ijms-logo

Journal Browser

Journal Browser

Nutrition, Epigenetics, and Diseases 2.0

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

Deadline for manuscript submissions: closed (25 October 2022) | Viewed by 13943

Special Issue Editors


E-Mail
Guest Editor

E-Mail Website
Guest Editor

Special Issue Information

Dear Colleagues,

It has been largely reported that diets/nutrients may modify the epigenetic regulation of gene expression. Moreover, it is now well documented that they may play a key role in the pathogenesis/prevention of non-communicable diseases (NCDs), including cardiovascular diseases, metabolic syndrome, diabetes mellitus type II, and cancer. This is interesting, since the development of NCDs has been related to epigenetic changes, including modifications of the DNA methylation state, histone tail methylation, acetylation, ubiquitination and phosphorylation, as well as non-coding RNA changes. Thus, nutraceuticals may represent a valid prevention strategy for NCDs through epigenetic mechanisms. A very important emerging field in this context is the beneficial role that nutraceuticals may exert in NCD patients showing a higher risk of SARS-CoV-2 infection due to their compromised immune responses. In this case, it has been suggested that nutraceuticals may act through epigenetic modifications to normalize the expression of unregulated inflammatory/cytokine markers and lead to the enhancement of adaptive immune responses.

We invite authors to contribute either original research articles or review articles to this Special Issue aiming to provide a comprehensive overview on the current knowledge about the role played by diet and nutrients as regulators of complex disease-related epigenetic modifications.

Dr. Gabriella Calviello
Dr. Simona Serini
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. 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

  • epigenetics
  • nutrition
  • diet
  • non-communicable diseases
  • cancer
  • neurodegenerative diseases
  • cardiovascular diseases
  • autoimmune diseases
  • metabolic diseases
  • respiratory diseases

Related Special Issue

Published Papers (9 papers)

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

Research

Jump to: Review

10 pages, 1707 KiB  
Article
Histone Acetylation of Immune Regulatory Genes in Human Placenta in Association with Maternal Intake of Olive Oil and Fish Consumption
by Nathalie Acevedo, Paolo Frumento, Hani Harb, Bilal Alashkar Alhamwe, Catharina Johansson, Lisa Eick, Johan Alm, Harald Renz, Annika Scheynius and Daniel P. Potaczek
Int. J. Mol. Sci. 2019, 20(5), 1060; https://doi.org/10.3390/ijms20051060 - 01 Mar 2019
Cited by 42 | Viewed by 4979
Abstract
Maternal diet modifies epigenetic programming in offspring, a potentially critical factor in the immune dysregulation of modern societies. We previously found that prenatal fish oil supplementation affects neonatal T-cell histone acetylation of genes implicated in adaptive immunity including PRKCZ, IL13, and [...] Read more.
Maternal diet modifies epigenetic programming in offspring, a potentially critical factor in the immune dysregulation of modern societies. We previously found that prenatal fish oil supplementation affects neonatal T-cell histone acetylation of genes implicated in adaptive immunity including PRKCZ, IL13, and TBX21. In this study, we measured H3 and H4 histone acetylation levels by chromatin immunoprecipitation in 173 term placentas collected in the prospective birth cohort, ALADDIN, in which information on lifestyle and diet is thoroughly recorded. In anthroposophic families, regular olive oil usage during pregnancy was associated with increased H3 acetylation at FOXP3 (p = 0.004), IL10RA (p = 0.008), and IL7R (p = 0.007) promoters, which remained significant after adjustment by offspring gender. Furthermore, maternal fish consumption was associated with increased H4 acetylation at the CD14 gene in placentas of female offspring (p = 0.009). In conclusion, prenatal olive oil intake can affect placental histone acetylation in immune regulatory genes, confirming previously observed pro-acetylation effects of olive oil polyphenols. The association with fish consumption may implicate ω-3 polyunsaturated fatty acids present in fish oil. Altered histone acetylation in placentas from mothers who regularly include fish or olive oil in their diets could influence immune priming in the newborn. Full article
(This article belongs to the Special Issue Nutrition, Epigenetics, and Diseases)
Show Figures

Figure 1

11 pages, 978 KiB  
Communication
Maternal High Fat Diet-Induced Obesity Modifies Histone Binding and Expression of Oxtr in Offspring Hippocampus in a Sex-Specific Manner
by Kelly A. Glendining and Christine L. Jasoni
Int. J. Mol. Sci. 2019, 20(2), 329; https://doi.org/10.3390/ijms20020329 - 15 Jan 2019
Cited by 31 | Viewed by 5917
Abstract
Maternal obesity during pregnancy increases risk for neurodevelopmental disorders in offspring, although the underlying mechanisms remain unclear. Epigenetic deregulation associates with many neurodevelopmental disorders, and recent evidence indicates that maternal nutritional status can alter chromatin marks in the offspring brain. Thus, maternal obesity [...] Read more.
Maternal obesity during pregnancy increases risk for neurodevelopmental disorders in offspring, although the underlying mechanisms remain unclear. Epigenetic deregulation associates with many neurodevelopmental disorders, and recent evidence indicates that maternal nutritional status can alter chromatin marks in the offspring brain. Thus, maternal obesity may disrupt epigenetic regulation of gene expression during offspring neurodevelopment. Using a C57BL/6 mouse model, we investigated whether maternal high fat diet (mHFD)-induced obesity alters the expression of genes previously implicated in the etiology of neurodevelopmental disorders within the Gestational Day 17.5 (GD 17.5) offspring hippocampus. We found significant two-fold upregulation of oxytocin receptor (Oxtr) mRNA in the hippocampus of male, but not female, GD 17.5 offspring from mHFD-induced obese dams (p < 0.05). To determine whether altered histone binding at the Oxtr gene promoter may underpin these transcriptional changes, we then performed chromatin immunoprecipitation (ChIP). Consistent with the Oxtr transcriptional changes, we observed increased binding of active histone mark H3K9Ac at the Oxtr transcriptional start site (TSS) in the hippocampus of mHFD male (p < 0.05), but not female, offspring. Together, these data indicate an increased vulnerability of male offspring to maternal obesity-induced changes in chromatin remodeling processes that regulate gene expression in the developing hippocampus, and contributes to our understanding of how early life nutrition affects the offspring brain epigenome. Full article
(This article belongs to the Special Issue Nutrition, Epigenetics, and Diseases)
Show Figures

Figure 1

14 pages, 1287 KiB  
Article
Resveratrol Modulates SIRT1 and DNMT Functions and Restores LINE-1 Methylation Levels in ARPE-19 Cells under Oxidative Stress and Inflammation
by Andrea Maugeri, Martina Barchitta, Maria Grazia Mazzone, Francesco Giuliano, Guido Basile and Antonella Agodi
Int. J. Mol. Sci. 2018, 19(7), 2118; https://doi.org/10.3390/ijms19072118 - 20 Jul 2018
Cited by 80 | Viewed by 5811
Abstract
The role of epigenetic alterations in the pathogenesis of retinal degenerative diseases, including age-related macular degeneration (AMD), has been pending so far. Our study investigated the effect of oxidative stress and inflammation on DNA methyltransferases (DNMTs) and Sirtuin 1 (SIRT1) functions, as well [...] Read more.
The role of epigenetic alterations in the pathogenesis of retinal degenerative diseases, including age-related macular degeneration (AMD), has been pending so far. Our study investigated the effect of oxidative stress and inflammation on DNA methyltransferases (DNMTs) and Sirtuin 1 (SIRT1) functions, as well as on long interspersed nuclear element-1 (LINE-1) methylation, in human retinal pigment epithelial (ARPE-19) cells. Therefore, we evaluated whether treatment with resveratrol may modulate DNMT and SIRT1 functions and restore changes in LINE-1 methylation. Cells were treated with 25 mU/mL glucose oxidase (GOx) or 10 µg/mL lipopolysaccharide (LPS) to mimic oxidative or inflammatory conditions, respectively. Oxidative stress decreased DNMT1, DNMT3a, DNMT3b, and SIRT1 expression (p-values < 0.05), as well as total DNMTs (−28.5%; p < 0.0001) and SIRT1 (−29.0%; p < 0.0001) activities. Similarly, inflammatory condition decreased DNMT1 and SIRT1 expression (p-values < 0.05), as well as total DNMTs (−14.9%; p = 0.007) and SIRT1 (−20.1%; p < 0.002) activities. Interestingly, GOx- and LPS-treated cells exhibited lower LINE-1 methylation compared to controls (p-values < 0.001). We also demonstrated that treatment with 10 μM resveratrol for 24 h counteracted the detrimental effect on DNMT and SIRT1 functions, and LINE-1 methylation, in cells under oxidative and inflammatory conditions. However, further studies should explore the perspectives of resveratrol as a suitable strategy for the prevention and/or treatment of retinal degenerative diseases. Full article
(This article belongs to the Special Issue Nutrition, Epigenetics, and Diseases)
Show Figures

Figure 1

Review

Jump to: Research

21 pages, 870 KiB  
Review
An Overview of Epigenetics in Obesity: The Role of Lifestyle and Therapeutic Interventions
by Abeer M. Mahmoud
Int. J. Mol. Sci. 2022, 23(3), 1341; https://doi.org/10.3390/ijms23031341 - 25 Jan 2022
Cited by 43 | Viewed by 13252
Abstract
Obesity has become a global epidemic that has a negative impact on population health and the economy of nations. Genetic predispositions have been demonstrated to have a substantial role in the unbalanced energy metabolism seen in obesity. However, these genetic variations cannot entirely [...] Read more.
Obesity has become a global epidemic that has a negative impact on population health and the economy of nations. Genetic predispositions have been demonstrated to have a substantial role in the unbalanced energy metabolism seen in obesity. However, these genetic variations cannot entirely explain the massive growth in obesity over the last few decades. Accumulating evidence suggests that modern lifestyle characteristics such as the intake of energy-dense foods, adopting sedentary behavior, or exposure to environmental factors such as industrial endocrine disruptors all contribute to the rising obesity epidemic. Recent advances in the study of DNA and its alterations have considerably increased our understanding of the function of epigenetics in regulating energy metabolism and expenditure in obesity and metabolic diseases. These epigenetic modifications influence how DNA is transcribed without altering its sequence. They are dynamic, reflecting the interplay between the body and its surroundings. Notably, these epigenetic changes are reversible, making them appealing targets for therapeutic and corrective interventions. In this review, I discuss how these epigenetic modifications contribute to the disordered energy metabolism in obesity and to what degree lifestyle and weight reduction strategies and pharmacological drugs can restore energy balance by restoring normal epigenetic profiles. Full article
(This article belongs to the Special Issue Nutrition, Epigenetics, and Diseases 2.0)
Show Figures

Figure 1

25 pages, 1554 KiB  
Review
Vitamin D as A Protector of Arterial Health: Potential Role in Peripheral Arterial Disease Formation
by Smriti Murali Krishna
Int. J. Mol. Sci. 2019, 20(19), 4907; https://doi.org/10.3390/ijms20194907 - 03 Oct 2019
Cited by 25 | Viewed by 13061
Abstract
Atherosclerotic occlusive diseases and aneurysms that affect large and medium-sized arteries outside the cardiac and cerebral circulation are collectively known as peripheral arterial disease (PAD). With a rise in the rate of aging population worldwide, the number of people diagnosed with PAD is [...] Read more.
Atherosclerotic occlusive diseases and aneurysms that affect large and medium-sized arteries outside the cardiac and cerebral circulation are collectively known as peripheral arterial disease (PAD). With a rise in the rate of aging population worldwide, the number of people diagnosed with PAD is rapidly increasing. The micronutrient vitamin D is an important steroid hormone that acts on many crucial cellular mechanisms. Experimental studies suggest that optimal levels of vitamin D have beneficial effects on the heart and blood vessels; however, high vitamin D concentrations have been implicated in promoting vascular calcification and arterial stiffness. Observations from various clinical studies shows that deficiency of vitamin D has been associated with a greater risk of PAD. Epidemiological studies have often reported an inverse relation between circulating vitamin D status measured in terms of 25-hydroxivitamin D [25(OH)D] levels and increased cardiovascular disease risk; however, randomized controlled trials did not show a consistent positive effect of vitamin D supplementation on cardiovascular disease risk or events. Even though PAD shares all the major risk factors with cardiovascular diseases, the effect of vitamin D deficiency in PAD is not clear. Current evidence suggests a strong role of vitamin D in promoting genomic and epigenomic changes. This review summarises the current literature that supports the notion that vitamin D deficiency may promote PAD formation. A better understanding of underlying pathological mechanisms will open up new therapeutic possibilities which is the main unmet need in PAD management. Furthermore, epigenetic evidence shows that a more holistic approach towards PAD prevention that incorporates a healthy lifestyle, adequate exercise and optimal nutrition may be more effective in protecting the genome and maintaining a healthy vasculature. Full article
(This article belongs to the Special Issue Nutrition, Epigenetics, and Diseases)
Show Figures

Figure 1

17 pages, 653 KiB  
Review
Nutritional Factors, DNA Methylation, and Risk of Type 2 Diabetes and Obesity: Perspectives and Challenges
by Luca Parrillo, Rosa Spinelli, Antonella Nicolò, Michele Longo, Paola Mirra, Gregory Alexander Raciti, Claudia Miele and Francesco Beguinot
Int. J. Mol. Sci. 2019, 20(12), 2983; https://doi.org/10.3390/ijms20122983 - 19 Jun 2019
Cited by 29 | Viewed by 6622
Abstract
A healthy diet improves life expectancy and helps to prevent common chronic diseases such as type 2 diabetes (T2D) and obesity. The mechanisms driving these effects are not fully understood, but are likely to involve epigenetics. Epigenetic mechanisms control gene expression, maintaining the [...] Read more.
A healthy diet improves life expectancy and helps to prevent common chronic diseases such as type 2 diabetes (T2D) and obesity. The mechanisms driving these effects are not fully understood, but are likely to involve epigenetics. Epigenetic mechanisms control gene expression, maintaining the DNA sequence, and therefore the full genomic information inherited from our parents, unchanged. An interesting feature of epigenetic changes lies in their dynamic nature and reversibility. Accordingly, they are susceptible to correction through targeted interventions. Here we will review the evidence supporting a role for nutritional factors in mediating metabolic disease risk through DNA methylation changes. Special emphasis will be placed on the potential of using DNA methylation traits as biomarkers to predict risk of obesity and T2D as well as on their response to dietary and pharmacological (epi-drug) interventions. Full article
(This article belongs to the Special Issue Nutrition, Epigenetics, and Diseases)
Show Figures

Figure 1

15 pages, 309 KiB  
Review
Epigenetic Effects of n-3 LCPUFAs: A Role in Pediatric Metabolic Syndrome
by Matilde Amatruda, Giulio Ippolito, Sara Vizzuso, Giulia Vizzari, Giuseppe Banderali and Elvira Verduci
Int. J. Mol. Sci. 2019, 20(9), 2118; https://doi.org/10.3390/ijms20092118 - 29 Apr 2019
Cited by 20 | Viewed by 4779
Abstract
Childhood obesity represents an important public health issue worldwide and is strongly linked to metabolic alterations such as hypertension, insulin resistance, and dyslipidemia. The constellation of these conditions is commonly known as Metabolic Syndrome (MetS). Metabolic syndrome is not just a simple cluster [...] Read more.
Childhood obesity represents an important public health issue worldwide and is strongly linked to metabolic alterations such as hypertension, insulin resistance, and dyslipidemia. The constellation of these conditions is commonly known as Metabolic Syndrome (MetS). Metabolic syndrome is not just a simple cluster of metabolic complications due to excess of adipose tissue, but is considered a risk factor for cardiovascular diseases. Evidence from several human and animal studies suggests that environmental and nutritional exposure during pregnancy may affect the newborn development and future health through epigenetic changes, playing a potential role in determining obesity and obesity-related complications. Understanding how nutritional epigenetic mechanisms contribute to the “transgenerational risk” for obesity and metabolic dysfunction is crucial in order to develop early prevention strategies for children’s health. Nutrigenetics is the science that studies the role of nutrients in gene expression. Long Chain Polyunsaturated Fatty Acids (LCPUFAs) are known for their health benefits, especially in relation to their ability to modulate inflammation and improve some obesity-associated comorbidities, mainly by decreasing plasma triglycerides. Recent nutrigenetic research is focusing on the potential role of LCPUFAs in influencing epigenetic markers. In this review, we present the most recent updates about the possible interaction between n-3 LCPUFAs and epigenetic pathways in metabolic syndrome. Literature from MEDLINE® and the Cochrane database between May 2005 and December 2018 has been scanned. Full article
(This article belongs to the Special Issue Nutrition, Epigenetics, and Diseases)
14 pages, 261 KiB  
Review
The Impact of Caloric Restriction on the Epigenetic Signatures of Aging
by Noémie Gensous, Claudio Franceschi, Aurelia Santoro, Maddalena Milazzo, Paolo Garagnani and Maria Giulia Bacalini
Int. J. Mol. Sci. 2019, 20(8), 2022; https://doi.org/10.3390/ijms20082022 - 24 Apr 2019
Cited by 61 | Viewed by 9746
Abstract
Aging is characterized by an extensive remodeling of epigenetic patterns, which has been implicated in the physiopathology of age-related diseases. Nutrition plays a significant role in modulating the epigenome, and a growing amount of data indicate that dietary changes can modify the epigenetic [...] Read more.
Aging is characterized by an extensive remodeling of epigenetic patterns, which has been implicated in the physiopathology of age-related diseases. Nutrition plays a significant role in modulating the epigenome, and a growing amount of data indicate that dietary changes can modify the epigenetic marks associated with aging. In this review, we will assess the current advances in the relationship between caloric restriction, a proven anti-aging intervention, and epigenetic signatures of aging. We will specifically discuss the impact of caloric restriction on epigenetic regulation and how some of the favorable effects of caloric restriction on lifespan and healthspan could be mediated by epigenetic modifications. Full article
(This article belongs to the Special Issue Nutrition, Epigenetics, and Diseases)
15 pages, 897 KiB  
Review
Metabolic Signaling into Chromatin Modifications in the Regulation of Gene Expression
by Tian Gao, Zyanya Díaz-Hirashi and Francisco Verdeguer
Int. J. Mol. Sci. 2018, 19(12), 4108; https://doi.org/10.3390/ijms19124108 - 18 Dec 2018
Cited by 20 | Viewed by 8056
Abstract
The regulation of cellular metabolism is coordinated through a tissue cross-talk by hormonal control. This leads to the establishment of specific transcriptional gene programs which adapt to environmental stimuli. On the other hand, recent advances suggest that metabolic pathways could directly signal into [...] Read more.
The regulation of cellular metabolism is coordinated through a tissue cross-talk by hormonal control. This leads to the establishment of specific transcriptional gene programs which adapt to environmental stimuli. On the other hand, recent advances suggest that metabolic pathways could directly signal into chromatin modifications and impact on specific gene programs. The key metabolites acetyl-CoA or S-adenosyl-methionine (SAM) are examples of important metabolic hubs which play in addition a role in chromatin acetylation and methylation. In this review, we will discuss how intermediary metabolism impacts on transcription regulation and the epigenome with a particular focus in metabolic disorders. Full article
(This article belongs to the Special Issue Nutrition, Epigenetics, and Diseases)
Show Figures

Graphical abstract

Back to TopTop