ijms-logo

Journal Browser

Journal Browser

Nutrient-Gene Interactions

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Genetics and Genomics".

Deadline for manuscript submissions: closed (31 May 2021) | Viewed by 31297

Special Issue Editor


E-Mail Website
Guest Editor
Center for Obesity Research and Education (CORE), Robert Gordon University, Aberdeen AB10 7QB, UK
Interests: obesity; non-alcoholic fatty liver disease; cancer; molecular nutrition; natural product; oxidative stress
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The involvement of nutrition and the role of specific nutrients in health maintenance and disease prevention and treatment has been the subject of centuries of debate and research, even more now when obesity has reached epidemic proportions and obesity-associated diseases, such as cancer, non-alcoholic fatty liver disease (NAFLD), type 2 diabetes and cardiovascular disease, are cause of great concern for both physicians and healthcare systems. A major challenge to modern nutrition and to nutritional pharmacology is to understand the basis for differences in nutrient requirements between individuals and ethnic groups, and the role of nutrients as therapeutics in order to achieve optimal health and/or prevention or treatment of disease. This requires an appreciation not only of the traditional links between nutrients and endocrine status, but also of the less well-defined links between nutrition/intake of specific nutrients and gene expression (nutrigenomics).

This Special Issue will focus on research aimed at determining interactions between nutrients and gene expression and exploring, at the functional and mechanistic levels, how nutrients affect gene expression and subsequent cell function and physiology, in the context of obesity and associated diseases. In a broad sense, research that entails understanding how the role of nutrients in health and metabolism are linked to biochemical events that lead to gene expression, and aimed at prevention or treatment of disease, or identification of individuals with particular needs, will be considered for publication.

Dr. Giovanna Bermano
Guest Editor

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. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. 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

  • Nutrigenomics
  • Gene expression
  • Micro nutrients
  • Macro nutrients
  • Obesity
  • Non-alcoholic fatty liver disease
  • Cancer
  • Type 2 diabetes
  • Cardiovascular disease
  • In vitro studies
  • In vivo studies
  • Clinical studies
  • Prevention
  • Treatment

Published Papers (8 papers)

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

Research

Jump to: Review

17 pages, 1496 KiB  
Article
Regulation of the Fructose Transporter Gene Slc2a5 Expression by Glucose in Cultured Microglial Cells
by Tooru M. Mizuno, Pei San Lew and Gursagar Jhanji
Int. J. Mol. Sci. 2021, 22(23), 12668; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222312668 - 23 Nov 2021
Cited by 10 | Viewed by 2791
Abstract
Microglia play a role in the regulation of metabolism and pathogenesis of obesity. Microglial activity is altered in response to changes in diet and the body’s metabolic state. Solute carrier family 2 member 5 (Slc2a5) that encodes glucose transporter 5 (GLUT5) [...] Read more.
Microglia play a role in the regulation of metabolism and pathogenesis of obesity. Microglial activity is altered in response to changes in diet and the body’s metabolic state. Solute carrier family 2 member 5 (Slc2a5) that encodes glucose transporter 5 (GLUT5) is a fructose transporter primarily expressed in microglia within the central nervous system. However, little is known about the nutritional regulation of Slc2a5 expression in microglia and its role in the regulation of metabolism. The present study aimed to address the hypothesis that nutrients affect microglial activity by altering the expression of glucose transporter genes. Murine microglial cell line SIM-A9 cells and primary microglia from mouse brain were exposed to different concentrations of glucose and levels of microglial activation markers and glucose transporter genes were measured. High concentration of glucose increased levels of the immediate-early gene product c-Fos, a marker of cell activation, Slc2a5 mRNA, and pro-inflammatory cytokine genes in microglial cells in a time-dependent manner, while fructose failed to cause these changes. Glucose-induced changes in pro-inflammatory gene expression were partially attenuated in SIM-A9 cells treated with the GLUT5 inhibitor. These findings suggest that an increase in local glucose availability leads to the activation of microglia by controlling their carbohydrate sensing mechanism through both GLUT5-dependent and –independent mechanisms. Full article
(This article belongs to the Special Issue Nutrient-Gene Interactions)
Show Figures

Graphical abstract

16 pages, 1356 KiB  
Article
The Same Metabolic Response to FGF21 Administration in Male and Female Obese Mice Is Accompanied by Sex-Specific Changes in Adipose Tissue Gene Expression
by Elena Makarova, Antonina Kazantseva, Anastasia Dubinina, Tatiana Jakovleva, Natalia Balybina, Konstantin Baranov and Nadezhda Bazhan
Int. J. Mol. Sci. 2021, 22(19), 10561; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221910561 - 29 Sep 2021
Cited by 11 | Viewed by 2277
Abstract
The preference for high-calorie foods depends on sex and contributes to obesity development. Fibroblast growth factor 21 (FGF21) beneficially affects taste preferences and obesity, but its action has mainly been studied in males. The aim of this study was to compare the effects [...] Read more.
The preference for high-calorie foods depends on sex and contributes to obesity development. Fibroblast growth factor 21 (FGF21) beneficially affects taste preferences and obesity, but its action has mainly been studied in males. The aim of this study was to compare the effects of FGF21 on food preferences and glucose and lipid metabolism in C57Bl/6J male and female mice with diet-induced obesity. Mice were injected with FGF21 or vehicle for 7 days. Body weight, choice between standard (SD) and high-fat (HFD) diets, blood parameters, and gene expression in white (WAT) and brown (BAT) adipose tissues, liver, muscles, and the hypothalamus were assessed. Compared to males, females had a greater preference for HFD; less WAT; lower levels of cholesterol, glucose, and insulin; and higher expression of Fgf21, Insr, Ppara, Pgc1, Acca and Accb in the liver and Dio2 in BAT. FGF21 administration decreased adiposity; blood levels of cholesterol, glucose, and insulin; hypothalamic Agrp expression, increased SD intake, decreased HFD intake independently of sex, and increased WAT expression of Pparg, Lpl and Lipe only in females. Thus, FGF21 administration beneficially affected mice of both sexes despite obesity-associated sex differences in metabolic characteristics, and it induced female-specific activation of gene expression in WAT. Full article
(This article belongs to the Special Issue Nutrient-Gene Interactions)
Show Figures

Figure 1

17 pages, 1294 KiB  
Article
Perinatal High-Fat Diet Influences Ozone-Induced Responses on Pulmonary Oxidant Status and the Molecular Control of Mitophagy in Female Rat Offspring
by Sven H. Rouschop, Samantha J. Snow, Urmila P. Kodavanti, Marie-José Drittij, Lou M. Maas, Antoon Opperhuizen, Frederik J. van Schooten, Alexander H. Remels and Roger W. Godschalk
Int. J. Mol. Sci. 2021, 22(14), 7551; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147551 - 14 Jul 2021
Cited by 4 | Viewed by 2237
Abstract
Previous research has shown that a perinatal obesogenic, high-fat diet (HFD) is able to exacerbate ozone-induced adverse effects on lung function, injury, and inflammation in offspring, and it has been suggested that mitochondrial dysfunction is implicated herein. The aim of this study was [...] Read more.
Previous research has shown that a perinatal obesogenic, high-fat diet (HFD) is able to exacerbate ozone-induced adverse effects on lung function, injury, and inflammation in offspring, and it has been suggested that mitochondrial dysfunction is implicated herein. The aim of this study was to investigate whether a perinatal obesogenic HFD affects ozone-induced changes in offspring pulmonary oxidant status and the molecular control of mitochondrial function. For this purpose, female Long-Evans rats were fed a control diet or HFD before and during gestation, and during lactation, after which the offspring were acutely exposed to filtered air or ozone at a young-adult age (forty days). Directly following this exposure, the offspring lungs were examined for markers related to oxidative stress; oxidative phosphorylation; and mitochondrial fusion, fission, biogenesis, and mitophagy. Acute ozone exposure significantly increased pulmonary oxidant status and upregulated the molecular machinery that controls receptor-mediated mitophagy. In female offspring, a perinatal HFD exacerbated these responses, whereas in male offspring, responses were similar for both diet groups. The expression of the genes and proteins involved in oxidative phosphorylation and mitochondrial biogenesis, fusion, and fission was not affected by ozone exposure or perinatal HFD. These findings suggest that a perinatal HFD influences ozone-induced responses on pulmonary oxidant status and the molecular control of mitophagy in female rat offspring. Full article
(This article belongs to the Special Issue Nutrient-Gene Interactions)
Show Figures

Figure 1

17 pages, 5757 KiB  
Article
n-6 High Fat Diet Induces Gut Microbiome Dysbiosis and Colonic Inflammation
by Ornella I. Selmin, Andreas J. Papoutsis, Sabine Hazan, Christopher Smith, Nick Greenfield, Micah G. Donovan, Spencer N. Wren, Thomas C. Doetschman, Justin M. Snider, Ashley J. Snider, Sherry H.-H. Chow and Donato F. Romagnolo
Int. J. Mol. Sci. 2021, 22(13), 6919; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22136919 - 28 Jun 2021
Cited by 25 | Viewed by 5521
Abstract
Background: Concerns are emerging that a high-fat diet rich in n-6 PUFA (n-6HFD) may alter gut microbiome and increase the risk of intestinal disorders. Research is needed to model the relationships between consumption of an n-6HFD starting at [...] Read more.
Background: Concerns are emerging that a high-fat diet rich in n-6 PUFA (n-6HFD) may alter gut microbiome and increase the risk of intestinal disorders. Research is needed to model the relationships between consumption of an n-6HFD starting at weaning and development of gut dysbiosis and colonic inflammation in adulthood. We used a C57BL/6J mouse model to compare the effects of exposure to a typical American Western diet (WD) providing 58.4%, 27.8%, and 13.7% energy (%E) from carbohydrates, fat, and protein, respectively, with those of an isocaloric and isoproteic soybean oil-rich n-6HFD providing 50%E and 35.9%E from total fat and carbohydrates, respectively on gut inflammation and microbiome profile. Methods: At weaning, male offspring were assigned to either the WD or n-6HFD through 10–16 weeks of age. The WD included fat exclusively from palm oil whereas the n-6HFD contained fat exclusively from soybean oil. We recorded changes in body weight, cyclooxygenase-2 (COX-2) expression, colon histopathology, and gut microbiome profile. Results: Compared to the WD, the n-6HFD increased plasma levels of n-6 fatty acids; colonic expression of COX-2; and the number of colonic inflammatory and hyperplastic lesions. At 16 weeks of age, the n-6HFD caused a marked reduction in the gut presence of Firmicutes, Clostridia, and Lachnospiraceae, and induced growth of Bacteroidetes and Deferribacteraceae. At the species level, the n-6HFD sustains the gut growth of proinflammatory Mucispirillum schaedleri and Lactobacillus murinus. Conclusions: An n-6HFD consumed from weaning to adulthood induces a shift in gut bacterial profile associated with colonic inflammation. Full article
(This article belongs to the Special Issue Nutrient-Gene Interactions)
Show Figures

Figure 1

10 pages, 2494 KiB  
Article
Comparative Analysis of Histone H3K4me3 Distribution in Mouse Liver in Different Diets Reveals the Epigenetic Efficacy of Cyanidin-3-O-glucoside Dietary Intake
by Giuseppe Persico, Francesca Casciaro, Alessandra Marinelli, Chiara Tonelli, Katia Petroni and Marco Giorgio
Int. J. Mol. Sci. 2021, 22(12), 6503; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126503 - 17 Jun 2021
Cited by 4 | Viewed by 2040
Abstract
Background: Different diets result in significantly different phenotypes through metabolic and genomic reprogramming. Epigenetic marks, identified in humans and mouse models through caloric restriction, a high-fat diet or the intake of specific bioactives, suggest that genomic reprogramming drives this metabolic reprogramming and mediates [...] Read more.
Background: Different diets result in significantly different phenotypes through metabolic and genomic reprogramming. Epigenetic marks, identified in humans and mouse models through caloric restriction, a high-fat diet or the intake of specific bioactives, suggest that genomic reprogramming drives this metabolic reprogramming and mediates the effect of nutrition on health. Histone modifications encode the epigenetic signal, which adapts genome functions to environmental conditions, including diets, by tuning the structure and properties of chromatin. To date, the effect of different diets on the genome-wide distribution of critical histone marks has not been determined. Methods: Using chromatin immunoprecipitation sequencing, we investigated the distribution of the trimethylation of lysine 4 of histone H3 in the liver of mice fed for one year with five different diets, including: chow containing yellow corn powder as an extra source of plant bioactives or specifically enriched with cyanidin-3-O-Glucoside, high-fat-enriched obesogenic diets, and caloric-restricted pro-longevity diets. Conclusions: Comparison of the resulting histone mark profiles revealed that functional food containing cyanidin determines a broad effect. Full article
(This article belongs to the Special Issue Nutrient-Gene Interactions)
Show Figures

Figure 1

15 pages, 1802 KiB  
Article
Five Days Periodic Fasting Elevates Levels of Longevity Related Christensenella and Sirtuin Expression in Humans
by Stephanie Lilja, Carina Stoll, Ulrike Krammer, Berit Hippe, Kalina Duszka, Tewodros Debebe, Ingrid Höfinger, Jürgen König, Angelika Pointner and Alexander Haslberger
Int. J. Mol. Sci. 2021, 22(5), 2331; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052331 - 26 Feb 2021
Cited by 30 | Viewed by 8158
Abstract
Periodic fasting (PF) is an increasingly popular approach that assists in the management of metabolic and inflammatory diseases as well as in preventing mechanisms involved in aging. However, little is known about the effects of fasting on gut microbiota and its impact on [...] Read more.
Periodic fasting (PF) is an increasingly popular approach that assists in the management of metabolic and inflammatory diseases as well as in preventing mechanisms involved in aging. However, little is known about the effects of fasting on gut microbiota and its impact on the epigenetic regulation of metabolically relevant enzymes, especially sirtuins (SIRTs). We analyzed the effect of periodic fasting on the human gut microbiota, SIRTs expression, and mitochondrial content in 51 males and females. The participants fasted under supervision for five consecutive days following the Buchinger fasting guidelines. Ketogenesis, selected mRNAs, miRNAs, mitochondrial (mt) DNA, and gut composition were analyzed before and after PF. PF triggered a significant switch in metabolism, as indicated by the increase in ß-hydroxybutyrate (BHB) and pyruvate dehydrogenase kinase isoform 4 (PDK4) expression in the capillary blood. MtDNA, SIRT1, SIRT3, and miRlet7b-5p expression in blood cells were elevated, whereas SIRT6 and miR125b-5p were not affected. Following fasting, gut microbiota diversity increased, and a statistically significant correlation between SIRT1 gene expression and the abundance of Prevotella and Lactobacillus was detected. The abundance of longevity related Christensenella species increased after fasting and inversely correlated with age as well as body mass index (BMI). Thus, this represents the first study that showing that fasting not only changes the composition of the gut microbiota, making it more diverse, but also affects SIRT expression in humans. Full article
(This article belongs to the Special Issue Nutrient-Gene Interactions)
Show Figures

Graphical abstract

Review

Jump to: Research

15 pages, 5577 KiB  
Review
The Roles of Carbohydrate Response Element Binding Protein in the Relationship between Carbohydrate Intake and Diseases
by Katsumi Iizuka
Int. J. Mol. Sci. 2021, 22(21), 12058; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222112058 - 08 Nov 2021
Cited by 12 | Viewed by 3807
Abstract
Carbohydrates are macronutrients that serve as energy sources. Many studies have shown that carbohydrate intake is nonlinearly associated with mortality. Moreover, high-fructose corn syrup (HFCS) consumption is positively associated with obesity, cardiovascular disease, and type 2 diabetes mellitus (T2DM). Accordingly, products with equal [...] Read more.
Carbohydrates are macronutrients that serve as energy sources. Many studies have shown that carbohydrate intake is nonlinearly associated with mortality. Moreover, high-fructose corn syrup (HFCS) consumption is positively associated with obesity, cardiovascular disease, and type 2 diabetes mellitus (T2DM). Accordingly, products with equal amounts of glucose and fructose have the worst effects on caloric intake, body weight gain, and glucose intolerance, suggesting that carbohydrate amount, kind, and form determine mortality. Understanding the role of carbohydrate response element binding protein (ChREBP) in glucose and lipid metabolism will be beneficial for elucidating the harmful effects of high-fructose corn syrup (HFCS), as this glucose-activated transcription factor regulates glycolytic and lipogenic gene expression. Glucose and fructose coordinately supply the metabolites necessary for ChREBP activation and de novo lipogenesis. Chrebp overexpression causes fatty liver and lower plasma glucose levels, and ChREBP deletion prevents obesity and fatty liver. Intestinal ChREBP regulates fructose absorption and catabolism, and adipose-specific Chrebp-knockout mice show insulin resistance. ChREBP also regulates the appetite for sweets by controlling fibroblast growth factor 21, which promotes energy expenditure. Thus, ChREBP partly mimics the effects of carbohydrate, especially HFCS. The relationship between carbohydrate intake and diseases partly resembles those between ChREBP activity and diseases. Full article
(This article belongs to the Special Issue Nutrient-Gene Interactions)
Show Figures

Figure 1

13 pages, 777 KiB  
Review
Alterations of Gut Microbiota by Overnutrition Impact Gluconeogenic Gene Expression and Insulin Signaling
by Ling He
Int. J. Mol. Sci. 2021, 22(4), 2121; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22042121 - 20 Feb 2021
Cited by 18 | Viewed by 3295
Abstract
A high-fat, Western-style diet is an important predisposing factor for the onset of type 2 diabetes and obesity. It causes changes in gut microbial profile, reduction of microbial diversity, and the impairment of the intestinal barrier, leading to increased serum lipopolysaccharide (endotoxin) levels. [...] Read more.
A high-fat, Western-style diet is an important predisposing factor for the onset of type 2 diabetes and obesity. It causes changes in gut microbial profile, reduction of microbial diversity, and the impairment of the intestinal barrier, leading to increased serum lipopolysaccharide (endotoxin) levels. Elevated lipopolysaccharide (LPS) induces acetyltransferase P300 both in the nucleus and cytoplasm of liver hepatocytes through the activation of the IRE1-XBP1 pathway in the endoplasmic reticulum stress. In the nucleus, induced P300 acetylates CRTC2 to increase CRTC2 abundance and drives Foxo1 gene expression, resulting in increased expression of the rate-limiting gluconeogenic gene G6pc and Pck1 and abnormal liver glucose production. Furthermore, abnormal cytoplasm-appearing P300 acetylates IRS1 and IRS2 to disrupt insulin signaling, leading to the prevention of nuclear exclusion and degradation of FOXO1 proteins to further exacerbate the expression of G6pc and Pck1 genes and liver glucose production. Inhibition of P300 acetyltransferase activity by chemical inhibitors improved insulin signaling and alleviated hyperglycemia in obese mice. Thus, P300 acetyltransferase activity appears to be a therapeutic target for the treatment of type 2 diabetes and obesity. Full article
(This article belongs to the Special Issue Nutrient-Gene Interactions)
Show Figures

Figure 1

Back to TopTop