Next Article in Journal
A Recap of Heme Metabolism towards Understanding Protoporphyrin IX Selectivity in Cancer Cells
Next Article in Special Issue
Association of Polymorphic Variants in Argonaute Genes with Depression Risk in a Polish Population
Previous Article in Journal
Human SMILE-Derived Stromal Lenticule Scaffold for Regenerative Therapy: Review and Perspectives
Previous Article in Special Issue
Transcriptome Profiling of the Dorsomedial Prefrontal Cortex in Suicide Victims
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neurobiological Mechanisms Modulating Emotionality, Cognition and Reward-Related Behaviour in High-Fat Diet-Fed Rodents

1
Department of Behavioural Biology, University of Osnabrueck, Barbarastrasse 11, 49076 Osnabrueck, Germany
2
Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(14), 7952; https://doi.org/10.3390/ijms23147952
Submission received: 23 May 2022 / Revised: 15 July 2022 / Accepted: 16 July 2022 / Published: 19 July 2022

Abstract

:
Affective and substance-use disorders are associated with overweight and obesity-related complications, which are often due to the overconsumption of palatable food. Both high-fat diets (HFDs) and psychostimulant drugs modulate the neuro-circuitry regulating emotional processing and metabolic functions. However, it is not known how they interact at the behavioural level, and whether they lead to overlapping changes in neurobiological endpoints. In this literature review, we describe the impact of HFDs on emotionality, cognition, and reward-related behaviour in rodents. We also outline the effects of HFD on brain metabolism and plasticity involving mitochondria. Moreover, the possible overlap of the neurobiological mechanisms produced by HFDs and psychostimulants is discussed. Our in-depth analysis of published results revealed that HFDs have a clear impact on behaviour and underlying brain processes, which are largely dependent on the developmental period. However, apart from the studies investigating maternal exposure to HFDs, most of the published results involve only male rodents. Future research should also examine the biological impact of HFDs in female rodents. Further knowledge about the molecular mechanisms linking stress and obesity is a crucial requirement of translational research and using rodent models can significantly advance the important search for risk-related biomarkers and the development of clinical intervention strategies.

Graphical Abstract

1. Introduction

The rates of overweight and obesity are rising in children and adults in most parts of the world, representing a major global health challenge [1]. Excessive weight and obesity are partly due to a sedentary lifestyle combined with a high-calorie diet (i.e., a diet rich in sugars and fat, also referred to as a “Western diet”) [2] and can be associated with compulsive eating, binge eating disorders, as well as eating addiction [3,4]. Overweight and obesity increase the risk of developing metabolic diseases (e.g., heart disease, stroke, high blood pressure, diabetes), but also cancer and chronic diseases [5]. Although overweight- and obesity-related metabolic complications can be reduced in a subset of individuals by changes in lifestyle (i.e., balanced diet, healthy eating habits, regular physical exercise), pharmacotherapies and/or surgical procedures, the vast majority of patients exhibit only transient weight loss, which is followed by rebound effects [6,7].
In addition to physical complications, overweight and obesity have a strong impact on mental health [8,9]. In particular, they increase the risk for developing depressive and anxiety disorders as well as substance-use disorders (SUD) (e.g., illegal drugs, nicotine or alcohol) [10,11,12,13,14]. The latter disorders have a high prevalence in the population worldwide and result in a high degree of comorbidity themselves [15,16,17].
In recent years, there has been increasing interest in understanding the shared neurobiological substrates of obesity and SUD [18]. In both disorders, the saliency of a specific type of reward (food or drug) becomes exaggerated, leading to overeating or compulsive drug abuse, respectively [19]. Food and drug reward activate overlapping brain nuclei [20,21,22], which complicates research designed to understand the neurobiological underpinnings of behaviours motivated by rewards in obesity and SUD [23,24].
A theoretical mechanism that has dominated the field for many years is the dysregulation of the brain reward pathways involving the dopamine (DA) system [25], which responds to drugs of abuse and is critical in the development of SUD [26,27]. The consumption of palatable food leads to the activation of the DA reward circuitry [28]. Additionally, limiting the intake of rodent chow diet increased the sensitivity to cocaine and amphetamine, two psychostimulant drugs widely used and abused that increase dopaminergic transmission [29,30,31]. Furthermore, limiting the access to a high-calorie diet produced dopaminergic adaptations similar to those observed in responses to rewarding stimuli in the nucleus accumbens (NAc), a brain area involved in reward processing and drug addiction [32,33].
In brain imaging studies, obese patients present a downregulation of striatal DA function [34], with individuals with the largest body mass index having the lowest dopamine receptor 2 (D2R) availabilities [34,35]. The reduction in D2R availability was associated with the reduced metabolism of various prefrontal brain regions [35]. Because variations in striatal D2R availability have been linked to changes in the reinforcing value of both food and drugs of abuse [34,36], it has been proposed that obese patients and drug addicts may share neuroadaptations in dopaminergic pathways. These neuroadaptations may regulate neuronal systems associated not only with reward and motivation, but also with inhibitory control, salience attribution, and emotional reactivity. Furthermore, obese patients exhibited greater activation of brain regions involved in reward and attention in response to palatable food images or consumption compared to normal-weight subjects [37,38]. Thus, these neuroadaptations may produce a reduced state of reward, which is then possibly compensated for by the overconsumption of palatable food and/or the abuse of drugs [39].
Multiple intertwined genetic, psychosocial, and neuro-immuno-endocrine factors have been proposed to contribute to the association of obesity and SUD, including the gut–brain axis, inflammation and oxidative stress [40,41,42,43,44]. In particular, much evidence shows that overweight and obesity are associated with alterations in oxidative stress and mitochondrial functions in peripheral organs and in the brain [45,46,47,48]. In parallel, drugs of abuse have been shown to increase oxidative stress occurring in dopaminergic neurotransmission [49,50,51,52,53]. A better knowledge of the precise neurobiological mechanisms contributing to the comorbidity of obesity and SUD, and their independent association with depressive and anxiety disorders, is needed. This could help understand how palatable food and drugs of abuse disrupt the reward circuit and lead to changes in compulsive and affective behaviours. This may also inform novel therapeutic targets and preventive efforts for obesity and SUD.

2. Aim of the Present Review

In this context, the aims of this narrative review are the following.
First, we recapitulate some of the predominant behavioural changes caused by exposure to palatable food and in combination with psychostimulant drugs (i.e., cocaine and amphetamine) in juvenile, adolescent, and adult rodents. Because of the high prevalence of obesity in pregnant women [54] and the short- and long-term negative consequences of poor nutrition during pregnancy and lactation on both the mother and child [55], we also examine the impact of palatable food in dams and their offspring.
Second, we describe the neurobiological mechanisms that may contribute to disruptions in behaviour and metabolic functions by palatable food in rodents. At the molecular level, we focus on brain energetics, mitochondrial function, oxidative stress, neuroplasticity and neuro-inflammation. A more detailed analysis of the molecular mechanisms involving the microbiota–gut–brain axis falls outside the scope of the current review (for more information see [56,57,58,59] and discussion Section 7.2.2).
Finally, we attempt to address the issue of whether the consumption of high-energy diets and psychostimulant drugs leads to overlapping changes in brain metabolism and plasticity that alter emotional states and thus modulate subsequent behaviours.

3. Important Considerations on the Use of High-Fat Diet Treatments in Rodents

Over the past century, diet-induced obesity models have been used in rodents to investigate the body and brain alterations that occur during the progression of obesity and in the presence or in the absence of predetermined genetic alterations [60]. They include various approaches (i.e., diets rich in fat and/or sugar, genetic models, pharmacological models) and produce, in various degrees, alterations in energy metabolism resembling those found in obese patients. For example, high-energy diets markedly increase body weight and adiposity, and induce a pre-diabetic phenotype (e.g., hyperglycaemia, hyperinsulinemia, insulin resistance, glucose intolerance). Moreover, they produce hormonal dysregulation, hypothalamic neuropeptidergic adaptations and low-grade inflammation [61,62,63,64,65].
For the behavioural level, the current literature is filled with examples in which long-term exposure to high-energy diets alters cognitive performance and modulates emotionality [66,67,68,69,70,71,72]. The latter can be defined as the measure of emotional behaviour related to undirected escape, avoidance of specific stimulus or area, immobility and sympathetic nervous system activation (e.g., heart rate, urination, defecation) [73]. However, there are numerous discrepant findings, for instance in rodent models of depression [69]. The main theoretical premise behind the inconsistencies concerns variations in experimental designs (e.g., diet content and duration, exposure period; sex, age, species and genetic background of the animals used; behavioural tests).
Recent criticism of the preclinical (mainly rodent) literature indicates that the commonly used commercial high-energy diets may not necessarily model the dietary habits associated with overweight and obesity in humans [74]. For instance, rodent high-fat diets (HFDs) typically contain either ≈45% or 60% kcal from fat and are characterised by varying fatty acid composition [62,74]. Because the currently recommended total fat intake for adult humans ranges from 20 to 30% [75,76] and the average European/American diets contain approximately 28.5 to 46.2% kcal from fat [74,77,78], the use of rodent HFD containing ≈45% kcal from fat seems the most appropriate diet from a translational point of view.
Here, we aimed to reduce the heterogeneity of findings between studies and parallel the fat content found in the average European/American diets. Thus, we compare the behavioural effects of HFD treatments containing ≈25–50% kcal from fat (predominantly 45%) with control (CON) diets providing 3–18% fat (usually 10%). Studies that used a rodent HFD containing a very high amount of fat (≈60% kcal), which rapidly produce extreme obesity [62] and increase pup cannibalism in dams [79], were excluded from the present review.
With the exception of the studies on maternal HFD and a few other studies, all reviewed studies used male mice or rats. To assess biological parameters indicative of metabolic dysfunction, the negative impact of the HFD treatment on energy metabolism was almost systematically verified (e.g., body weight gain, increased calorie intake, hyperglycaemia, hyperinsulinemia, dyslipidaemia, peripheral inflammation) (see details in Supplementary Table S1).

4. Impact of a High-Fat Diet Treatment on Emotionality, Cognition and Reward-Related Behaviour

The impact of HFD treatment on rodent behaviour is summarized in Table 1.

4.1. Impact of a Maternal High-Fat Diet Treatment in Dams

HFD consumption during pre- and post-partum periods, two metabolically challenging periods, produced mixed changes in the gestational body weight, but generally increased adiposity and induced hyperleptinemia in dams (see Supplementary Table S1). The behavioural profile of the dams showed that maternal HFD had a negative impact on maternal behaviour, especially regarding nest building and pup licking/grooming [80,81].
Interestingly, one study showed that maternal HFD had no significant impact on nursing behaviour in breeding dams. However a reduction in the quality of maternal care was observed in dams from generation F3, which were selectively bred for diet-induced obesity or diet-induced resistance [82].
So far, few studies have investigated the link between obesity-related factors and increased risk for maternal diseases as well as the importance of inherited predisposition to HFD-induced obesity. Further work is therefore needed to understand how obesity during pregnancy and lactation influence the maternal metabolic and behavioural adaptations necessary to bring about new life.

4.2. Impact of a Maternal High-Fat Diet Treatment in Offsspring

To our knowledge, very few studies examined the impact of HFD treatment (containing ≈ 25–45% kcal from fat) applied to breeding dams during pregnancy and lactation on the behavioural and metabolic phenotypes of the offspring (see Table 1, Supplementary Table S1).
The offspring of mothers fed a HFD during pregnancy showed impaired learning in an operant conditioning paradigm when tested in adulthood [83]. This was indicated by an increased number of sessions required to press an operant lever and obtain a reward [83]. However, mice from HFD-fed dams and CON-fed dams did not significantly differ regarding their motivation to obtain a reward and their appetitive and/or consummatory drive (i.e., a similar number of rewards was received) [83]. They also did not significantly differ regarding locomotion and anxiety-related behaviour [83]. Because of the very limited data available regarding the long-term behavioural consequences of a HFD during prenatal development, further work is required to draw any conclusion.
A few studies investigated the effects of a HFD during the pre- and post-partum periods on the offspring’s development, behaviour and cognition. Juvenile offspring of mothers fed a HFD during pregnancy and lactation showed an increased preference for a 1% corn oil solution [84], changes in voluntary running wheel activity [85] and increased passive stress-coping behaviour [86]. No significant differences between the offspring of HFD-fed dam and CON-fed dams were seen in locomotion, anhedonia and learning and memory [86,87]. Regarding psychostimulant-related behaviour, the offspring displayed reduced amphetamine-induced locomotion and sensitization [87].
Compared to offspring of CON-fed mothers during pregnancy and lactation, adolescent offspring of mothers fed a HFD treatment during pregnancy and lactation showed no significant differences in locomotion and the rewarding or motivational effects of cocaine in the cocaine self-administration paradigm [88]. When tested in adulthood, a few studies failed to also detect an effect on a maternal HFD treatment on behavioural phenotypes, notably locomotion, anxiety-related behaviour and cognitive performance [83,89].
It is important to mention that the maternal HFD produced long-term changes in the reactivity of the hypothalamic–pituitary–adrenal axis, the core mediator of neuroendocrine stress responses [90]. In addition, cognitive deficits could be identified by a post-weaning exposure to HFD treatment [89], suggesting that a second hit stressor may be necessary to unmask programmed behavioural defects of the early nutritional environment.

4.3. Impact of a High-Fat Diet Treatment during the Onset of Puberty and Early Adolescence

4.3.1. Cognition and Emotionality

The consumption of a HFD during the onset of puberty and early adolescence period (here referred to as “juvenile HFD”, involving 4–6-week-old mice and 3–9-week-old rats [91] produced long-term negative consequences on hippocampus-dependent cognition (see Table 1). This was observed in several tasks assessing spatial memory, recognition memory and fear-aggravated memory [92,93,94,95,96]. Similar to the effects of a metabolic challenge (i.e., exposure to HFD), chronic exposure to physical and/or psychosocial stressors early in life produced long-term impairments in learning and memory in rodents, specifically for the spatial domain [97,98]. However, the biological and signalling pathways leading to alterations in cognitive performance for the different types of stress may differ.
Regarding emotionality, a juvenile HFD treatment did not reliably increase anxiety-related behaviour in various paradigms [92,93,95,99,100,101,102,103,104]. It also had no major impact on locomotion and stress-coping style in the forced swim test [92,93,94,95,100,101,102,103,105,106] (see Table 1).

4.3.2. Psychostimulant-Related Behaviour

Concerning psychostimulant-related behaviour (see Table 1), the outcomes of a juvenile HFD treatment were dependent on its schedule of availability. When given ad libitum, a juvenile HFD treatment weakened cocaine- and amphetamine-related memories in the conditioned place preference (CPP) paradigm, especially for the lower drug doses administered [99,107,108,109]. When time-restricted access was provided, juvenile HFD treatment leading to binge eating enhanced the acquisition and reinstatement of cocaine self-administration [107]. The results of these studies suggest that palatable food can modulate psychostimulant-rewarding and reinforcing properties under certain patterns of food administration. In particular, they highlight that animals that develop a pattern of binge eating on palatable food are more vulnerable to psychostimulant drugs (referred to as cross-sensitization) [111]. The neurobiological mechanisms by which HFD treatments and psychostimulant drugs act to promote compulsive behaviour warrant further investigation.
One crucial factor to consider is the importance of the social housing conditions of the animals. In fact, a juvenile intermittent HFD treatment led to binge eating in single-housed mice and reduced their cocaine-induced CPP, but increased cocaine-induced CPP in group-housed animals that did not show binge-eating behaviour [112]. This study suggests that palatable food may work as an alternative reward to psychostimulant drugs and stimulates the same brain pathways as psychostimulants do. However, the mechanisms by which social housing conditions modulate the rewarding properties of HFD treatment are presently unclear.
Regarding the behaviour of animals during the exposure to psychostimulant drugs (see Table 1), a juvenile HFD treatment was found to modulate the behavioural sensitization to amphetamine [108,113] and had a minor impact on cocaine-induced anxiolytic and “antidepressant” effects [106,107]. During cocaine withdrawal, HFD treatment facilitated the extinction of cocaine-induced CPP, increased cocaine-induced locomotion, and produced anxiolytic and antidepressant effects [99,106]. Interestingly, preventing access to the HFD treatment increased anxiety-related behaviour, cocaine-induced locomotion and cocaine-induced CPP for a subthreshold cocaine dose [107], suggesting that withdrawal from HFD could potentiate psychostimulant-rewarding properties.
The long-term consequences of chronic consumption of (and withdrawal from) palatable foods during the early adolescence period remain unclear but might lead to alterations of the brain reward system that have been associated with obesity and metabolic disorders.

4.4. Impact of a High-Fat Diet Treatment during Late Adolescence and Young Adulthood

Turning to the experimental evidence on the impact of HFD treatment during late adolescence and young adulthood (i.e., in 6–9-week-old mice and 9–10-week-old rats [91]), (see Table 1), the summarized data show that an adolescent HFD treatment increased anxiety-related and anhedonia-like responses, without reliably affecting stress-coping style [114,115,116,117,118]. It also reduced locomotion, exploration but not social behaviours [116,117,119,120]. In contrast to HFD treatment in juveniles, HFD treatment in adolescents had no major impact on hippocampus-dependent learning and memory [116,121,122]. Similar results of increased emotionality and reduced reward responsiveness have been reported in rodents exposed to physical and/or psychosocial stressors during adolescence [123,124].

4.5. Impact of a High-Fat Diet Treatment in Adulhood

HFD treatment in adulthood (beyond the age of 9 weeks in mice and 10 weeks in rats) reduced the hedonic response in the sucrose preference test [125], but it did not produce major behavioural alterations concerning anxiety, locomotion/exploration, and learning and memory [126,127]. However, with such a small number of studies, caution must be applied, as these findings might not reflect the overall impact of an adult HFD treatment on emotionality, cognition and reward-related behaviour in rodents.
HFD treatment in adults altered cocaine-induced locomotor and antidepressant effects [128], and increased amphetamine-induced locomotion, but without inducing a behavioural sensitization to amphetamine [129]. Interestingly, withdrawal from amphetamine significantly increased body weight gain and produced hyperphagia in HFD-fed animals, suggesting that HFD consumption may alleviate reward deficits mediated by psychostimulant withdrawal. Because the increase in food consumption in amphetamine-withdrawn rats was specific to the laboratory chow rather than the HFD [129], further work is needed to determine how psychostimulant drugs and HFD interact to produce long-lasting sensitization of reward-related behaviours.

4.6. Conclusions

In light of the reported findings, it is conceivable that the strong effects of HFD treatment on cognitive performance and reward-related behaviour were seen in both juvenile and adolescent rodents. Indeed, during the pre-pubertal phase and adolescence, rodents undergo dramatic hormonal, neurobiological and behavioural changes [130,131,132]. In particular, the limbic system (notably the hippocampus and amygdala) as well as cortical regions involved in emotional and learning processing undergo structural and functional maturation [133]. Data from several studies have identified these periods as critical time windows during which stress exposure produces short- and long-term effects on emotionality and cognition [134,135,136,137], as well as associated neural structure and function [138,139,140]. In humans, adolescence is considered a window of vulnerability to pathological development [141,142]. Adolescents are particularly sensitive to reward and often increase their consumption of palatable foods, such as a high-fat diet [143], which could lead to obesity.
Because pre-puberty and adolescence are critical periods of neurobehavioural reorganization necessary for life-long cognitive processes and reward processes, the possible mechanisms underlying the marked vulnerability to the detrimental effects of HFD treatment may involve changes in energy metabolism and neuronal plasticity. Such mechanisms are discussed in the next section.

5. Impact of a High-Fat Diet Treatment on Molecular Correlates of Energy Metabolism and Plasticity

As reviewed above, poor dietary choices during crucial stages of development can have a detrimental impact on emotionality, cognition and reward-related behaviours. Therefore, what are the effects of HFDs on underlying brain mechanisms?
The impact of HFD treatment on energy metabolism, molecular correlation of mitochondrial dysfunction (e.g., redox imbalance, oxidative damage and inflammation) and neuroplasticity in rodents is summarized in Table 2. Data are sorted by brain area and developmental period (see details in Supplementary Table S1).

5.1. Impact of a High-Fat Diet Treatment on the Cerebral Cortex

5.1.1. Energy Metabolism

Regardless of the time at which it was applied, HFD treatment had a minor impact on glucose, amino acid and phospholipid metabolism in the cortex [105,119,127]. However, it seemed to have induced a slight molecular insulin resistance, as indicated by increased insulin level and a lack of activation response of key players of the insulin/phosphatidylinositol 3-kinase (PI3K)/Akt/glycogen synthase kinase-3β (GSK-3β) cascade [119].

5.1.2. Mitochondria-Related Functions and Oxidative Stress

The data gathered show that HFD treatment in both juveniles and adults had a minor impact on cortical mitochondrial bioenergetics (i.e., expression of mitochondrial respiratory chain complexes, respiratory spare capacity, ATP production) [105,144] and the cell redox state [121,127,144]. One study in HFD-fed juvenile animals found a significant decrease in the level of superoxide dismutase (SOD), the first line of defence against oxidative stress, and the anti-oxidant glutathione (GSH) [144]. This was associated with enhanced lipid peroxidation, indicated by increased level of malondialdehyde, and modest neuro-inflammation shown by the variations in the expression of some cytokines (e.g., tumour necrosis factor alpha and interleukins 1β) [144]. Two other studies in HFD-fed juvenile or adolescent animals observed no significant changes in the level of GSH and hydroxynonenal (HNE), another indicator of lipid oxidation [121,127]. No significant changes were seen in the expression of glial fibrillary acidic protein (GFAP) and IBA1 (allograft inflammatory factor 1), a marker of activated microglia [127].
Interestingly, alterations in the redox balance were found in cortical synaptic mitochondria from HFD-fed animals, which also showed alterations in several mitochondrial respiration parameters (i.e., reduced basal respiration, ATP production, proton leak and increased degree of coupling) [144]. Thus, it is possible that HFD treatment produced detrimental effects particularly localized at the synapses.

5.1.3. Neuroplasticity and Neuroinflammation

Because synaptic mitochondria regulate synaptic transmission, variations in synaptic mitochondrial respiration accompanied by increased oxidative stress would be expected to be associated with changes in neurotransmission and neuroplasticity. However, HFD treatment had only a minor impact on synaptic plasticity in the cortex [115,121,127,144]. As for maternal HFD treatment, it had no significant impact on dopaminergic markers in the prefrontal cortex of juvenile offspring [87]. It is noteworthy that HFD treatment in adolescents modulated the expression of brain-derived neurotrophic factor (BDNF), a major regulator of synaptic transmission and plasticity [144]. Additionally, it reduced the synaptic BDNF protein synthesis and the phosphorylation of BDNF upstream factor cyclic AMP-responsive element-binding protein (CREB) in cortical synaptosomal fractions [144], again highlighting the marked effects of HFD treatment on BDNF-related molecular plasticity at the synaptic level.

5.2. Impact of a High-Fat Diet Treatment on the Hippocampus

5.2.1. Energy Metabolism

In the hippocampus, a critical brain area for learning and memory, HFD treatment had a minor influence on glucose metabolism and insulin signalling, and no significant impact on glucose transport, amino acid and phospholipid metabolism [105,119,122,127]. As observed for the cerebral cortex, an HFD in adolescence might be associated with a minor molecular insulin resistance, as shown by the lack of activation response of Akt [119]. It might also be associated with reduced glycolysis processes, as suggested by the significant increase in the expression of glucose-6-phosphate and the reduced activity of the pyruvate dehydrogenase enzyme [105].

5.2.2. Mitochondria-Related Functions and Oxidative Stress

The available evidence shows no major impact of HFD treatment in juveniles and adults on oxidative capacity and redox balance of the hippocampus. Indeed, mixed changes were seen in the protein expression of some respiratory chain complexes and no significant impact was found on citrate synthase activity and ATP production or anti-oxidant level [94,121,127]. It also revealed contrasting signs of lipid peroxidation, astrogliosis, microgliosis and discrepant changes in cytokines release [94,103,117,121,126,127].

5.2.3. Neuroplasticity and Neuroinflammation

In contrast to observations made in the cortex, HFD treatment had a major impact on hippocampal neurotransmission and synaptic plasticity in all periods examined, but especially during adolescence. For example, HFD treatment significantly affected serotonergic transmission [117,118]. HFD treatment reduced the expression of BDNF, as well as that of key synaptic proteins involved in vesicle trafficking and receptor anchoring; it also reduced the number of dendritic spines and produced some changes in the cyto-architecture of the hippocampus [117,121,122,127]. Similarly, maternal HFD treatment decreased the length of dendritic spines (without affecting the spine density) and altered astrocyte morphology [89,145].

5.3. Impact of a High-Fat Diet Treatment on the Hypothalamus

5.3.1. Energy Metabolism

Unfortunately, studies that examined the impact of HFD treatment on the hypothalamus during adolescence are lacking. Regarding HFD treatment in juveniles and adults, a close look at the data indicates that it did not affect hypothalamic glucose, amino acid or phospholipid metabolism [127,146,147]. Nonetheless, it potentially produced alterations in insulin signalling, as indicated by increased insulin receptor expression in the arcuate nucleus (ARC), an area of particular importance to energy homeostasis [146,147].
Table 2. Literature overview of the impact of a high-fat diet treatment on molecular correlates of energy metabolism and plasticity in the brain of rodents. Description: Studies are organized by period in which the high-fat diet (HFD) treatment was applied (i.e., before puberty, during adolescence or in adulthood). For the effects of a maternal HFD treatment, studies are organised by the developmental period (i.e., before puberty or early adolescence, during late adolescence or in adulthood) in which the offspring were tested. The numbered references refer to the bibliography section.
Table 2. Literature overview of the impact of a high-fat diet treatment on molecular correlates of energy metabolism and plasticity in the brain of rodents. Description: Studies are organized by period in which the high-fat diet (HFD) treatment was applied (i.e., before puberty, during adolescence or in adulthood). For the effects of a maternal HFD treatment, studies are organised by the developmental period (i.e., before puberty or early adolescence, during late adolescence or in adulthood) in which the offspring were tested. The numbered references refer to the bibliography section.
Brain Area & Ref.Overall
Function
Outcomes

(Increased)

(Decreased)

(Not Changed)
Maternal HFD treatment
Testing in offspring
PFC
[87]
Neuronal func. Level of DA, DOPAC
HC
[89,145]
Neuronal func. Dendritic spine lengthDendritic spine density
DCX-positive cells
Neuroinflam.Astrocyte process number and total length
AMY
[145]
Neuronal func. Dendritic spine lengthDendritic spine density
NAc
[84,87]
Neuronal func.Level of DA, DOPAC
Ddr2 mRNA
Htr1a mRNA
TH density fibres
Expression of TH, DAT, D1/2R
STR
[85,87]
Neuronal func. Expression of TH, DAT, D1/2R
VTA
[84,87]
Neuronal func.Htr1a mRNATH positive neurons §
Th mRNA §
TH positive neurons §
Th mRNA §
Juvenile HFD treatment
CC
[105]
Glucose metab. Glucose transport, glycolysis
Insulin signalingInsulin level Insulin sensitivity
OXPHOSExpression of ETC CIV-VSpare respiratory capacity Expression of ETC CI-III, basal OCR, State 3 OCR, level of ATP
HC
[103,104,105]
Glucose metab. GlycolysisGlucose transport
Insulin signaling Insulin level and sensitivity
OXPHOS Expression of ETC CI-IIExpression of ETC CI-V, level of ATP
MT biogenesis Expression of PGC1α, PPARγ
Oxidative stressLipid peroxidation Anti-oxidant defenses (SOD, GPX)
Neuronal func. Dendritic spines, expression of SYPExpression of BDNF
Neuroinflam.Level of IL10, micro/astrogliosisLevel of IL6Level of IL6, IL1β, TNFα, microgliosis
HYP
[104,147,148]
Insulin signalingIns mRNA Mtor, Irs1 mRNA
OXPHOS Expression of ETC CI-V, level of ATP
Oxidative stress Anti-oxidant defenses (SOD, CAT), lipid peroxidation
Neuroinflam.Microgliosis Tgfβ mRNA, microgliosis
AMY
[103,104]
Neuroinflam.Level of TNFα, microgliosis Level of IL6
NAc
[107,108,113]
Insulin signalingMtor mRNA
Neuronal func.Expression of D1RExpression of DATBasal DA release; level of DA, DOPAC; expression of TH, DAT, D1/2R
VTA
[113]
Neuronal func. Spontaneous/bursting DA activity
Expression of TH, DAT
HFD treatment in late adolescence
CC
[115,117,119,121]
Insulin signaling Insulin sensitivity
Oxidative stress Anti-oxidant defenses (GSH, GSSG), lipid peroxidation
Neuronal func.Ddr2 mRNAGabbr1/2 mRNAExpression of BDNF, SYP
Drd1 mRNA
Neuroinflam. Il1β, Il2 mRNAIl10, Il4, Il6, Tnfα, Tgfβ, Ifnγ mRNA
HC
[117,118,119,121]
Insulin signaling Insulin sensitivity
Oxidative stress Anti-oxidant defenses (GSH, GSSG), lipid peroxidation
Neuronal func. Nissl staining, Bdnf mRNA, level of 5-HT; Htr1a, Slc6a4, Ido2 mRNACA1 pyramidal layer thickness, CA1 LTP, expression of PSD95, BDNF, SYP
Neuroinflam.Il1β, Il2, Il6, Il17 mRNAIl10 mRNAIl4, Tnfα, Tgfβ, Ifnγ mRNA
HYP [149]Neuroinflam. IL1β mRNA, astrogliosis
Adult HFD treatment
CC
[127,144]
Glucose metab. Level of glucose, lactate
AA/PL metab. Level of PEALevel of Gln, Glu, GABA
OXPHOS State 3 OCR, Spare respiratory capacity
Oxidative stressLipid peroxidationAnti-oxidant defenses (GSH, GSSG, SOD)Anti-oxidant defenses (GSH, As)
Neuronal func. Expression of BDNFLevel of NAA, expression of PSD95, SYP, SYN, VGLUT1/2, VGAT
Neuroinflam.Level of TNFα, IL1β Micro/astrogliosis
HC
[126,127]
Glucose metab. Level of glucose, lactate
AA/PL metab. Level of Gln, Glu, GABA, PEA
Oxidative stress Anti-oxidant defenses (GSH, As)
Neuronal func. Expression of SYN, VGLUT1, VGATLevel of NAA, expression of PSD95, SYP, VGLUT2
Neuroinflam. Tnfα, mRNA, microgliosis
HYP
[127,146]
Glucose metab. Level of glucose, lactate
Insulin signalingIns mRNA
AA/PL metab. Level of Gln, Glu, GABA, PEA
Oxidative stress Anti-oxidant defenses (GSH, As)
Neuronal func. Expression of VGLUT1/2, VGATLevel of NAA, expression of PSD95, SYP, SYN
Neuroinflam.Level of TNFα, IL1β, IL6, astrogliosis Microgliosis
Abbreviations: 5-HT: serotonin; AA: amino-acid; AMY: amygdala; As: ascorbate; ATP: adenosine triphosphate; BDNF/Bdnf: protein/gene coding for brain-derived neurotrophic factor; CA1: region 1 of the cornu ammonis; CAT: catalase; CC: cerebral cortex; D1R/Drd1: protein/gene coding for dopamine receptor 1; D2R/Drd2: protein/gene coding for dopamine receptor 2; DA: dopamine; DAT: dopamine transporter; DCX: doublecortin; DOPAC: 3,4-dihydroxyphenylacetic acid; ETC CI-V: electron transport chain complex I-V; GABA: gamma-aminobutyric acid; Gabbr1/2: gene coding for gamma-aminobutyric acid type B receptor subunit 1 or 2; Gln: glutamine; Glu: glutamate; GPX: glutathione peroxidase; GSH: glutathione; GSSG: oxidised glutathione; HC: hippocampus; HFD: high-fat diet; Htr1a: gene coding for 5-hydroxytryptamine receptor 1A; HYP: hypothalamus; Ido2: gene coding for indoleamine 2,3-dioxygenase 2; Ifnγ: gene coding for interferon; IL/Il: protein/gene coding for interleukin; Ins: gene coding for insulin; Irs1: gene coding for insulin receptor substrate 1; LTP: long-term potentiation; metab.: metabolism; MT: mitochondria; Mtor: gene coding for mechanistic target of rapamycin kinase; NAA: N-acetyl aspartate; NAc: nucleus accumbens; Neuroinflam: neuro-inflammation; Neuronal func.: neuronal function; OCR: oxygen consumption rate; OXPHOS: oxidative phosphorylation; PEA: palmitoylethanolamide; PFC: prefrontal cortex; PL: phospholipid; PGC1α: peroxisome proliferator-activated receptor gamma coactivator 1-alpha; PPARγ: peroxisome proliferator-activated receptor gamma; PSD95: postsynaptic density protein 95; Slc6a4: gene coding for sodium-dependent serotonin transporter; SOD: superoxide dismutase; STR: striatum; SYN: syntaxin; SYP: synaptophysin; Tgfβ: gene coding for transforming growth factor beta; TH/Th: protein/gene coding for tyrosine hydroxylase; TNFα/Tnfα: protein/gene coding for tumour necrosis factor; VGAT: vesicular GABA transporter; VGLUT1/2: vesicular glutamate transporter 1 or 2; VTA: ventral tegmental area. Symbols: §: outcomes differing based on the age of animals at the time of testing.

5.3.2. Mitochondria-Related Functions and Oxidative Stress

HFD treatment in juveniles had no significant impact on hypothalamic mitochondrial respiration and energy storage during oxidative phosphorylation. This was shown by the absence of significant changes in the activity of respiratory chain complexes and ATP production [147,148]. It also had no significant impact on the cell redox state [147,148]. For instance, it did not alter the expression of first-line antioxidant defences, SOD, catalase (CAT) and GSH [147]. Additionally, it did not alter the total anti-oxidant status and did not increase oxidative damage to lipids [147,148].
HFD treatment in juveniles produced slight alterations in hypothalamic mitochondrial morphology [147,150]. A decreased mitochondrial surface area was reported in the paraventricular nucleus of the hypothalamus (PVN), although no significant changes were seen in the mitochondria type, aspect ratio and surface density coverage [147]. Thaler and colleagues (2012) found divergent mitochondria ultrastructure in pro-opiomelanocortin (POMC) neurons of the ARC. In this study, the morphological examination of mitochondria displayed homogenous, compact electron-dense lumens with well-organised, parallel-oriented cristae in mitochondria in chow-fed rats but not in HFD-treated rats [150].

5.3.3. Neuroplasticity and Neuroinflammation

In the hypothalamus, HFD treatment in adults was found to produce astrogliosis as well as reduce the protein expression of vesicular glutamate transporter and GABA transporters, possibly reducing the availability of excitatory and inhibitory vesicles for neurotransmission [127,149]. However, no significant impact of HFD treatment was seen on the expression of synaptic proteins, neuronal integrity and microgliosis [104,127,147].

5.4. Impact of a High-Fat Diet Treatment on the VTA-NAc DA System

5.4.1. Impact of a High-Fat Diet Treatment

To our surprise, there has been little research on the impact of HFD treatment on brain metabolism and plasticity within brain regions that are potentially involved in the behavioural overlap of consumption of HFD and psychostimulant-related behaviour.
For example, maternal HFD treatment had a significant impact on the basal functionality of the ventral tegmental area (VTA)–nucleus accumbens (NAc) DA pathway, which plays a critical role in reward-relevant behaviours and emotional behaviours, especially following stress exposure [151]. For example, juvenile offspring from HFD-fed dams showed a significant increase in the level of the DA neurotransmitter and in 3,4-Dihydroxyphenylacetic acid (DOPAC, a metabolite of DA) in the NAc [87]. No significant changes were seen in the expression of the DA transporter (DAT), DA receptors and tyrosine hydroxylase (TH), the rate-limiting step in this synthesis of DA [87]. In the VTA, juveniles from HFD-fed dams exhibited changes in the expression of TH and the number of TH-positive neurons [87]. Further studies are therefore needed to determine how HFD treatment produces neuro-adaptations in the mesolimbic DA system during the pre- and the post-partum periods to influence reward-related behaviours.
By contrast to observations in offspring from HFD-fed dams, HFD treatment applied during early life had a minor impact on the VTA-NAc DA pathway. For example, HFD treatment produced no significant changes in DA neuronal function of the VTA, DA release in the NAc, and it generated contrasting variations in the expression of DAT and DA receptors [108,113]. Further work is therefore needed to characterise the impact of HFD treament impact the activity of the mesolimbic DA system.

5.4.2. Combined Effecs of a High-Fat Diet Treatment with Psychostimulant Drugs

When combined with psychostimulant drugs, HFD treatment significantly increased the activity of VTA DA neurons in response to amphetamine; it also potentiated NAc DA release and induced the recruitment of postsynaptic DA receptors, which resulted in increased amphetamine-induced locomotor activity in HFD-fed animals [113]. In the NAc and the striatum, a juvenile HFD treatment modulated the expression of the cannabinoid receptor 1 and the opioid mu receptor 1 [107,110], which may reflect variations in the release of endogenous peptides. Together, the results provided in these studies suggest that the neuroadaptations in the brain reward pathway in HFD-fed animals may contribute to the changes observed in psychostimulant-related behaviour.

5.5. Conclusions

Taken together, the reviewed data from the literature call for further studies aiming at understanding the relationship between the effects of HFD treatment in different developmental periods on behavioural endpoints and the changes in neurotransmission and molecular plasticity in specific brain areas. In addition, further work is crucially needed to characterise the impact of HFD treatment on VTA–NAc signalling elements that play pivotal roles in reward/reinforcement circuits of the mesolimbic system. Because the homeostatic and reward circuits are often studied separately, the impact of HFD treatment on reward circuits is a question that remains mostly unanswered.

6. Combined Impact of High-Fat Diet and Stress Exposure in Rodents

As reviewed above, HFD treatment (25–50% kcal from fat, predominantly 45%) in rodents produced some impairments in emotionality, cognition and reward-related behaviour, which were accompanied by distinctive neuroanatomical and molecular changes according to the period in which the HFD treatment was applied (see summary in Figure 1). However, these observations were made under basal, non-stressed conditions.
Because both poor dietary choices and stress can lead to disruptions in emotional behaviour, cognitive performance and reward-related behaviour, it is important to consider their effects combined.
The analysis of the literature examining the impact of HFD treatment in animals exposed to chronic stress and showing depression-related phenotypes reveals mixed findings. Some studies reported that adolescent stress and ad libitum HFD interacted to produce a greater vulnerability to the detrimental effects of HFD on emotionality and reward sensitivity in male rodents exposed to psychological stressors. For example, exposure to predator odour threat increased anxiety-related behaviours in HFD-fed rats and reduced brain volume, especially the hippocampal volume, compared to HFD-fed unstressed rats [152]. In another study, HFD-fed mice subjected to a vicarious social defeat stress, wherein one mouse witnesses the physical defeat of a conspecific from the safety of an adjacent compartment (i.e., uncoupling of emotional and physical stress), showed increased social avoidance and reduced hedonic response [153].
Some studies reported that the interaction of stress and HFD reduced the vulnerability to the unfavourable effects of HFD on behaviour and energy metabolism in male rodents exposed to combined psychological and physical stressors. For example, HFD-fed adult mice exposed to social defeat stress showed a reduced body weight gain compared to HFD-fed unstressed mice despite a significant increased caloric intake, an effect due to increased energy expenditure and especially increased fat oxidation [154]. As another example, limited access to HFD treatment reduced the anxiety-related behaviour of adolescent rats subjected to social defeat stress as well as changed their behaviour during the direct confrontation with residents [155]. Together, these two studies confirm the stress-buffering/comfort properties of palatable food against stressors [156].
It is important to mention that a few studies fail to observe an interaction of stress and diet in rodents. For instance, HFD-fed male mice exposed to early life stress in the form of neonatal maternal separation or to unpredictable chronic mild stress showed, in adulthood, no significant differences in metabolic parameters [157] or self-care behaviour and hedonic response [158] compared to HFD-fed unstressed mice.
Taken together, these studies highlight that the chronic activation of the stress response in rodents interfered with HFD treatment to produce behavioural and metabolic impairments that depended on the type and the duration of stress exposure. Interestingly, HFD treatment could reduce the antidepressant efficacy of fluoxetine, a selective serotonin reuptake inhibitor, on stress-induced depression-like behaviour [153,158]. These data suggest that diet plays a role in antidepressant efficacy and parallel some clinical findings showing poor responsiveness to antidepressant therapy in patients suffering from obesity and related metabolic disorders [159,160].

7. Discussion

7.1. Overview of the Impact of a High-Fat Diet Treatment on Behaviour and Brain in Rodents

In the present review, we have provided an overview of important behavioural and molecular correlates of HFD-induced impairments in rodents (see Figure 1). While the use of varying diets, developmental stages, and behavioural tests make the comparison between studies difficult, HFD treatment generally produced a long-lasting detrimental impact on emotionality, cognition and psychostimulant-related behaviours (see summary in Table 1). In addition, HFD treatment produced marked alterations in neuroplasticity and neuro-inflammation, but only slight impairments in energy metabolism and mitochondrial functions (see summary in Table 2). However, only a few studies have measured the effects of HFD treatment on brain energy metabolism and plasticity in the attempt to link behavioural alterations with molecular phenotypes. In addition, when molecular studies were performed, they were limited to one brain area or a small number of brain regions. Therefore, the molecular, cellular, and neural mechanisms underlying the HFD-induced alterations in emotionality, cognition and reward-related behaviour remain to be elucidated.

7.2. Limitations

7.2.1. Lack of Systematic Assessment of the Metabolic Status

One important point to consider is that the assessment of dietary-induced alterations in the peripheral metabolic status was not systematic (see Supplementary Table S1). Therefore, it is unclear whether the HFD-induced changes in brain and behaviour result from the dietary manipulation (i.e., the diet, the diet composition or the high caloric density) or are instead the consequence of metabolic dysfunctions that develop with weight gain, adiposity and peripheral/central metabolic changes. In fact, a few studies observed that HFD treatment induced alterations in behaviour without producing adverse metabolic effects. For example, both ad libitum and restricted HFD treatments produced major changes in psychostimulant-related behaviours without increasing the body weight and altering the insulin and leptin signalling [107,108,110,161].
While the verification of the body weight gain has performed almost consistently, few studies have investigated whether HFD treatment produced insulin resistance, as measured by elevated fasting plasma glucose and insulin, and the measure of leptin levels has also not been routinely done (see Supplementary Table S1). Because insulin and leptin have a critical role not only in whole-body energy homeostasis but also in regulating neuronal structure and function (e.g., through regulation of synaptic plasticity and trafficking of neurotransmitter receptors) [162,163], further work is needed to link behavioural alterations with changes in insulin and leptin signalling in brain areas associated with emotionality, cognition and reward. Because of the close interaction of mitochondria, leptin and insulin signalling in the brain, a better understanding of the underlying mechanisms by which insulin and leptin resistance modify mitochondrial functions may help identify novel therapeutic strategies to combat obesity and associated comorbidities.

7.2.2. Lack of Evaluation of the Neuroendocrine and Immune Systems

It is also important to highlight that relatively little is known about the impact of HFD treatment on neuroendocrine function and peripheral mediators of inflammation from the studies reviewed. In fact, contrasting effects of HFD treatment were found on corticosterone release in offspring of HFD-fed dams [89,90,145,164] and HFD treatment had no major impact on corticosterone release in HFD-fed animals [102,107,112,120,127]. In addition, discrepant findings were found on peripheral measures of oxidative stress and inflammation [101,114,144,147]. Further investigation is needed to determine whether and by which mechanisms HFD treatment could disturb hypothalamic–pituitary–adrenal axis function and produce systemic immune dysregulation.
Although the description of the effects of HFD treatment on gut microbiota falls out of the scope of this review, it appears important to highlight the importance of this topic. It is well documented that the food consumed affects the bacteria composition within the gut microbiome, which plays a crucial role in food absorption, nutrient and energy extraction as well as low-grade inflammation [56]. In particular, high-calorie diets and specific dietary components play a role in shifting the microbiota composition (e.g., alteration of gut integrity, community profiling or metabolite production, inflammation) [56]. Such changes in gut microbiome composition have been recently shown to contribute, in rodents, to diet-induced anxiety- and depression-like behaviour as well as cognitive impairments, especially in hippocampus-dependent tasks [57]. Thus, understanding the roles of the gut microbiota in emotionality, cognition and reward may hold exciting prospects for the treatment of obesity and associated comorbidities (e.g., SUD).

7.2.3. Lack of Studies on the Sensitivity to Drugs of Abuse and Dopaminergic Neurotransmission

Surprisingly, the impact of HFD on psychostimulant-related behaviours has been very scarcely studied, despite the substantial evidence that obesity and SUD share common biological substrates [18]. Only a few rodent studies have investigated the HFD-induced alterations in addiction-relevant behaviours and the associated cellular and molecular mechanisms in reward-related areas (see Table 1 and Supplementary Table S1). Consequently, much remains to be investigated with respect to the behavioural evidence and underlying mechanisms of the interaction between HFD and psychostimulant drugs. Importantly, both the schedule of HFD consumption and the social housing conditions (single- versus group-housed animals) seem to modulate the outcomes of psychostimulant-related behaviours. A recent study suggested that the post-weaning housing condition could modulate the susceptibility to HFD-induced body weight gain, adiposity and thermoregulation in male mice [165], highlighting the need to consider the housing density in the interpretation of the behavioural and metabolic data.
Interestingly, some neuroadaptations produced by HFD treatment parallel those observed with cocaine (see summary in Supplementary Table S2). For example, cocaine exposure produced slight changes in glucose and phospholipid metabolism as well as neurotransmission and increased oxidative stress in the NAc [166,167]. In addition, similar to observations made with HFD treatment [144], cocaine exposure increased oxidative stress and reduced mitochondrial activity in isolated synaptosomes from the whole brain [168]. Moreover, it produced significant changes in the gene expression pattern of electron transport chain complexes in the cingulate cortex, which connects the prefrontal cortex and the limbic system [169,170]. Because the mitochondrial DNA (mtDNA) copy number was reported to be increased in the prefrontal cortex and hippocampus of rats with a history of cocaine self-administration [170], perhaps the variation in mtDNA compensated a decreased number of mitochondria or mitochondrial synthesis in order to maintain a normal level of mitochondrial transcription to provide ATP [171]. A history of cocaine self-administration induced neuroplasticity in the NAc through alterations in the pattern of genes important in several mitochondrial functions (e.g., mitochondrial transcription and replication, mitochondrial dynamics and energy production) [166,172,173]. The possible mechanisms underlying the changes in molecular correlates of mitochondrial dysfunction by cocaine have yet to be determined, but they may occur in response to the massive DA release due to cocaine binding to transporter sites of monoamines, as well as from the oxidative potential of cocaine metabolites [49].
Importantly, cocaine oppositely modulated the accumbal mitochondrial morphology and the mRNA expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Pgc1α), a master regulator of mitochondrial biogenesis, and dynamin-1 like (Drp1), a regulator of mitochondrial division, in dopamine receptor D1-expressing medium spiny neurons (D1-MSNs) and dopamine receptor D2-expressing MSNs (D2-MSNs) [166,172,173]. Because cocaine has been reported to produce contrasting effects on dendritic spine density, excitatory plasticity, signalling processes and transcriptional activity in D1-MSNs and D2-MSNs [174,175], further work is needed to understand the importance of mitochondria in cocaine behavioural plasticity in specific neuronal subtypes.

7.2.4. Lack of Studies on Sex Differences

Apart from the studies investigating the effects of a maternal HFD treatment in dams and/or their offspring, male rodents have typically been used to examine the behavioural and molecular effects of HFD treatment. Thus, only very few studies have examined the effects of HFD treatment in female rodents. Therefore, sex differences were not explicitly addressed in this review. One study found an increased anxiety-related behaviour in HFD-fed female mice [176], but the experimental design lacked a direct comparison between the sexes. Another study found no signs of molecular alterations in the hypothalamus of HFD-fed female and male rats [148]. Interestingly, Rodenas-Gonzalez and colleagues observed that certain patterns of HFD treatment blocked the reinstatement of cocaine-induced CPP in male mice but not in female mice, suggesting that female animals might be less sensitive to the protective effects of HFD treatment [110]. Given the global gender disparities in stress-related disorders [177] as well as obesity and metabolic diseases [178,179,180], more studies in female rodents are imperative to determine the possible sex differences in response to dietary intervention.

7.2.5. Remaining Challenges in Translating Rodent High-Fat Diet Treatments to Human Obesity

Finally, it is worth mentioning that we have reviewed preclinical studies that used a forced exposure of rodents to HFD treatment. While such dietary manipulation allows studying the behavioural, physiological and neural responses to unhealthy diets, this deviates from the human situation where individuals can make food choice. In addition, it does not mimic the importance of the high caloric intake in the form of fluids in humans. Studies that examined how HFD composition, pattern and form (i.e., solid/liquid) affects behaviour and associated brain function and plasticity are lacking. Available studies show that rodents given free choice to consume a high-energy diet typically show hyperphagia, snacking behaviour and increased food-motivated behaviour, leading to accelerated behavioural and metabolic alterations similar to those observed in human obesity [181,182,183,184,185,186,187]. Interestingly, strong sex differences in food choice were observed in adult mice that were exposed to an early life stress paradigm (i.e., limited nesting and bedding material) combined with acute stress exposure in adulthood [188]. The preference for fat in female mice was accompanied by sex differences in the physiological stress response as well the brain circuits regulating food intake and reward [188]. Given the importance of the issue of sex differences in stress responses, further research is warranted and clearly needed to determine the sex-specific motivational aspects of HFD feeding and the underlying precise mechanistic bases.

8. Conclusions

In conclusion, our in-depth analysis of the published literature revealed that high-energy diets had a pronounced impact on emotionality, cognition and reward-related behaviour as well as underlying brain processes that depended on the developmental period. Because most of the studies reviewed involved only male rodents (apart from those examining the effects of a maternal HFD treatment in dams and/or their offspring), future research should extend these observations to female rodents. This would help to elucidate the sex-specific mechanisms linking stress and obesity. Profound knowledge about the molecular mechanisms linking stress and obesity is crucially needed in translational research using rodent models and can significantly advance the important search for risk-biomarkers and the development of clinical intervention strategies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23147952/s1.

Author Contributions

Conceptualization, V.R.; writing—original draft preparation, V.R.; writing—review and editing, D.Z., C.T. and V.R. All authors have read and agreed to the published version of the manuscript.

Funding

We acknowledge the support by Deutsche Forschungsgemeinschaft (DFG) and Open Access Publishing Fund of Osnabrueck University. The university had no further role in the writing of the paper and in the decision to submit the article for publication.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Lars Wilmes (University College Cork, Ireland) and Neele Meyer (University of Osnabrück, Germany) for their insightful comments on this review paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ng, M.; Fleming, T.; Robinson, M.; Thomson, B.; Graetz, N.; Margono, C.; Mullany, E.C.; Biryukov, S.; Abbafati, C.; Abera, S.F.; et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet 2014, 384, 766–781. [Google Scholar] [CrossRef]
  2. Drewnowski, A.; Kurth, C.; Holden-Wiltse, J.; Saari, J. Food preferences in human obesity: Carbohydrates versus fats. Appetite 1992, 18, 207–221. [Google Scholar] [CrossRef]
  3. Hebebrand, J.; Albayrak, Ö.; Adan, R.; Antel, J.; Dieguez, C.; de Jong, J.; Leng, G.; Menzies, J.; Mercer, J.G.; Murphy, M.; et al. “Eating addiction”, rather than “food addiction”, better captures addictive-like eating behavior. Neurosci. Biobehav. Rev. 2014, 47, 295–306. [Google Scholar] [CrossRef] [PubMed]
  4. Corsica, J.A.; Pelchat, M.L. Food addiction: True or false? Curr. Opin. Gastroenterol. 2010, 26, 165–169. [Google Scholar] [CrossRef]
  5. Pi-Sunyer, X. The Medical Risks of Obesity. Postgrad. Med. 2009, 121, 21–33. [Google Scholar] [CrossRef]
  6. Bray, G.A.; Frühbeck, G.; Ryan, D.H.; Wilding, J. Management of obesity. Lancet 2016, 387, 1947–1956. [Google Scholar] [CrossRef]
  7. Kim, G.W.; Lin, J.E.; Blomain, E.S.; Waldman, S.A. Antiobesity Pharmacotherapy: New Drugs and Emerging Targets. Clin. Pharmacol. Ther. 2014, 95, 53–66. [Google Scholar] [CrossRef]
  8. GBD 2015 Obesity Collaborators; Afshin, A.; Forouzanfar, M.H.; Reitsma, M.B.; Sur, P.; Estep, K.; Lee, A.; Marczak, L.; Mokdad, A.H.; Moradi-Lakeh, M.; et al. Health Effects of Overweight and Obesity in 195 Countries over 25 Years. N. Engl. J. Med. 2017, 377, 13–27. [Google Scholar] [CrossRef]
  9. Di Angelantonio, E.; Bhupathiraju, S.N.; Wormser, D.; Gao, P.; Kaptoge, S.; de Gonzalez, A.B.; Cairns, B.J.; Huxley, R.; Jackson, C.L.; Joshy, G.; et al. Body-mass index and all-cause mortality: Individual-participant-data meta-analysis of 239 prospective studies in four continents. Lancet 2016, 388, 776–786. [Google Scholar] [CrossRef]
  10. Citrome, L. Binge eating disorder revisited: What’s new, what’s different, what’s next. CNS Spectr. 2019, 24, 4–13. [Google Scholar] [CrossRef]
  11. Gariepy, G.; Nitka, D.; Schmitz, N. The association between obesity and anxiety disorders in the population: A systematic review and meta-analysis. Int. J. Obes. 2010, 34, 407–419. [Google Scholar] [CrossRef] [PubMed]
  12. Petry, N.M.; Barry, D.; Pietrzak, R.H.; Wagner, J.A. Overweight and Obesity are Associated with Psychiatric Disorders: Results From the National Epidemiologic Survey on Alcohol and Related Conditions. Psychosom. Med. 2008, 70, 288–297. [Google Scholar] [CrossRef]
  13. Preiss, K.; Brennan, L.; Clarke, D. A systematic review of variables associated with the relationship between obesity and depression. Obes. Rev. 2013, 14, 906–918. [Google Scholar] [CrossRef] [PubMed]
  14. Simon, G.E.; Von Korff, M.; Saunders, K.; Miglioretti, D.L.; Crane, P.; Van Belle, G.; Kessler, R.C. Association Between Obesity and Psychiatric Disorders in the US Adult Population. Arch. Gen. Psychiatry 2006, 63, 824–830. [Google Scholar] [CrossRef] [PubMed]
  15. Jane-Llopis, E.; Matytsina, I. Mental health and alcohol, drugs and tobacco: A review of the comorbidity between mental disorders and the use of alcohol, tobacco and illicit drugs. Drug Alcohol Rev. 2006, 25, 515–536. [Google Scholar] [CrossRef] [PubMed]
  16. Onaemo, V.N.; Fawehinmi, T.O.; D’Arcy, C. Comorbid Cannabis Use Disorder with Major Depression and Generalized Anxiety Disorder: A Systematic Review with Meta-analysis of Nationally Representative Epidemiological Surveys. J. Affect. Disord. 2021, 281, 467–475. [Google Scholar] [CrossRef] [PubMed]
  17. Rappeneau, V.; Bérod, A. Reconsidering depression as a risk factor for substance use disorder: Insights from rodent models. Neurosci. Biobehav. Rev. 2017, 77, 303–316. [Google Scholar] [CrossRef]
  18. Rapaka, R.; Schnur, P.; Shurtleff, D. Obesity and addiction: Common neurological mechanisms and drug development. Physiol. Behav. 2008, 95, 2–9. [Google Scholar] [CrossRef]
  19. Volkow, N.D.; Wang, G.-J.; Tomasi, D.; Baler, R.D. Obesity and addiction: Neurobiological overlaps. Obes. Rev. 2013, 14, 2–18. [Google Scholar] [CrossRef]
  20. Miñarro, J.; Rodríguez-Arias, M. Common Neural Mechanisms of Palatable Food Intake and Drug Abuse: Knowledge Obtained with Animal Models. Curr. Pharm. Des. 2020, 26, 2372–2384. [Google Scholar] [CrossRef]
  21. DiLeone, R.J.; Taylor, J.R.; Picciotto, M.R. The drive to eat: Comparisons and distinctions between mechanisms of food reward and drug addiction. Nat. Neurosci. 2012, 15, 1330–1335. [Google Scholar] [CrossRef] [PubMed]
  22. Volkow, N.D.; Wang, G.J.; Fowler, J.S.; Tomasi, D.; Baler, R. Food and Drug Reward: Overlapping Circuits in Human Obesity and Addiction. Brain Imaging Behav. Neurosci. 2012, 11, 1–24. [Google Scholar] [CrossRef]
  23. McTeague, L.M.; Rosenberg, B.M.; Lopez, J.W.; Carreon, D.M.; Huemer, J.; Jiang, Y.; Chick, C.F.; Eickhoff, S.B.; Etkin, A. Identification of Common Neural Circuit Disruptions in Emotional Processing Across Psychiatric Disorders. Am. J. Psychiatry 2020, 177, 411–421. [Google Scholar] [CrossRef] [PubMed]
  24. Sweeney, P.; Yang, Y. Neural Circuit Mechanisms Underlying Emotional Regulation of Homeostatic Feeding. Trends Endocrinol. Metab. 2017, 28, 437–448. [Google Scholar] [CrossRef] [PubMed]
  25. Trinko, R.; Sears, R.M.; Guarnieri, D.J.; DiLeone, R.J. Neural mechanisms underlying obesity and drug addiction. Physiol. Behav. 2007, 91, 499–505. [Google Scholar] [CrossRef]
  26. Hyman, S.E.; Malenka, R.C.; Nestler, E.J. Neural mechanisms of addiction: The Role of Reward-Related Learning and Memory. Annu. Rev. Neurosci. 2006, 29, 565–598. [Google Scholar] [CrossRef]
  27. Nestler, E.J. The Neurobiology of Cocaine Addiction. Sci. Pr. Perspect. 2005, 3, 4–10. [Google Scholar] [CrossRef]
  28. Macedo, I.C.; de Freitas, J.S.; Torres, I.L.D.S. The Influence of Palatable Diets in Reward System Activation: A Mini Review. Adv. Pharmacol. Sci. 2016, 2016, 7238679. [Google Scholar] [CrossRef]
  29. Carr, K.D. Augmentation of drug reward by chronic food restriction: Behavioral evidence and underlying mechanisms. Physiol. Behav. 2002, 76, 353–364. [Google Scholar] [CrossRef]
  30. Deroche-Gamonet, V.; Piazza, P.V.; Casolini, P.; Le Moal, M.; Simon, H. Sensitization to the psychomotor effects of amphetamine and morphine induced by food restriction depends on corticosterone secretion. Brain Res. 1993, 611, 352–356. [Google Scholar] [CrossRef]
  31. Stamp, J.A.; Mashoodh, R.; van Kampen, J.M.; Robertson, H.A. Food restriction enhances peak corticosterone levels, cocaine-induced locomotor activity, and ΔFosB expression in the nucleus accumbens of the rat. Brain Res. 2008, 1204, 94–101. [Google Scholar] [CrossRef] [PubMed]
  32. Sulzer, D.; Cragg, S.J.; Rice, M.E. Striatal dopamine neurotransmission: Regulation of release and uptake. Basal Ganglia 2016, 6, 123–148. [Google Scholar] [CrossRef] [PubMed]
  33. Wanat, M.J.; Willuhn, I.; Clark, J.J.; Phillips, P. Phasic Dopamine Release in Appetitive Behaviors and Drug Addiction. Curr. Drug Abus. Rev. 2009, 2, 195–213. [Google Scholar] [CrossRef]
  34. Wang, G.-J.; Volkow, N.D.; Logan, J.; Pappas, N.R.; Wong, C.T.; Zhu, W.; Netusll, N.; Fowler, J.S. Brain dopamine and obesity. Lancet 2001, 357, 354–357. [Google Scholar] [CrossRef]
  35. Volkow, N.D.; Wang, G.-J.; Telang, F.; Fowler, J.S.; Thanos, P.K.; Logan, J.; Alexoff, D.; Ding, Y.-S.; Wong, C.; Ma, Y.; et al. Low dopamine striatal D2 receptors are associated with prefrontal metabolism in obese subjects: Possible contributing factors. NeuroImage 2008, 42, 1537–1543. [Google Scholar] [CrossRef]
  36. Volkow, N.D.; Wang, G.-J.; Fowler, J.S.; Telang, F. Overlapping neuronal circuits in addiction and obesity: Evidence of systems pathology. Philos. Trans. R. Soc. B Biol. Sci. 2008, 363, 3191–3200. [Google Scholar] [CrossRef] [PubMed]
  37. Stice, E.; Spoor, S.; Bohon, C.; Veldhuizen, M.G.; Small, D.M. Relation of reward from food intake and anticipated food intake to obesity: A functional magnetic resonance imaging study. J. Abnorm. Psychol. 2008, 117, 924–935. [Google Scholar] [CrossRef]
  38. Nummenmaa, L.; Hirvonen, J.; Hannukainen, J.; Immonen, H.; Lindroos, M.M.; Salminen, P.; Nuutila, P. Dorsal Striatum and Its Limbic Connectivity Mediate Abnormal Anticipatory Reward Processing in Obesity. PLoS ONE 2012, 7, e31089. [Google Scholar] [CrossRef]
  39. Stice, E.; Figlewicz, D.P.; Gosnell, B.A.; Levine, A.S.; Pratt, W.E. The contribution of brain reward circuits to the obesity epidemic. Neurosci. Biobehav. Rev. 2013, 37, 2047–2058. [Google Scholar] [CrossRef]
  40. Colon-Perez, L.; Montesinos, J.; Monsivais, M. The future of neuroimaging and gut-brain axis research for substance use disorders. Brain Res. 2022, 1781, 147835. [Google Scholar] [CrossRef]
  41. Gupta, A.; Osadchiy, V.; Mayer, E.A. Brain–gut–microbiome interactions in obesity and food addiction. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 655–672. [Google Scholar] [CrossRef] [PubMed]
  42. de Araujo, I.E.; Schatzker, M.; Small, D.M. Rethinking Food Reward. Annu. Rev. Psychol. 2020, 71, 139–164. [Google Scholar] [CrossRef] [PubMed]
  43. O’Brien, C.J.; Haberman, E.R.; Domingos, A.I. A Tale of Three Systems: Toward a Neuroimmunoendocrine Model of Obesity. Annu. Rev. Cell Dev. Biol. 2021, 37, 549–573. [Google Scholar] [CrossRef]
  44. Tobore, T.O. Towards a comprehensive theory of obesity and a healthy diet: The causal role of oxidative stress in food addiction and obesity. Behav. Brain Res. 2020, 384, 112560. [Google Scholar] [CrossRef] [PubMed]
  45. Bournat, J.C.; Brown, C.W. Mitochondrial dysfunction in obesity. Curr. Opin. Endocrinol. Diabetes Obes. 2010, 17, 446–452. [Google Scholar] [CrossRef]
  46. Fernández-Sánchez, A.; Madrigal-Santillán, E.; Bautista, M.; Esquivel-Soto, J.; Morales-González, Á.; Esquivel-Chirino, C.; Durante-Montiel, I.; Sánchez-Rivera, G.; Valadez-Vega, C.; Morales-González, J.A. Inflammation, Oxidative Stress, and Obesity. Int. J. Mol. Sci. 2011, 12, 3117–3132. [Google Scholar] [CrossRef]
  47. Furukawa, S.; Fujita, T.; Shimabukuro, M.; Iwaki, M.; Yamada, Y.; Nakajima, Y.; Nakayama, O.; Makishima, M.; Matsuda, M.; Shimomura, I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J. Clin. Investig. 2004, 114, 1752–1761. [Google Scholar] [CrossRef]
  48. Savini, I.; Gasperi, V.; Catani, M.V. Oxidative Stress and Obesity. In Obesity: A Practical Guide; Springer International Publishing: Cham, Switzerland, 2016; pp. 65–86. [Google Scholar]
  49. Beiser, T.; Yaka, R. The Role of Oxidative Stress in Cocaine Addiction. J. Neurol. Neuromed. 2019, 4, 17–21. [Google Scholar] [CrossRef]
  50. Jîtcă, G.; Ősz, B.; Tero-Vescan, A.; Vari, C. Psychoactive Drugs—From Chemical Structure to Oxidative Stress Related to Dopaminergic Neurotransmission. A Review. Antioxidants 2020, 10, 381. [Google Scholar] [CrossRef]
  51. Olguín, H.J.; Guzmán, D.C.; García, E.H.; Mejía, G.B. The Role of Dopamine and Its Dysfunction as a Consequence of Oxidative Stress. Oxidative Med. Cell. Longev. 2016, 2016, 9730467. [Google Scholar] [CrossRef]
  52. Kovacic, P. Unifying mechanism for addiction and toxicity of abused drugs with application to dopamine and glutamate mediators: Electron transfer and reactive oxygen species. Med. Hypotheses 2005, 65, 90–96. [Google Scholar] [CrossRef] [PubMed]
  53. Thornton, C.; Grad, E.; Yaka, R. The role of mitochondria in cocaine addiction. Biochem. J. 2021, 478, 749–764. [Google Scholar] [CrossRef] [PubMed]
  54. Chen, C.; Xu, X.; Yan, Y. Estimated global overweight and obesity burden in pregnant women based on panel data model. PLoS ONE 2018, 13, e0202183. [Google Scholar] [CrossRef]
  55. Gawlińska, K.; Gawliński, D.; Filip, M.; Przegaliński, E. Relationship of maternal high-fat diet during pregnancy and lactation to offspring health. Nutr. Rev. 2020, 79, 709–725. [Google Scholar] [CrossRef] [PubMed]
  56. Beam, A.; Clinger, E.; Hao, L. Effect of Diet and Dietary Components on the Composition of the Gut Microbiota. Nutrients 2021, 13, 2795. [Google Scholar] [CrossRef] [PubMed]
  57. Leigh, S.-J.; Morris, M.J. Diet, inflammation and the gut microbiome: Mechanisms for obesity-associated cognitive impairment. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2020, 1866, 165767. [Google Scholar] [CrossRef] [PubMed]
  58. Gómez-Zorita, S.; Aguirre, L.; Milton-Laskibar, I.; Fernández-Quintela, A.; Trepiana, J.; Kajarabille, N.; Mosqueda-Solís, A.; González, M.; Portillo, M.P. Relationship between Changes in Microbiota and Liver Steatosis Induced by High-Fat Feeding—A Review of Rodent Models. Nutrients 2019, 11, 2156. [Google Scholar] [CrossRef]
  59. Mokkala, K.; Houttu, N.; Cansev, T.; Laitinen, K. Interactions of dietary fat with the gut microbiota: Evaluation of mechanisms and metabolic consequences. Clin. Nutr. 2020, 39, 994–1018. [Google Scholar] [CrossRef]
  60. Ingle, D.J. A Simple Means of Producing Obesity in the Rat. Exp. Biol. Med. 1949, 72, 604–605. [Google Scholar] [CrossRef]
  61. Buettner, R.; Schölmerich, J.; Bollheimer, L.C. High-fat Diets: Modeling the Metabolic Disorders of Human Obesity in Rodents. Obesity 2007, 15, 798–808. [Google Scholar] [CrossRef]
  62. Hariri, N.; Thibault, L. High-fat diet-induced obesity in animal models. Nutr. Res. Rev. 2010, 23, 270–299. [Google Scholar] [CrossRef] [PubMed]
  63. Pandit, R.; Mercer, J.G.; Overduin, J.; la Fleur, S.E.; Adan, R.A. Dietary Factors Affect Food Reward and Motivation to Eat. Obes. Facts 2012, 5, 221–242. [Google Scholar] [CrossRef] [PubMed]
  64. Preguiça, I.; Alves, A.; Nunes, S.; Fernandes, R.; Gomes, P.; Viana, S.D.; Reis, F. Diet-induced rodent models of obesity-related metabolic disorders—A guide to a translational perspective. Obes. Rev. 2020, 21, e13081. [Google Scholar] [CrossRef] [PubMed]
  65. Rosini, T.C.; da Silva, A.S.R.; de Moraes, C. Diet-Induced Obesity: Rodent Model for the Study of Obesity-Related Disorders. Rev. Assoc. Med. Bras. 2012, 58, 383–387. [Google Scholar]
  66. Abbott, K.N.; Arnott, C.K.; Westbrook, R.F.; Tran, D.M. The effect of high fat, high sugar, and combined high fat-high sugar diets on spatial learning and memory in rodents: A meta-analysis. Neurosci. Biobehav. Rev. 2019, 107, 399–421. [Google Scholar] [CrossRef]
  67. Cordner, Z.A.; Tamashiro, K.L. Effects of high-fat diet exposure on learning & memory. Physiol. Behav. 2015, 152, 363–371. [Google Scholar] [CrossRef]
  68. Freeman, L.R.; Haley-Zitlin, V.; Rosenberger, D.S.; Granholm, A.-C. Damaging effects of a high-fat diet to the brain and cognition: A review of proposed mechanisms. Nutr. Neurosci. 2014, 17, 241–251. [Google Scholar] [CrossRef]
  69. Liang, Y.; Zou, L.; Tian, Y.; Zhou, S.; Chen, X.; Lin, C. Dietary and metabolic risk of neuropsychiatric disorders: Insights from animal models. Br. J. Nutr. 2021, 126, 1771–1787. [Google Scholar] [CrossRef]
  70. Morris, M.J.; Beilharz, J.E.; Maniam, J.; Reichelt, A.C.; Westbrook, R.F. Why is obesity such a problem in the 21st century? The intersection of palatable food, cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. 2015, 58, 36–45. [Google Scholar] [CrossRef]
  71. Murphy, M.; Mercer, J.G. Diet-Regulated Anxiety. Int. J. Endocrinol. 2013, 2013, 701967. [Google Scholar] [CrossRef]
  72. Sarangi, M.; Dus, M. Crème de la Créature: Dietary Influences on Behavior in Animal Models. Front. Behav. Neurosci. 2021, 15. [Google Scholar] [CrossRef]
  73. Archer, J. Tests for emotionality in rats and mice: A review. Anim. Behav. 1973, 21, 205–235. [Google Scholar] [CrossRef]
  74. Speakman, J.R. Use of high-fat diets to study rodent obesity as a model of human obesity. Int. J. Obes. 2019, 43, 1491–1492. [Google Scholar] [CrossRef] [PubMed]
  75. Fats and Fatty Acids in Human Nutrition. Report of an Expert Consultation. FAO Food Nutr. Pap. 2010, 91, 1–166. [Google Scholar]
  76. Trumbo, P.; Schlicker, S.; Yates, A.A.; Poos, M.; Food and Nutrition Board of the Institute of Medicine, The National Academies. 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]
  77. Ritchie, H.; Roser, M. Diet Compositions. Available online: https://ourworldindata.org/diet-compositions?fbclid=IwAR3k-_TpvIw2zZUdZiBB3xZJHsgjfJTM0rtIrptoMSwxcTNtoajIaMnJkmw (accessed on 15 July 2022).
  78. Eilander, A.; Harika, R.K.; Zock, P.L. Intake and sources of dietary fatty acids in Europe: Are current population intakes of fats aligned with dietary recommendations? Eur. J. Lipid Sci. Technol. 2015, 117, 1370–1377. [Google Scholar] [CrossRef] [PubMed]
  79. Bellisario, V.; Panetta, P.; Balsevich, G.; Baumann, V.; Noble, J.; Raggi, C.; Nathan, O.; Berry, A.; Seckl, J.; Schmidt, M.; et al. Maternal high-fat diet acts as a stressor increasing maternal glucocorticoids’ signaling to the fetus and disrupting maternal behavior and brain activation in C57BL/6J mice. Psychoneuroendocrinology 2015, 60, 138–150. [Google Scholar] [CrossRef] [PubMed]
  80. Baptissart, M.; Lamb, H.E.; To, K.; Bradish, C.; Tehrani, J.; Reif, D.; Cowley, M. Neonatal mice exposed to a high-fat diet in utero influence the behaviour of their nursing dam. Proc. R. Soc. B Boil. Sci. 2018, 285, 20181237. [Google Scholar] [CrossRef]
  81. Connor, K.L.; Vickers, M.H.; Beltrand, J.; Meaney, M.J.; Sloboda, D.M. Nature, nurture or nutrition? Impact of maternal nutrition on maternal care, offspring development and reproductive function. J. Physiol. 2012, 590, 2167–2180. [Google Scholar] [CrossRef]
  82. Leuthardt, A.S.; Bayer, J.; Rodríguez, J.M.M.; Boyle, C.N. Influence of High Energy Diet and Polygenic Predisposition for Obesity on Postpartum Health in Rat Dams. Front. Physiol. 2021, 12, 772707. [Google Scholar] [CrossRef]
  83. Rodriguez, J.; Rodríguez-González, G.; Reyes-Castro, L.; Ibáñez, C.; Ramírez, A.; Chavira, R.; Larrea, F.; Nathanielsz, P.; Zambrano, E. Maternal obesity in the rat programs male offspring exploratory, learning and motivation behavior: Prevention by dietary intervention pre-gestation or in gestation. Int. J. Dev. Neurosci. 2012, 30, 75–81. [Google Scholar] [CrossRef] [PubMed]
  84. Paradis, J.; Boureau, P.; Moyon, T.; Nicklaus, S.; Parnet, P.; Paillé, V. Perinatal Western Diet Consumption Leads to Profound Plasticity and GABAergic Phenotype Changes within Hypothalamus and Reward Pathway from Birth to Sexual Maturity in Rat. Front. Endocrinol. 2017, 8, 216. [Google Scholar] [CrossRef]
  85. Cunha, F.D.S.; Molle, R.D.; Portella, A.K.; Benetti, C.D.S.; Noschang, C.; Goldani, M.Z.; Silveira, P.P. Both Food Restriction and High-Fat Diet during Gestation Induce Low Birth Weight and Altered Physical Activity in Adult Rat Offspring: The “Similarities in the Inequalities” Model. PLoS ONE 2015, 10, e0118586. [Google Scholar] [CrossRef] [PubMed]
  86. Gawlińska, K.; Gawliński, D.; Korostyński, M.; Borczyk, M.; Frankowska, M.; Piechota, M.; Filip, M.; Przegaliński, E. Maternal dietary patterns are associated with susceptibility to a depressive-like phenotype in rat offspring. Dev. Cogn. Neurosci. 2021, 47, 100879. [Google Scholar] [CrossRef] [PubMed]
  87. Naef, L.; Srivastava, L.; Gratton, A.; Hendrickson, H.; Owens, S.M.; Walker, C.-D. Maternal high fat diet during the perinatal period alters mesocorticolimbic dopamine in the adult rat offspring: Reduction in the behavioral responses to repeated amphetamine administration. Psychopharmacology 2008, 197, 83–94. [Google Scholar] [CrossRef]
  88. 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]
  89. Lépinay, A.L.; Larrieu, T.; Joffre, C.; Acar, N.; Gárate, I.; Castanon, N.; Ferreira, G.; Langelier, B.; Guesnet, P.; Brétillon, L.; et al. Perinatal high-fat diet increases hippocampal vulnerability to the adverse effects of subsequent high-fat feeding. Psychoneuroendocrinology 2015, 53, 82–93. [Google Scholar] [CrossRef]
  90. Niu, X.; Wu, X.; Ying, A.; Shao, B.; Li, X.; Zhang, W.; Lin, C.; Lin, Y. Maternal high fat diet programs hypothalamic-pituitary-adrenal function in adult rat offspring. Psychoneuroendocrinology 2019, 102, 128–138. [Google Scholar] [CrossRef]
  91. Schneider, M. Adolescence as a vulnerable period to alter rodent behavior. Cell Tissue Res. 2013, 354, 99–106. [Google Scholar] [CrossRef]
  92. Aslani, S.; Vieira, N.; Marques, F.; Costa, P.; Sousa, N.; Palha, J.A. The effect of high-fat diet on rat’s mood, feeding behavior and response to stress. Transl. Psychiatry 2015, 5, e684. [Google Scholar] [CrossRef]
  93. Blanco-Gandía, M.C.; Miñarro, J.; Rodríguez-Arias, M. Behavioral profile of intermittent vs. continuous access to a high fat diet during adolescence. Behav. Brain Res. 2019, 368, 111891. [Google Scholar] [CrossRef] [PubMed]
  94. Busquets, O.; Ettcheto, M.; Eritja, À.; Espinosa-Jiménez, T.; Verdaguer, E.; Olloquequi, J.; Beas-Zarate, C.; Castro-Torres, R.D.; Casadesús, G.; Auladell, C.; et al. c-Jun N-terminal Kinase 1 ablation protects against metabolic-induced hippocampal cognitive impairments. J. Mol. Med. 2019, 97, 1723–1733. [Google Scholar] [CrossRef] [PubMed]
  95. Del Rio, D.; Morales, L.; Ruiz-Gayo, M.; Del Olmo, N. Effect of high-fat diets on mood and learning performance in adolescent mice. Behav. Brain Res. 2016, 311, 167–172. [Google Scholar] [CrossRef] [PubMed]
  96. Valladolid-Acebes, I.; Stucchi, P.; Cano, V.; Fernández-Alfonso, M.; Merino, B.; Gil-Ortega, M.; Fole, A.; Morales, L.; Ruiz-Gayo, M.; Del Olmo, N. High-fat diets impair spatial learning in the radial-arm maze in mice. Neurobiol. Learn. Mem. 2011, 95, 80–85. [Google Scholar] [CrossRef]
  97. Isgor, C.; Kabbaj, M.; Akil, H.; Watson, S.J. Delayed effects of chronic variable stress during peripubertal-juvenile period on hippocampal morphology and on cognitive and stress axis functions in rats. Hippocampus 2004, 14, 636–648. [Google Scholar] [CrossRef]
  98. Tzanoulinou, S.; Gantelet, E.; Sandi, C.; Márquez, C. Programming effects of peripubertal stress on spatial learning. Neurobiol. Stress 2020, 13, 100282. [Google Scholar] [CrossRef]
  99. Blanco-Gandia, M.; Aracil-Fernández, A.; Romero, S.M.; Aguilar, M.; Manzanares, J.; Miñarro, J.; Rodríguez-Arias, M. Changes in gene expression and sensitivity of cocaine reward produced by a continuous fat diet. Psychopharmacology 2017, 234, 2337–2352. [Google Scholar] [CrossRef]
  100. de Noronha, S.R.; Campos, G.V.; Abreu, A.R.; de Souza, A.A.; Chianca, D.A.; de Menezes, R.C. High fat diet induced-obesity facilitates anxiety-like behaviors due to GABAergic impairment within the dorsomedial hypothalamus in rats. Behav. Brain Res. 2017, 316, 38–46. [Google Scholar] [CrossRef]
  101. Del Rosario, A.; McDermott, M.M.; Panee, J. Effects of a high-fat diet and bamboo extract supplement on anxiety- and depression-like neurobehaviours in mice. Br. J. Nutr. 2012, 108, 1143–1149. [Google Scholar] [CrossRef]
  102. Finger, B.; Dinan, T.; Cryan, J. High-fat diet selectively protects against the effects of chronic social stress in the mouse. Neuroscience 2011, 192, 351–360. [Google Scholar] [CrossRef]
  103. Noronha, S.; Lima, P.; Campos, G.; Chírico, M.; Abreu, A.; Figueiredo, A.; Silva, F.; Chianca, D.; Lowry, C.; De Menezes, R. Association of high-fat diet with neuroinflammation, anxiety-like defensive behavioral responses, and altered thermoregulatory responses in male rats. Brain Behav. Immun. 2019, 80, 500–511. [Google Scholar] [CrossRef] [PubMed]
  104. White, K.A.; Hutton, S.R.; Weimer, J.M.; Sheridan, P.A. Diet-induced obesity prolongs neuroinflammation and recruits CCR2 + monocytes to the brain following herpes simplex virus (HSV)-1 latency in mice. Brain Behav. Immun. 2016, 57, 68–78. [Google Scholar] [CrossRef] [PubMed]
  105. Głombik, K.; Detka, J.; Góralska, J.; Kurek, A.; Solnica, B.; Budziszewska, B. Brain Metabolic Alterations in Rats Showing Depression-Like and Obesity Phenotypes. Neurotox. Res. 2020, 37, 406–424. [Google Scholar] [CrossRef] [PubMed]
  106. Loebens, M.; Barros, H. Diet influences cocaine withdrawal behaviors in the forced swimming test. Pharmacol. Biochem. Behav. 2003, 74, 259–267. [Google Scholar] [CrossRef]
  107. Blanco-Gandia, M.; Cantacorps, L.; Aracil-Fernández, A.; Romero, S.M.; Aguilar, M.; Manzanares, J.; Valverde, O.; Miñarro, J.; Rodríguez-Arias, M. Effects of bingeing on fat during adolescence on the reinforcing effects of cocaine in adult male mice. Neuropharmacology 2017, 113, 31–44. [Google Scholar] [CrossRef]
  108. Hryhorczuk, C.; Florea, M.; Rodaros, D.; Poirier, I.; Daneault, C.; Rosiers, C.D.; Arvanitogiannis, A.; Alquier, T.; Fulton, S. Dampened Mesolimbic Dopamine Function and Signaling by Saturated but not Monounsaturated Dietary Lipids. Neuropsychopharmacology 2016, 41, 811–821. [Google Scholar] [CrossRef]
  109. Morales, L.; Del Olmo, N.; Valladolid-Acebes, I.; Fole, A.; Cano, V.; Merino, B.; Stucchi, P.; Ruggieri, D.; López, L.; Alguacil, L.F.; et al. Shift of Circadian Feeding Pattern by High-Fat Diets Is Coincident with Reward Deficits in Obese Mice. PLoS ONE 2012, 7, e36139. [Google Scholar] [CrossRef]
  110. Ródenas-González, F.; Blanco-Gandía, M.D.C.; Pascual, M.; Molari, I.; Guerri, C.; López, J.M.; Rodríguez-Arias, M. A limited and intermittent access to a high-fat diet modulates the effects of cocaine-induced reinstatement in the conditioned place preference in male and female mice. Psychopharmacology 2021, 238, 2091–2103. [Google Scholar] [CrossRef]
  111. Blanco-Gandia, M.C.; Montagud-Romero, S.; Rodríguez-Arias, M. Binge eating and psychostimulant addiction. World J. Psychiatry 2021, 11, 517–529. [Google Scholar] [CrossRef]
  112. Blanco-Gandía, M.C.; Montagud-Romero, S.; Aguilar, M.A.; Miñarro, J.; Rodríguez-Arias, M. Housing conditions modulate the reinforcing properties of cocaine in adolescent mice that binge on fat. Physiol. Behav. 2018, 183, 18–26. [Google Scholar] [CrossRef]
  113. Naneix, F.; Tantot, F.; Glangetas, C.; Kaufling, J.; Janthakhin, Y.; Boitard, C.; De Smedt-Peyrusse, V.; Pape, J.R.; Vancassel, S.; Trifilieff, P.; et al. Impact of Early Consumption of High-Fat Diet on the Mesolimbic Dopaminergic System. ENeuro 2017, 4, 4. [Google Scholar] [CrossRef] [PubMed]
  114. Ganji, A.; Salehi, I.; Sarihi, A.; Shahidi, S.; Komaki, A. Effects of Hypericum Scabrum extract on anxiety and oxidative stress biomarkers in rats fed a long-term high-fat diet. Metab. Brain Dis. 2017, 32, 503–511. [Google Scholar] [CrossRef] [PubMed]
  115. De Carvalho, L.M.; Gonçalves, J.L.; Pedersen, A.S.B.; Damasceno, S.; Júnior, R.E.M.; Maioli, T.U.; de Faria, A.M.C.; Godard, A.L.B. High-fat diet withdrawal modifies alcohol preference and transcription of dopaminergic and GABAergic receptors. J. Neurogenet. 2019, 33, 10–20. [Google Scholar] [CrossRef] [PubMed]
  116. Takase, K.; Tsuneoka, Y.; Oda, S.; Kuroda, M.; Funato, H. High-fat diet feeding alters olfactory-, social-, and reward-related behaviors of mice independent of obesity. Obesity 2016, 24, 886–894. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, H.; Lv, W.; Pan, Q.; Kalavagunta, P.K.; Liu, Q.; Qin, G.; Cai, M.; Zhou, L.; Wang, T.; Xia, Z.; et al. Simvastatin therapy in adolescent mice attenuates HFD-induced depression-like behavior by reducing hippocampal neuroinflammation. J. Affect. Disord. 2019, 243, 83–95. [Google Scholar] [CrossRef] [PubMed]
  118. Zemdegs, J.; Quesseveur, G.; Jarriault, D.; Pénicaud, L.; Fioramonti, X.; Guiard, B.P. High-fat diet-induced metabolic disorders impairs 5-HT function and anxiety-like behavior in mice. J. Cereb. Blood Flow Metab. 2016, 173, 2095–2110. [Google Scholar] [CrossRef]
  119. Arnold, S.E.; Lucki, I.; Brookshire, B.R.; Carlson, G.C.; Browne, C.A.; Kazi, H.; Bang, S.; Choi, B.-R.; Chen, Y.; McMullen, M.F.; et al. High fat diet produces brain insulin resistance, synaptodendritic abnormalities and altered behavior in mice. Neurobiol. Dis. 2014, 67, 79–87. [Google Scholar] [CrossRef]
  120. Finger, B.C.; Dinan, T.G.; Cryan, J.F. The temporal impact of chronic intermittent psychosocial stress on high-fat diet-induced alterations in body weight. Psychoneuroendocrinology 2012, 37, 729–741. [Google Scholar] [CrossRef]
  121. Li, L.; Wang, Z.; Zuo, Z. Chronic Intermittent Fasting Improves Cognitive Functions and Brain Structures in Mice. PLoS ONE 2013, 8, e66069. [Google Scholar] [CrossRef]
  122. Mielke, J.G.; Nicolitch, K.; Avellaneda, V.; Earlam, K.; Ahuja, T.; Mealing, G.; Messier, C. Longitudinal study of the effects of a high-fat diet on glucose regulation, hippocampal function, and cerebral insulin sensitivity in C57BL/6 mice. Behav. Brain Res. 2006, 175, 374–382. [Google Scholar] [CrossRef]
  123. Novick, A.M.; Levandowski, M.L.; Laumann, L.E.; Philip, N.S.; Price, L.H.; Tyrka, A.R. The effects of early life stress on reward processing. J. Psychiatr. Res. 2018, 101, 80–103. [Google Scholar] [CrossRef] [PubMed]
  124. Schroeder, A.; Notaras, M.; Du, X.; Hill, R.A. On the Developmental Timing of Stress: Delineating Sex-Specific Effects of Stress across Development on Adult Behavior. Brain Sci. 2018, 8, 121. [Google Scholar] [CrossRef] [PubMed]
  125. Hersey, M.; Woodruff, J.L.; Maxwell, N.; Sadek, A.T.; Bykalo, M.K.; Bain, I.; Grillo, C.A.; Piroli, G.G.; Hashemi, P.; Reagan, L.P. High-fat diet induces neuroinflammation and reduces the serotonergic response to escitalopram in the hippocampus of obese rats. Brain Behav. Immun. 2021, 96, 63–72. [Google Scholar] [CrossRef] [PubMed]
  126. Duffy, C.; Hofmeister, J.; Nixon, J.; Butterick, T. High fat diet increases cognitive decline and neuroinflammation in a model of orexin loss. Neurobiol. Learn. Mem. 2019, 157, 41–47. [Google Scholar] [CrossRef]
  127. Lizarbe, B.; Soares, A.F.; Larsson, S.; Duarte, J.M.N.; Lizarbe, B.; Soares, A.F.; Larsson, S.; Duarte, J.M.N. Neurochemical Modifications in the Hippocampus, Cortex and Hypothalamus of Mice Exposed to Long-Term High-Fat Diet. Front. Neurosci. 2018, 12, 985. [Google Scholar] [CrossRef]
  128. Erhardt, E.; Zibetti, L.; Godinho, J.; Bacchieri, B.; Barros, H. Behavioral changes induced by cocaine in mice are modified by a hyperlipidic diet or recombinant leptin. Braz. J. Med Biol. Res. 2006, 39, 1625–1635. [Google Scholar] [CrossRef]
  129. 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]
  130. Casey, B.; Jones, R.M.; Levita, L.; Libby, V.; Pattwell, S.S.; Ruberry, E.J.; Soliman, F.; Somerville, L.H. The storm and stress of adolescence: Insights from human imaging and mouse genetics. Dev. Psychobiol. 2010, 52, 225–235. [Google Scholar] [CrossRef]
  131. Romeo, R.D.; Patel, R.; Pham, L.; So, V.M. Adolescence and the ontogeny of the hormonal stress response in male and female rats and mice. Neurosci. Biobehav. Rev. 2016, 70, 206–216. [Google Scholar] [CrossRef]
  132. Spear, L. The adolescent brain and age-related behavioral manifestations. Neurosci. Biobehav. Rev. 2000, 24, 417–463. [Google Scholar] [CrossRef]
  133. Brydges, N.M. Pre-pubertal stress and brain development in rodents. Curr. Opin. Behav. Sci. 2016, 7, 8–14. [Google Scholar] [CrossRef]
  134. Burke, A.R.; McCormick, C.M.; Pellis, S.M.; Lukkes, J.L. Impact of adolescent social experiences on behavior and neural circuits implicated in mental illnesses. Neurosci. Biobehav. Rev. 2017, 76, 280–300. [Google Scholar] [CrossRef] [PubMed]
  135. McCormick, C.; Green, M. From the stressed adolescent to the anxious and depressed adult: Investigations in rodent models. Neuroscience 2013, 249, 242–257. [Google Scholar] [CrossRef] [PubMed]
  136. Green, M.R.; McCormick, C.M. Effects of stressors in adolescence on learning and memory in rodent models. Horm. Behav. 2013, 64, 364–379. [Google Scholar] [CrossRef] [PubMed]
  137. McCormick, C.M.; Mathews, I.Z.; Thomas, C.; Waters, P. Investigations of HPA function and the enduring consequences of stressors in adolescence in animal models. Brain Cogn. 2010, 72, 73–85. [Google Scholar] [CrossRef]
  138. Eiland, L.; Romeo, R. Stress and the developing adolescent brain. Neuroscience 2013, 249, 162–171. [Google Scholar] [CrossRef]
  139. Romeo, R.D. The impact of stress on the structure of the adolescent brain: Implications for adolescent mental health. Brain Res. 2017, 1654, 185–191. [Google Scholar] [CrossRef]
  140. Tottenham, N.; Galván, A. Stress and the Adolescent Brain: Amygdala-Prefrontal Cortex Circuitry and Ventral Striatum as Developmental Targets. Neurosci. Biobehav. Rev. 2016, 70, 217–227. [Google Scholar] [CrossRef]
  141. Andersen, S.L. Trajectories of brain development: Point of vulnerability or window of opportunity? Neurosci. Biobehav. Rev. 2003, 27, 3–18. [Google Scholar] [CrossRef]
  142. Paus, T.; Keshavan, M.; Giedd, J.N. Why do many psychiatric disorders emerge during adolescence? Nat. Rev. Neurosci. 2008, 9, 947–957. [Google Scholar] [CrossRef]
  143. Crews, F.; He, J.; Hodge, C. Adolescent cortical development: A critical period of vulnerability for addiction. Pharmacol. Biochem. Behav. 2007, 86, 189–199. [Google Scholar] [CrossRef] [PubMed]
  144. Cavaliere, G.; Trinchese, G.; Penna, E.; Cimmino, F.; Pirozzi, C.; Lama, A.; Annunziata, C.; Catapano, A.; Mattace Raso, G.; Meli, R.; et al. High-Fat Diet Induces Neuroinflammation and Mitochondrial Impairment in Mice Cerebral Cortex and Synaptic Fraction. Front. Cell. Neurosci. 2019, 13, 509. [Google Scholar] [CrossRef] [PubMed]
  145. Janthakhin, Y.; Rincel, M.; Costa, A.M.; Darnaudéry, M.; Ferreira, G. Maternal high-fat diet leads to hippocampal and amygdala dendritic remodeling in adult male offspring. Psychoneuroendocrinology 2017, 83, 49–57. [Google Scholar] [CrossRef]
  146. De Souza, C.T.; Araujo, E.P.; Bordin, S.; Ashimine, R.; Zollner, R.D.L.; Boschero, A.C.; Saad, M.J.A.; Velloso, L.A. Consumption of a Fat-Rich Diet Activates a Proinflammatory Response and Induces Insulin Resistance in the Hypothalamus. Endocrinology 2005, 146, 4192–4199. [Google Scholar] [CrossRef] [PubMed]
  147. Terrien, J.; Seugnet, I.; Seffou, B.; Herrero, M.J.; Bowers, J.; Chamas, L.; Decherf, S.; Duvernois-Berthet, E.; Djediat, C.; Ducos, B.; et al. Reduced central and peripheral inflammatory responses and increased mitochondrial activity contribute to diet-induced obesity resistance in WSB/EiJ mice. Sci. Rep. 2019, 9, 19696. [Google Scholar] [CrossRef] [PubMed]
  148. Toniazzo, A.P.; Arcego, D.M.; Lazzaretti, C.; Mota, C.; Schnorr, C.E.; Pettenuzzo, L.F.; Krolow, R.; Moreira, J.C.F.; Dalmaz, C. Sex-dependent effect on mitochondrial and oxidative stress parameters in the hypothalamus induced by prepubertal stress and access to high fat diet. Neurochem. Int. 2019, 124, 114–122. [Google Scholar] [CrossRef]
  149. Lorenzo, P.I.; Vazquez, E.M.; López-Noriega, L.; Fuente-Martín, E.; Mellado-Gil, J.M.; Franco, J.M.; Cobo-Vuilleumier, N.; Martínez, J.A.G.; Romero-Zerbo, S.Y.; Perez-Cabello, J.A.; et al. The metabesity factor HMG20A potentiates astrocyte survival and reactive astrogliosis preserving neuronal integrity. Theranostics 2021, 11, 6983–7004. [Google Scholar] [CrossRef]
  150. Thaler, J.P.; Yi, C.-X.; Schur, E.A.; Guyenet, S.J.; Hwang, B.H.; Dietrich, M.; Zhao, X.; Sarruf, D.A.; Izgur, V.; Maravilla, K.R.; et al. Obesity is associated with hypothalamic injury in rodents and humans. J. Clin. Investig. 2012, 122, 153–162. [Google Scholar] [CrossRef]
  151. Koob, G.F.; Volkow, N.D. Neurocircuitry of Addiction. Neuropsychopharmacology 2010, 35, 217–238. [Google Scholar] [CrossRef]
  152. Kalyan-Masih, P.; Vega-Torres, J.D.; Miles, C.; Haddad, E.; Rainsbury, S.; Baghchechi, M.; Obenaus, A.; Figueroa, J.D. Western High-Fat Diet Consumption during Adolescence Increases Susceptibility to Traumatic Stress while Selectively Disrupting Hippocampal and Ventricular Volumes. Eneuro 2016, 3. [Google Scholar] [CrossRef]
  153. Sial, O.K.; Gnecco, T.; Cardona-Acosta, A.M.; Vieregg, E.; Cardoso, E.A.; Parise, L.F.; Bolaños-Guzmán, C.A. Exposure to Vicarious Social Defeat Stress and Western-Style Diets during Adolescence Leads to Physiological Dysregulation, Decreases in Reward Sensitivity, and Reduced Antidepressant Efficacy in Adulthood. Front. Neurosci. 2021, 15, 701919. [Google Scholar] [CrossRef] [PubMed]
  154. Coccurello, R.; Romano, A.; Giacovazzo, G.; Tempesta, B.; Fiore, M.; Giudetti, A.M.; Marrocco, I.; Altieri, F.; Moles, A.; Gaetani, S. Increased intake of energy-dense diet and negative energy balance in a mouse model of chronic psychosocial defeat. Eur. J. Nutr. 2018, 57, 1485–1498. [Google Scholar] [CrossRef]
  155. MacKay, J.; Kent, P.; James, J.; Cayer, C.; Merali, Z. Ability of palatable food consumption to buffer against the short- and long-term behavioral consequences of social defeat exposure during juvenility in rats. Physiol. Behav. 2017, 177, 113–121. [Google Scholar] [CrossRef]
  156. Lutter, M.; Nestler, E.J. Homeostatic and Hedonic Signals Interact in the Regulation of Food Intake. J. Nutr. 2009, 139, 629–632. [Google Scholar] [CrossRef] [PubMed]
  157. Eller, O.C.; Morris, E.M.; Thyfault, J.P.; Christianson, J.A. Early life stress reduces voluntary exercise and its prevention of diet-induced obesity and metabolic dysfunction in mice. Physiol. Behav. 2020, 223, 113000. [Google Scholar] [CrossRef] [PubMed]
  158. Isingrini, E.; Camus, V.; Le Guisquet, A.-M.; Pingaud, M.; Devers, S.; Belzung, C. Association between Repeated Unpredictable Chronic Mild Stress (UCMS) Procedures with a High Fat Diet: A Model of Fluoxetine Resistance in Mice. PLoS ONE 2010, 5, e10404. [Google Scholar] [CrossRef] [PubMed]
  159. McIntyre, R.S.; Park, K.Y.; Law, C.W.Y.; Sultan, F.; Adams, A.; Lourenço, M.T.; Lo, A.K.S.; Soczynska, J.K.; Woldeyohannes, H.; Alsuwaidan, M.; et al. The Association between Conventional Antidepressants and the Metabolic Syndrome: A Review of the Evidence and Clinical Implications. CNS Drugs 2010, 24, 741–753. [Google Scholar] [CrossRef]
  160. Woo, Y.S.; Seo, H.-J.; McIntyre, R.S.; Bahk, W.-M. Obesity and Its Potential Effects on Antidepressant Treatment Outcomes in Patients with Depressive Disorders: A Literature Review. Int. J. Mol. Sci. 2016, 17, 80. [Google Scholar] [CrossRef]
  161. Ditzen, C.; Jastorff, A.M.; Kessler, M.S.; Bunck, M.; Teplytska, L.; Erhardt, A.; Krömer, S.A.; Varadarajulu, J.; Targosz, B.-S.; Sayan-Ayata, E.F.; et al. Protein Biomarkers in a Mouse Model of Extremes in Trait Anxiety. Mol. Cell. Proteom. 2006, 5, 1914–1920. [Google Scholar] [CrossRef]
  162. Kullmann, S.; Kleinridders, A.; Small, D.M.; Fritsche, A.; Häring, H.-U.; Preissl, H.; Heni, M. Central nervous pathways of insulin action in the control of metabolism and food intake. Lancet Diabetes Endocrinol. 2020, 8, 524–534. [Google Scholar] [CrossRef]
  163. Morrison, C.D. Leptin signaling in brain: A link between nutrition and cognition? Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2009, 1792, 401–408. [Google Scholar] [CrossRef] [PubMed]
  164. D’Asti, E.; Long, H.; Tremblay-Mercier, J.; Grajzer, M.; Cunnane, S.C.; Di Marzo, V.; Walker, C.-D. Maternal Dietary Fat Determines Metabolic Profile and the Magnitude of Endocannabinoid Inhibition of the Stress Response in Neonatal Rat Offspring. Endocrinology 2010, 151, 1685–1694. [Google Scholar] [CrossRef] [PubMed]
  165. Schipper, L.; van Heijningen, S.; Karapetsas, G.; van der Beek, E.M.; van Dijk, G. Individual housing of male C57BL/6J mice after weaning impairs growth and predisposes for obesity. PLoS ONE 2020, 15, e0225488. [Google Scholar] [CrossRef]
  166. Chandra, R.; Engeln, M.; Schiefer, C.; Patton, M.H.; Martin, J.A.; Werner, C.T.; Riggs, L.M.; Francis, T.C.; McGlincy, M.; Evans, B.; et al. Drp1 Mitochondrial Fission in D1 Neurons Mediates Behavioral and Cellular Plasticity during Early Cocaine Abstinence. Neuron 2017, 96, 1327–1341.e6. [Google Scholar] [CrossRef] [PubMed]
  167. Li, Y.; Yan, G.-Y.; Zhou, J.-Q.; Bu, Q.; Deng, P.-C.; Yang, Y.-Z.; Lv, L.; Deng, Y.; Zhao, J.-X.; Shao, X.; et al. 1H NMR-based metabonomics in brain nucleus accumbens and striatum following repeated cocaine treatment in rats. Neuroscience 2012, 218, 196–205. [Google Scholar] [CrossRef]
  168. Vitcheva, V.; Simeonova, R.; Kondeva-Burdina, M.; Mitcheva, M. Selective Nitric Oxide Synthase Inhibitor 7-Nitroindazole Protects against Cocaine-Induced Oxidative Stress in Rat Brain. Oxidative Med. Cell. Longev. 2015, 2015, 157876. [Google Scholar] [CrossRef]
  169. Dietrich, J.-B.; Poirier, R.; Aunis, M.; Zwiller, J. Cocaine Downregulates the Expression of the Mitochondrial Genome in Rat Brain. Ann. N. Y. Acad. Sci. 2004, 1025, 345–350. [Google Scholar] [CrossRef] [PubMed]
  170. Sadakierska-Chudy, A.; Kotarska, A.; Frankowska, M.; Jastrzębska, J.; Wydra, K.; Miszkiel, J.; Przegaliński, E.; Filip, M. The Alterations in Mitochondrial DNA Copy Number and Nuclear-Encoded Mitochondrial Genes in Rat Brain Structures after Cocaine Self-Administration. Mol. Neurobiol. 2017, 54, 7460–7470. [Google Scholar] [CrossRef]
  171. Castellani, C.A.; Longchamps, R.J.; Sun, J.; Guallar, E.; Arking, D.E. Thinking outside the nucleus: Mitochondrial DNA copy number in health and disease. Mitochondrion 2020, 53, 214–223. [Google Scholar] [CrossRef]
  172. Chandra, R.; Engeln, M.; Francis, T.C.; Konkalmatt, P.; Patel, D.; Lobo, M.K. A Role for Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1α in Nucleus Accumbens Neuron Subtypes in Cocaine Action. Biol. Psychiatry 2017, 81, 564–572. [Google Scholar] [CrossRef] [PubMed]
  173. Cole, S.L.; Chandra, R.; Harris, M.; Patel, I.; Wang, T.; Kim, H.; Jensen, L.; Russo, S.J.; Turecki, G.; Gancarz-Kausch, A.M.; et al. Cocaine-induced neuron subtype mitochondrial dynamics through Egr3 transcriptional regulation. Mol. Brain 2021, 14, 101. [Google Scholar] [CrossRef] [PubMed]
  174. Bobadilla, A.-C.; Heinsbroek, J.A.; Gipson, C.D.; Griffin, W.C.; Fowler, C.D.; Kenny, P.J.; Kalivas, P.W. Corticostriatal plasticity, neuronal ensembles, and regulation of drug-seeking behavior. Prog. Brain. Res. 2017, 235, 93–112. [Google Scholar] [CrossRef] [PubMed]
  175. Kim, J.; Park, B.-H.; Lee, J.H.; Park, S.K.; Kim, J.-H. Cell Type-Specific Alterations in the Nucleus Accumbens by Repeated Exposures to Cocaine. Biol. Psychiatry 2011, 69, 1026–1034. [Google Scholar] [CrossRef] [PubMed]
  176. Griffin, T.M.; Fermor, B.; Huebner, J.L.; Kraus, V.B.; Rodriguiz, R.M.; Wetsel, W.C.; Cao, L.; Setton, L.A.; Guilak, F. Diet-induced obesity differentially regulates behavioral, biomechanical, and molecular risk factors for osteoarthritis in mice. Arthritis Res. Ther. 2010, 12, R130. [Google Scholar] [CrossRef]
  177. Bangasser, D.A.; Valentino, R.J. Sex differences in stress-related psychiatric disorders: Neurobiological perspectives. Front. Neuroendocr. 2014, 35, 303–319. [Google Scholar] [CrossRef]
  178. Kanter, R.; Caballero, B. Global Gender Disparities in Obesity: A Review. Adv. Nutr. Int. Rev. J. 2012, 3, 491–498. [Google Scholar] [CrossRef]
  179. Mauvais-Jarvis, F.; Merz, N.B.; Barnes, P.J.; Brinton, R.D.; Carrero, J.-J.; DeMeo, D.L.; De Vries, G.J.; Epperson, C.N.; Govindan, R.; Klein, S.L.; et al. Sex and gender: Modifiers of health, disease, and medicine. Lancet 2020, 396, 565–582. [Google Scholar] [CrossRef]
  180. Murphy, M.O.; Loria, A.S. Sex-specific effects of stress on metabolic and cardiovascular disease: Are women at higher risk? Am. J. Physiol. Integr. Comp. Physiol. 2017, 313, R1–R9. [Google Scholar] [CrossRef]
  181. Ahmed, H.; Hannan, J.L.; Apolzan, J.; Osikoya, O.; Cushen, S.; Romero, S.A.; Goulopoulou, S. A free-choice high-fat, high-sucrose diet induces hyperphagia, obesity, and cardiovascular dysfunction in female cycling and pregnant rats. Am. J. Physiol. Integr. Comp. Physiol. 2019, 316, R472–R485. [Google Scholar] [CrossRef]
  182. Apolzan, J.W.; Harris, R.B. Differential effects of chow and purified diet on the consumption of sucrose solution and lard and the development of obesity. Physiol. Behav. 2012, 105, 325–331. [Google Scholar] [CrossRef]
  183. Harris, R.B.S.; Apolzan, J.W. Changes in glucose tolerance and leptin responsiveness of rats offered a choice of lard, sucrose, and chow. Am. J. Physiol. Integr. Comp. Physiol. 2012, 302, R1327–R1339. [Google Scholar] [CrossRef] [PubMed]
  184. La Fleur, S.E.; Luijendijk, M.C.M.; Van Rozen, A.J.; Kalsbeek, A.; Adan, R.A.H. A free-choice high-fat high-sugar diet induces glucose intolerance and insulin unresponsiveness to a glucose load not explained by obesity. Int. J. Obes. 2011, 35, 595–604. [Google Scholar] [CrossRef] [PubMed]
  185. La Fleur, S.E.; Vanderschuren, L.; Luijendijk, M.C.; Kloeze, B.M.; Tiesjema, B.; Adan, R.A.H. A reciprocal interaction between food-motivated behavior and diet-induced obesity. Int. J. Obes. 2007, 31, 1286–1294. [Google Scholar] [CrossRef] [PubMed]
  186. Rolls, B.J.; Van Duijvenvoorde, P.; Rowe, E.A. Variety in the diet enhances intake in a meal and contributes to the development of obesity in the rat. Physiol. Behav. 1983, 31, 21–27. [Google Scholar] [CrossRef]
  187. Slomp, M.; Belegri, E.; Blancas-Velazquez, A.S.; Diepenbroek, C.; Eggels, L.; Gumbs, M.C.; Joshi, A.; Koekkoek, L.L.; Lamuadni, K.; Ugur, M.; et al. Stressing the importance of choice: Validity of a preclinical free-choice high-caloric diet paradigm to model behavioural, physiological and molecular adaptations during human diet-induced obesity and metabolic dysfunction. J. Neuroendocr. 2019, 31, e12718. [Google Scholar] [CrossRef]
  188. Ruigrok, S.; Kotah, J.; Kuindersma, J.; Speijer, E.; van Irsen, A.; la Fleur, S.; Korosi, A. Adult food choices depend on sex and exposure to early-life stress: Underlying brain circuitry, adipose tissue adaptations and metabolic responses. Neurobiol. Stress 2021, 15, 100360. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the impact of a high-fat diet (HFD) treatment on behavioural phenotypes (i.e., emotionality, cognition and reward-related behaviour) in rodents, as well as associated brain mechanisms. Treatment with a HFD leads to alterations in energy metabolism (i.e., glucose metabolism, insulin signalling), molecular correlates of mitochondrial functions (e.g., OXPHOS, oxidative stress and neuroinflammation) and neuronal function (i.e., neurotransmission, synaptic plasticity) in a subset of brain areas, depending on the developmental period at which the HFD is administered. Abbreviations: AMY: amygdala; CC: cerebral cortex; HC: hippocampus; HFD: high-fat diet; HYP: hypothalamus; NAc: nucleus accumbens.
Figure 1. Overview of the impact of a high-fat diet (HFD) treatment on behavioural phenotypes (i.e., emotionality, cognition and reward-related behaviour) in rodents, as well as associated brain mechanisms. Treatment with a HFD leads to alterations in energy metabolism (i.e., glucose metabolism, insulin signalling), molecular correlates of mitochondrial functions (e.g., OXPHOS, oxidative stress and neuroinflammation) and neuronal function (i.e., neurotransmission, synaptic plasticity) in a subset of brain areas, depending on the developmental period at which the HFD is administered. Abbreviations: AMY: amygdala; CC: cerebral cortex; HC: hippocampus; HFD: high-fat diet; HYP: hypothalamus; NAc: nucleus accumbens.
Ijms 23 07952 g001
Table 1. Literature overview of the impact of a high-fat diet treatment on emotionality, cognition and reward-related behaviours in rodents, taking into account the developmental period. Description: Studies are organised by the developmental period (i.e., before puberty or early adolescence, during late adolescence or in adulthood) in which the high-fat diet (HFD) treatment was applied. For the effects of a maternal HFD treatment, studies are organised by the developmental period (i.e., before puberty or early adolescence, during late adolescence or in adulthood) in which the offspring were tested. All studies reviewed used male (♂) mice (m) or rats (r), otherwise indicated by the female (♀) symbol. The numbered references refer to the bibliography section.
Table 1. Literature overview of the impact of a high-fat diet treatment on emotionality, cognition and reward-related behaviours in rodents, taking into account the developmental period. Description: Studies are organised by the developmental period (i.e., before puberty or early adolescence, during late adolescence or in adulthood) in which the high-fat diet (HFD) treatment was applied. For the effects of a maternal HFD treatment, studies are organised by the developmental period (i.e., before puberty or early adolescence, during late adolescence or in adulthood) in which the offspring were tested. All studies reviewed used male (♂) mice (m) or rats (r), otherwise indicated by the female (♀) symbol. The numbered references refer to the bibliography section.
Behavioural TestAssessementOutcomes & Ref.

(Increased)

(Decreased)

(Not Changed)
Conclusion
Maternal HFD treatment (pre-partum)
Testing in adult offspring
EPMAnxiety ♂♀ (pooled) [83r]
(not changed)
OFTLocomotion/exploration ♂♀ (pooled) [83r]
(not changed)
OBTLearning & memory ♂♀ (pooled) [83r]
(decreased)
Maternal HFD treatment (pre- and post-partum)
Testing in juvenile offspring
EZMAnxiety ♂♀ [86r]
(not changed)
FSTPassive stress-coping♂♀ [86r]
(increased)
ACLocomotion/exploration ♂♀ [86r, 87r]
(not changed)
RWVolontary exercise♀ [85r]♂ [85r] Δ
(sign. changes)
SPTAnhedonia ♂♀ [86r]
(not changed)
NORTLearning & memory ♂♀ [86r]
(not changed)
ACAMPH locomotion [87r]
(decreased)
ACAMPH sensitization[87r]
Maternal HFD treatment (pre- and post-partum)
Testing in adolescent offspring
ACLocomotion/exploration ♀ [88r]
(not changed)
ACQCOC- self-administration ♀ [88r]
(not changed)
EXT♀ [88r]
RST♀ [88r]
Maternal HFD treatment (pre- and post-partum)
Testing in adult offspring
EZMAnxiety ♂♀ (pooled) [83r]
(not changed)
AC/OFTLocomotion/exploration ♂♀ (pooled) [83r, 145r]
(not changed)
OBTLearning & memory ♂♀ (pooled) [83r]
(inconcl.)
MWM[89r]
Juvenile HFD treatment
EPM/ETM/MBT/
OFT/DaLi
Anxiety[92r, 100r, 101m, 103r, 104m][104m][93m, 94m, 95m, 99m, 102m, 104m]
(inconcl.)
FSTPassive stress-coping[92r][95m, 105r][101m, 102m, 106r]
(inconcl.)
OFT/CALocomotion/exploration [93m, 94m, 95m, 100r, 101m, 102m,103r]
(not changed)
HBT[101m]
FUST/SITAnhedonia [102m]
(not changed)
NORT/RAM/HWM/Learning & memory [93m, 94m, 95m, 96m]
(decreased)
MWM/PAT [92r, 109m]
EXPCOC/AMPH-ind. CPP[112m *$][108r *, 109m, 112m *$]♂♀ [107m *, 108r, 109m, 110m *]Δ
(sign. changes)
EXT ♂♀ [107m *, 110m *, 112m *]
RST[99m][110m *][107m *, 112m *]
ACQ/RSTCOC self-administration[107m *]
(increased)
OFT/ACCOC/AMPH locomotion [99m, 113r]Δ
(sign. changes)
ACAMPH sensitization[113 r][108r]
EPMCOC-ind. anxiety[107m *]
FSTCOC-ind. immobility [106r]
HFD treatment in late adolescence
EPM/EZM/MBT/OFT/
NSF/DaLi
Anxiety[114r, 115m, 118m, 177m] [116m, 120m]
(increased)
FST/TSTPassive stress-coping[117m] [102m, 116m, 118m]
(inconcl.)
OFT/TMLocomotion/exploration [117m, 119m]
(decreased)
SPT/SxB/FUSTAnhedonia[116m] [102m]
(inconcl.)
SIT[116m] [120m]
SRT [116m]
NORT/MWM/BMLearning & memory [116m, 121m, 122m]
(not changed)
OBT[122m]
Adult HFD treatment
OFTAnxiety [126m]
(not changed)
OFT/YMLocomotion/exploration [127m][126m]
(inconcl.)
SPTAnhedonia[125r]
(not changed)
TWATLearning & memory [126m]
(not changed)
OFT/ACCOC/AMPH locomotion[129r][128r] Δ
(sign. changes)
EPMCOC-ind. anxiolysis [128r]
FSTCOC-ind. active coping[128r]
Abbreviations: AC: activity cage; ACQ: acquisition; AMPH: amphetamine; BM: Barnes maze test; COC: cocaine; CPP: conditioned place preference test; DaLi: dark–light box test; EPM: elevated plus-maze test; ETM: elevated T-maze test; EZM: elevated zero-maze test; EXP: expression; EXT: extinction; FST: forced swim test; FUST: female urine sniffing test; HBT: hole board test; HFD: high-fat diet; HWM: Hebb Williams maze test; m: mice; ind.: induced; MBT: marble burying test; MWM: Morris water maze test; NORT: novel object recognition test; NSF: novelty-suppressed feeding test; OBT: operant bar-pressing task; OFT: open-field test; PAT: passive avoidance task; r: rats; RAM: radial arm maze test; RST: reinstatement; RW: running wheel; SIT: social interaction test; SPT: sucrose preference test; SRT: social recognition test; SxB: sexual behaviour; TM: T-maze test; TST: tail suspension test; TWAT: two-way avoidance task; YM: Y-maze test. Symbols: * limited access to a high-fat diet; $: outcomes differing based on the housing conditions.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ziemens, D.; Touma, C.; Rappeneau, V. Neurobiological Mechanisms Modulating Emotionality, Cognition and Reward-Related Behaviour in High-Fat Diet-Fed Rodents. Int. J. Mol. Sci. 2022, 23, 7952. https://doi.org/10.3390/ijms23147952

AMA Style

Ziemens D, Touma C, Rappeneau V. Neurobiological Mechanisms Modulating Emotionality, Cognition and Reward-Related Behaviour in High-Fat Diet-Fed Rodents. International Journal of Molecular Sciences. 2022; 23(14):7952. https://doi.org/10.3390/ijms23147952

Chicago/Turabian Style

Ziemens, Dorothea, Chadi Touma, and Virginie Rappeneau. 2022. "Neurobiological Mechanisms Modulating Emotionality, Cognition and Reward-Related Behaviour in High-Fat Diet-Fed Rodents" International Journal of Molecular Sciences 23, no. 14: 7952. https://doi.org/10.3390/ijms23147952

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