, also resulting from a diet rich in added sugars [43]. In contrast, in a rat infarction model, an increase in glucose levels in the ischaemic area was observed [44]. Interestingly, in addition to changes in sugar metabolism in the nervous system, sugar itself could exert a protective effect by increasing glucose uptake by nerve cells and restoring adequate metabolic homeostasis [45,46]. Excessive consumption of fructose can lead to inflammation in the nervous system, microglial overactivation, and oxidative stress. Preclinical research has also suggested cognitive and memory impairment in rodents fed high concentrations of fructose [47] or sugar [48,49]. Sugar consumption impacts brain diseases or disorders, such as anxiety, depression, neurogenesis, fear, and changes in molecular and neurochemical expression [50].
The basic animal research model for examining the effects of sugar intake is to provide them with sweet food. Such modified diet consists predominantly of food enriched with sweetened (with sucrose, glucose, or fructose) beverages or HFCS or, to a lesser extent, food enriched only with a high content of sucrose, fructose, glucose, or other sugars in feed [20]. Animal models of excess sugar intake in the diet produce obesity, and this effect is mainly under investigation [51,52]. In animal modelling studies (but not in human research), it is possible to measure the amount of sweet food consumption and energy balance due to strictly restrictive limits on types of food, liquids, or number of calories.

1.3. Carbohydrate ADME Processes

The fate of carbohydrates in an organism can be described by the absorption, distribution, metabolism, and excretion (ADME) process. In mammals, dietary di-, oligo- and polysaccharides are digested in the mouth, stomach, and intestine and are broken down into monosaccharides by special (salivary amylase, stomach acid, and specific carbohydrases—glycoside hydrolases, respectively) enzymes, and finally, the small intestine absorbs them into the bloodstream. Cellulose dietary fibres are indigestible carbohydrates fermented in the large intestine using the presence of bacteria [53]. Monosaccharides are transported through the hepatic portal vein to the liver. Fructose and galactose are phosphorylated to glucose by fructo- and galactokinase. Glucose is metabolized in all body cells by glycolysis [54]. This metabolic pathway converts 1 glucose molecule into 2 pyruvic acid molecules—the substrate for the citric acid cycle (CAC). Glycolysis also releases 2 high-energy adenosine triphosphate (ATP) molecules and 2 molecules of reduced nicotinamide adenine dinucleotide. The complete breakdown of one glucose molecule by aerobic respiration (glycolysis + CAC) forms approximately 30–33 molecules of ATP [55]. Glucose serves as a substrate of cellular respiration or is stored as glycogen in glycogenesis [14].
Monosaccharides, including glucose and fructose, enter mammalian cells via two different types of membrane-associated carrier proteins: sodium-glucose linked transporters (SGLTs) and facilitated diffusion glucose transporters (GLUTs) [56,57,58]. SGLT and GLUT expression occurs in tissues (e.g., in the intestine, kidney, and liver) where energy requirements or sugar biotransformation is necessary for other metabolic pathways (Table 2). Additionally, adult brain neurons are in high energy demand; therefore, transporter presence is found in the brain [59] (Table 2). Neurons consume approximately 20% of glucose-derived energy, making them the main consumer of glucose. Glucose enters the mammalian brain from the blood across the blood–brain barrier (BBB), which galactose and fructose are believed to cross. The large blood-brain concentration gradient drives the facilitating transport of glucose across endothelial membranes via GLUT1 glucose transporters into the extracellular fluid. GLUT1 further mediates glucose uptake from the extracellular fluid into astrocytes, oligodendroglia, and microglia, whereas GLUT3, which has a much higher transport rate than GLUT1, facilitates neuronal glucose uptake [45]. The expression of GLUT1 is much higher than that of GLUT3 in the developing and prenatal brain. In neurons and astrocytes, glucose is the source of pyruvate to fuel the citric acid cycle for the production of ATP. The SGLTs that mediate secondary active transport are likely not active in healthy conditions but rather in pathologic situations [60]. In contrast to glucose (80–120 mg/dL) [61,62], circulating levels of fructose in blood plasma are extremely low (<0.050 mM) [63,64]. The process of transporting fructose from the blood to the brain is unclear, and the data collected remain inconclusive. Among the expression of active fructose transporters (Table 2), such as SGLT4, GLUT5, and GLUT8 [4,64,65,66], only GLUT5 has been identified in the human and rat BBB [67,68].
The most important organ regulating carbohydrate metabolism in mammals is the liver. Here, the control of energy homeostasis is mainly reduced to regulation of the level of glucose in the blood. Glucose delivered to hepatocytes leads to de novo lipogenesis (DNL), the synthesis of lipids (glycogen) stored in complex particles—lipoproteins in the liver. The decrease in glucose levels during starvation leads to the breakdown of glycogen stores or the synthesis of glucose as a result of gluconeogenesis [69]. Epidemiological studies have indicated that diets rich in simple sugars increase both DNL and the growth of peripheral, subcutaneous, and internal adipose tissue [70]. Below, several factors, including pancreatic hormones, diet, and the body’s energy balance, are discussed in the context of their roles in the conversion of glucose into lipids and lipids into glucose.

2. Hormonal and Neuronal Regulation of SUGAR Intake

The homeostasis of blood glucose or fructose levels requires strong interaction of not only the endocrine system (pancreatic hormones, stress hormones) but also communication of the nervous system.

2.1. Pancreatic Hormones Control Blood Glucose Homeostasis

The glucoregulation process is responsible for maintaining a constant level of glucose in the body [71]. Hormones released from pancreatic cells play an important role in this negative feedback regulation, and their release is controlled by the amount of glucose present [72]. When blood glucose levels drop, leading to hypoglycaemia, pancreatic alpha cells are activated, and glucagon is released. Glucagon is transported with the blood, e.g., to the liver, where it activates glucagon receptors, enhances the process of glycogenolysis and inhibits glycogenesis. Glycogenolysis breaks down spare glycogen into glucose and releases it into the bloodstream. During hyperglycaemia (i.e., an increase in glucose levels in the blood), insulin, another pancreatic hormone, is released. Similar to glucagon, insulin is transported with the blood to the liver but enhances the process of glycogenesis and accelerates the uptake of glucose from the blood by muscle and adipose tissue cells [73,74,75].

2.2. Sweetness Perception and Regulation

The processing of sweet taste information is related to the presence of taste receptor cells (TRCs) expressing G-protein coupled receptors (GPCRs) concentrated in the taste buds of the tongue [76]. The taste 1 receptor family and the taste 2 receptor family of type 2 TRC receptor cells are capable of detecting sweet tastes. Upon activation with a sweet molecule ligand, the GPCR receptor activates the chemosensing signalling pathway [77]. Finally, ATP is released by the semichannels, activates the purinergic receptors present on the afferent fibres of the cranial nerves of the taste buds, and sends signals to the brain taste perception areas located in the gustatory cortex [78]. In the intestines, sweet taste receptors are concentrated mainly on enteroendocrine cells that secrete bioactive molecules, e.g., hormones. Sweet taste receptors in the gastrointestinal tract are responsible for nutrient detection, glucose homeostasis, and the secretion of gastrointestinal peptides [79]. The taste information from the intestines via the vagus nerve moves to the rostral division of the nucleus of the tractus solitarius (in rodents also to the parabrachial nucleus) and then to the parvicellular part of the ventral posteromedial nucleus of the thalamus, via the ventral path to the amygdala and to lateral areas of the hypothalamus [80,81].

2.3. Energy-Balanced Peptide Hormones

Sugar intake and metabolism are controlled by peptide hormones, such as leptin and ghrelin. Leptin is produced by adipocytes, whereas ghrelin is secreted by gastric enteroendocrine cells [82,83]. Leptin and ghrelin, together with blood, penetrate the BBB and regulate the activity of the hypothalamic melanocortin system in the area of the arcuate nucleus (ARC) [84]. One group of ARC neurons—anorexigenic—expresses the precursor peptide pro-opiomelanocortin (POMC) and CART (cocaine and amphetamine-regulated transcript). POMC is converted into α-melanocyte-stimulating hormone (α-MSH) and serves as a melanocortin 4 receptor (MC4R) agonist. Activation of POMC/CART neurons by leptin causes anorexic effects, including reduced food intake, weight loss, the release of α-MSH, and decreased release of NPY. The second group of neurons–orexigenic–expresses neuropeptide Y (NPY) and the MC4R antagonist agouti-associated protein (AgRP). Activation of NPY/AGRP neurons by ghrelin causes increased food consumption, weight gain, AgRP release, and decreased α-MSH release [85,86]. Leptin induces weight loss by suppressing food intake, whereas ghrelin acts as an appetite stimulant signal. An HSD (rich in carbohydrates) causes an increase in leptin concentrations and a decrease in circulating ghrelin levels, which are greater than with a high-fat diet (HFD) [87].

2.4. Stress-Induced Bingeing “Comfort Food”

As discussed above, exposure to several stressors alters the metabolic and behavioural status of the body [88]. Stress factors activate feedback interactions along the hypothalamic–pituitary–adrenal (HPA) axis. The response to stress is release of the peptide corticotropin-releasing hormone (CRH) in the paraventricular nucleus of the hypothalamus. CRH is transferred through the hypothalamo-hypophyseal portal system to the anterior pituitary gland and stimulates corticotrophic cells to release adrenocorticotropic hormone (ACTH) from POMC. Via the bloodstream, ACTH travels to the adrenal glands, where it stimulates the secretion of cortisol, which is the most important stress-induced hormone [89,90]. Cortisol regulates glucose and lipid metabolism, appetite, food consumption, and weight gain [91,92]. Exposure to stress (prenatal and postnatal) can alter both the amount and the quality of calories consumed, and stress-induced changes in food consumption and energy balance can interact with the emotional state of humans [93,94]. The high reactivity of cortisol to stress can increase susceptibility in humans to eating palatable products rich in fat or sugar, called “comfort food” [95,96,97,98,99]. Similar to clinical observations, enhancement of palatable food intake has been reported in rodents during stress. Experimental animals choose “comfort food” to reduce depression and/or anxiety status [97,100]. As demonstrated in the limited sucrose intake paradigm, sucrose solution reduced stress responses [101,102,103]. Consumption of sucrose or highly sweet foods can limit activation of the stress system by an effect on the reward circuits in the brain [104,105]. Conversely, enhanced sugar intake can promote stress-driven emotional and addictive behaviours [50,106].

2.5. Changes in the Reward Brain System Following HSD

The reward brain system (the mesocorticolimbic pathway) is based on brain structures and neural pathways related to rewards, motivation, and the desire for pleasure [107,108]. The reward system contains numerous interrelated structures, such as the ventral tegmental area, nucleus accumbens (NAc), prefrontal cortex (PFC), hippocampus, and amygdala [109,110]. In humans, excessive consumption of glucose or fructose predisposed individuals to changes in activity in areas of the brain known to be associated with reward, learning reward, and eating behaviour. The fructose effect was limited to the activity of neurons, whereas glucose had a wide impact on the brain [111,112,113]. A diet high in sugar, including glucose, may cause excessive distribution of glucose to the brain [114], contributing to behavioural changes in food intake [115].
Recent preclinical data indicate that excessive sugar intake changed the reward circuitry at neurochemical and cellular levels as well as developed addictive-like behaviours and emotional states [116,117,118,119,120,121,122]. The changes depended on sugar dietary patterns. Thus, at the neurochemical level intermittent access to sucrose enhanced the dopaminergic, opioid and cholinergic neurotransmission in the mesocorticolimbic system [123,124]. Binge-like sugar consumption facilitated dopamine (DA) release in the NAc, similarly to drugs of abuse [125]. Long-term consumption of sucrose altered nicotinic acetylcholine receptor (nAChR) expression in the NAc, whereas nAChR compounds evoked different effects on sucrose intake depending on its long-term vs. short-term exposure [126,127]. Sucrose solution given chronically (7 or 21 days) to male rats increased the level of accumbal DA neurotransmission [124,125,128]. Additionally, a 25% glucose solution over 31 days increased the expression of accumbal D1 and μ-opioid receptors in female rats [129]. Finally, a maternal HSD changed the MC4R expression in brain reward structures in male and female offspring [130,131,132].
Moreover, at the cellular levels a binge-like sucrose intake by rats for long-term (12 weeks) period, but not for short-term (4 weeks) period, resulted in alterations within the medium spiny neurons in the NAc shell (but not core) with a significant reduction in dendritic length and increased distal dendritic spine density [133]. Sucrose served as a potent modulator of neuron morphology following prolonged heavy use, as its consumption enhanced excitatory synaptic strength onto NAc DA neurons [127]. In addition to sucrose, previous studies have demonstrated similarities following acute exposure to another non-caloric sweetener saccharin at the level of the NAc [134,135,136]. Very recent study indicated that chronic (12 weeks) 5% sucrose consumption to mice resulted in a reduction in the serotonin (5-HT) receptors innervation within the PFC and dentate gyrus of the hippocampus, a reduction in the number of microglia in the latter brain structure as well as a decrease in the density of the vesicular glutamate transporter (VGLUT3) and of 5-HT/VGUT3 varicosities but not in the number of oligodendrocyte progenitor cells [137].

2.6. Behavioural Consequences of HSD

In humans, HSD let to memory impairment and cognitive deficits [49,138], whereas it does not increase the improvement of semantic memory [139]. An observational study in children indicated that sucrose consumption was associated with the incidence of impulsivity and attention deficit hyperactivity disorder (ADHD) among 6-year-old boys and that persistent, high consumption or an increase in sugar consumption between 6 and 11 years of age was not associated with a higher prevalence of ADHD between 6 and 11 years of age [140].
Preclinical research reported that a higher (25%) but not low (5%) sucrose concentration given for a long time (12 weeks) to adolescent mice resulted in enhancement of hyperlocomotor response to novelty and defects in episodic and spatial memory in adulthood without modulation of learning processes [141]. In rodents, an HSD had a negative impact on aspects of behavioural tasks involving decision making and behaviour selection. A 2 h intake of sucrose over a 24-day period induced a significant spatial memory deficit as measured by site recognition in rats. Moreover, rats entering the delay discounting task had behavioural evidence of hippocampal dysfunction [142]. Hippocampus-dependent alteration in behaviours was accompanied with altered hippocampal neurogenesis such as a reduction in the proliferation and differentiation of newborn neurons in the dentate gyrus [141], which raises a positive correlation between high- and long-term exposure to sucrose during adolescent and neurocognitive defects in adulthood. Furthermore, anxiety-like or depressive-like symptoms being observed simultaneously following long-term 8–25% sucrose consumption [123,143,144,145,146,147,148,149], but not with 5% sucrose consumption [137]. In addition, a high sucrose intake (30% wt/vol) in pregnant mice induced behavioural phenotypes similar to ADHD in the offspring, manifested by increased motor activity, and impulsivity. As well as, high fructose consumption (30% wt/vol) induced hyperactive behaviour, similar to the results obtained with sucrose treatment [150]. These findings suggest that the maternal HSD (sucrose or fructose) consumption could be considered a potentially new risk factor in neurobehavioural dysregulation, such as development of ADHD and/or possibly addiction-like behaviours (see below).

3. Maternal HSD Overabundance as Offspring Disease Factor

Increased consumption of added sugars is visible in developing societies, especially among children and adolescents. In pregnant women limited sugar intake is recommended. The period of pregnancy and lactation is a special time for the foetus. Consistent with the developmental origins of health and disease hypothesis, exposure to external environmental factors in early life influences the proper growth and development of every organism and could play a crucial role in determining the risk of developing diseases in adulthood [151,152,153]. One important external factor is the composition and type of the maternal diet consumed during pregnancy and lactation [154]. Recently, animal studies and clinical trials have confirmed the involvement of maternal nutrition in the early life of the offspring. Improper nutrition could increase the susceptibility of offspring to diseases in early life or in adulthood [155,156].
Clinical observations have indicated that sugar-rich foods are desirable during pregnancy and were overeaten during the study period, suggesting that many reproductive women are exposed to the adverse effects of an HSD [118]. Carbohydrates in the maternal diet during pregnancy should make up 45–64% of daily calories, including approximately 6–9 servings of whole grains per day. Breastfeeding women require approximately 500 additional kcal/day in addition to what is recommended for nonpregnant women [157]. A serious risk to the health of the mother and the foetus is incurred by being overweight and obese. Among women of reproductive age (20–39 years old), 31.8% were obese in 2011–2012. Women with a higher body mass index before pregnancy have a greater risk of adverse perinatal outcomes [158]. HSDs during pregnancy and lactation leading to overweight and obesity can increase the risk of gestational diabetes mellitus, high blood pressure, and abnormal growth of the foetus [159,160]. Furthermore, human observational studies have suggested that lactation influences insulin and glucose homeostasis [161]. In follow-up studies, lactating diabetic women showed improved glucose tolerance and lowered fasting glucose [162]. In obese women during lactation, insulin levels and the insulin-to-glucose ratio were significantly lower, and carbohydrate utilization and total energy expenditure were higher [163]. It is also of interest that maternal diseases, promoted by HSDs in pregnancy and lactation, can be transferred to offspring with the manifestation of metabolic or neurological diseases [164,165,166,167,168,169].
In preclinical studies, HSDs during pregnancy and lactation caused effects similar to those observed in clinical trials. Apart from metabolic syndrome (weight gain and increased levels of triglycerides, cholesterol, leptin, glucose, and insulin), HSDs induced cardiac diseases and increased blood pressure in rodents [170,171]. In other studies, a maternal diet rich in fats and/or sugars during pregnancy and lactation predisposed the offspring to developing the metabolic disorders observed in the mother or promoted neurobehavioural disorders [172,173,174,175,176,177,178,179,180,181]. Maternal HSD (64% kcal) affect the memory processes, such as impaired recognition memory and spatial memory, and also disrupting the glutamate NMDA receptor in the PFC of rat offspring [182]. Moreover, “junk food” rich in sugar, eaten during pregnancy and lactation, predisposes individuals to hyperphagia due to enhancement of the choice of palatable food, also in offspring [183].

4. HSD as a Risk Factor to Develop SUD and Food Addiction

SUD is a serious complex disorder that affects the brain and behaviour and leads to uncontrolled use of a legal or illegal substance despite harmful consequences and unsolved issues from the perspective of public health [184]. Many factors influence SUD development contain social-age and economic situation, biological-genetic predisposition or parental drug history, and psychological-high impulsivity or sensation-seeking, determinants. Currently, the prevention of SUD is quite costly and ineffective due to the lack of appropriate therapies, the resistance and the relapse of patients to SUD. Broader SUD education and effective psychotherapy are required, and pharmacotherapy is currently being sought.
According to the World Drug Report, in 2018 nearly 1 in every 19 people (269 million people worldwide) had used drugs at least once in the previous year, and the number of users of any drug globally increased by approximately 28% over the 2009–2018 period. Of these people, in the past year, approximately 35 million had drug use disorder symptoms, corresponding to 0.7% of the population aged 15–64 years old [185]. Several drugs can involve addictive behaviour in both humans and animals. These drugs consist of psychostimulants, opioids, cannabinoids, nicotine, and alcohol. Their behavioural outcomes are realized through distinct effector mechanisms, such as neurotransmitter transporters, ion channels, and receptor proteins. As a consequence of long-term intake of all drugs of abuse—after initial positive emotions—they generate compulsive and uncontrollable motivations to seek and reuse them despite negative consequences, e.g., psychic, somatic, and/or vegetative disturbances) [186]. It is well documented that drugs of abuse—despite different pharmacological mechanisms of action via neurotransmitter transporters, ion channels or receptor proteins—share the common feature that they trigger addictive potential. The psychostimulant—cocaine rapidly increases DA neurotransmission within the mesocorticolimbic circuitry of the brain from the ventral tegmental area to the ventral striatum (=NAc) and the PFC, and induces synaptic plasticity in DA neurons after acute, passive administration and in cocaine self-administered rats [187,188]. Notably, addiction-like behaviour has also been associated with disrupted homoeostasis of other monoamine or amino acid neurocircuitries.
It should also be indicated that the consumption of sugar or HSD can be addictive [189,190] and can predispose individuals to the risk of food addiction. At the neurochemical level, in both animals and humans there are significant similarities and overlaps between drugs of abuse and sugar, from brain neurochemistry to behaviour [189]. Long-term sugar consumption produced cocaine-like effects, changes in striatal D1 and D2 receptors, and changes in mood, possibly through its ability to induce reward and pleasure [105,119,120,122,143]. Additionally, the availability of DA precursors may be altered by prolonged consumption of HSD [191]. Sugar consumption produces cocaine-like effects, changes in striatal D1 and D2 receptors, and changes in mood, possibly through its ability to induce reward and pleasure, leading to sugar-seeking behaviour [119,120,121,123]. Sugar acts as an addictive molecule, causing overeating and withdrawal; it also affects mood, behaviour, learning, and memory [192]. These features are also characteristic for drugs of abuse [193]. More importantly, food addiction, such as SUD, is characterized by seeking and compulsive behaviours. Thus, bingeing sweet food or long-term exposure of HSD to rodents let to overweight or obesity [194], whereas sucrose or sugar withdrawal predispose them to behaviour similar to depression and anxiety [120,143]. Fructose (8% solution) given for 21 days to male rats increased bingeing behaviour following a long-term intermittent access model and decreased NAc shell neuron activation [195].
Moreover, recent data have indicated that diet patterns, especially maternal HFD, can increase addiction-like behaviours in offspring [130,196,197,198], but there are few sets of data on the impact of an HSD in the prenatal and postnatal periods [50,131]. As shown, a maternal HSD during pregnancy and lactation (Table 3) or postnatal HSD consumption (Table 4) impacts sensitivity to addiction-like behaviours. For example, our studies indicated that maternal HSD increased cocaine-seeking behaviour in offspring in a rat model of cocaine self-administration [130,131]. Maternal sucrose and HFCS intake increased alcohol intake in females and caused hyperactivity in response to amphetamine in male offspring [199]. Similar to the prenatal period, postnatal high-sugar intake promoted locomotor hyperactivity to cocaine [200] and amphetamine [201,202,203], whereas sugar either increased or decreased cocaine [204] and amphetamine [203,205] behaviour in the conditioned place preference. Furthermore, palatable food in amphetamine administration provoked memory impairment [206] and food intake dysregulation in rats [207] and baboons [208]. In addition, a diet rich in corn starch enhanced depressive-like behaviour in male rats after cocaine treatment [209]. The available laboratory data strongly suggest that a maternal HSD or high-sugar postnatal bingeing predisposes to changes in the sensitivity to addictive drugs; however, the behavioural outcomes depend on diet time-frame exposure and drug-dependent measurements.

5. HSD Evokes Behavioural Predictors of Drug of Abuse

Several factors have been proposed to be potentially involved in the onset and development of SUD. SUD—development and abuse—is correlated with physical and mental illnesses such as anxiety, depression, personality disorders, eating disorders, and abnormal mood changes. Among them, mood disturbances are important predictors of frequent use of drugs of abuse and problems. Anxiety disorders are a group of mental disorders characterized by a significant feeling of anxiety or fear [210]. Women are 1.5 to 2 times more likely than men to receive a diagnosis of an anxiety disorder. The significant mediators of anxiety in the central nervous system are norepinephrine, 5-HT, DA, and gamma-aminobutyric acid [211]. In fact, anxiety (especially social anxiety disorder (=social phobia)), characterized by sentiments of fear and subjectively unpleasant feelings in the face of anticipated events, was found to be a trigger for drug use in adolescence or young adulthood [212,213,214,215,216,217]. Higher levels of anxiety symptoms were associated with earlier alcohol consumption by humans [218,219] and in a rodent model of depression [220]. Moreover, anxiety triggers a greater desire for cocaine [221,222] or nicotine [223,224,225] addiction, as observed in human studies.
Similarly, depressive symptoms contribute to the use of addictive drugs. According to the WHO, 280 million people worldwide have depression, and an estimated 3.8% of the population affected, including 5.0% among adults, suffers from depression. Depression affects an estimated 15 adults (6.7%) in any given year. Women are more likely to be affected by depression than men. Among depressive symptoms, the strongest predictors of depression are crying, pessimism, changes in appetite and loss of interest, and they enhance the frequency of cocaine use or greater severity of cocaine use in current cocaine users [226,227,228,229,230]. Clinical findings have also revealed that anhedonia (=lack of interest or pleasure in activities) was associated with lifetime cocaine use and dependence [231] and enhanced the addictive effects of cocaine, leading to greater use and poorer treatment outcomes [232]. Another important predictor involved in the onset and development of SUD is impulsivity [233], which refers to a tendency to act without careful thinking or to react prematurely [234,235]. This feature is a marker of psychiatric symptoms in general and in SUD. As shown in many clinical and preclinical studies, impulsivity is a vulnerability trait for SUD and a property of SUD [236,237,238]. Moreover, variability in impulsivity could increase the odds of both heavy drinking and alcohol-related problems [239], and a significant relationship between impaired response inhibition or impulsivity and high-risk alcohol use in nonclinical populations was demonstrated [240]. Bozkurt and colleagues [241] found that—among others—self-reported trait impulsivity predicted alcohol use severity in treatment-seeking patients [242]. There have also been epidemiological studies showing a direct association between the sensation/novelty-seeking trait and SUD, especially cocaine use disorder (CUD) [243,244,245,246]. Sensation/novelty seekers are more prone to experimenting with addictive drugs [247,248]. The laboratory data support the clinical observations since sensation/novelty-seeking has been implicated in vulnerability to CUD in rodent models [249]. Thus, the sensation/novelty-seeking trait (recognized as a high-responder (HR)) trait in novelty-induced locomotor activity or high-novelty-preference trait (HNP) in novelty-induced place preference (=the propensity to choose a new environment in a free choice procedure) in rats is associated with differential sensitivity to psychostimulant drugs of abuse [250,251]. As demonstrated by Belin and colleagues, HR rats represent a “drug use prone” phenotype that causes an individual to develop CUD (but not compulsive cocaine self-administration), whereas HNP rats copy with an “addiction prone” phenotype that enhances the shift from sustained to compulsive drug intake and addiction [249].
HSDs have also been linked with emotional or mood disturbances, such as stress, anxiety, and depression. Stress—a normal, useful reaction to trigger motivation, adaptation, and reaction to the surrounding environment—at high levels can result in biological, psychological, and social problems and even serious harm to people and animals [252]. As shown, nine weeks of exposure to 15% fructose solution resulted in passive stress-coping behaviour in female mice, whereas 15–30% fructose evoked lower self-care behaviour and short-term impairment of spatial memory [253]. Interestingly, the induction of chronic social stress led to a decrease in glucose uptake, particularly in the brain tissue, which could contribute to a reduction in the incentive to reward sweet tastes [254]. The inductors of anxiety symptoms seem to be—among others—simple sugars, refined carbohydrates, and sweeteners consumed in large amounts. Limited animal studies have demonstrated an increase in anxiety-like symptoms after a chronic 7-week diet rich in sweeteners [255]. In a human study, in turn, an improvement was shown in anxiety symptoms among obese participants following a low-carbohydrate diet [256].
Depression (major depressive disorder) is a mental disorder that negatively affects feeling and thinking [257]. It is caused by a combination of genetic, biological, environmental, and psychological factors [258]. Adult female rats fed a high fructose diet (55% kcal) showed increased depressive-like behaviours in the forced swim test [259], whereas preadolescent, but not adult, male rats showed increased activity in the open field test and reduced time spent in the open arms on the elevated plus maze test. Moreover, fructose-fed rats spent more time immobile in the forced swim test [145]. Human research has revealed that high consumption of foods rich in added sugars and sweetened drinks increased the risk of depression over the next few years [260] and resulted in a high incidence and recurrence of mood disorders [261]. More recent studies have confirmed that excessive consumption of sugar-sweetened soft drinks was associated with an increased risk of depression in adults and Asian adolescents [262]. In overweight people, increased consumption of SSBs was associated with an increased incidence of a diagnosis of depression [263]. The results of studies on repetitive body-centred behaviour found that high-sugar consumption was associated with greater impulsivity without planning, whereas diets high in both saturated fat and sugar were associated with greater motor impulsiveness among study participants [264]. Large amounts of added sugar in food induced greater sensitivity to delays, greater preference for larger size, and increased size sensitivity [265], according to an earlier study in rats in the impulsive choice task [266].
Correlation between foetal programming and metabolic health of the offspring in human epidemiological studies of maternal HFD and HSD is strong, especially in long-term offspring implications such as obesity and diabetes. However, no data from clinical trials exist linking maternal consumption of HSD during pregnancy or postnatal consumption and predisposition to SUD. Preclinical studies emphasize the role of HSD in behavioural, neural, and neurochemical changes causing a higher vulnerability to addiction-like behaviours leading to an increased preference to consume palatable foods and sensitivity to drugs of abuse, but many more clinical trials are necessary to understand this important mechanism.

6. Conclusions and Perspective

This paper has emphasized the latest data showing that HSD, added sugar, and sugar components are some predictive factors in changing behaviour. Epidemiological studies have shown that sugar intake is too high in industrialized countries. An increasing number of adults and children consume added sugar, SSBs, HFCS, and refined sugar. Preclinical studies have pointed out that a maternal HSD during pregnancy and lactation is strongly correlated with offspring’s metabolic and mental disorder development (mood disorders and compulsive or impulsive dysfunction). HSD and sugar can be addictive and can predispose an individual to the risk of food addiction. Recent limited animal studies have demonstrated that perinatal or postnatal chronic exposure to HSD or sugar (glucose and/or fructose solutions) changed the sensitivity to drugs of abuse in addiction-like behaviour models. In addition to increasing the locomotor activity of the animals, sugar in combination with the drug enhanced drug preferences and sensitivity. Then, HSD promoted an increase in depressive-like behaviour in rodents with a history of addiction-like behaviours. Finally, in vivo functional studies have confirmed that dietary sugar and drugs of abuse use the same brain structures, being a part of the reward system pathways.
Clinical observations support preclinical findings and suggest a link between diet patterns and the risk of mental disorders. Epidemiological data showed increases in depressive and anxiety behaviours in patients with HSD histories and drug users. Sugar overconsumption can also affect mood, causing compulsivity and impulsivity repetitive behaviours, which are SUD predictors. The current clinical nonrandomized data require better exploration and research, especially since SUD is an incurable disease and a social and economic community problem, and it affects an increasing number of people of all ages, regardless of gender.
In summary, in this paper, we discussed the current knowledge on maternal and postnatal sugar intake as predictor of the development of mental disorders, including SUD. Based on the data, it is strongly recommended limitation in sugar consumption in an individual’s everyday diet, especially for children and pregnant women. Future epidemiological analyses and clinical research are needed to better explain the effect of sugar intake on the SUD development.

Author Contributions

Conceptualization and supervision, M.F.; methodology, K.W. (Kacper Witek) and K.W. (Karolina Wydra); writing—original draft preparation, M.F., K.W. (Kacper Witek) and K.W. (Karolina Wydra); writing—review and editing, M.F., K.W. (Kacper Witek). All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the statutory fund of the Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Asif, M.; Akram, M.; Profile, S.; Saeed, T.; Naveed, A.; Asif, H.M.; Khan, M.I.; Akhtar, N.; Ur Rehman, R.; Shah, S.M.A.; et al. Carbohydrates. Int. Res. J. Biochem. Bioinform. 2011, 1, 1–5. [Google Scholar]
  2. Lee, C.K. The Chemistry and Biochemistry of the Sweetness of Sugars. Adv. Carbohydr. Chem. Biochem. 1987, 45, 199–351. [Google Scholar] [CrossRef] [PubMed]
  3. Cummings, J.H.; Stephen, A.M. Carbohydrate Terminology and Classification. Eur. J. Clin. Nutr. 2007, 61, S5–S18. [Google Scholar] [CrossRef] [Green Version]
  4. Hannou, S.A.; Haslam, D.E.; McKeown, N.M.; Herman, M.A. Fructose Metabolism and Metabolic Disease. J. Clin. Investig. 2018, 128, 545. [Google Scholar] [CrossRef] [PubMed]
  5. Rippe, J.M.; Angelopoulos, T.J. Relationship between Added Sugars Consumption and Chronic Disease Risk Factors: Current Understanding. Nutrients 2016, 8, 697. [Google Scholar] [CrossRef] [Green Version]
  6. Slavin, J.; Carlson, J. Carbohydrates. Adv. Nutr. 2014, 5, 760. [Google Scholar] [CrossRef] [Green Version]
  7. U.S. Department of Health and Human Services; U.S. Department of Agriculture. 2015–2020 Dietary Guidelines for Americans, 8th Ed.; December 2015. Available online: http://health.gov/dietaryguidelines/2015/guidelines/ (accessed on 13 June 2021).
  8. Dietary Guidelines Advisory Committee. Scientif ic Report of the 2020 Dietary Guidelines Advisory Committee: Advisory Report to the Secretary of Agriculture and the Secretary of Health Human Services; U.S. Department of Agriculture, Agricultural Research Service: Washington, DC, USA, 2020. [Google Scholar]
  9. Toop, C.R.; Gentili, S. Fructose Beverage Consumption Induces a Metabolic Syndrome Phenotype in the Rat: A Systematic Review and Meta-Analysis. Nutrients 2016, 8, 577. [Google Scholar] [CrossRef] [Green Version]
  10. National Research Council (US) Subcommittee on Laboratory Animal Nutrition. Nutrient Requirements of Laboratory Animals, 4th ed.; National Academies Press: Washington, DC, USA, 1995. [Google Scholar] [CrossRef]
  11. Ricci, M. Laboratory Animal Control Diets: Very Important, Often Neglected. Lab Anim. 2015, 44, 240–241. [Google Scholar] [CrossRef]
  12. Bruce, H.M.; Parkes, A.S. Feeding and Breeding of Laboratory Animals: IX. A Complete Cubed Diet for Mice and Rats. J. Hyg. 1949, 47, 202. [Google Scholar] [CrossRef] [Green Version]
  13. Tuck, C.J.; de Palma, G.; Takami, K.; Brant, B.; Caminero, A.; Reed, D.E.; Muir, J.G.; Gibson, P.R.; Winterborn, A.; Verdu, E.F.; et al. Nutritional Profile of Rodent Diets Impacts Experimental Reproducibility in Microbiome Preclinical Research. Sci. Rep. 2020, 10, 17784. [Google Scholar] [CrossRef]
  14. Welsh, J.A.; Sharma, A.J.; Grellinger, L.; Vos, M.B. Consumption of Added Sugars Is Decreasing in the United States. Am. J. Clin. Nutr. 2011, 94, 726–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. McGuire, S.; Ervin, R.B.; Kit, B.K.; Carroll, M.D.; Ogden, C.L. Consumption of Added Sugar among U.S. Children and Adolescents, 2005–2008. NCHS Data Brief No 87. Hyattsville, MD: National Center for Health Statistics. Adv. Nutr. 2012, 3, 534. [Google Scholar] [CrossRef] [Green Version]
  16. Herrick, K.A.; Fryar, C.D.; Hamner, H.C.; Park, S.; Ogden, C.L. Added Sugars Intake among US Infants and Toddlers. J. Acad. Nutr. Diet. 2020, 120, 23. [Google Scholar] [CrossRef] [PubMed]
  17. Bowman, S.A.; Clemens, J.C.; Friday, J.E.; Moshfegh, A.J. 2020. Food Patterns Equivalent Intakes from Food: Mean Amounts Consumed per Individual, What We Eat in America, NHANES 2017–2018, Tables 1–4. Worldwide Web Site: Food Surveys Research Group. Available online: https://www.ars.usda.gov/nea/bhnrc/fsrg/fped (accessed on 10 April 2022).
  18. Rosinger, A.; Herrick, K.; Gahche, J.; Park, S. Sugar-Sweetened Beverage Consumption Among U.S. Youth, 2011–2014. NCHS Data Brief. 2017, 271, 1–8. [Google Scholar]
  19. Lundeen, E.A.; Park, S.; Pan, L.; Blanck, H.M. Daily Intake of Sugar-Sweetened Beverages among US Adults in 9 States, by State and Sociodemographic and Behavioral Characteristics, 2016. Prev. Chronic Dis. 2018, 15, E154. [Google Scholar] [CrossRef] [PubMed]
  20. Trumbo, P.; Schlicker, S.; Yates, A.A.; Poos, M. Dietary Reference Intakes for Energy, Carbohydrate, Fiber, Fat, Fatty Acids, Cholesterol, Protein and Amino Acids. J. Am. Diet Assoc. 2002, 102, 1621–1630. [Google Scholar] [CrossRef]
  21. U.S. Department of Agriculture; U.S. Department of Health and Human Services. Dietary Guidelines for Americans, 2020–2025. 9th Edition. December 2020. Available online: DietaryGuidelines.gov (accessed on 10 April 2022).
  22. Yang, Q.; Zhang, Z.; Gregg, E.W.; Flanders, W.D.; Merritt, R.; Hu, F.B. Added Sugar Intake and Cardiovascular Diseases Mortality Among US Adults. JAMA Int. Med. 2014, 174, 516–524. [Google Scholar] [CrossRef] [Green Version]
  23. Popkin, B.M.; Adair, L.S.; Ng, S.W. Global Nutrition Transition and the Pandemic of Obesity in Developing Countries. Nutr. Rev. 2012, 70, 3–21. [Google Scholar] [CrossRef] [Green Version]
  24. Bandy, L.K.; Scarborough, P.; Harrington, R.A.; Rayner, M.; Jebb, S.A. Reductions in Sugar Sales from Soft Drinks in the UK from 2015 to 2018. BMC Med. 2020, 18, 20. [Google Scholar] [CrossRef] [Green Version]
  25. Stephan, B.C.M.; Wells, J.C.K.; Brayne, C.; Albanese, E.; Siervo, M. Increased Fructose Intake as a Risk Factor for Dementia. J. Gerontol. Ser. A 2010, 65A, 809–814. [Google Scholar] [CrossRef] [Green Version]
  26. Malik, V.S.; Hu, F.B. Fructose and Cardiometabolic Health: What the Evidence from Sugar-Sweetened Beverages Tells Us. J. Am. Coll. Cardiol. 2015, 66, 1615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Stanhope, K.L.; Bremer, A.A.; Medici, V.; Nakajima, K.; Ito, Y.; Nakano, T.; Chen, G.; Fong, T.H.; Lee, V.; Menorca, R.I.; et al. Consumption of Fructose and High Fructose Corn Syrup Increase Postprandial Triglycerides, LDL-Cholesterol, and Apolipoprotein-B in Young Men and Women. J. Clin. Endocrinol. Metab. 2011, 96, E1596–E1605. [Google Scholar] [CrossRef]
  28. Vos, M.B.; Kimmons, J.E.; Gillespie, C.; Welsh, J.; Blank, H.M. Dietary Fructose Consumption Among US Children and Adults: The Third National Health and Nutrition Examination Survey. Medscape J. Med. 2008, 10, 160. [Google Scholar] [PubMed]
  29. Elliott, S.S.; Keim, N.L.; Stern, J.S.; Teff, K.; Havel, P.J. Fructose, Weight Gain, and the Insulin Resistance Syndrome. Am. J. Clin. Nutr. 2002, 76, 911–922. [Google Scholar] [CrossRef]
  30. Bray, G.A. Energy and Fructose from Beverages Sweetened With Sugar or High-Fructose Corn Syrup Pose a Health Risk for Some People. Adv. Nutr. 2013, 4, 220. [Google Scholar] [CrossRef] [Green Version]
  31. Bray, G.A.; Nielsen, S.J.; Popkin, B.M. Consumption of High-Fructose Corn Syrup in Beverages May Play a Role in the Epidemic of Obesity. Am. J. Clin. Nutr. 2004, 79, 537–543. [Google Scholar] [CrossRef] [PubMed]
  32. Stanhope, K.L. Sugar Consumption, Metabolic Disease and Obesity: The State of the Controversy. Crit. Rev. Clin. Lab. Sci. 2016, 53, 52–67. [Google Scholar] [CrossRef]
  33. Macdonald, I.A. A Review of Recent Evidence Relating to Sugars, Insulin Resistance and Diabetes. Eur. J. Nutr. 2016, 55, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Johnson, R.J.; Sánchez-Lozada, L.G.; Andrews, P.; Lanaspa, M.A. Perspective: A Historical and Scientific Perspective of Sugar and Its Relation with Obesity and Diabetes. Adv. Nutr. 2017, 8, 412–422. [Google Scholar] [CrossRef] [PubMed]
  35. Yoshida, Y.; Simoes, E.J. Sugar-Sweetened Beverage, Obesity, and Type 2 Diabetes in Children and Adolescents: Policies, Taxation, and Programs. Curr. Diab. Rep. 2018, 18, 1–10. [Google Scholar] [CrossRef]
  36. Schetz, M.; de Jong, A.; Deane, A.M.; Druml, W.; Hemelaar, P.; Pelosi, P.; Pickkers, P.; Reintam-Blaser, A.; Roberts, J.; Sakr, Y.; et al. Obesity in the Critically Ill: A Narrative Review. Intensive Care Med. 2019, 45, 757–769. [Google Scholar] [CrossRef] [PubMed]
  37. Faruque, S.; Tong, J.; Lacmanovic, V.; Agbonghae, C.; Minaya, D.M.; Czaja, K. The Dose Makes the Poison: Sugar and Obesity in the United States—A Review. Pol. J. Food Nutr. Sci. 2019, 69, 219–233. [Google Scholar] [CrossRef] [PubMed]
  38. Haththotuwa, R.N.; Wijeyaratne, C.N.; Senarath, U. Worldwide Epidemic of Obesity; Elsevier: Oxford, UK, 2013; Volume 1, pp. 3–11. [Google Scholar] [CrossRef]
  39. Iadecola, C. Sugar and Alzheimer’s Disease: A Bittersweet. Nat. Neurosci. 2015, 18, 477. [Google Scholar] [CrossRef] [Green Version]
  40. Johnson, R.J.; Gomez-Pinilla, F.; Nagel, M.; Nakagawa, T.; Rodriguez-Iturbe, B.; Sanchez-Lozada, L.G.; Tolan, D.R.; Lanaspa, M.A. Cerebral Fructose Metabolism as a Potential Mechanism Driving Alzheimer’s Disease. Front. Aging Neurosci. 2020, 12, 560865. [Google Scholar] [CrossRef]
  41. Mohamed, H.E.; Asker, M.E.; Shaheen, M.A.; Eissa, R.G.; Younis, N.N. Alleviation of Fructose-Induced Alzheimer’s Disease in Rats by Pioglitazone and Decaffeinated Green Coffee Bean Extract. J. Food Biochem. 2021, 45, e13715. [Google Scholar] [CrossRef]
  42. Marques, A.; Dutheil, F.; Durand, E.; Rieu, I.; Mulliez, A.; Fantini, M.L.; Boirie, Y.; Durif, F. Glucose Dysregulation in Parkinson’s Disease: Too Much Glucose or Not Enough Insulin? Parkinsonism Relat. Disord. 2018, 55, 122–127. [Google Scholar] [CrossRef]
  43. Palavra, N.C.; Lubomski, M.; Flood, V.M.; Davis, R.L.; Sue, C.M. Increased Added Sugar Consumption Is Common in Parkinson’s Disease. Front. Nutr. 2021, 8, 628845. [Google Scholar] [CrossRef]
  44. Zhang, S.; Lachance, B.B.; Mattson, M.P.; Jia, X. Glucose Metabolic Crosstalk and Regulation in Brain Function and Diseases. Prog. Neurobiol. 2021, 204, 102089. [Google Scholar] [CrossRef]
  45. Mergenthaler, P.; Lindauer, U.; Dienel, G.A.; Meisel, A. Sugar for the Brain: The Role of Glucose in Physiological and Pathological Brain Function. Trends Neurosci. 2013, 36, 587. [Google Scholar] [CrossRef] [Green Version]
  46. Tang, B.L. Glucose, Glycolysis, and Neurodegenerative Diseases. J. Cell. Physiol. 2020, 235, 7653–7662. [Google Scholar] [CrossRef] [PubMed]
  47. Spagnuolo, M.S.; Iossa, S.; Cigliano, L. Sweet but Bitter: Focus on Fructose Impact on Brain Function in Rodent Models. Nutrients 2021, 13, 1. [Google Scholar] [CrossRef] [PubMed]
  48. Molteni, R.; Barnard, R.J.; Ying, Z.; Roberts, C.K.; Gomez-Pinilla, F. A High-Fat, Refined Sugar Diet Reduces Hippocampal Brain-Derived Neurotrophic Factor, Neuronal Plasticity, and Learning. Neuroscience 2002, 112, 803–814. [Google Scholar] [CrossRef] [Green Version]
  49. Beilharz, J.E.; Maniam, J.; Morris, M.J. Diet-Induced Cognitive Deficits: The Role of Fat and Sugar, Potential Mechanisms and Nutritional Interventions. Nutrients 2015, 7, 6719–6738. [Google Scholar] [CrossRef] [Green Version]
  50. Jacques, A.; Chaaya, N.; Beecher, K.; Ali, S.A.; Belmer, A.; Bartlett, S. The Impact of Sugar Consumption on Stress Driven, Emotional and Addictive Behaviors. Neurosci. Biobehav. Rev. 2019, 103, 178–199. [Google Scholar] [CrossRef] [PubMed]
  51. Bagnol, D.; Al-Shamma, H.A.; Behan, D.; Whelan, K.; Grottick, A.J. Diet-Induced Models of Obesity (DIO) in Rodents. Curr. Protoc. Neurosci. 2012, 59, 9–38. [Google Scholar] [CrossRef] [PubMed]
  52. Rosini, T.C.; Ramos da Silva, A.S.; de Moraes, C. Diet-Induced Obesity: Rodent Model for the Study of Obesity-Related Disorders. Rev. Assoc. Méd. Bras. 2012, 58, 383–387. [Google Scholar] [CrossRef] [Green Version]
  53. Dashty, M. A Quick Look at Biochemistry: Carbohydrate Metabolism. Clin. Biochem. 2013, 46, 1339–1352. [Google Scholar] [CrossRef]
  54. Nakrani, M.N.; Wineland, R.H.; Anjum, F. Physiology, Glucose Metabolism; StatPearls Publishing LLC: Tampa, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK560599/ (accessed on 10 April 2022).
  55. Berg, J.M.; Tymoczko, J.L.; Stryer, L. Biochemistry, 5th ed.; Freeman, W.H., Ed.; Madison Avenue: New York, NY, USA, 2002; Chapter 17; pp. 497–514. ISBN 9788131248850. [Google Scholar]
  56. Deng, D.; Yan, N. GLUT, SGLT, and SWEET: Structural and Mechanistic Investigations of the Glucose Transporters. Protein Sci. 2016, 25, 546–558. [Google Scholar] [CrossRef] [Green Version]
  57. Navale, A.M.; Paranjape, A.N. Glucose Transporters: Physiological and Pathological Roles. Biophys. Rev. 2016, 8, 5. [Google Scholar] [CrossRef] [Green Version]
  58. Scheepers, A.; Joost, H.G.; Schurmann, A. The Glucose Transporter Families SGLT and GLUT: Molecular Basis of Normal and Aberrant Function. JPEN J. Parenter. Enter. Nutr. 2004, 28, 364–371. [Google Scholar] [CrossRef] [Green Version]
  59. Koepsell, H. Glucose Transporters in Brain in Health and Disease. Pflug. Arch. Eur. J. Physiol. 2020, 472, 1299–1343. [Google Scholar] [CrossRef]
  60. Benarroch, E.E. Brain Glucose Transporters. Neurology 2014, 82, 1374–1379. [Google Scholar] [CrossRef] [PubMed]
  61. McMillin, J.M. Blood Glucose. In Clinical Methods: The History, Physical, and Laboratory Examinations, 3rd ed.; Walker, H.K., Hall, W.D., Hurst, J.W., Eds.; Butterworths: Boston, MA, USA, 1990; Chapter 141. [Google Scholar]
  62. Georga, E.I.; Fotiadis, D.I.; Tigas, S.K. Pathophysiology and Management of Type 1 Diabetes. In Personalized Predictive Modeling in Type 1 Diabetes; Academic Press: Cambridge, MA, USA, 2018; pp. 15–35. [Google Scholar] [CrossRef]
  63. Douard, V.; Ferraris, R.P. Regulation of the Fructose Transporter GLUT5 in Health and Disease. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Merino, B.; Fernández-Díaz, C.M.; Cózar-Castellano, I.; Perdomo, G. Intestinal Fructose and Glucose Metabolism in Health and Disease. Nutrients 2020, 12, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Regnault, T.R.; Gentili, S.; Sarr, O.; Toop, C.R.; Sloboda, D.M. Fructose, Pregnancy and Later Life Impacts. Clin. Exp. Pharmacol. Physiol. 2013, 40, 824–837. [Google Scholar] [CrossRef]
  66. Chadt, A.; Al-Hasani, H. Glucose Transporters in Adipose Tissue, Liver, and Skeletal Muscle in Metabolic Health and Disease. Pflug. Arch. 2020, 472, 1273. [Google Scholar] [CrossRef]
  67. Hassel, B.; Elsais, A.; Frøland, A.S.; Taubøll, E.; Gjerstad, L.; Quan, Y.; Dingledine, R.; Rise, F. Uptake and Metabolism of Fructose by Rat Neocortical Cells in Vivo and by Isolated Nerve Terminals in Vitro. J. Neurochem. 2015, 133, 572–581. [Google Scholar] [CrossRef] [Green Version]
  68. Chiba, Y.; Murakami, R.; Matsumoto, K.; Wakamatsu, K.; Nonaka, W.; Uemura, N.; Yanase, K.; Kamada, M.; Ueno, M. Glucose, Fructose, and Urate Transporters in the Choroid Plexus Epithelium. Int. J. Mol. Sci. 2020, 21, 7230. [Google Scholar] [CrossRef]
  69. Glimcher, L.H.; Lee, A.H. From Sugar to Fat: How the Transcription Factor XBP1 Regulates Hepatic Lipogenesis. Ann. N. Y. Acad. Sci. 2009, 1173, E2. [Google Scholar] [CrossRef]
  70. Schwarz, J.M.; Clearfield, M.; Mulligan, K. Conversion of Sugar to Fat: Is Hepatic de Novo Lipogenesis Leading to Metabolic Syndrome and Associated Chronic Diseases? J. Am. Osteopath. Assoc. 2017, 117, 520–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Wolever, T.M.S.; Miller, J.B. Sugars and Blood Glucose Control. Am. J. Clin. Nutr. 1995, 62, 212S–221S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Röder, P.v.; Wu, B.; Liu, Y.; Han, W. Pancreatic Regulation of Glucose Homeostasis. Exp. Mol. Med. 2016, 48, e219. [Google Scholar] [CrossRef] [Green Version]
  73. Jerry Kaneko, J.; Harvey, J.J.; Bruss, M.L. Clinical Biochemistry of Domestic Animals; Academic Press: Cambridge, MA, USA, 2008. [Google Scholar] [CrossRef]
  74. Nadkarni, P.; Chepurny, O.G.; Holz, G.G. Regulation of Glucose Homeostasis by GLP-1. Prog. Mol. Biol. Transl. Sci. 2014, 121, 23–65. [Google Scholar] [CrossRef] [Green Version]
  75. Melmed, S.; Koenig, R.; Rosen, C.; Auchus, R.; Goldfine, A. Shlomo Melmed Ronald Koenig Clifford Rosen Richard Auchus Allison Goldfine 37. Complications of Diabetes Mellitus. In Williams Textbook of Endocrinology; Elsevier Health Sciences: Amsterdam, The Netherlands, 2019; pp. 1438–1520. [Google Scholar]
  76. Lee, A.A.; Owyang, C. Sugars, Sweet Taste Receptors, and Brain Responses. Nutrients 2017, 9, 653. [Google Scholar] [CrossRef] [Green Version]
  77. Chaudhari, N.; Roper, S.D. The Cell Biology of Taste. J. Cell Biol. 2010, 190, 285–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Depoortere, I. Taste Receptors of the Gut: Emerging Roles in Health and Disease. Gut 2014, 63, 179–190. [Google Scholar] [CrossRef]
  79. Furness, J.B.; Rivera, L.R.; Cho, H.J.; Bravo, D.M.; Callaghan, B. The Gut as a Sensory Organ. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 729–740. [Google Scholar] [CrossRef]
  80. Fernstrom, J.D.; Munger, S.D.; Sclafani, A.; de Araujo, I.E.; Roberts, A.; Molinary, S. Mechanisms for Sweetness. J. Nutr. 2012, 142, 1134S. [Google Scholar] [CrossRef]
  81. Scott, T.R.; Small, D.M. The Role of the Parabrachial Nucleus in Taste Processing and Feeding. Ann. N. Y. Acad. Sci. 2009, 1170, 372–377. [Google Scholar] [CrossRef]
  82. Abdalla, M.M.I. Ghrelin—Physiological Functions and Regulation. Eur. Endocrinol. 2015, 11, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zhang, F.; Chen, Y.; Heiman, M.; DiMarchi, R. Leptin: Structure, Function and Biology. Vitam. Horm. 2005, 71, 345–372. [Google Scholar] [CrossRef]
  84. Cummings, D.E.; Foster, K.E. Ghrelin-Leptin Tango in Body-Weight Regulation. Gastroenterology 2003, 124, 1532–1535. [Google Scholar] [CrossRef]
  85. Klok, M.D.; Jakobsdottir, S.; Drent, M.L. The Role of Leptin and Ghrelin in the Regulation of Food Intake and Body Weight in Humans: A Review. Obes. Rev. 2007, 8, 21–34. [Google Scholar] [CrossRef] [PubMed]
  86. García, A.S.E.; Moreno, A.G.M.; Reyes Castillo, Z. Papel de La Grelina y la Leptina en el Comportamiento Alimentario: Evidencias Genéticas y Moleculares. Endocrinol. Diabetes Nutr. 2021, 68, 654–663. [Google Scholar] [CrossRef]
  87. Nogueiras, R.; Tschöp, M.H.; Zigman, J.M. CNS Regulation of Energy Metabolism: Ghrelin versus Leptin. Ann. N. Y. Acad. Sci. 2008, 1126, 14. [Google Scholar] [CrossRef] [Green Version]
  88. Dallman, M.F.; Pecoraro, N.; Akana, S.F.; la Fleur, S.E.; Gomez, F.; Houshyar, H.; Bell, M.E.; Bhatnagar, S.; Laugero, K.D.; Manalo, S. Chronic Stress and Obesity: A New View of “Comfort Food”. Proc. Natl. Acad. Sci. USA 2003, 100, 11696. [Google Scholar] [CrossRef] [Green Version]
  89. Smith, S.M.; Vale, W.W. The Role of the Hypothalamic-Pituitary-Adrenal Axis in Neuroendocrine Responses to Stress. Dialogues Clin. Neurosci. 2006, 8, 383. [Google Scholar] [CrossRef]
  90. Miller, W.L. The Hypothalamic-Pituitary-Adrenal Axis: A Brief History. Horm. Res. Paediatr. 2018, 89, 212–223. [Google Scholar] [CrossRef]
  91. Hiller-Sturmhöfel, S.; Bartke, A. The Endocrine System: An Overview. Alcohol Health Res. World 1998, 22, 153. [Google Scholar]
  92. Tsigos, C.; Kyrou, I.; Kassi, E.; Chrousos, G.P.; Feingold, K.R.; Anawalt, B.; Boyce, A.; Chrousos, G.; de Herder, W.W.; Dhatariya, K.; et al. (Eds.) Stress: Endocrine Physiology and Pathophysiology. [Updated 2020 Oct 17]; Endotext [Internet], com, Inc.: South Dartmouth, MA, USA, 2000. Available online: https://www.ncbi.nlm.nih.gov/books/NBK278995/#strss-endo-pathophys.STRESS_ENDOCRINE_PA (accessed on 10 April 2022).
  93. Kinsella, M.T.; Monk, C. Impact of Maternal Stress, Depression & Anxiety on Fetal Neurobehavioral Development. Clin. Obstet. Gynecol. 2009, 52, 425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ulrich-Lai, Y.M.; Fulton, S.; Wilson, M.; Petrovich, G.; Rinaman, L. Stress Exposure, Food Intake, and Emotional State. Stress 2015, 18, 381. [Google Scholar] [CrossRef] [PubMed]
  95. Wardle, J.; Steptoe, A.; Oliver, G.; Lipsey, Z. Stress, Dietary Restraint and Food Intake. J. Psychosom. Res. 2000, 48, 195–202. [Google Scholar] [CrossRef]
  96. Dallman, M.F.; la Fleur, S.E.; Pecoraro, N.C.; Gomez, F.; Houshyar, H.; Akana, S.F. Minireview: Glucocorticoids—Food Intake, Abdominal Obesity, and Wealthy Nations in 2004. Endocrinology 2004, 145, 2633–2638. [Google Scholar] [CrossRef] [Green Version]
  97. Ulrich-Lai, Y.M.; Christiansen, A.M.; Ostrander, M.M.; Jones, A.A.; Jones, K.R.; Choi, D.C.; Krause, E.G.; Evanson, N.K.; Furay, A.R.; Davis, J.F.; et al. Pleasurable Behaviors Reduce Stress via Brain Reward Pathways. Proc. Natl. Acad. Sci. USA 2010, 107, 20529–20534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Nagy, M.R.; Gill, A.; Adams, T.; Gerras, J.; Mazin, L.; Leung, C.; Hasson, R.E. Stress-Induced Suppression of Food Intake in Overweight and Obese Adolescents. Psychosom. Med. 2019, 81, 814–820. [Google Scholar] [CrossRef]
  99. Herhaus, B.; Ullmann, E.; Chrousos, G.; Petrowski, K. High/Low Cortisol Reactivity and Food Intake in People with Obesity and Healthy Weight. Transl. Psychiatry 2020, 10, 40. [Google Scholar] [CrossRef] [Green Version]
  100. Inam, Q.U.A.; Ikram, H.; Shireen, E.; Haleem, D.J. Effects of Sugar Rich Diet on Brain Serotonin, Hyperphagia and Anxiety in Animal Model of Both Genders. Pak. J. Pharm. Sci. 2016, 29, 757–763. [Google Scholar]
  101. Egan, A.E.; Seemiller, L.R.; Packard, A.E.B.; Solomon, M.B.; Ulrich-Lai, Y.M. Palatable Food Reduces Anxiety-like Behaviors and HPA Axis Responses to Stress in Female Rats in an Estrous-Cycle Specific Manner. Horm. Behav. 2019, 115, 104557. [Google Scholar] [CrossRef]
  102. Packard, A.E.B.; Di, S.; Egan, A.E.; Fourman, S.M.; Tasker, J.G.; Ulrich-Lai, Y.M. Sucrose-Induced Plasticity in the Basolateral Amygdala in a ‘Comfort’ Feeding Paradigm. Brain Struct. Funct. 2017, 222, 4035. [Google Scholar] [CrossRef]
  103. Dess, N.K. Divergent Responses to Saccharin vs. Sucrose Availability after Stress in Rats. Physiol. Behav. 1992, 52, 115–125. [Google Scholar] [CrossRef]
  104. Gordon, E.L.; Ariel-Donges, A.H.; Bauman, V.; Merlo, L.J. What Is the Evidence for “Food Addiction?” A Systematic Review. Nutrients 2018, 10, 477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Hoebel, B.G.; Avena, N.M.; Bocarsly, M.E.; Rada, P. A Behavioral and Circuit Model Based on Sugar Addiction in Rats. J. Addict. Med. 2009, 3, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Singh, M. Mood, Food and Obesity. Front. Psychol. 2014, 5, 925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Yamaguchi, T.; Wang, H.L.; Li, X.; Ng, T.H.; Morales, M. Mesocorticolimbic Glutamatergic Pathway. J. Neurosci. 2011, 31, 8476–8490. [Google Scholar] [CrossRef] [Green Version]
  108. Volkow, N.D.; Michaelides, M.; Baler, R. The Neuroscience of Drug Reward and Addiction. Physiol. Rev. 2019, 99, 2115. [Google Scholar] [CrossRef]
  109. Arias-Carrión, O.; Stamelou, M.; Murillo-Rodríguez, E.; Menéndez-Gonzlez, M.; Pöppel, E. Dopaminergic Reward System: A Short Integrative Review. Int. Arch. Med. 2010, 3, 24. [Google Scholar] [CrossRef] [Green Version]
  110. Blaess, S.; Stott, S.R.W.; Ang, S.-L. The Generation of Midbrain Dopaminergic Neurons. In Patterning and Cell Type Specification in the Developing CNS and PNS; Academic Press: Cambridge, MA, USA, 2020; pp. 369–398. [Google Scholar] [CrossRef]
  111. Van Opstal, A.M.; Kaal, I.; van den Berg-Huysmans, A.A.; Hoeksma, M.; Blonk, C.; Pijl, H.; Rombouts, S.A.R.B.; van der Grond, J. Dietary Sugars and Non-Caloric Sweeteners Elicit Different Homeostatic and Hedonic Responses in the Brain. Nutrition 2019, 60, 80–86. [Google Scholar] [CrossRef]
  112. Van Opstal, A.M.; Hafkemeijer, A.; van den Berg-Huysmans, A.A.; Hoeksma, M.; Blonk, C.; Pijl, H.; Rombouts, S.A.R.B.; van der Grond, J. Brain Activity and Connectivity Changes in Response to Glucose Ingestion. Nutr. Neurosci. 2020, 23, 110–117. [Google Scholar] [CrossRef]
  113. Van Opstal, A.M.; Hafkemeijer, A.; van den Berg-Huysmans, A.A.; Hoeksma, M.; Mulder, T.P.J.; Pijl, H.; Rombouts, S.A.R.B.; van der Grond, J. Brain Activity and Connectivity Changes in Response to Nutritive Natural Sugars, Non-Nutritive Natural Sugar Replacements and Artificial Sweeteners. Nutr. Neurosci. 2021, 24, 395–405. [Google Scholar] [CrossRef] [Green Version]
  114. Shestov, A.A.; Emir, U.E.; Kumar, A.; Henry, P.G.; Seaquist, E.R.; Öz, G. Simultaneous Measurement of Glucose Transport and Utilization in the Human Brain. Am. J. Physiol. Endocrinol. Metab. 2011, 301, E1040–E1049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Peters, R.; White, D.J.; Scholey, A. Resting State FMRI Reveals Differential Effects of Glucose Administration on Central Appetite Signalling in Young and Old Adults. J. Psychopharmacol. 2020, 34, 304–314. [Google Scholar] [CrossRef] [PubMed]
  116. Benton, D. The Plausibility of Sugar Addiction and Its Role in Obesity and Eating Disorders. Clin. Nutr. 2010, 29, 288–303. [Google Scholar] [CrossRef] [PubMed]
  117. Ventura, T.; Santander, J.; Torres, R.; Contreras, A.M. Neurobiologic Basis of Craving for Carbohydrates. Nutrition 2014, 30, 252–256. [Google Scholar] [CrossRef] [PubMed]
  118. Goran, M.I.; Plows, J.F.; Ventura, E.E. Effects of Consuming Sugars and Alternative Sweeteners during Pregnancy on Maternal and Child Health: Evidence for a Secondhand Sugar Effect. Proc. Nutr. Soc. 2019, 78, 262–271. [Google Scholar] [CrossRef]
  119. Wiss, D.A.; Avena, N.; Rada, P. Sugar Addiction: From Evolution to Revolution. Front. Psychiatry 2018, 9, 545. [Google Scholar] [CrossRef] [Green Version]
  120. Avena, N.M.; Rada, P.; Hoebel, B.G. Evidence for Sugar Addiction: Behavioral and Neurochemical Effects of Intermittent, Excessive Sugar Intake. Neurosci. Biobehav. Rev. 2008, 32, 20. [Google Scholar] [CrossRef] [Green Version]
  121. Avena, N.M.; Long, K.A.; Hoebel, B.G. Sugar-Dependent Rats Show Enhanced Responding for Sugar after Abstinence: Evidence of a Sugar Deprivation Effect. Physiol. Behav. 2005, 84, 359–362. [Google Scholar] [CrossRef]
  122. Spangler, R.; Wittkowski, K.M.; Goddard, N.L.; Avena, N.M.; Hoebel, B.G.; Leibowitz, S.F. Opiate-like Effects of Sugar on Gene Expression in Reward Areas of the Rat Brain. Brain Res. Mol. Brain Res. 2004, 124, 134–142. [Google Scholar] [CrossRef]
  123. Avena, N.M.; Bocarsly, M.E.; Rada, P.; Kim, A.; Hoebel, B.G. After Daily Bingeing on a Sucrose Solution, Food Deprivation Induces Anxiety and Accumbens Dopamine/Acetylcholine Imbalance. Physiol. Behav. 2008, 94, 309–315. [Google Scholar] [CrossRef] [Green Version]
  124. Avena, N.M.; Rada, P.; Moise, N.; Hoebel, B.G. Sucrose Sham Feeding on a Binge Schedule Releases Accumbens Dopamine Repeatedly and Eliminates the Acetylcholine Satiety Response. Neuroscience 2006, 139, 813–820. [Google Scholar] [CrossRef] [PubMed]
  125. Rada, P.; Avena, N.M.; Hoebel, B.G. Daily Bingeing on Sugar Repeatedly Releases Dopamine in the Accumbens Shell. Neuroscience 2005, 134, 737–744. [Google Scholar] [CrossRef] [PubMed]
  126. Shariff, M.; Quik, M.; Holgate, J.; Morgan, M.; Patkar, O.L.; Tam, V.; Belmer, A.; Bartlett, S.E. Neuronal Nicotinic Acetylcholine Receptor Modulators Reduce Sugar Intake. PLoS ONE 2016, 11, e0150270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Stuber, G.D.; Sparta, D.R.; Stamatakis, A.M.; van Leeuwen, W.A.; Hardjoprajitno, J.E.; Cho, S.; Tye, K.M.; Kempadoo, K.A.; Zhang, F.; Deisseroth, K.; et al. Excitatory Transmission from the Amygdala to Nucleus Accumbens Facilitates Reward Seeking. Nature 2011, 475, 377–382. [Google Scholar] [CrossRef]
  128. Hajnal, A.; Smith, G.P.; Norgren, R. Oral Sucrose Stimulation Increases Accumbens Dopamine in the Rat. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004, 286, R31–R37. [Google Scholar] [CrossRef]
  129. Colantuoni, C.; Schwenker, J.; McCarthy, J.; Rada, P.; Ladenheim, B.; Cadet, J.L.; Schwartz, G.J.; Moran, T.H.; Hoebel, B.G. Excessive Sugar Intake Alters Binding to Dopamine and Mu-Opioid Receptors in the Brain. Neuroreport 2001, 12, 3549–3552. [Google Scholar] [CrossRef]
  130. Gawliński, D.; Gawlińska, K.; Frankowska, M.; Filip, M. Maternal Diet Influences the Reinstatement of Cocaine-Seeking Behavior and the Expression of Melanocortin-4 Receptors in Female Offspring of Rats. Nutrients 2020, 12, 1462. [Google Scholar] [CrossRef]
  131. Gawliński, D.; Gawlińska, K.; Frankowska, M.; Filip, M. Maternal High-Sugar Diet Changes Offspring Vulnerability to Reinstatement of Cocaine-Seeking Behavior: Role of Melanocortin-4 Receptors. FASEB J. 2020, 34, 9192–9206. [Google Scholar] [CrossRef]
  132. Archer, Z.A.; Rayner, D.v.; Duncan, J.S.; Bell, L.M.; Mercer, J.G. Introduction of a High-Energy Diet Acutely up-Regulates Hypothalamic Cocaine and Amphetamine-Regulated Transcript, Mc4R and Brown Adipose Tissue Uncoupling Protein-1 Gene Expression in Male Sprague-Dawley Rats. J. Neuroendocrinol. 2005, 17, 10–17. [Google Scholar] [CrossRef]
  133. Klenowski, P.M.; Shariff, M.R.; Belmer, A.; Fogarty, M.J.; Mu, E.W.H.; Bellingham, M.C.; Bartlett, S.E. Prolonged Consumption of Sucrose in a Binge-Like Manner, Alters the Morphology of Medium Spiny Neurons in the Nucleus Accumbens Shell. Front. Behav. Neurosci. 2016, 10, 54. [Google Scholar] [CrossRef] [Green Version]
  134. Scheggi, S.; Secci, M.E.; Marchese, G.; de Montis, M.G.; Gambarana, C. Influence of Palatability on Motivation to Operate for Caloric and Non-Caloric Food in Non Food-Deprived and Food-Deprived Rats. Neuroscience 2013, 236, 320–331. [Google Scholar] [CrossRef] [PubMed]
  135. Tukey, D.S.; Ferreira, J.M.; Antoine, S.O.; D’Amour, J.A.; Ninan, I.; de Vaca, S.C.; Incontro, S.; Wincott, C.; Horwitz, J.K.; Hartner, D.T.; et al. Sucrose Ingestion Induces Rapid AMPA Receptor Trafficking. J. Neurosci. 2013, 33, 6123–6132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Carelli, R.M.; West, E.A. When a Good Taste Turns Bad: Neural Mechanisms Underlying the Emergence of Negative Affect and Associated Natural Reward Devaluation by Cocaine. Neuropharmacology 2014, 76, 360–369. [Google Scholar] [CrossRef] [Green Version]
  137. Beecher, K.; Wang, J.; Jacques, A.; Chaaya, N.; Chehrehasa, F.; Belmer, A.; Bartlett, S.E. Sucrose Consumption Alters Serotonin/Glutamate Co-Localisation Within the Prefrontal Cortex and Hippocampus of Mice. Front. Mol. Neurosci. 2021, 14, 133. [Google Scholar] [CrossRef]
  138. Chong, C.P.; Shahar, S.; Haron, H.; Din, N.C. Habitual Sugar Intake and Cognitive Impairment among Multi-Ethnic Malaysian Older Adults. Clin. Interv. Aging 2019, 14, 1331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Stollery, B.; Christian, L. Glucose and Memory: The Influence of Drink, Expectancy, and Beliefs. Psychopharmacology 2013, 228, 685–697. [Google Scholar] [CrossRef] [PubMed]
  140. Del-Ponte, B.; Anselmi, L.; Assunção, M.C.F.; Tovo-Rodrigues, L.; Munhoz, T.N.; Matijasevich, A.; Rohde, L.A.; Santos, I.S. Sugar Consumption and Attention-Deficit/Hyperactivity Disorder (ADHD): A Birth Cohort Study. J. Affect. Disord. 2019, 243, 290–296. [Google Scholar] [CrossRef]
  141. Beecher, K.; Alvarez Cooper, I.; Wang, J.; Walters, S.B.; Chehrehasa, F.; Bartlett, S.E.; Belmer, A. Long-Term Overconsumption of Sugar Starting at Adolescence Produces Persistent Hyperactivity and Neurocognitive Deficits in Adulthood. Front. Neurosci. 2021, 15, 670430. [Google Scholar] [CrossRef]
  142. Wong, A.; Dogra, V.R.; Reichelt, A.C. High-Sucrose Diets in Male Rats Disrupt Aspects of Decision Making Tasks, Motivation and Spatial Memory, but Not Impulsivity Measured by Operant Delay-Discounting. Behav. Brain Res. 2017, 327, 144–154. [Google Scholar] [CrossRef]
  143. Colantuoni, C.; Rada, P.; McCarthy, J.; Patten, C.; Avena, N.M.; Chadeayne, A.; Hoebel, B.G. Evidence That Intermittent, Excessive Sugar Intake Causes Endogenous Opioid Dependence. Obes. Res. 2002, 10, 478–488. [Google Scholar] [CrossRef]
  144. Parylak, S.L.; Cottone, P.; Sabino, V.; Rice, K.C.; Zorrilla, E.P. Effects of CB1 and CRF1 Receptor Antagonists on Binge-like Eating in Rats with Limited Access to a Sweet Fat Diet: Lack of Withdrawal-like Responses. Physiol. Behav. 2012, 107, 231–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Harrell, C.S.; Burgado, J.; Kelly, S.D.; Johnson, Z.P.; Neigh, G.N. High-Fructose Diet during Periadolescent Development Increases Depressive-like Behavior and Remodels the Hypothalamic Transcriptome in Male Rats. Psychoneuroendocrinology 2015, 62, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Eudave, D.M.; BeLow, M.N.; Flandreau, E.I. Effects of High Fat or High Sucrose Diet on Behavioral-Response to Social Defeat Stress in Mice. Neurobiol. Stress 2018, 9, 1–8. [Google Scholar] [CrossRef] [PubMed]
  147. Kim, S.; Shou, J.; Abera, S.; Ziff, E.B. Sucrose Withdrawal Induces Depression and Anxiety-like Behavior by Kir2.1 Upregulation in the Nucleus Accumbens. Neuropharmacology 2018, 130, 10–17. [Google Scholar] [CrossRef]
  148. Santos, C.J.; Ferreira, A.V.M.; Oliveira, A.L.; Oliveira, M.C.; Gomes, J.S.; Aguiar, D.C. Carbohydrate-Enriched Diet Predispose to Anxiety and Depression-like Behavior after Stress in Mice. Nutr. Neurosci. 2018, 21, 33–39. [Google Scholar] [CrossRef]
  149. Gueye, A.B.; Vendruscolo, L.F.; de Avila, C.; le Moine, C.; Darnaudéry, M.; Cador, M. Unlimited Sucrose Consumption during Adolescence Generates a Depressive-like Phenotype in Adulthood. Neuropsychopharmacology 2018, 43, 2627–2635. [Google Scholar] [CrossRef] [Green Version]
  150. Choi, C.S.; Kim, P.; Park, J.H.; Gonzales, E.L.T.; Kim, K.C.; Cho, K.S.; Ko, M.J.; Yang, S.M.; Seung, H.; Han, S.H.; et al. High Sucrose Consumption during Pregnancy Induced ADHD-like Behavioral Phenotypes in Mice Offspring. J. Nutr. Biochem. 2015, 26, 1520–1526. [Google Scholar] [CrossRef]
  151. Barker, D.J.P. The Origins of the Developmental Origins Theory. J. Intern. Med. 2007, 261, 412–417. [Google Scholar] [CrossRef]
  152. Wadhwa, P.D.; Buss, C.; Entringer, S.; Swanson, J.M. Developmental Origins of Health and Disease: Brief History of the Approach and Current Focus on Epigenetic Mechanisms. Semin. Reprod. Med. 2009, 27, 358–368. [Google Scholar] [CrossRef] [Green Version]
  153. Heindel, J.J.; Vandenberg, L.N. Developmental Origins of Health and Disease: A Paradigm for Understanding Disease Cause and Prevention. Curr. Opin. Pediatr. 2015, 27, 248–253. [Google Scholar] [CrossRef] [Green Version]
  154. Netting, M.J.; Middleton, P.F.; Makrides, M. Does Maternal Diet during Pregnancy and Lactation Affect Outcomes in Offspring? A Systematic Review of Food-Based Approaches. Nutrition 2014, 30, 1225–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Gluckman, P.D.; Hanson, M.A.; Cooper, C.; Thornburg, K.L. Effect of in Utero and Early-Life Conditions on Adult Health and Disease. N. Engl. J. Med. 2008, 359, 61–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Montalvo-Martínez, L.; Maldonado-Ruiz, R.; Cárdenas-Tueme, M.; Reséndez-Pérez, D.; Camacho, A. Maternal Overnutrition Programs Central Inflammation and Addiction-Like Behavior in Offspring. Biomed Res. Int. 2018, 2018, 8061389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Kominiarek, M.A.; Rajan, P. Nutrition Recommendations in Pregnancy and Lactation. Med. Clin. N. Am. 2016, 100, 1199–1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. ACOG Practice Bulletin No 156: Obesity in Pregnancy. Obstet. Gynecol. 2015, 126, e112–e126. [CrossRef] [PubMed]
  159. Torloni, M.R.; Betran, A.P.; Horta, B.L.; Nakamura, M.U.; Atallah, A.N.; Moron, A.F.; Valente, O. Prepregnancy BMI and the Risk of Gestational Diabetes: A Systematic Review of the Literature with Meta-Analysis. Obes. Rev. 2009, 10, 194–203. [Google Scholar] [CrossRef]
  160. Metzger, B.E.; Lowe, L.P.; Dyer, A.R.; Trimble, E.R.; Sheridan, B.; Hod, M.; Chen, R.; Yogev, Y.; Coustan, D.R.; Catalano, P.M.; et al. Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study: Associations with Neonatal Anthropometrics. Diabetes 2009, 58, 453–459. [Google Scholar] [CrossRef] [Green Version]
  161. Stuebe, A.M.; Rich-Edwards, J.W. The Reset Hypothesis: Lactation and Maternal Metabolism. Am. J. Perinatol. 2009, 26, 81–88. [Google Scholar] [CrossRef] [Green Version]
  162. Kjos, S.L.; Henry, O.A.; Montoro, M.; Buchanan, T.A.; Mestman, J.H. Insulin-Requiring Diabetes in Pregnancy: A Randomized Trial of Active Induction of Labor and Expectant Management. Am. J. Obstet. Gynecol. 1993, 169, 611–615. [Google Scholar] [CrossRef]
  163. Diniz, J.M.M.; da Costa, T.H.M. Independent of Body Adiposity, Breast-Feeding Has a Protective Effect on Glucose Metabolism in Young Adult Women. Br. J. Nutr. 2004, 92, 905–912. [Google Scholar] [CrossRef] [Green Version]
  164. Blackmore, H.L.; Ozanne, S.E. Maternal Diet-Induced Obesity and Offspring Cardiovascular Health. J. Dev. Orig. Health Dis. 2013, 4, 338–347. [Google Scholar] [CrossRef] [PubMed]
  165. Williams, C.B.; MacKenzie, K.C.; Gahagan, S. The Effect of Maternal Obesity on the Offspring. Clin. Obstet. Gynecol. 2014, 57, 508. [Google Scholar] [CrossRef]
  166. Francis, E.C.; Dabelea, D.; Shankar, K.; Perng, W. Maternal Diet Quality during Pregnancy Is Associated with Biomarkers of Metabolic Risk among Male Offspring. Diabetologia 2021, 64, 2478–2490. [Google Scholar] [CrossRef] [PubMed]
  167. Giordano, F.; Carbone, P.; Nori, F.; Mantovani, A.; Taruscio, D.; Figà-Talamanca, I. Maternal Diet and the Risk of Hypospadias and Cryptorchidism in the Offspring. Paediatr. Perinat. Epidemiol. 2008, 22, 249–260. [Google Scholar] [CrossRef] [PubMed]
  168. Radzicka-Mularczyk, S.A.; Pietryga, M.; Brazert, J. How Mother’s Obesity May Affect the Pregnancy and Offspring. Ginekol. Polska 2020, 91, 769–772. [Google Scholar] [CrossRef] [PubMed]
  169. Godfrey, K.M.; Reynolds, R.M.; Prescott, S.L.; Nyirenda, M.; Jaddoe, V.W.V.; Eriksson, J.G.; Broekman, B.F.P. Influence of Maternal Obesity on the Long-Term Health of Offspring. Lancet Diabetes Endocrinol. 2017, 5, 53–64. [Google Scholar] [CrossRef] [Green Version]
  170. Sharma, N.; Okere, I.C.; Barrows, B.R.; Lei, B.; Duda, M.K.; Yuan, C.L.; Previs, S.F.; Sharov, V.G.; Azimzadeh, A.M.; Ernsberger, P.; et al. High-Sugar Diets Increase Cardiac Dysfunction and Mortality in Hypertension Compared to Low-Carbohydrate or High-Starch Diets. J. Hypertens. 2008, 26, 1402–1410. [Google Scholar] [CrossRef] [Green Version]
  171. Panchal, S.K.; Poudyal, H.; Iyer, A.; Nazer, R.; Alam, A.; Diwan, V.; Kauter, K.; Sernia, C.; Campbell, F.; Ward, L.; et al. High-Carbohydrate High-Fat Diet–Induced Metabolic Syndrome and Cardiovascular Remodeling in Rats. J. Cardiovasc. Pharmacol. 2011, 57, 51–64. [Google Scholar] [CrossRef]
  172. Toop, C.R.; Muhlhausler, B.S.; O’Dea, K.; Gentili, S. Impact of Perinatal Exposure to Sucrose or High Fructose Corn Syrup (HFCS-55) on Adiposity and Hepatic Lipid Composition in Rat Offspring. J. Physiol. 2017, 595, 4379–4398. [Google Scholar] [CrossRef]
  173. Tobar-Bernal, F.A.; Zamudio, S.R.; Quevedo-Corona, L. The High-Fructose Intake of Dams during Pregnancy and Lactation Exerts Sex-Specific Effects on Adult Rat Offspring Metabolism. J. Dev. Orig. Health Dis. 2021, 12, 411–419. [Google Scholar] [CrossRef]
  174. Ornellas, F.; Carapeto, P.V.; Aguila, M.B.; Mandarim-de-Lacerda, C.A. Sex-Linked Changes and High Cardiovascular Risk Markers in the Mature Progeny of Father, Mother, or Both Father and Mother Consuming a High-Fructose Diet. Nutrition 2020, 71, 110612. [Google Scholar] [CrossRef] [PubMed]
  175. Trujillo-Villarreal, L.A.; Romero-Díaz, V.J.; Marino-Martínez, I.A.; Fuentes-Mera, L.; Ponce-Camacho, M.A.; Devenyi, G.A.; Mallar Chakravarty, M.; Camacho-Morales, A.; Garza-Villarreal, E.E. Maternal Cafeteria Diet Exposure Primes Depression-like Behavior in the Offspring Evoking Lower Brain Volume Related to Changes in Synaptic Terminals and Gliosis. Transl. Psychiatry 2021, 11, 53. [Google Scholar] [CrossRef] [PubMed]
  176. Ribaroff, G.A.; Wastnedge, E.; Drake, A.J.; Sharpe, R.M.; Chambers, T.J.G. Animal Models of Maternal High Fat Diet Exposure and Effects on Metabolism in Offspring: A Meta-Regression Analysis. Obes. Rev. 2017, 18, 673–686. [Google Scholar] [CrossRef]
  177. Wang, D.; Zhang, H.; Zeng, M.; Tang, X.; Zhu, X.; Guo, Y.; Qi, L.; Xie, Y.; Zhang, M.; Chen, D. Maternal High Sugar and Fat Diet Benefits Offspring Brain Function via Targeting on the Gut-Brain Axis. Aging 2021, 13, 10240–10274. [Google Scholar] [CrossRef] [PubMed]
  178. Andreas, E.; Reid, M.; Zhang, W.; Moley, K.H. The Effect of Maternal High-Fat/High-Sugar Diet on Offspring Oocytes and Early Embryo Development. Mol. Hum. Reprod. 2019, 25, 717–728. [Google Scholar] [CrossRef]
  179. Gawlińska, K.; Gawliński, D.; Filip, M.; Przegaliński, E. Relationship of Maternal High-Fat Diet during Pregnancy and Lactation to Offspring Health. Nutr. Rev. 2021, 79, 709–725. [Google Scholar] [CrossRef] [PubMed]
  180. Menting, M.D.; Mintjens, S.; van de Beek, C.; Frick, C.J.; Ozanne, S.E.; Limpens, J.; Roseboom, T.J.; Hooijmans, C.R.; van Deutekom, A.W.; Painter, R.C. Maternal Obesity in Pregnancy Impacts Offspring Cardiometabolic Health: Systematic Review and Meta-Analysis of Animal Studies. Obes. Rev. 2019, 20, 675–685. [Google Scholar] [CrossRef] [Green Version]
  181. Menting, M.D.; van de Beek, C.; Mintjens, S.; Wever, K.E.; Korosi, A.; Ozanne, S.E.; Limpens, J.; Roseboom, T.J.; Hooijmans, C.; Painter, R.C. The Link between Maternal Obesity and Offspring Neurobehavior: A Systematic Review of Animal Experiments. Neurosci. Biobehav. Rev. 2019, 98, 107–121. [Google Scholar] [CrossRef]
  182. Mizera, J.; Kazek, G.; Niedzielska-Andres, E.; Pomierny-Chamiolo, L. Maternal High-Sugar Diet Results in NMDA Receptors Abnormalities and Cognitive Impairment in Rat Offspring. FASEB J. 2021, 35, e21547. [Google Scholar] [CrossRef]
  183. Bayol, S.A.; Farrington, S.J.; Stickland, N.C. A Maternal “junk Food” Diet in Pregnancy and Lactation Promotes an Exacerbated Taste for “Junk Food” and a Greater Propensity for Obesity in Rat Offspring. Br. J. Nutr. 2007, 98, 843–851. [Google Scholar] [CrossRef] [Green Version]
  184. Volkow, N.D.; Poznyak, V.; Saxena, S.; Gerra, G. Drug Use Disorders: Impact of a Public Health Rather than a Criminal Justice Approach. World Psychiatry 2017, 16, 213. [Google Scholar] [CrossRef] [PubMed]
  185. The United Nations Office on Drugs and Crime. World Drug Report 2021; United Nations Publication: Vienna, Austria, 2021; Sales No. E.21.XI.8; ISBN 9789211483611. [Google Scholar]
  186. Castilla-Ortega, E.; Serrano, A.; Blanco, E.; Araos, P.; Suárez, J.; Pavón, F.J.; de Fonseca, F.R.; Santín, L.J. A Place for the Hippocampus in the Cocaine Addiction Circuit: Potential Roles for Adult Hippocampal Neurogenesis. Neurosci. Biobehav. Rev. 2016, 66, 15–32. [Google Scholar] [CrossRef] [PubMed]
  187. Venton, B.J.; Seipel, A.T.; Phillips, P.E.M.; Wetsel, W.C.; Gitler, D.; Greengard, P.; Augustine, G.J.; Wightman, R.M. Cocaine Increases Dopamine Release by Mobilization of a Synapsin-Dependent Reserve Pool. J. Neurosci. 2006, 26, 3206–3209. [Google Scholar] [CrossRef]
  188. Egilmez, Y.; Jung, M.E.; Lane, J.D.; Emmett-Oglesby, M.W. Dopamine Release during Cocaine Self-Administration in Rats: Effect of SCH23390. Brain Res. 1995, 701, 142–150. [Google Scholar] [CrossRef]
  189. Ahmed, S.H.; Guillem, K.; Vandaele, Y. Sugar Addiction: Pushing the Drug-Sugar Analogy to the Limit. Curr. Opin. Clin. Nutr. Metab. Care 2013, 16, 434–439. [Google Scholar] [CrossRef]
  190. Onaolapo, A.Y.; Onaolapo, O.J.; Olowe, O.A. An Overview of Addiction to Sugar. In Dietary Sugar, Salt and Fat in Human Health; Academic Press: Cambridge, MA, USA, 2020; pp. 195–216. [Google Scholar] [CrossRef]
  191. Hartmann, H.; Pauli, L.K.; Janssen, L.K.; Huhn, S.; Ceglarek, U.; Horstmann, A. Preliminary Evidence for an Association between Intake of High-Fat High-Sugar Diet, Variations in Peripheral Dopamine Precursor Availability and Dopamine-Dependent Cognition in Humans. J. Neuroendocrinol. 2020, 32, e12917. [Google Scholar] [CrossRef]
  192. Lenoir, M.; Serre, F.; Cantin, L.; Ahmed, S.H. Intense Sweetness Surpasses Cocaine Reward. PLoS ONE 2007, 2, e698. [Google Scholar] [CrossRef] [Green Version]
  193. Lopes, F.M.; Boos, F.Z.; Zanini, A.M.; Czermainski, F.R. Neuropsychological and Behavioral Aspects of Drug Use and Abuse: Theory, Research, and Intervention. Drugs Hum. Behav. 2021, 1, 151–170. [Google Scholar] [CrossRef]
  194. DiNicolantonio, J.J.; O’Keefe, J.H.; Wilson, W.L. Sugar Addiction: Is It Real? A Narrative Review. Br. J. Sports Med. 2018, 52, 910–913. [Google Scholar] [CrossRef]
  195. Rorabaugh, J.M.; Stratford, J.M.; Zahniser, N.R. A Relationship between Reduced Nucleus Accumbens Shell and Enhanced Lateral Hypothalamic Orexin Neuronal Activation in Long-Term Fructose Bingeing Behavior. PLoS ONE 2014, 9, e95019. [Google Scholar] [CrossRef]
  196. Peleg-Raibstein, D.; Sarker, G.; Litwan, K.; Krämer, S.D.; Ametamey, S.M.; Schibli, R.; Wolfrum, C. Enhanced Sensitivity to Drugs of Abuse and Palatable Foods Following Maternal Overnutrition. Transl. Psychiatry 2016, 6, e911. [Google Scholar] [CrossRef] [PubMed]
  197. Duart-Castells, L.; Cantacorps, L.; López-Arnau, R.; Montagud-Romero, S.; Puster, B.; Mera, P.; Serra, D.; Camarasa, J.; Pubill, D.; Valverde, O.; et al. Effects of High-Fat Diet and Maternal Binge-Like Alcohol Consumption and Their Influence on Cocaine Response in Female Mice Offspring. Int. J. Neuropsychopharmacol. 2021, 24, 77. [Google Scholar] [CrossRef] [PubMed]
  198. Wijenayake, S.; Rahman, M.F.; Lum, C.M.W.; de Vega, W.C.; Sasaki, A.; McGowan, P.O. Maternal High-Fat Diet Induces Sex-Specific Changes to Glucocorticoid and Inflammatory Signaling in Response to Corticosterone and Lipopolysaccharide Challenge in Adult Rat Offspring. J. Neuroinflamm. 2020, 17, 116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Bocarsly, M.E.; Barson, J.R.; Hauca, J.M.; Hoebel, B.G.; Leibowitz, S.F.; Avena, N.M. Effects of Perinatal Exposure to Palatable Diets on Body Weight and Sensitivity to Drugs of Abuse in Rats. Physiol. Behav. 2012, 107, 568–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Collins, G.T.; Chen, Y.; Tschumi, C.; Rush, E.L.; Mensah, A.; Koek, W.; France, C.P. Effects of Consuming a Diet High in Fat and/or Sugar on the Locomotor Effects of Acute and Repeated Cocaine in Male and Female C57BL/6J Mice. Exp. Clin. Psychopharmacol. 2015, 23, 228–237. [Google Scholar] [CrossRef]
  201. Avena, N.M.; Hoebel, B.G. Amphetamine-Sensitized Rats Show Sugar-Induced Hyperactivity (Cross-Sensitization) and Sugar Hyperphagia. Pharmacol. Biochem. Behav. 2003, 74, 635–639. [Google Scholar] [CrossRef]
  202. Avena, N.M.; Hoebel, B.G. A Diet Promoting Sugar Dependency Causes Behavioral Cross-Sensitization to a Low Dose of Amphetamine. Neuroscience 2003, 122, 17–20. [Google Scholar] [CrossRef]
  203. Moore, C.F.; Leonard, M.Z.; Micovic, N.M.; Miczek, K.A.; Sabino, V.; Cottone, P. Reward Sensitivity Deficits in a Rat Model of Compulsive Eating Behavior. Neuropsychopharmacology 2019, 45, 589–596. [Google Scholar] [CrossRef]
  204. Rorabaugh, J.M.; Stratford, J.M.; Zahniser, N.R. Differences in Bingeing Behavior and Cocaine Reward Following Intermittent Access to Sucrose, Glucose or Fructose Solutions. Neuroscience 2015, 301, 213–220. [Google Scholar] [CrossRef] [Green Version]
  205. Vitale, M.A.; Chen, D.; Kanarek, R.B. Chronic Access to a Sucrose Solution Enhances the Development of Conditioned Place Preferences for Fentanyl and Amphetamine in Male Long-Evans Rats. Pharmacol. Biochem. Behav. 2003, 74, 529–539. [Google Scholar] [CrossRef]
  206. Bruggeman, E.C.; Li, C.; Ross, A.P.; Doherty, J.M.; Williams, B.F.; Frantz, K.J.; Parent, M.B. A High Fructose Diet Does Not Affect Amphetamine Self-Administration or Spatial Water Maze Learning and Memory in Female Rats. Pharmacol. Biochem. Behav. 2011, 99, 356–364. [Google Scholar] [CrossRef] [PubMed]
  207. Orsini, C.A.; Ginton, G.; Shimp, K.G.; Avena, N.M.; Gold, M.S.; Setlow, B. Food Consumption and Weight Gain after Cessation of Chronic Amphetamine Administration. Appetite 2014, 78, 76–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Foltin, R.W. Consumption of Palatable Food Decreases the Anorectic Effects of Serotonergic, but Not Dopaminergic Drugs in Baboons. Physiol. Behav. 2011, 103, 493–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Loebens, M.; Barros, H.M.T. Diet Influences Cocaine Withdrawal Behaviors in the Forced Swimming Test. Pharmacol. Biochem. Behav. 2003, 74, 259–267. [Google Scholar] [CrossRef]
  210. Ströhle, A.; Gensichen, J.; Domschke, K. The Diagnosis and Treatment of Anxiety Disorders. Deutsches Ärzteblatt Int. 2018, 115, 611. [Google Scholar] [CrossRef] [PubMed]
  211. Bandelow, B.; Baldwin, D.; Abelli, M.; Bolea-Alamanac, B.; Bourin, M.; Chamberlain, S.R.; Cinosi, E.; Davies, S.; Domschke, K.; Fineberg, N.; et al. Biological Markers for Anxiety Disorders, OCD and PTSD: A Consensus Statement. Part II: Neurochemistry, Neurophysiology and Neurocognition. World J. Biol. Psychiatry 2017, 18, 162–214. [Google Scholar] [CrossRef] [PubMed]
  212. Smith, J.P.; Book, S.W. Anxiety and Substance Use Disorders: A Review. Psychiatr. Times 2008, 25, 19. [Google Scholar] [PubMed]
  213. Stathopoulou, G.; Gold, A.K.; Hoyt, D.L.; Milligan, M.; Hearon, B.A.; Otto, M.W. Does Anxiety Sensitivity Predict Addiction Severity in Opioid Use Disorder? Addict. Behav. 2021, 112, 106644. [Google Scholar] [CrossRef]
  214. Horigian, V.E.; Schmidt, R.D.; Feaster, D.J. Loneliness, Mental Health, and Substance Use among US Young Adults during COVID-19. J. Psychoact. Drugs 2021, 53, 1–9. [Google Scholar] [CrossRef]
  215. Vorspan, F.; Mehtelli, W.; Dupuy, G.; Bloch, V.; Lépine, J.P. Anxiety and Substance Use Disorders: Co-Occurrence and Clinical Issues. Curr. Psychiatry Rep. 2015, 17, 1–7. [Google Scholar] [CrossRef]
  216. Goodwin, R.D.; Stein, D.J. Anxiety Disorders and Drug Dependence: Evidence on Sequence and Specificity among Adults. Psychiatry Clin. Neurosci. 2013, 67, 167–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Zvolensky, M.J.; Schmidt, N.B. Anxiety and Substance Use Disorders: Introduction to the Special Series. J. Anxiety Disord. 2004, 18, 1–6. [Google Scholar] [CrossRef]
  218. Smith, J.P.; Randall, C.L. Anxiety and Alcohol Use Disorders: Comorbidity and Treatment Considerations. Alcohol Res. Curr. Rev. 2012, 34, 414. [Google Scholar]
  219. Johannessen, E.L.; Andersson, H.W.; Bjørngaard, J.H.; Pape, K. Anxiety and Depression Symptoms and Alcohol Use among Adolescents—A Cross Sectional Study of Norwegian Secondary School Students. BMC Public Health 2017, 17, 494. [Google Scholar] [CrossRef] [PubMed]
  220. Grasing, M.J.; Xu, H.; Idowu, J.Y.; Grasing, K. Changes Depression- and Anxiety- like Behaviors Following Selective Breeding for Cocaine Reinforcement. Psychiatry Res. 2021, 295, 113637. [Google Scholar] [CrossRef] [PubMed]
  221. Markou, A.; Kosten, T.R.; Koob, G.F. Neurobiological Similarities in Depression and Drug Dependence: A Self-Medication Hypothesis. Neuropsychopharmacology 1998, 18, 135–174. [Google Scholar] [CrossRef] [Green Version]
  222. DiGirolamo, G.J.; Gonzalez, G.; Smelson, D.; Guevremont, N.; Andre, M.I.; Patnaik, P.O.; Zaniewski, Z.R. Increased Depression and Anxiety Symptoms Are Associated with More Breakdowns in Cognitive Control to Cocaine Cues in Veterans with Cocaine Use Disorder. J. Dual Diagn. 2017, 13, 298–304. [Google Scholar] [CrossRef] [Green Version]
  223. Ayran, G.; Köse, S.; Küçükoğlu, S.; Özdemir, A.A. The Effect of Anxiety on Nicotine Dependence among University Students during the COVID-19 Pandemic. Perspect. Psychiatr. Care 2022, 58, 114–123. [Google Scholar] [CrossRef]
  224. Benowitz, N.L. Nicotine Addiction. N. Engl. J. Med. 2010, 362, 2295. [Google Scholar] [CrossRef]
  225. Kobiella, A.; Ulshöfer, D.E.; Vollmert, C.; Bühler, M.; Esslinger, C.; Smolka, M.N. Nicotine Increases Neural Response to Unpleasant Stimuli and Anxiety in Non-Smokers. Addict. Biol. 2011, 16, 285–295. [Google Scholar] [CrossRef]
  226. Conner, K.R.; Pinquart, M.; Holbrook, A.P. Meta-Analysis of Depression and Substance Use and Impairment among Cocaine Users. Drug Alcohol Depend. 2008, 98, 13–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Poling, J.; Kosten, T.R.; Sofuoglu, M. Treatment Outcome Predictors for Cocaine Dependence. Am. J. Drug Alcohol Abuse 2007, 33, 191–206. [Google Scholar] [CrossRef] [PubMed]
  228. Rounsaville, B.J. Treatment of Cocaine Dependence and Depression. Biol. Psychiatry 2004, 56, 803–809. [Google Scholar] [CrossRef] [PubMed]
  229. Leventhal, A.M.; Mooney, M.E.; DeLaune, K.A.; Schmitz, J.M. Using Addiction Severity Profiles to Differentiate Cocaine-Dependent Patients with and without Comorbid Major Depression. Am. J. Addict. Am. Acad. Psychiatr. Alcohol. Addict. 2006, 15, 362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Ford, J.D.; Gelernter, J.; DeVoe, J.S.; Zhang, W.; Weiss, R.D.; Brady, K.; Farrer, L.; Kranzler, H.R. Association of Psychiatric and Substance Use Disorder Comorbidity with Cocaine Dependence Severity and Treatment Utilization in Cocaine-Dependent Individuals. Drug Alcohol Depend. 2009, 99, 193–203. [Google Scholar] [CrossRef] [Green Version]
  231. Leventhal, A.M.; Brightman, M.; Ameringer, K.J.; Greenberg, J.; Mickens, L.; Ray, L.A.; Sun, P.; Sussman, S. Anhedonia Associated with Stimulant Use and Dependence in a Population-Based Sample of American Adults. Exp. Clin. Psychopharmacol. 2010, 18, 562–569. [Google Scholar] [CrossRef] [Green Version]
  232. Wardle, M.C.; Vincent, J.N.; Suchting, R.; Green, C.E.; Lane, S.D.; Schmitz, J.M. Anhedonia Is Associated with Poorer Outcomes in Contingency Management for Cocaine Use Disorder. J. Subst. Abuse Treat. 2017, 72, 32–39. [Google Scholar] [CrossRef] [Green Version]
  233. Lee, R.S.C.; Hoppenbrouwers, S.; Franken, I. A Systematic Meta-Review of Impulsivity and Compulsivity in Addictive Behaviors. Neuropsychol. Rev. 2019, 29, 14–26. [Google Scholar] [CrossRef]
  234. Dawe, S.; Loxton, N.J. The Role of Impulsivity in the Development of Substance Use and Eating Disorders. Neurosci. Biobehav. Rev. 2004, 28, 343–351. [Google Scholar] [CrossRef]
  235. Whiteside, S.P.; Lynam, D.R. The Five Factor Model and Impulsivity: Using a Structural Model of Personality to Understand Impulsivity. Personal. Individ. Differ. 2001, 30, 669–689. [Google Scholar] [CrossRef]
  236. Homberg, J.R.; Karel, P.; Verheij, M.M.M. Individual Differences in Cocaine Addiction: Maladaptive Behavioural Traits. Addict. Biol. 2014, 19, 517–528. [Google Scholar] [CrossRef] [PubMed]
  237. Davis, C.; Cohen, A.; Davids, M.; Rabindranath, A. Attention-Deficit/Hyperactivity Disorder in Relation to Addictive Behaviors: A Moderated-Mediation Analysis of Personality-Risk Factors and Sex. Front. Psychiatry 2015, 6, 47. [Google Scholar] [CrossRef] [PubMed]
  238. Dalley, J.W.; Robbins, T.W. Fractionating Impulsivity: Neuropsychiatric Implications. Nat. Rev. Neurosci. 2017, 18, 158–171. [Google Scholar] [CrossRef] [PubMed]
  239. Stamates, A.L.; Linden-Carmichael, A.N.; Preonas, P.D.; Lau-Barraco, C. Testing Daily Associations between Impulsivity, Affect, and Alcohol Outcomes: A Pilot Study. Addict. Res. Theory 2019, 27, 242–248. [Google Scholar] [CrossRef] [PubMed]
  240. Lees, B.; Mewton, L.; Stapinski, L.A.; Squeglia, L.M.; Rae, C.D.; Teesson, M. Neurobiological and Cognitive Profile of Young Binge Drinkers: A Systematic Review and Meta-Analysis. Neuropsychol. Rev. 2019, 29, 357–385. [Google Scholar] [CrossRef]
  241. Bozkurt, M.; Evren, C.; Umut, G.; Evren, B. Relationship of Attention-Deficit/Hyperactivity Disorder Symptom Severity with Severity of Alcohol-Related Problems in a Sample of Inpatients with Alcohol Use Disorder. Neuropsychiatr. Dis. Treat. 2016, 12, 1661–1667. [Google Scholar] [CrossRef] [Green Version]
  242. Andersson, H.W.; Lauvsnes, A.D.F.; Nordfjærn, T. Emerging Adults in Inpatient Substance Use Treatment: A Prospective Cohort Study of Patient Characteristics and Treatment Outcomes. Eur. Addict. Res. 2021, 27, 206–215. [Google Scholar] [CrossRef]
  243. Franques, P.; Auriacombe, M.; Piquemal, E.; Verger, M.; Brisseau-Gimenez, S.; Grabot, D.; Tignol, J. Sensation Seeking as a Common Factor in Opioid Dependent Subjects and High Risk Sport Practicing Subjects. A Cross Sectional Study. Drug Alcohol Depend. 2003, 69, 121–126. [Google Scholar] [CrossRef]
  244. Kreek, M.J.; Nielsen, D.A.; Butelman, E.R.; LaForge, K.S. Genetic Influences on Impulsivity, Risk Taking, Stress Responsivity and Vulnerability to Drug Abuse and Addiction. Nat. Neurosci. 2005, 8, 1450–1457. [Google Scholar] [CrossRef]
  245. Zuckerman, M.; Eysenck, S.B.; Eysenck, H.J. Sensation Seeking in England and America: Cross-Cultural, Age, and Sex Comparisons. J. Consult. Clin. Psychol. 1978, 46, 139–149. [Google Scholar] [CrossRef]
  246. Zuckerman, M.; Ball, S.; Black, J. Influences of Sensation Seeking, Gender, Risk Appraisal, and Situational Motivation on Smoking. Addict. Behav. 1990, 15, 209–220. [Google Scholar] [CrossRef]
  247. Pelloux, Y.; Costentin, J.; Duterte-Boucher, D. Differential Effects of Novelty Exposure on Place Preference Conditioning to Amphetamine and Its Oral Consumption. Psychopharmacology 2004, 171, 277–285. [Google Scholar] [CrossRef] [PubMed]
  248. Wingo, T.; Nesil, T.; Choi, J.S.; Li, M.D. Novelty Seeking and Drug Addiction in Humans and Animals: From Behavior to Molecules. J. Neuroimmune Pharmacol. 2015, 11, 456–470. [Google Scholar] [CrossRef] [Green Version]
  249. Belin, D.; Berson, N.; Balado, E.; Piazza, P.V.; Deroche-Gamonet, V. High-Novelty-Preference Rats Are Predisposed to Compulsive Cocaine Self-Administration. Neuropsychopharmacology 2011, 36, 569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Yetnikoff, L.; Arvanitogiannis, A. Differential Sensitivity to the Acute and Sensitizing Behavioral Effects of Methylphenidate as a Function of Strain in Adolescent and Young Adult Rats. Behav. Brain Funct. 2013, 9, 38. [Google Scholar] [CrossRef] [Green Version]
  251. Zakharova, E.; Leoni, G.; Kichko, I.; Izenwasser, S. Differential Effects of Methamphetamine and Cocaine on Conditioned Place Preference and Locomotor Activity in Adult and Adolescent Male Rats. Behav. Brain Res. 2009, 198, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Mofatteh, M. Risk Factors Associated with Stress, Anxiety, and Depression among University Undergraduate Students. AIMS Public Health 2021, 8, 36. [Google Scholar] [CrossRef]
  253. de Souza, L.; Barros, W.d.M.; de Souza, R.M.; Delanogare, E.; Machado, A.E.; Braga, S.P.; Rosa, G.K.; Nardi, G.M.; Rafacho, A.; Speretta, G.F.F.; et al. Impact of Different Fructose Concentrations on Metabolic and Behavioral Parameters of Male and Female Mice. Physiol. Behav. 2021, 228, 113187. [Google Scholar] [CrossRef]
  254. Carneiro-Nascimento, S.; Opacka-Juffry, J.; Costabile, A.; Boyle, C.N.; Herde, A.M.; Ametamey, S.M.; Sigrist, H.; Pryce, C.R.; Patterson, M. Chronic Social Stress in Mice Alters Energy Status Including Higher Glucose Need but Lower Brain Utilization. Psychoneuroendocrinology 2020, 119, 104747. [Google Scholar] [CrossRef]
  255. Iemolo, A.; Valenza, M.; Tozier, L.; Knapp, C.M.; Kornetsky, C.; Steardo, L.; Sabino, V.; Cottone, P. Withdrawal from Chronic, Intermittent Access to a Highly Palatable Food Induces Depressive-like Behavior in Compulsive Eating Rats. Behav. Pharmacol. 2012, 23, 593–602. [Google Scholar] [CrossRef] [Green Version]
  256. Aucoin, M.; Lachance, L.; Naidoo, U.; Remy, D.; Shekdar, T.; Sayar, N.; Cardozo, V.; Rawana, T.; Chan, I.; Cooley, K. Diet and Anxiety: A Scoping Review. Nutrients 2021, 13, 4418. [Google Scholar] [CrossRef] [PubMed]
  257. Kanter, J.W.; Rusch, L.C.; Brondino, M.J. Depression Self-Stigma: A New Measure and Preliminary Findings. J. Nerv. Ment. Dis. 2008, 196, 663–670. [Google Scholar] [CrossRef] [PubMed]
  258. McCarron, R.M.; Vanderlip, E.R.; Rado, J. Depression. Ann. Intern. Med. 2016, 165, ITC49–ITC63. [Google Scholar] [CrossRef]
  259. Kloster, A.; Hyer, M.M.; Dyer, S.; Salome-Sanchez, C.; Neigh, G.N. High Fructose Diet Induces Sex-Specific Modifications in Synaptic Respiration and Affective-like Behaviors in Rats. Neuroscience 2021, 454, 40–50. [Google Scholar] [CrossRef] [PubMed]
  260. Reis, D.J.; Ilardi, S.S.; Namekata, M.S.; Wing, E.K.; Fowler, C.H. The Depressogenic Potential of Added Dietary Sugars. Med. Hypotheses 2020, 134, 109421. [Google Scholar] [CrossRef]
  261. Xu, Y.; Zeng, L.; Zou, K.; Shan, S.; Wang, X.; Xiong, J.; Zhao, L.; Zhang, L.; Cheng, G. Role of Dietary Factors in the Prevention and Treatment for Depression: An Umbrella Review of Meta-Analyses of Prospective Studies. Transl. Psychiatry 2021, 11, 478. [Google Scholar] [CrossRef]
  262. Huang, Q.; Liu, H.; Suzuki, K.; Ma, S.; Liu, C. Linking What We Eat to Our Mood: A Review of Diet, Dietary Antioxidants, and Depression. Antioxidants 2019, 8, 376. [Google Scholar] [CrossRef] [Green Version]
  263. Pérez-Ara, M.Á.; Gili, M.; Visser, M.; Penninx, B.W.J.H.; Brouwer, I.A.; Watkins, E.; Owens, M.; García-Toro, M.; Hegerl, U.; Kohls, E.; et al. Associations of Non-Alcoholic Beverages with Major Depressive Disorder History and Depressive Symptoms Clusters in a Sample of Overweight Adults. Nutrients 2020, 12, 3202. [Google Scholar] [CrossRef]
  264. Grant, J.E.; Valle, S.; Chamberlain, S.R. Nutrition in Skin Picking Disorder and Trichotillomania. Front. Psychiatry 2021, 12, 761321. [Google Scholar] [CrossRef]
  265. Steele, C.C.; Steele, T.J.; Gwinner, M.; Rosenkranz, S.K.; Kirkpatrick, K. The Relationship between Dietary Fat Intake, Impulsive Choice, and Metabolic Health. Appetite 2021, 165, 105292. [Google Scholar] [CrossRef]
  266. Steele, C.C.; Pirkle, J.R.A.; Davis, I.R.; Kirkpatrick, K. Dietary Effects on the Determinants of Food Choice: Impulsive Choice, Discrimination, Incentive Motivation, Preference, and Liking in Male Rats. Appetite 2019, 136, 160–172. [Google Scholar] [CrossRef] [PubMed]
Table 1. Classification of dietary carbohydrates. Saccharides are classified according to degree of polymerization (DP) by indication of the length of the carbohydrate chain. The sugar term includes the sum of all the mono- and di-saccharides (DP1, and DP2). Higher DPs classified as carbohydrates, such as oligo- and polysaccharides. Functional and structure divisions include subgroups with major dietary components and main natural sources.
Table 1. Classification of dietary carbohydrates. Saccharides are classified according to degree of polymerization (DP) by indication of the length of the carbohydrate chain. The sugar term includes the sum of all the mono- and di-saccharides (DP1, and DP2). Higher DPs classified as carbohydrates, such as oligo- and polysaccharides. Functional and structure divisions include subgroups with major dietary components and main natural sources.
Types (DP)SubgroupMajor ComponentsNatural Sources
Sugars
(DP1, and DP2)
monosaccharidesglucose, galactose, fructosefruit, vegetables, honey, seeds
disaccharidessucrose, maltose, lactosetable sugar (sugar cane or sugar beet), grains, dairy products
Oligosaccharides
(DP3–9)
maltooligosaccharidesmaltodextrinscorn starch
Polysaccharides
(DP ≥ 10)
starchamylose, amylopectincereals, vegetables, processed flour products
non-starchcellulose, hemicellulose, pectins, glycogenvegetables, fruits, seeds, meat
Table 2. Major sodium–glucose linked transporters (SGLTs) and facilitated diffusion glucose transporters (GLUTs) in several mammals’ organs expression. Transfer for glucose and galactose include possibly the same transporters. Fructose requires other transport proteins (pointed by *).
Table 2. Major sodium–glucose linked transporters (SGLTs) and facilitated diffusion glucose transporters (GLUTs) in several mammals’ organs expression. Transfer for glucose and galactose include possibly the same transporters. Fructose requires other transport proteins (pointed by *).
LocalizationSGLTsGLUTs
IntestineSGLT1, SGLT3, SGLT4 *, SGLT6GLUT2 *, GLUT5 *, GLUT7 *, GLUT12
KidneySGLT1, SGLT2 *, SGLT3, SGLT4 *, SGLT5 *, SGLT6GLUT1, GLUT2 *, GLUT3, GLUT5 *, GLUT9, GLUT10, GLUT11 *, GLUT13
LiverSGLT2 *, SGLT3, SGLT4 *GLUT1, GLUT2 *, GLUT3, GLUT5 *, GLUT7, GLUT8 *, GLUT9
BrainSGLT1, SGLT2, SGLT3, SGLT4 *, SGLT6GLUT1, GLUT2 *, GLUT3, GLUT4, GLUT5 *, GLUT6, GLUT8 *, GLUT13
Table 3. Preclinical studies on the prenatal HSD consumption on behavioural and molecular or neurochemical consequences after drugs of abuse in offspring.
Table 3. Preclinical studies on the prenatal HSD consumption on behavioural and molecular or neurochemical consequences after drugs of abuse in offspring.
Prenatal ExposureAnimalsDrug of AbuseConsequences Enhanced by HSDRef
HSD ModelPeriodBehaviouralMolecular/Neurochemical
70% carbohydrates (44% sucrose)Pregnancy and lactationFemale WR offspringCocaine hydrochloride↑ CUE- and cocaine reinstatement relapse in cocaine SA
↑ cocaine-seeking behaviour
↑ MC4R expression in the NAc, and dorsal striatum[130]
Male WR offspring↓ MC4R expression in the PFC, NAc, and dorsal striatum
↑ MC4R expression in the amygdala
[131]
10% sucrose or 16% HFCS solutionMale and female SD offspringAmphetamine↑ female alcohol intake during alcohol training
↑ male hyperactivity in LAM
-[199]
HSD: High-Sugar Diet; HFCS: high-fructose corn syrup; WR: Wistar rat strain; SD: Sprague Dawley rat strain; CUE: tone + light—conditional stimulus; SA: self-administration; LAM: locomotor activity measurement; NAc: nucleus accumbens; PFC: prefrontal cortex; MC4R: melanocortin 4 receptor; ↓: decrease; ↑: increase; -: no data.
Table 4. Preclinical studies on the postnatal HSD consumption on behavioural and molecular or neurochemical consequences after drugs of abuse.
Table 4. Preclinical studies on the postnatal HSD consumption on behavioural and molecular or neurochemical consequences after drugs of abuse.
Postnatal ExposureAnimalsDrug of AbuseConsequences Enhanced by HSDRef
HSD ModelPeriod (Days)BehaviouralMolecular/Neurochemical
10% wt/vol sucrose84Male and female C57BL/6J miceCocaine↑ female postcocaine tests locomotor activity,
↑ female sensitization to cocaine in LAM
-[200]
0.2% saccharin (0.3 mL/trial)15Male WR↓ sensitization to cocaine in discrete-trials choice procedures-[192]
70% from corn starch38Male WR↓ immobility, head shake, and locomotion after drug treatment before FST
↓ immobility, climbing, swim, dive, and locomotion in cocaine withdrawal FST
-[209]
8% fructose or glucose or sucrose9Male SD↑ cocaine-CPP after sucrose and fructose,
↓ cocaine-CPP after glucose bingeing
-[204]
10% wt/v sucrose21Female SDAmphetamine↑ activity and sensitivity to low dose of drug in LAM-[202]
10% sucrose5Male SD↑ activity in LAM-[201]
32% (w/v) sucrose solution21Male LE↑ conditioning scores in CPP-[205]
50% kcal from sucrose>35Male WR↑ locomotor activity in LAM, ↓ sensitivity to drug in CPP, and ICSS↓ NAc DA
↑ VTA DAT mRNA
↓ extracellular DA
[203]
60% fructose47, 88, and 144Female SD↑↓SA, and spatial memory testing in water maze test-[206]
64.3% sugar in palatable food30Male LE↑ chow intake after SA-[207]
75% sugar in Skittles candy28Male baboons (Papio cynocephalus anubis)↓ sensitivity to candy reward during pellet meal sessions-[208]
32% (w/v) sucrose solution21Male LEFentanyl↑ conditioning scores
↑ time spent in the previously drug-paired side in CPP
-[205]
HSD: High-Sugar Diet; WR: Wistar rat strain; SD: Sprague Dawley rat strain; CUE: tone + light—conditional stimulus; SA: self-administration; LAM: locomotor activity measurement; LE: Long-Evans rat strain; FST: Forced Swimming Test; CPP: Conditioned Place Preference Test; HFHS: High-Fat High-Sugar Diet; ICSS: Intracranial Self-Stimulation; NAc: nucleus accumbens; VTA: ventral tegmental area; DAT: dopamine transporter; DA: dopamine; ↓: decrease; ↑: increase; -: no data.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Witek, K.; Wydra, K.; Filip, M. A High-Sugar Diet Consumption, Metabolism and Health Impacts with a Focus on the Development of Substance Use Disorder: A Narrative Review. Nutrients 2022, 14, 2940. https://doi.org/10.3390/nu14142940

AMA Style

Witek K, Wydra K, Filip M. A High-Sugar Diet Consumption, Metabolism and Health Impacts with a Focus on the Development of Substance Use Disorder: A Narrative Review. Nutrients. 2022; 14(14):2940. https://doi.org/10.3390/nu14142940

Chicago/Turabian Style

Witek, Kacper, Karolina Wydra, and Małgorzata Filip. 2022. "A High-Sugar Diet Consumption, Metabolism and Health Impacts with a Focus on the Development of Substance Use Disorder: A Narrative Review" Nutrients 14, no. 14: 2940. https://doi.org/10.3390/nu14142940

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop