nutrients-logo

Journal Browser

Journal Browser

Nutrition, Taste, Reward and Bariatric Surgery

A special issue of Nutrients (ISSN 2072-6643). This special issue belongs to the section "Nutrition and Obesity".

Deadline for manuscript submissions: closed (5 April 2024) | Viewed by 7166

Special Issue Editors


E-Mail Website
Guest Editor
Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
Interests: bariatric surgery; animal models of obesity; food reward; taste; addiction

E-Mail Website
Co-Guest Editor
Department of Surgery, Division of Minimally Invasive Surgery/Bariatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
Interests: bariatric surgery; weight management; obesity; minimally invasive surgery
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Bariatric surgery is the most effective and lasting treatment for severe obesity and associated metabolic disorders, but various procedures have differential outcomes with individual differences with respect to durability of weight loss, in addition to potentially conferring increased risk for substance use. The neural and hormonal mechanisms underlying decreased high calorie food intake and altered nutrient and taste preferences with the potential of reward transfer to non-food substances after bariatric surgery remain largely unexplored. Thus, this Special Issue solicits original studies and reviews on the recent progress in the field in order to promote a better understanding of pre-surgical and post-surgical factors contributing to metabolic surgery’s long-term success as well as individual differences in quality-of-life outcomes. We invite submissions from studies in human and animal models with a broad area of focus including but not limited to nutrient preferences, individual taste and reward reactivity, metabolic, gut-and-brain hormonal and neural (e.g., vagal) mechanisms, as well as the contribution of changes in gut microbial–host metabolic crosstalk, and inflammatory processes.   

Prof. Dr. Andras Hajnal
Prof. Dr. Ann M Rogers
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Nutrients is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • bariatric surgery
  • high-calorie diets
  • post-operative nutrition
  • food reward
  • food preferences
  • taste
  • substance use disorders
  • gut–brain signaling
  • gut microbiota
  • gut–neuroimmune system

Published Papers (5 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

17 pages, 3706 KiB  
Article
Binge Eating (BE) and Obesity: Brain Activity and Psychological Measures before and after Roux-En-Y Gastric Bypass (RYGB)
by Shaunte Baboumian, Lauren Puma, Charles Swencionis, Nerys M. Astbury, Jennifer Ho, Spiro P. Pantazatos and Allan Geliebter
Nutrients 2023, 15(17), 3808; https://doi.org/10.3390/nu15173808 - 31 Aug 2023
Viewed by 1040
Abstract
Brain activity in response to food cues following Roux-En-Y Gastric Bypass (RYGB) in binge eating (BE) or non-binge eating (NB) individuals is understudied. Here, 15 RYGB (8 BE; 7 NB) and 13 no treatment (NT) (7 BE; 6 NB) women with obesity underwent [...] Read more.
Brain activity in response to food cues following Roux-En-Y Gastric Bypass (RYGB) in binge eating (BE) or non-binge eating (NB) individuals is understudied. Here, 15 RYGB (8 BE; 7 NB) and 13 no treatment (NT) (7 BE; 6 NB) women with obesity underwent fMRI imaging while viewing high and low energy density food (HEF and LEF, respectively) and non-food (NF) visual cues. A region of interest (ROI) analysis compared BE participants to NB participants in those undergoing RYGB surgery pre-surgery and 4 months post. Results were corrected for multiple comparisons using liberal (p < 0.006 uncorrected) and stringent (p < 0.05 FDR corrected) thresholds. Four months following RYGB (vs. no treatment (NT) control), both BE and NB participants showed greater reductions in blood oxygen level-dependent (BOLD) signals (a proxy of local brain activity) in the dorsomedial prefrontal cortex in response to HEF (vs. LEF) cues (p < 0.006). BE (vs. NB) participants showed greater increases in the precuneus (p < 0.006) and thalamic regions (p < 0.05 corrected) to food (vs. NF). For RYGB (vs. NT) participants, BE participants, but not NB participants, showed lower BOLD signal in the middle occipital gyrus (p < 0.006), whilst NB participants, but not BE participants, showed lower signal in inferior frontal gyrus (p < 0.006) in response to HEF (vs. LEF). Results suggest distinct neural mechanisms of RGYB in BE and may help lead to improved clinical treatments. Full article
(This article belongs to the Special Issue Nutrition, Taste, Reward and Bariatric Surgery)
Show Figures

Figure 1

14 pages, 1020 KiB  
Article
Weight Regain and Ingestive Behavior in Women after Metabolic Surgery
by Jessica G. Nicanor-Carreón, Neda Seyedsadjadi, Blair Rowitz and Marta Yanina Pepino
Nutrients 2023, 15(17), 3670; https://doi.org/10.3390/nu15173670 - 22 Aug 2023
Cited by 1 | Viewed by 1162
Abstract
This study investigated associations between maladaptive ingestive behaviors and weight regain in women who underwent metabolic surgery 2–10 years ago. Using a web-based survey, we assessed emotional, external, and restrained eating (Dutch Eating Behavior Questionnaire—DEBQ), food cravings (Food-Craving Inventory—FCI), and other behaviors (e.g., [...] Read more.
This study investigated associations between maladaptive ingestive behaviors and weight regain in women who underwent metabolic surgery 2–10 years ago. Using a web-based survey, we assessed emotional, external, and restrained eating (Dutch Eating Behavior Questionnaire—DEBQ), food cravings (Food-Craving Inventory—FCI), and other behaviors (e.g., Eating Disorder Examination Questionnaire—EDE-Q; Alcohol Use Disorder Identification Test-Concise—AUDIT-C) in 36 women (42.9 ± 9.5 years old) post-surgery. We found that weight regain was specifically associated with increased frequency of cravings for sweets (r = 0.43), higher global scores in the EDE-Q (r = 0.38), and time elapsed since surgery (r = 0.35; all p’s < 0.04). Multiple regression analysis revealed that the association between weight regain and sweet cravings interacted with time after surgery (p = 0.04), with the strongest association observed in women assessed closer to the surgery (i.e., 2.0–2.8 years). The combination of time after surgery and its interaction with sweet cravings accounted for 31% of the individual variations in weight regain (p = 0.005). Notably, among participants who reported alcohol consumption (31 of 36), 55% had an AUDIT-C score indicating hazardous drinking. These findings highlight the relevance of attending to patients’ reports of frequent sweet cravings and screening for alcohol use to enhance strategies tailored to prevent weight regain and alcohol-related health problems post-surgery. Full article
(This article belongs to the Special Issue Nutrition, Taste, Reward and Bariatric Surgery)
Show Figures

Figure 1

12 pages, 694 KiB  
Article
Associations of Food Addiction Symptomatology and Disordered Eating Behaviors in a Pre-Surgical Bariatric Population
by Melissa Butt, Paddy Ssentongo, Ann M. Rogers and Andrea Rigby
Nutrients 2023, 15(15), 3474; https://doi.org/10.3390/nu15153474 - 06 Aug 2023
Viewed by 1149
Abstract
The construct of food addiction (FA) has been highly debated in recent years particularly in the fields of disordered eating, medical weight management, and bariatric surgery. Some researchers have argued that FA symptoms are distinct, highly prevalent, and present a barrier for patients [...] Read more.
The construct of food addiction (FA) has been highly debated in recent years particularly in the fields of disordered eating, medical weight management, and bariatric surgery. Some researchers have argued that FA symptoms are distinct, highly prevalent, and present a barrier for patients seeking medical treatment for obesity. The purpose of this study is to evaluate the cross-sectional associations between FA symptomatology, binge eating disorder (BED) and other appetitive traits, as well as dietary quality in a sample of adults with obesity seeking bariatric surgery. This post hoc analysis was conducted on a prospectively collected dataset from August 2020 to August 2022 at a single academic medical center. Descriptive statistics were used to characterize the sample. Additional analyses included: correlation coefficients, multivariable linear regression, and analysis of variance. A total of 587 patients were included in this analysis with low average scores for FA symptoms (mean: 1.48; standard deviation (SD): 2.15). Those with no BED symptoms had the lowest average FA symptoms scores (mean: 0.87; SD: 1.52) and those with both bingeing and LOCE had the highest average scores (mean: 3.35; SD: 2.81). This finding supports the hypothesis that, while related, FA and BED may represent different cognitions and behaviors. Full article
(This article belongs to the Special Issue Nutrition, Taste, Reward and Bariatric Surgery)
Show Figures

Figure 1

20 pages, 3215 KiB  
Article
Reduction of Plasma BCAAs following Roux-en-Y Gastric Bypass Surgery Is Primarily Mediated by FGF21
by Harsh Shah, Alyssa Kramer, Caitlyn A. Mullins, Marie Mattern, Ritchel B. Gannaban, R. Leigh Townsend, Shawn R. Campagna, Christopher D. Morrison, Hans-Rudolf Berthoud and Andrew C. Shin
Nutrients 2023, 15(7), 1713; https://doi.org/10.3390/nu15071713 - 31 Mar 2023
Cited by 1 | Viewed by 2012
Abstract
Type 2 diabetes (T2D) is a challenging health concern worldwide. A lifestyle intervention to treat T2D is difficult to adhere, and the effectiveness of approved medications such as metformin, thiazolidinediones (TZDs), and sulfonylureas are suboptimal. On the other hand, bariatric procedures such as [...] Read more.
Type 2 diabetes (T2D) is a challenging health concern worldwide. A lifestyle intervention to treat T2D is difficult to adhere, and the effectiveness of approved medications such as metformin, thiazolidinediones (TZDs), and sulfonylureas are suboptimal. On the other hand, bariatric procedures such as Roux-en-Y gastric bypass (RYGB) are being recognized for their remarkable ability to achieve diabetes remission, although the underlying mechanism is not clear. Recent evidence points to branched-chain amino acids (BCAAs) as a potential contributor to glucose impairment and insulin resistance. RYGB has been shown to effectively lower plasma BCAAs in insulin-resistant or T2D patients that may help improve glycemic control, but the underlying mechanism for BCAA reduction is not understood. Hence, we attempted to explore the mechanism by which RYGB reduces BCAAs. To this end, we randomized diet-induced obese (DIO) mice into three groups that underwent either sham or RYGB surgery or food restriction to match the weight of RYGB mice. We also included regular chow-diet-fed healthy mice as an additional control group. Here, we show that compared to sham surgery, RYGB in DIO mice markedly lowered serum BCAAs most likely by rescuing BCAA breakdown in both liver and white adipose tissues. Importantly, the restored BCAA metabolism following RYGB was independent of caloric intake. Fasting insulin and HOMA-IR were decreased as expected, and serum valine was strongly associated with insulin resistance. While gut hormones such as glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) are postulated to mediate various surgery-induced metabolic benefits, mice lacking these hormonal signals (GLP-1R/Y2R double KO) were still able to effectively lower plasma BCAAs and improve glucose tolerance, similar to mice with intact GLP-1 and PYY signaling. On the other hand, mice deficient in fibroblast growth factor 21 (FGF21), another candidate hormone implicated in enhanced glucoregulatory action following RYGB, failed to decrease plasma BCAAs and normalize hepatic BCAA degradation following surgery. This is the first study using an animal model to successfully recapitulate the RYGB-led reduction of circulating BCAAs observed in humans. Our findings unmasked a critical role of FGF21 in mediating the rescue of BCAA metabolism following surgery. It would be interesting to explore the possibility of whether RYGB-induced improvement in glucose homeostasis is partly through decreased BCAAs. Full article
(This article belongs to the Special Issue Nutrition, Taste, Reward and Bariatric Surgery)
Show Figures

Figure 1

Review

Jump to: Research

35 pages, 2745 KiB  
Review
Influence of Bariatric Surgery on Gut Microbiota Composition and Its Implication on Brain and Peripheral Targets
by Sevag Hamamah, Andras Hajnal and Mihai Covasa
Nutrients 2024, 16(7), 1071; https://doi.org/10.3390/nu16071071 - 05 Apr 2024
Viewed by 1255
Abstract
Obesity remains a significant global health challenge, with bariatric surgery remaining as one of the most effective treatments for severe obesity and its related comorbidities. This review highlights the multifaceted impact of bariatric surgery beyond mere physical restriction or nutrient malabsorption, underscoring the [...] Read more.
Obesity remains a significant global health challenge, with bariatric surgery remaining as one of the most effective treatments for severe obesity and its related comorbidities. This review highlights the multifaceted impact of bariatric surgery beyond mere physical restriction or nutrient malabsorption, underscoring the importance of the gut microbiome and neurohormonal signals in mediating the profound effects on weight loss and behavior modification. The various bariatric surgery procedures, such as Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG), act through distinct mechanisms to alter the gut microbiome, subsequently impacting metabolic health, energy balance, and food reward behaviors. Emerging evidence has shown that bariatric surgery induces profound changes in the composition of the gut microbiome, notably altering the Firmicutes/Bacteroidetes ratio and enhancing populations of beneficial bacteria such as Akkermansia. These microbiota shifts have far-reaching effects beyond gut health, influencing dopamine-mediated reward pathways in the brain and modulating the secretion and action of key gut hormones including ghrelin, leptin, GLP-1, PYY, and CCK. The resultant changes in dopamine signaling and hormone levels contribute to reduced hedonic eating, enhanced satiety, and improved metabolic outcomes. Further, post-bariatric surgical effects on satiation targets are in part mediated by metabolic byproducts of gut microbiota like short-chain fatty acids (SCFAs) and bile acids, which play a pivotal role in modulating metabolism and energy expenditure and reducing obesity-associated inflammation, as well as influencing food reward pathways, potentially contributing to the regulation of body weight and reduction in hedonic eating behaviors. Overall, a better understanding of these mechanisms opens the door to developing non-surgical interventions that replicate the beneficial effects of bariatric surgery on the gut microbiome, dopamine signaling, and gut hormone regulation, offering new avenues for obesity treatment. Full article
(This article belongs to the Special Issue Nutrition, Taste, Reward and Bariatric Surgery)
Show Figures

Figure 1

Back to TopTop