Next Article in Journal
Food Patterns among Chinese Immigrants Living in the South of Spain
Previous Article in Journal
The Impact of Decaffeinated Green Tea Extract on Fat Oxidation, Body Composition and Cardio-Metabolic Health in Overweight, Recreationally Active Individuals
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nutrition and Rheumatoid Arthritis in the ‘Omics’ Era

by
Manuela Cassotta
1,†,
Tamara Y. Forbes-Hernandez
2,†,
Danila Cianciosi
3,
Maria Elexpuru Zabaleta
1,
Sandra Sumalla Cano
1,
Irma Dominguez
1,
Beatriz Bullon
4,
Lucia Regolo
3,
Josè Miguel Alvarez-Suarez
5,6,
Francesca Giampieri
3,7,* and
Maurizio Battino
3,8,*
1
Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain
2
Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain
3
Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, 60131 Ancona, Italy
4
Department of Periodontology, Dental School, University of Sevilla, 41004 Sevilla, Spain
5
AgroScience & Food Research Group, Universidad de Las Américas, Quito 170125, Ecuador
6
King Fahd Medical Research Center, King Abdulaziz University, Jedda 21589, Saudi Arabia
7
Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
8
International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2021, 13(3), 763; https://doi.org/10.3390/nu13030763
Submission received: 7 January 2021 / Revised: 16 February 2021 / Accepted: 24 February 2021 / Published: 26 February 2021

Abstract

:
Modern high-throughput ‘omics’ science tools (including genomics, transcriptomics, proteomics, metabolomics and microbiomics) are currently being applied to nutritional sciences to unravel the fundamental processes of health effects ascribed to particular nutrients in humans and to contribute to more precise nutritional advice. Diet and food components are key environmental factors that interact with the genome, transcriptome, proteome, metabolome and the microbiota, and this life-long interplay defines health and diseases state of the individual. Rheumatoid arthritis (RA) is a chronic autoimmune disease featured by a systemic immune-inflammatory response, in genetically susceptible individuals exposed to environmental triggers, including diet. In recent years increasing evidences suggested that nutritional factors and gut microbiome have a central role in RA risk and progression. The aim of this review is to summarize the main and most recent applications of ‘omics’ technologies in human nutrition and in RA research, examining the possible influences of some nutrients and nutritional patterns on RA pathogenesis, following a nutrigenomics approach. The opportunities and challenges of novel ‘omics technologies’ in the exploration of new avenues in RA and nutritional research to prevent and manage RA will be also discussed.

Graphical Abstract

1. Introduction

Recent advances in high-throughput/high-content techniques have led to a new framework in biomedical research, the so-called ‘Omics era’, which combines the opportunity to gather great amounts of data and details at the molecular level together with the evolution of new computational models and statistical tools that are able to analyze and filter such data. Then, progresses in next generation sequencing (NGS), high-throughput platforms, mass-spectrometry and bioinformatic equipment, enabled the synchronous extensive study of thousands of genes (genomics), epigenetic factors (epigenomics), RNA (transcriptomics), metabolites (metabolomics) proteins (proteomics), and human-microbiota (microbiomics), with the potential of combining diverse categories of ‘omics’ data (‘multi-omics’ or ‘system biology’). Those novel ‘omics’ approaches and techniques have revolutionized the study of complex human diseases, providing an unparalleled genome-wide view of genetic variation, gene expression, interaction with microbiota, and environmentally responsive epigenetic changes [1,2,3]. Moreover, nutritional research has shifted from traditional physiology and epidemiology to molecular genetics and biology. Applications of the above mentioned ‘omics’ facilitated molecular nutrition understanding. Pursuing this course, nutrigenomics has developed as a pluridisciplinary research field in nutrition-science that intends to clarify how nutrition can affect human health. While the first meaning of nutrigenomics concerned only with studies on nutrients or bioactive food compounds influencing gene expression of a person, currently, this definition has been extended and, in recent times, nutrigenomics refers to the employment of transcriptomics, genomics, proteomics, metabolomics and epigenomics to find out and elucidate the existing mutual relations between genes and nutrients at a molecular level, also encompassing nutrigenetic studies exploring the relationships between genetic variants and diet in modulating disease risk [4]. In this review, we use the term “nutrigenomics” in its broadest definition. Rheumatoid arthritis (RA) is a persistent inflammatory autoimmune disorder which affects roughly 1% of the global population. It is characterized by extensive synovitis, systemic inflammation and various degrees of cartilage and bone erosion. Multiple genetic and environmental factors, including modifiable lifestyle factors such as cigarette smoking and dietary habits have been linked with an increased risk for RA. Cumulative evidences suggested that nutrition has a central role in RA risk and progression [5,6]. It is already widely-known that bioactive food components can interact with genes impacting transcription factors, protein expression, metabolite production and microbiota. On the other hand, the genetic makeup of the single person can delineate the nutritional state, metabolic responses, and predisposition to diet-associated health disorders. Research in this field can help us to comprehend why some people respond differently from others to the same foods, beverages and supplements. Recent evidence has shown that the host microbiota, and especially the intestinal microbiota, play a central role in the onset and progression of several diseases, including RA [7,8]. Accelerated progress of high-throughput molecular technologies has allowed to conduct meticulous studies of the microbiota in humans, exploring the biological signatures that are connected with definite illnesses, environmental conditions, or pharmacological/nutritional interventions [9,10,11,12,13]. Nutrition is the main modulator of bacterial composition and abundance in the gastrointestinal tract, suggesting the opportunities for therapeutic nutritional approaches to manipulate microbiota composition and diversity [14,15]. Nutrigenomics and nutritional microbiomics approaches have received increasing attention and are currently being used to study respectively the mutual interactions between food and genes and between diet and microbiota in several diseases including cancer [16,17], cardiovascular [18,19], metabolic [20], and autoimmune diseases [21,22,23,24] in order to enable their better understanding, prevention and treatment through optimization of individuals’ dietary intakes.
Despite important research advancements and clinical improvements, RA still represents a public health challenge, in terms of both epidemiological and economic burden: many patients still experience premature work disability, co-morbidities, and important adverse effects caused by medications. Although with availability of several disease-modifying antirheumatic drugs and biologic therapies the outcomes for patients with RA have significantly improved, RA remains a long-standing condition for which there is currently no effective cure [25]. Extensive use of the ever expanding, novel ‘omics’ technologies will facilitate both the understanding of RA pathogenesis and the identification and modification of nutritional-related risk factors, allowing to establish a prevention strategy for RA in susceptible population as well as to complement the present treatment strategies for a better disease management. The combined-use of several ‘omics’ technologies will also enable the discovery of novel biomarkers related with specific food or dietary intake, greatly facilitating human nutritional studies. After presenting an overview of the most recent genomics, transcriptomics, proteomics, metabolomics, epigenomics, and microbiomics approaches and their implementation within human nutrition research, in this paper we review the role of ‘omics’ technologies in elucidating the pathogenesis of RA and the possible influence of some nutrients, food bioactive compounds, nutritional patterns and microbiota on RA pathogenesis.
The aim of this review is to present the main novel ‘omics’ approaches and their current and possible future application in RA and nutrition research, with the ultimate goal to promote a human-relevant nutrigenomics approach to RA for disease prevention and better disease management. The opportunities and challenges of novel ‘omics technologies’ will be also discussed.
Literature search was conducted using PubMed (between August–November 2020) and different combinations of search terms and Boolean operators (see Supplementary Table S1). Only human-based studies (e.g., observational and interventional studies in RA and inflammation) were reviewed.

2. Applications of ‘Omics’ Approaches and Technologies within Nutritional Research

A crucial goal of nutritional research is to establish the role of nutrition in metabolic control and to boost health. Nutrition and health correlations have been classically explained in terms of energetic and structural necessity of the body, as assured by nutrients. Nevertheless, foods also include several secondary biologically active non-nutrient compounds which can also help in the prevention and even in the treatment of various chronic diseases. In fact many epidemiological studies have shown a correlation between nutrition and the incidence of different disorders, in particular type 2 diabetes [26,27], cardiovascular diseases [28,29,30], cancer [31,32,33,34], neurodegenerative disorders [35,36], and RA [5,37,38]. However, the understanding of the exact components and the mechanisms underlying their supposed beneficial or detrimental effects is still insufficient. Newly introduced modern high throughput ‘omics’ approaches are greatly contributing to the elucidation of the connections between dietary exposure and health at the molecular level [39,40]. Genomics, transcriptomics, proteomics, and metabolomics are four main platforms of comprehensive ‘omics’ approach in nutritional science. They respectively investigate the whole set of DNA, RNA, proteins and metabolites in a cell, tissue or entire organism. The start of the 21st century was featured by prompt progress in high-throughput technologies, high-content- and single-cell approaches, mass spectrometry, bioinformatics, and computing capacities. These tools are widely used in an effort to identify molecular events implicated in the health effects of nutritional components or in diet-related diseases [41]. Nutrigenetics is the science that detects and describes gene variants linked with differential response to nutrients and relating this variation to different disease states. Next generation sequencing (NGS), also known as ‘second-generation sequencing’, makes it possible to read the code of great amounts of small fragments of DNA or RNA in parallel, enabling more rapid sequencing with higher throughput at dropping costs. Genome-wide association studies together with NGS have contributed to identify novel genomic variants (for e.g., genetic polymorphisms) with the aim of understanding complex disease pathobiology as well as examining the influences of dietary exposure and genetic variants in humans. The integration of genetic polymorphisms into nutritional epidemiological studies has allowed to tackle several limitations intrinsic in such studies, such as genetic variability affecting either the absorption, biotransformation, metabolism, distribution or elimination of a nutrient or bioactive food compound [42,43]. One example of how nutrigenomics and NGS technologies has been employed to elucidate the involvement of precise dietary factors results from an investigation on coffee and heart disease [44]. Numerous studies had examined this relationship and deduced that coffee either decreases risk, has no impact, or increases risk [45]. Even tough coffee is a complex mixture consisting of a large amount of bioactive compounds, it is an important source of caffeine and it has always been recognized that caffeine might be particularly deleterious for the cardiovascular system. Caffeinated-coffee was found to increase the risk of heart infarction among persons who carry a gene variant that makes them ‘slow’ caffeine metabolizers, but do not affect individuals who are ‘fast’ caffeine metabolizers [46]. NGS and omics-based readouts applied to nutrigenetics, will give crucially significant information that will support clinicians in ensuring the optimal diet for a particular individual, namely personalized nutrition.
Transcriptomics studies have become routine, thanks to technologies such as real-time-PCR and robust microarrays. Moreover, RNA sequencing has developed as a robust alternative for transcriptome investigation as it is covering wider spectrum of RNAs ensuring more useful information [47,48]. Transcriptomics analysis provides opportunities to investigate the transcriptome at a given nutritional condition, affording a comprehensive view of intracellular RNA expression [49]. For example, transcription profiling has been widely applied to assess the possible effects of anthocyanins, pigments naturally occurring in many comestible vegetables and fruits, on obesity associated gene expression in human adipocytes [50] and also for exploring the potentialities of gene expression profiling in blood to investigate the influences of nutritional exposure in human intervention studies [51]. Nutri-miromics explores the impact of diet on gene expression as a result of epigenetic processes related to microRNAs (miRNAs), which may influence the risk for the development of chronic diseases, including RA [52,53]. MiRNAs are small non-coding endogenous RNA molecules that functions in post-transcriptional regulation of gene expression by causing mRNA degradation or translational suppression through binding to a target messenger RNA. They may be regulated by environmental and nutritional factors, mainly by single nutrients or bioactive food components [54], suggesting that diet manipulation may have the potential to serve as a therapeutic approach in controlling the risk of chronic diseases. It has been suggested that miRNAs not only are synthesized endogenously, but also might be acquired from diet [55]. Although such subject is still debated, it has been proposed that exogenous miRNAs may modulate serum miRNAs profiles possibly influencing biological processes [56]. Next-generation sequencing and omics-based analyses provides a powerful tool to identify dietary miRNAs as well as to understand the complex crosstalk between nutrition, miRNAs, gene targets, and human health and disease [57]. Extensive availability of NGS and ‘multi-omics’ technologies has allowed genome-wide clarification of the epigenomic makeup in dozens of cell types, throughout developmental times, in several species, including humans [58].
Nutritional epigenomics deals with assessing the influence of nutrition and bioactive food compounds on global epigenetic mechanisms that regulate gene activity and expression, for e.g., DNA methylation, histone modifications (histone methylation, acetylation, and phosphorylation), chromatin remodelling, and noncoding RNAs. Epigenetic mechanisms have been implicated in pathogenesis of several diseases, including autoimmune conditions [59,60,61,62,63]. Since nutrition is among the most significant environmental factors influencing the epigenetic profile, nutritional-epigenomics powered by novel technologies is emerging as a promising approach in nutritional research and for personalized nutrition [64]. In the past few years, new technologies that allow to sequence longer strands of nucleic acids by reading single DNA or RNA molecules have made progress and become more eminent [65]. These technologies, which can also be called ‘long-read sequencing’ or ‘third generation sequencing’, together with the development in bioinformatics equipment and single-cell sequencing methodods, will allow to decipher human genome, transcriptome, microbiome and epigenome to a greater depth [66,67,68,69,70]. These technologies are already being successful applied in nutritional sciences to investigate, for e.g., the effect of dietary interventions on human microbiome [71].
Metabolomics is referred to the study of the overall metabolites set or small molecules (metabolome) present in biological samples. Conventionally, Nuclear Magnetic Resonance (NMR), Proton Nuclear Magnetic Resonance (1HNMR) Spectroscopy, and Mass Spectrometry (MS) have represented the principal technologies employed in metabolomics studies. Progress in these tools, in particular Matrix-Assisted Laser Desorption Ionization Time Of Flight (MALDI-TOF), Secondary Ion Mass Spectrometry (SIMS), and Fourier transform ion cyclotron resonance MS, along with new approaches for compound identification [72] have opened numerous opportunities for quantitative, non-invasive analysis for metabolites in human-body fluids and tissues as well as changes thereon in response to diet. Nutritional metabolomics has emerged as high-performance and sensitive method for the identification and characterization of biochemical pathways [73]. Moreover, it has been used in several studies for assessing metabolite profiles as a result of specific dietary intake [74,75,76,77] and several metabolic perturbations were detected. To refine the assessment of a person’s food intake, and therefore elucidate suggested correlations between diet and disease, relevant and accurate dietary evaluation methods are crucial. Dietary biomarkers have arisen as a complementary tool to the conventional methods in nutrition studies, and in the few past years, metabolomics has established as an important approach for the identification of new dietary biomarkers [78]. Proteomics is the large-scale, high-throughput study of the expression, structure, function, modifications, and interactions of proteins, within a biofluid or tissue sample. A broad spectrum of different approaches and methods are being utilized for proteomic studies, particularly microarray-based tools, mass-spectrometry, nuclear-magnetic resonance, and the most modern single-cell and high-sensitivity protein analyses [79,80,81]. Nutritional proteomics or nutriproteomics harnesses proteomics technologies to identify molecular and cellular variations in protein expression and function on a comprehensive level as well as evaluating the interaction of proteins with food components. Food components may interact with endogenous proteins inducing post-translational modifications and modulating their original functions. The characterization of such modifications will allow a better understanding of the interplay between bioactive dietary components and diet-related diseases [82,83]. For e.g., nutriproteomics could help in elucidating the possible relationships between food antigens and autoimmune disorders [84]. Tsuda et al. (2015) have demonstrated that monoclonal autoantibodies derived from RA patients cross-react not only with various autoantigens but also with numerous food proteins. The authors proposed that such dietary proteins may trigger the generation of RA-specific autoantibodies to induce autoimmunity in at risk individuals [85]. Interestingly, it has been also hypothesized that chemical alterations of food proteins by different toxic agents in food may lead to immune reaction against altered food proteins that cross-react with tissue antigens, causing autoimmune reactions [84]. Understanding the possible link between specific food consumption and autoimmunity in humans may lead to prevention of autoimmune diseases through precise dietary advices in at risk individuals. From birth, humans interact and coevolve with trillions of microbes residing in most body surfaces and cavities, referred to as the human microbiota. Advances in ‘omics’ technologies and computational methods have driven the investigation of the microbiota’s contribution to human health and disease, led by massive efforts such as the Human Microbiome Project and the Europe-based MetaHit Consortium [86]. Microbiomics is an emerging rapidly-growing field in which all the microbes of a particular community (for e.g., gut microbiota) are analyzed together, harnessing ‘omics’ approaches and technologies, including metagenomics, metatranscriptomics, metaproteomics and metabolomics. These technologies, which investigate respectively the collective genome, transcriptome, proteome, and metabolome of microorganisms from a sample (e.g., human stools or saliva), are providing information concerning the structure and function of the entire microbial community as well as the identification and assessment of regulatory and metabolic machinery by which host and microbes interact among themselves to determine a healthy or diseased state in the human host [87]. Every single microbial genus in the gut comprises numerous species and strains that may harbour important dissimilarities in their genomes and functional characteristics and it has been documented that strain-level diversity may result in inconsistencies in genus and species associations with dietary interventions, health or disease [88,89]. Our data frequently are based on a genus- or species-level taxonomic allocations that, even if helpful, may not be adequate for an exhaustive comprehension of the complex relations between the gut microbiome, diet and human health. As sequencing technologies continue to evolve, novel strain-level understandings can be achieved in the study of the relationships between gut microbiota, diet and human health [90]. Microbiome composition has been linked to disease also by way of modulation of diet-derived specific metabolites and signaling pathways [91,92]. Thus, nutritional microbiomics and metabolomics studies hold promise for the discovery of pathways linked to disease processes. Nutritional microbiomics is a promising approach to investigate the interaction between diet and gut microbiota and the potential of modulating these interactions for the prevention of human diseases [93]. Major ‘omics’ approaches and emerging technologies already employed for nutritional research are overviewed in Table 1.

3. The Contribution of ‘Omics’ in Elucidating Rheumatoid Arthritis Pathogenesis

RA is an autoimmune condition that impacts predominantly the lining of the synovial joints and is characterized by chronic synovitis, systemic inflammation and various degrees of bone and cartilage erosion. Systemic inflammation characterizing RA is related with different extra-articular conditions, including cardiovascular diseases, resulting in higher mortality in patients with RA. In industrialized countries, RA is the most frequent form of inflammatory arthritis. The disorder is more common in women and the age of onset is typically between 25 and 50, in the midst of working life, with important social and economic burden [104]. Genetic factors can explain 50–60% of the risk [105,106] while environmental modifiable factors, such as infectious diseases, tobacco smoking, air pollution, dust, and nutrition, account for the remainder [107,108]. Thanks to the rapid progresses in the field of ‘omics’ technologies, the past decade has resulted in tremendous advancement in our capacity to interpret genetic and molecular reasons underlying complex conditions such as autoimmune diseases [109]. The etiopathology of RA is not fully understood, however, NGS combined with the application of innovative multi-omics approaches, cell profiling-technologies and bioinformatics tools has enabled a wider investigation and deeper insight into the pathogenesis and disease variants of RA, including the definition of RA-associated cell populations [110], specific gene expression profiles [111,112,113,114,115], susceptibility loci, gene-environment interactions, as well as genetic loci associated with subsets of patients and those linked with response to therapy and/or dietary components [116,117,118,119,120]. Genomics studies of RA, including recent application of genome wide association studies (GWAS), have discovered over 100 genetic loci linked with RA risk and/or severity [121]. Amid all the genetic susceptibility elements found so far, the human leukocyte antigen (HLA) locus is the more important one. A strong link between RA and HLA-DRB1 alleles encoding an amino acid sequence pattern known as the ‘shared epitope’ has already been recognized for a long time. The shared epitope has been associated with a higher risk of anti-citrullinated protein antibodies (ACPAs)-positive RA while ACPAs positivity has been linked with more aggressive and destructive RA, systemic manifestations and cardiovascular complications [117]. It has been documented that the production of autoantibodies, including rheumatoid factor and ACPAs is an early and asymptomatic event that may precede for several years the onset of clinical apparent symptoms in RA. In the symptomatic RA phase, compensatory pathways that keep the disease asymptomatic may fail, promoting the transition from preclinical to clinically apparent disease [122]. These findings indicate an emerging disease paradigm where both genetic and environmental factors trigger a preclinical systemic autoimmune state, possibly originated at mucosal sites [123], followed by genetic and environmental factors that may propagate this silent autoimmune state to clinically overt RA [122,124].
Among the non-HLA gene variants identified there are many that involve immune system, citrullination, cytokine, and inflammation genes and/or known targets of approved therapy [121,125]. Loci showing stronger association with disease risk include genes encoding for protein tyrosine phosphatase, non-receptor type 22 (PTPN22), tumor necrosis factor (TNF) receptor-associated factor 1 (TRAF1), signal transducer and activator of transcription 4 (STAT4), interleukin 6 signal transducer (IL6ST), interleukin 2 receptor subunits alpha and beta, and CD40 [125]. The list of candidate genes is rapidly growing [126]. By combining the whole-genome sequencing and transcription profiles data of RA patients it is becoming possible to reveal the potential molecular pathways and crucial genes which play important roles in RA development as well as to identify the relevant cell types in RA pathogenesis [127,128,129]. Several pathways within the inflammatory cascade are activated in RA. All of them result in the upregulation of transcription factors and proinflammatory cytokines including nuclear factor kappa B (NFkB), interleukin (IL)-1, IL-6, IL-17, monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor (TNF)- α. Diverse genes in RA loci are concerned with the NF-kB signaling pathway, the Janus kinase (JAK)-signal transducers and activators of transcription (STAT), or cytokines signaling pathways [130,131,132]. The release of cytokines promotes the activation of fibroblast-like synoviocytes (FLSs), macrophages, neutrophils, adhesion molecules, and clotting factors as well as the release of macrophage colony-stimulating factors, which increase macrophages and also induce osteoclast activation, promoting active bone destruction. Matrix metalloproteinases (MMPs), prostaglandins, cyclooxygenase-2 (COX-2) and free radicals are also produced in excessive amounts which enhance the inflammatory pathways, resulting in the breakdown of extracellular matrix and cartilage loss [133,134]. The local and systemic immune responses lead to the development of a persistent low-intensity inflammatory condition which is related to important RA comorbidities and complications, such as cardiovascular diseases, vascular damage, insulin-resistance, and amyloidosis [135,136,137,138]. Recent high-throughput and epigenome-wide analysis have allowed to demonstrate that epigenetics play vital roles in the pathogenesis of RA [49,139], providing a possible crossing point by which genetic and environmental risk factors interact each other to influence the susceptibility, onset, and development of RA. Indeed, DNA methylation, post-translational histone modifications and variations in gene expression induced by non-coding RNAs are implicated in adaptive and innate immune cell differentiation, migration, proliferation, apoptosis, and FLSs activation in RA patients [140,141]. Proteomics-based approaches have been utilized for the identification of key protein and peptide mediators in RA [142], as well as to detect and quantify cytokines, allowing the discovery of new potential biomarkers [143], which may serve for early diagnosis, as indicators for clinical observation on the disease progression and to monitor responses to therapeutic interventions. By studying biomarkers in patient populations, the disease could be sorted into different subcategories that show different outcomes and responses to specific drugs or nutritional interventions [120,144,145,146]. Multi-omics analysis integrated with advanced bioinformatic tools and machine learning are already yielding a more comprehensive understanding of molecular mechanisms underlying RA pathogenesis and therapeutic response, paving the way for personalized-medicine [147,148].

4. Nutrigenomics Approach to Rheumatoid Arthritis

It is already well documented from epidemiological studies that nutritional patterns could play a role both as disease risk and protective factor, on the basis of the properties of particular foods. Certain nutritional factors can indeed exhibit pro-inflammatory outcomes (for e.g., red meat, salt, excessive caloric intake) or conversely mitigate inflammation (for e.g., fruit and vegetables, flaxseeds, low caloric intake) [5,6]. Nutritional components derived from diet, such as glucids, amino acids, fatty acids, vitamins, minerals, and other natural compounds occurring in small quantities in food, can not only fulfil a structural role in the cell, but also represent molecular signals which may impact biochemical pathways and alter gene expression. Indeed, they can directly interrelate with key factors and regulate the pathways involved in inflammation cascades related to RA pathogenesis, or affect intra- and extracellular microenvironments, thereby indirectly altering cellular activities [149]. Christensen et al. have recently demonstrated the association between diet and immune cell-related gene expression patterns in humans [150]. Interestingly, reduction in pain and higher physical function in RA patients, as well as a lower risk of the disease in individuals carrying the HLA-DRB1 shared epitope allele, have been associated with adherence to the Mediterranean diet (MedDiet) [37,151]. The assigned health benefits of the MedDiet could be justified by a modulating effect on genes related to inflammation and oxidative stress [152]. Although nutrigenomic studies on RA patients are still scarce, the influence of several dietary patterns and bioactive compounds on inflammation-or other RA-related pathways has been investigated. For e.g., Camargo et al. (2013) showed that MedDiet is able to reduce the expression of NF-κB, TNF-α, MCP-1 and MMP-9 in human peripheral blood mononuclear cells [153]. Consumption of a MedDiet for 4 weeks has been associated with reduced IL-1β gene expression in human mononuclear cells in fasting and postprandial states [154]. It has also been shown that adherence to MedDiet is linked with changes of methylation-state in inflammation-related genes in peripheral blood cells [155]. In addition, recent studies have shown that the Mediterranean-based nutritional interventions are able to induce changes in the expression of inflammation-related miRNAs [100], possibly influencing RA risk by means of epigenetic mechanisms. MedDiet is characterized by frequent consumption of food from vegetable sources, whole grains, beans, nuts, seeds, legumes, fruit, and spices, while olive oil represents the principal source of fat. MedDiet also involves an equilibrate intake of fish and wine, as well as a lower consumption of red meat, sweets, and dairy products [156] Components of the MedDiet such as olive oil, vegetables, fruits, fatty-fish, and tree-nuts provide a model for functional foods, on the basis of their natural contents of bioactive compounds and nutraceuticals, such as flavonoids, alkaloids, polyphenols, terpenoids, sterols, pigments, and polyunsaturated fatty-acids of omega-3 series (omega-3 PUFAs). Olive oil holds a high nutritional value and a peculiar composition, which is particularly relevant in the case of Extra Virgin Olive Oil (EVOO). EVOO is considered an important bioactive food with several valuable properties and it may be efficacious in the management of inflammatory and autoimmune diseases, including RA [157,158]. It has been shown that the phenolic fraction of EVOO modulates cytokines production, including IL-6, TNF-α, and IL-1β, perhaps via NF-κB signaling pathway, exerting an anti-inflammatory and immunomodulatory action in systemic lupus erythematosus patients [159]. Castaner et al. (2012) have shown that three weeks ingestion of high-polyphenol-containing olive oil, reduce the activation of the CD40/CD40-ligand system and its downstream products in healthy subjects, as compared to olive oil containing less polyphenol [160]. About that, it would be worth noting that the inflammatory CD40/CD40-ligand pathway seems to be implicated in the progression from undifferentiated arthritis or ACPAs-positive arthralgia to established RA [161]. Moreover, a recent study has demonstrated that EVOO phenolic compounds may have a favorable effect on bone by modulating osteoblast-related gene expression, which would explain their beneficial effect against bone pathologies [162]. Omega-3 PUFAs could have a protective and preventive effect in RA, given their anti-inflammatory and pain-killer properties [163,164]. Epidemiological studies have reported a substantial inverse association between oily fish consumption and RA [165,166,167], suggesting that omega-3 PUFAs found in fish oil (eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)) may be a protective factor against RA progression. In favour of this hypothesis, a case-control study established that RA cases had important decreased levels of EPA, and EPA+DHA in their erythrocyte membranes (a biomarker of omega-3 PUFAs status) compared with controls [168]. Moreover, Gan et al. (2017) showed that both omega-3 PUFAs supplement use and omega-3 PUFAs levels in red blood cell membranes were inversely linked with ACPAs positivity in people without RA, but at genetic risk for future RA [119]. A further study by the same authors found that the potential protective effect of omega-3 PUFAs on RA-related autoimmunity may be most pronounced in those who exhibit HLA-DRB1 shared-epitope genetic variant [169]. Emerging evidences from epigenomic-wide association studies indicate that one of the mechanisms responsible for the omega-3 PUFAs-related anti-inflammatory modulation of gene expression within the cells involves the alteration of epigenetic markers, such as DNA methylation [97]. Inverse associations with risk of disease development and disease activity has been also associated with high intakes of fruit and vegetables, suggesting a protective factor [170,171]. Fruits, such as berries, pomegranates and citrus fruits, as well as vegetables and whole grains, are rich sources of a variety of vitamins and other bioactive compounds, especially the polyphenolic flavonoids that have been associated with anti-inflammatory, anti-oxidant and analgesic effects. Vegetarian and vegan dietary patterns are particularly high in such compounds and have been proven useful in the management of RA [172]. Moreover, several studies have demonstrated that patients with arthritis consume less fruits, whole grains and plant proteins, compared to those without arthritis [173,174,175]. Nutrigenomic effects of numerous dietary bioactive compounds naturally occurring in fruits, vegetables, and their derivative products, have been explored in humans. For e.g., resveratrol, a phenolic compound normally present in some fruits including red grapes, mulberries as well as in peanuts and red wine, has been shown to downregulate NF-κB, TNF-α, IL-β, IL-6 and COX-2 [176,177]. In addition, according to recent nutri-miromics studies, resveratrol is capable of downregulating miRNA-21, and miRNA-155 in different human cell culture models [98,99]. MiRNA-21 and miRNA-155 are highly expressed in RA, and are known to play critical roles in disease pathogenesis [178,179,180]. Moderate consumption of alcohol was negatively associated with the risk of developing ACPA-positive RA, especially in smokers carrying HLA-DRB1 SE alleles [181], while excessive sodium chloride consumption among cigarette-smokers increases by more than two the risk of developing HLA-DRB1 SE-positive RA, suggesting a significant additive effect between smoking, diet and genetic risk factors [182]. It has been shown that caffeine downregulates inflammatory pathways involved in autoimmunity. In vitro experiments showed significant downregulation at the mRNA levels of key inflammatory genes including STAT1 and TNF, and also cytokine levels were decreased significantly with caffeine treatment [183]. Another study has found that coffee decreases methotrexate (i.e., the first-line disease-modifying antirheumatic drug used to treat RA) intolerance and increases treatment compliance among RA patients [184]. Soukup et al. scrutinized the effect of coffee intake on the therapeutic impact of methotrexate in RA patients grouped based on different genotypes related to adenosine pathway. They found that genotypes and coffee consumption affect risk of RA and effectiveness of methotrexate treatment [118]. Based on genome-wide gene expression response data, Van Bussel et al. [185] have theorised that a brief term caloric restriction is effective in downregulating gene sets involved in the immune response in young men. Moreover, a recent multi-omics study has found that caloric restriction ameliorates insulin sensitivity in humans [186], while studies in human obesity and insulin resistance have shown a clear relationship between decreased insulin sensitivity, the chronic activation of pro-inflammatory signaling pathways and RA activity. Indeed, elevated levels of TNF-α and IL-6 have all been recorded in several diabetic and insulin-resistant conditions [187,188], while insulin-resistant pathways have been linked with disease activity in RA [189]. Various in vivo studies have stated the nutrigenomic impacts of single nutrient supplementations/withdrawal in mouse or rat models of RA, namely magnesium [190], methionine [191], nobiletin (a citrus flavone) [192], omega-3 PUFAs-derived metabolites [193], Equol (a major soybean phenolic metabolite) [194], epigallocatechin-3-gallate (a green tea polyphenol) [195] and numerous others, and all these interventions usually showed promise of favorably modulating some of the genes or pathways implicated in inflammation or arthritis in rodents. However, considering the differences between RA, as it occurs in humans, and the RA-like condition currently replicated in RA animal models [196], translatability of these in vivo studies to humans in order to predict the effects of bioactive compounds or nutritional interventions, may be questionable. Additional studies designed to identify the genetic susceptibility to RA and the interaction between particular genetic variants, RNAs and proteins expression, metabolites production and nutrient intake in humans, will be crucial in planning patient-specific nutritional interventions and preventive strategies based on patient genetic characteristics.

5. Rheumatoid Arthritis, Microbiome and Nutrition

It is well known that microbiome can affect the inflammatory state of an individual by influencing both the host innate and adaptive immune system and its metabolic potential [197]. In recent times, thanks also to the progress in microbiomics techniques which has allowed a deep characterization of microbial communities, the gut microbiota has been given an important role in RA aetiology and development [3,198,199].
Zhang et al. carried out metagenomic sequencing and a metagenome-wide association study on stool, salivary and dental specimens from a wide cohort of treatment-naïve RA patients and healthy controls. Consequently, they found that the gut microbiome and the oral microbiome displayed noteworthy dissimilarities between RA patients and control subjects, and, the altered gut microbiome and oral microbiome of RA patients were in part restored by DMARDs [200]. Other human microbiomics studies have revealed that RA patients display a decreased gut microbiota diversity if compared with healthy controls [201]. Patients with RA, especially erosive patients, have a characteristic enterotype of gut microbiota with a decreased abundance of bacteria belonging to the family Bifidobacterium and Bacteroides [202], an expansion of certain rare bacterial lineages [203], and, at least in the preclinic phase of the disease, an abundance of Prevotella copri in shared-epitope positive individuals [204]. It is now accepted that diet composition has a crucial role in the control of gut microbial populations and, thus, in the potential prevention, management and treatment of many human diseases, including RA [205]. During the past few years, numerous works have associated diet/nutrients, gut microbial communities and the expression of genes involved in immune responses [206]. Considering this relation, there may be important therapeutic benefit in manipulating microbial composition by dietary interventions. A fresh study by De Filippis et al. [88] has suggested that diet affect P. copri at the strain level. The authors analyzed the gut metagenomes of individuals with different dietary patterns, checking for the presence of distinct P. copri strains. They found that a diet high in fiber were linked to P. copri types with better ability in carbohydrate catabolism, while P. copri strains associated with an omnivore dietary pattern had a higher preponderance of genes involved in branched-chain amino acids (BCAA) biosynthesis. Whether particular strains are more prone to trigger RA than others, and whether different nutritional patterns could affect this risk, remains to be elucidated by further studies. However, it has been shown that high BCAA is a risk factor for type 2 diabetes, glucose intolerance, and promote pro-oxidant and pro-inflammatory activities in peripheral blood mononuclear cells [207], suggesting a possible relationship between P. copri, diet, and RA in genetically susceptible individuals, paving the way for precise nutritional interventions aiming to prevent RA. While the pathogenesis of microbiome-mediated diseases remains to be completely clarified, one mechanism may be linked to microbial metabolism. There is evidence that some foods have pro- or anti-inflammatory effects mediated by diet-related metabolites [208]. Metabolites generated by gut commensal microbes in response to host diet, may influence both microbiota and host homeostasis, with potentially beneficial or detrimental effects. Tang et al. have identified dietary compounds and phytochemicals that may affect microbiota abundance within the gut and interact with microbial community composition to change host metabolism. In particular long-term intake of plant-derived foods as well as consumption of artificial sweeteners were linked to important disparities in circulating metabolites, especially bile acids, which were linked on gut enterotype, suggesting that microbiome makeup and structure mediate the effect of nutrition on host physiology [91]. This could be interesting considering that bile acids are increasingly recognised as important signalling molecules in the regulation of immune homeostasis and inflammation [209]. Several metabolomics studies and genome-based analysis of bacteria have reported that the microbial metabolites regulate immune system and inflammation. For e.g., short chain fatty acids (SCFA), produced by bacteria that ferment fiber, are known to mediate immune functions and inflammation [210,211]. Moreover long-term vegetarian diet has been linked with an enrichment in butyrate-producing bacteria [212], while a short-term high-fiber nutritional intervention study in RA patients has been shown to increase anti-inflammatory SCFA and decrease pro-arthritic cytokines, along with a durable shift in the microbiome composition [102]. In addition, evidence suggests that SCFA may act as histone deacetylase inhibitors in human cells, thereby epigenetically modulating inflammation-related gene expression [213]. Trimethylamine N-oxide (TMAO), is an important microbiota-generated metabolite derived from dietary choline, betaine, and L-carnitine, which are contained in great amounts, in red meat, eggs, and dairy. TMAO is known to exert an impact on several significant mechanisms in the atherosclerosis pathogenesis pathway and vascular disfunction, promoting the etiological mechanisms of cardiovascular disease in RA [214]. Persistent red meat consumption increases systemic TMAO levels while high intake of meat and processed-meat products has been associated with an increased risk of RA [215,216], suggesting that this association could be mediated by the host-microbiome and microbial metabolome. Recently, it has been found that RA patients have a distinct oral microbiome. In addition, RA oral microbiota showed greater micobial diversity in comparison with healthy subjects, suggesting that there could be more potentially pathogenic microbes in the oral cavity of patients with RA and that this could negatively affect the outcome of the disease [217]. Tong et al. have observed a typical compositional modification of salivary microbiome in persons at increased risk for RA, indicating that oral microbiota dysbiosis arise in the preclinic stage of RA and are linked with systemic autoimmune features [218]. For e.g., it has been shown that Porphyromonas gingivalis, a bacterium which can be found the oral cavity, may generate an enzyme that citrullinates proteins. A close connection between the resulting inflammatory disorder of the oral mucosa (periodontitis) and a higher susceptibility to seropositive RA has been suggested [219]. Interestingly, some studies revealed that dietary habits can influence periodontal health, both directly, and indirectly by altering oral microbiota composition, [103,220,221], suggesting that some benefit might possibly be achieved through dietary modulation in at risk subjects.

6. Discussion

The beginning of the 21st century was featured by fast progress in high-throughput ‘omics’ approaches, high-content-technologies, bioinformatics and computational power. Genomics, transcriptomics, proteomics, metabolomics, and microbiomics analysis now make it possible to improve the understanding of the pathogenesis of complex diseases such as RA as well as to study the relationship between nutrition, microbiota, health and disease at a molecular level. Thanks to the ‘omics’ methods and approaches, researchers are now experiencing the opportunity of connecting food components, diet, individual genetic background, health, and disease. Novel ‘omics’ technologies offer unprecedented opportunities to overcome the limitations inherent in traditional nutrition and RA research. Although robust associations between dietary intake and population health or disease are evident from conventional observational epidemiology, the outcomes of large-scale intervention studies examining the causality of those links have often proved unconvincing or have failed to demonstrate causality, including nutritional interventional studies related to RA [222,223]. This apparent conflict is most likely due to the well-known difficulty in assessing nutritional status and measuring habitual dietary intake which may lead to confounding in observational epidemiology. Indeed, dietary intake assessment is usually established by self-reported food intake questionnaires, which have intrinsic limitations. Metabolomics and proteomics benefit from the accessibility to the advanced high-sensitivity analytical tools to provide new opportunities for dietary biomarker development and application. Biomarkers of food or nutrient intake may allow to assess food consumption accurately and objectively by measuring blood/urine/fecal metabolites and avoiding the subjective errors that self-reporting of intake may introduce. There are several proteomics and metabolomics studies that have detected candidate biomarkers for diverse dietary habits, as well as for several kinds of foods, including meat, vegetables and fruits. Numerous studies have also described metabolites associated to specific dietary patterns, like MedDiet, high fat, or Western diets [208]. Interestingly, some studies revealed increased levels of carnitine and taurine in patients with active RA [224,225]. Carnitine and taurine are known to be potential biomarkers of meat intake thus this might support the evidences from epidemiological studies which correlate high meat consumption with RA [215]. The identification of food biomarkers is an ongoing process. The application of biomarkers in nutritional research will be crucial to ameliorate the assessment of dietary intake, exposure to particular nutritional components, and of compliance to nutritional interventions, as well providing information on interindividual differences in response to diet. There is a large need for further studies to better understand the influence of diet or dietary bioactive components on RA risk, possibly encompassing a nutrigenomics approach. This approach will lead to precision nutrition, whose goals are the prevention and management of chronic diseases, such as RA, by adjusting nutritional interventions or recommendations according to a specific genetic background or metabolic profile. Even if dietary biomarkers usually allow for a more realistic measure of nutritional intake, some factors which are absent in the conventional methods of dietary assessment could bias the measurement of dietary intake biomarkers. Such factors include genetic variations, nutritional factors (e.g., nutrient-nutrient interaction), lifestyle/physiological conditions (e.g., smoking), biological sampling and analytical methodology. However, existing research on this issue is still scarce. Currently there is no agreement as to which metabolites would be the most suitable biomarkers for distinct kind of foods. By the way, some metabolites are indicators of groups of food, not being capable to distinguish between the exact kinds of foods being examined (for e.g., a metabolite typically found in meat may not be useful in discerning between different types of meat). Possibly, for several foods, an association of diverse metabolites would be more suitable as a marker than a single metabolite. Unfortunately, the multi-omics studies in RA have not gathered dietary intake data nor employed the same ‘omics’ platforms so far, hampering the connection of specific food intake with metabolic changes in RA patients. Thus it is essential to evaluate a biomarker’s reliability, reproducibility, ability to reveal modifications over time and robustness throughout different populations, as well as advantages and disadvantages to warrant it is assessed using the adequate techniques [208,226]. Since conducting nutrition research with human subjects may be often challenging, many studies aimed to explore the mutual relationship between autoimmune diseases, including RA, nutrition and microbiota have been- and are being carried out in animal models. However, while animal models have been helpful in clarifying the fundamental mechanisms underpinning RA pathogenesis and immune responses, the use of non-human models to replicate the complexity of the relationship between RA, diet, microbiota as well as to evaluate the effects of nutritional interventions, may possibly be misleading, taking into account the several interspecies differences characterizing for e.g., physiologic responses to nutrients and systemic inflammatory challenges [227,228], gastrointestinal physiology and microbiome composition [229]. Recent developments in stem cell biology and three-dimensional complex fluidic in vitro systems offer exciting opportunities for developing new human biology-based models for use in nutrigenomics, microbiota and RA omics-based research [198,229]. Such emerging tools, which are already being applied in biomedical research, are likely to be better models of the complexity of the human in vivo situation. The integration of these tools with nutrigenomics and microbiomics could provide nutrition researchers with huge opportunities to undertake well-controlled experiments using very manageable human-relevant models to investigate the mechanisms through which food components modulate gene and protein expression, metabolite production and epigenetic pathways in both health and disease. Individually, ‘omics’ technologies have promoted a critical shift in biomedical and nutrition sciences. Anyhow, each approach individually cannot grab the entire biological complexity of nutrition-disease relationships. Combination of multiple technologies, referred to as ‘multi-omics approach or systems biology’ is emerging as an approach to allow a broader view of biology, disease and the influences of environmental factors, including diet [230]. The integration of data from different ‘omics’ platforms (system biology) could provide multidimensional insight into the relationship between pathogenetic processes and the influence of nutrition, allowing the retrieval of comprehensive and holistic biological information. Although an “inflammatory” dietary pattern may have a role in the switch from preclinical to clinical RA, and early nutritional intervention might possibly result in a delay or prevention of the onset of RA, it requires both an early diagnosis and the identification of at risk population [174,231]. Since current diagnostic tests are not sufficiently sensitive or accurate in the very early stages of the disease, RA is typically diagnosed only once damage to the joints has already begun, a time at which the window for optimal interventions may have been missed. The multi-omics approach has the potential to identify multiple biomarkers that can be used to revolutionize the management of RA by mean of enabling a timely diagnosis. Furthermore, through the analysis of biomarkers in patient populations, the disease could be stratified into distinct subsets that exhibit differential risks, outcomes, and eventually different responses to specific foods or dietary interventions. The integration of diverse complex ‘omics’ datasets may be considered one of the key challenges of today’s bioinformatics, due to different data formats, high data dimensionality and need for data normalization [232,233]. The constant exponential growth in ‘omics’ data requires a parallel development in computing power and software systems for handling this challenge. New bioinformatic stuffs for the integration of data from several ‘omics’ fields continue to arise, and will assist researchers to faithfully decode data in the context of biological systems, but harmonized actions are essential to encourage this process [234]. The multi-omics approach will provide the broader scientific community with a valuable resource to address many questions about RA pathogenesis and disease-nutrition interactions at a system-biology level, resulting in development of new diagnostic tools, therapeutic strategies, and targeted nutritional interventions, resulting in better disease prevention and management (Figure 1).

7. Conclusions

The continuous advancement in ‘omics’ technologies has a huge potential to transform nutrition and RA research. The application and integration of novel ‘omics’ technologies together with advanced computational tools will enable a better understanding of RA pathogenetic mechanisms at a molecular level, the discovery of new biomarkers, the identification and characterization of food bioactive compounds and their impact on RA-related pathways. This will lead to a more human-relevant approach to nutrition- and RA- research that ultimately will lead to a nutrigenomics approach and precision nutrition, for future better disease prevention and management.

Supplementary Materials

The following are available online at https://www.mdpi.com/2072-6643/13/3/763/s1, Table S1: Supplementary information about applied search terms in PubMed.

Author Contributions

Conceptualization: M.C. and T.Y.F.-H.; Software, L.R. and D.C.; Data curation, I.D. and B.B.; Writing—original draft: M.C. and T.Y.F.-H.; Visualization: M.E.Z. and S.S.C.; Writing—review & editing: J.M.A.-S. and F.G.; Supervision: F.G. and M.B.; Project administration: M.B. All authors have read and agreed to the published version of the manuscript.

Funding

No external funds supported this work.

Data Availability Statement

Data sharing is not applicable to this article. No new data were created in this study.

Acknowledgments

T.Y.F.-H. is supported by a “Juan de la Cierva-Formación” post-doctoral contract.

Conflicts of Interest

The authors have no relevant interests to declare.

References

  1. Donlin, L.T.; Park, S.-H.; Giannopoulou, E.; Ivovic, A.; Park-Min, K.-H.; Siegel, R.M.; Ivashkiv, L.B. Insights into rheumatic diseases from next-generation sequencing. Nat. Rev. Rheumatol. 2019, 15, 327–339. [Google Scholar] [CrossRef] [PubMed]
  2. Nemtsova, M.V.; Zaletaev, D.V.; Bure, I.V.; Mikhaylenko, D.S.; Kuznetsova, E.B.; Alekseeva, E.A.; Beloukhova, M.I.; Deviatkin, A.A.; Lukashev, A.N.; Zamyatnin, A.A. Epigenetic Changes in the Pathogenesis of Rheumatoid Arthritis. Front. Genet. 2019, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kishikawa, T.; Maeda, Y.; Nii, T.; Motooka, D.; Matsumoto, Y.; Matsushita, M.; Matsuoka, H.; Yoshimura, M.; Kawada, S.; Teshigawara, S.; et al. Metagenome-wide association study of gut microbiome revealed novel aetiology of rheumatoid arthritis in the Japanese population. Ann. Rheum. Dis. 2020, 79, 103. [Google Scholar] [CrossRef] [PubMed]
  4. Sales, N.M.R.; Pelegrini, P.B.; Goersch, M.C. Nutrigenomics: Definitions and advances of this new science. J. Nutr. Metab. 2014, 2014, 202759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gioia, C.; Lucchino, B.; Tarsitano, M.G.; Iannuccelli, C.; Di Franco, M. Dietary Habits and Nutrition in Rheumatoid Arthritis: Can Diet Influence Disease Development and Clinical Manifestations? Nutrients 2020, 12, 1456. [Google Scholar] [CrossRef] [PubMed]
  6. He, J.; Wang, Y.; Feng, M.; Zhang, X.; Jin, Y.-B.; Li, X.; Su, L.-C.; Liu, S.; Wang, A.-X.; Chen, X.-M.; et al. Dietary intake and risk of rheumatoid arthritis-a cross section multicenter study. Clin. Rheumatol. 2016, 35, 2901–2908. [Google Scholar] [CrossRef]
  7. Maeda, Y.; Takeda, K. Host–microbiota interactions in rheumatoid arthritis. Exp. Mol. Med. 2019, 51, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Horta-Baas, G.; Romero-Figueroa, M.d.S.; Montiel-Jarquín, A.J.; Pizano-Zárate, M.L.; García-Mena, J.; Ramírez-Durán, N. Intestinal Dysbiosis and Rheumatoid Arthritis: A Link between Gut Microbiota and the Pathogenesis of Rheumatoid Arthritis. J. Immunol. Res. 2017, 2017, 4835189. [Google Scholar] [CrossRef] [PubMed]
  9. Sela, D.A.; Mills, D.A. The marriage of nutrigenomics with the microbiome: The case of infant-associated bifidobacteria and milk. Am. J. Clin. Nutr. 2014, 99, 697S–703S. [Google Scholar] [CrossRef] [Green Version]
  10. Di Bella, J.M.; Bao, Y.; Gloor, G.B.; Burton, J.P.; Reid, G. High throughput sequencing methods and analysis for microbiome research. J. Microbiol. Methods 2013, 95, 401–414. [Google Scholar] [CrossRef]
  11. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef] [PubMed]
  12. Dei-Cas, I.; Giliberto, F.; Luce, L.; Dopazo, H.; Penas-Steinhardt, A. Metagenomic analysis of gut microbiota in non-treated plaque psoriasis patients stratified by disease severity: Development of a new Psoriasis-Microbiome Index. Sci. Rep. 2020, 10, 12754. [Google Scholar] [CrossRef] [PubMed]
  13. Eetemadi, A.; Rai, N.; Pereira, B.M.P.; Kim, M.; Schmitz, H.; Tagkopoulos, I. The Computational Diet: A Review of Computational Methods Across Diet, Microbiome, and Health. Front. Microbiol. 2020, 11, 393. [Google Scholar] [CrossRef] [Green Version]
  14. Leeming, E.R.; Johnson, A.J.; Spector, T.D.; Le Roy, C.I. Effect of Diet on the Gut Microbiota: Rethinking Intervention Duration. Nutrients 2019, 11, 2862. [Google Scholar] [CrossRef] [Green Version]
  15. Kolodziejczyk, A.A.; Zheng, D.; Elinav, E. Diet–microbiota interactions and personalized nutrition. Nat. Rev. Microbiol. 2019, 17, 742–753. [Google Scholar] [CrossRef]
  16. Nasir, A.; Bullo, M.M.H.; Ahmed, Z.; Imtiaz, A.; Yaqoob, E.; Jadoon, M.; Ahmed, H.; Afreen, A.; Yaqoob, S. Nutrigenomics: Epigenetics and cancer prevention: A comprehensive review. Crit. Rev. Food Sci. Nutr. 2020, 60, 1375–1387. [Google Scholar] [CrossRef]
  17. De Angelis, M.; Ferrocino, I.; Calabrese, F.M.; De Filippis, F.; Cavallo, N.; Siragusa, S.; Rampelli, S.; Di Cagno, R.; Rantsiou, K.; Vannini, L.; et al. Diet influences the functions of the human intestinal microbiome. Sci. Rep. 2020, 10, 4247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Yoshida, N.; Yamashita, T.; Hirata, K.-I. Gut Microbiome and Cardiovascular Diseases. Diseases 2018, 6, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Ferguson Jane, F.; Allayee, H.; Gerszten Robert, E.; Ideraabdullah, F.; Kris-Etherton Penny, M.; Ordovás José, M.; Rimm Eric, B.; Wang Thomas, J.; Bennett Brian, J. Nutrigenomics, the Microbiome, and Gene-Environment Interactions: New Directions in Cardiovascular Disease Research, Prevention, and Treatment. Circ. Cardiovasc. Genet. 2016, 9, 291–313. [Google Scholar] [CrossRef] [Green Version]
  20. Dongiovanni, P.; Valenti, L. A Nutrigenomic Approach to Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2017, 18, 1534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Al Theyab, A.; Almutairi, T.; Al-Suwaidi, A.M.; Bendriss, G.; McVeigh, C.; Chaari, A. Epigenetic Effects of Gut Metabolites: Exploring the Path of Dietary Prevention of Type 1 Diabetes. Front. Nutr. 2020, 7. [Google Scholar] [CrossRef]
  22. Ferguson, L.R. Nutrigenetics, nutrigenomics and inflammatory bowel diseases. Expert Rev. Clin. Immunol. 2013, 9, 717–726. [Google Scholar] [CrossRef] [PubMed]
  23. Berná, G.; Oliveras-López, M.J.; Jurado-Ruíz, E.; Tejedo, J.; Bedoya, F.; Soria, B.; Martín, F. Nutrigenetics and nutrigenomics insights into diabetes etiopathogenesis. Nutrients 2014, 6, 5338–5369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ren, X.; Li, X. Advances in Research on Diabetes by Human Nutriomics. Int. J. Mol. Sci. 2019, 20, 5375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Taylor, P.C.; Moore, A.; Vasilescu, R.; Alvir, J.; Tarallo, M. A structured literature review of the burden of illness and unmet needs in patients with rheumatoid arthritis: A current perspective. Rheumatol. Int. 2016, 36, 685–695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sami, W.; Ansari, T.; Butt, N.S.; Hamid, M.R.A. Effect of diet on type 2 diabetes mellitus: A review. Int. J. Health Sci. (Qassim) 2017, 11, 65–71. [Google Scholar]
  27. Popkin, B.M. Nutrition Transition and the Global Diabetes Epidemic. Curr. Diabetes Rep. 2015, 15, 64. [Google Scholar] [CrossRef] [PubMed]
  28. Casas, R.; Castro-Barquero, S.; Estruch, R.; Sacanella, E. Nutrition and Cardiovascular Health. Int. J. Mol. Sci. 2018, 19, 3988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Becerra-Tomás, N.; Paz-Graniel, I.; WC Kendall, C.; Kahleova, H.; Rahelić, D.; Sievenpiper, J.L.; Salas-Salvadó, J. Nut consumption and incidence of cardiovascular diseases and cardiovascular disease mortality: A meta-analysis of prospective cohort studies. Nutr. Rev. 2019, 77, 691–709. [Google Scholar] [CrossRef]
  30. Marklund, M.; Wu, J.H.Y.; Imamura, F.; Del Gobbo, L.C.; Fretts, A.; de Goede, J.; Shi, P.; Tintle, N.; Wennberg, M.; Aslibekyan, S.; et al. Biomarkers of Dietary Omega-6 Fatty Acids and Incident Cardiovascular Disease and Mortality. Circulation 2019, 139, 2422–2436. [Google Scholar] [CrossRef] [Green Version]
  31. Shirlina, N.G.; Vilms, E.A.; Stasenko, V.L. Nutrition as a possible risk factor for breast cancer. Gig. Sanit. 2015, 94, 48–52. [Google Scholar] [PubMed]
  32. Thanikachalam, K.; Khan, G. Colorectal Cancer and Nutrition. Nutrients 2019, 11, 164. [Google Scholar] [CrossRef] [Green Version]
  33. Key, T.J.; Bradbury, K.E.; Perez-Cornago, A.; Sinha, R.; Tsilidis, K.K.; Tsugane, S. Diet, nutrition, and cancer risk: What do we know and what is the way forward? BMJ 2020, 368, m511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Gonzales, J.F.; Barnard, N.D.; Jenkins, D.J.; Lanou, A.J.; Davis, B.; Saxe, G.; Levin, S. Applying the precautionary principle to nutrition and cancer. J. Am. Coll. Nutr. 2014, 33, 239–246. [Google Scholar] [CrossRef]
  35. Pistollato, F.; Iglesias, R.C.; Ruiz, R.; Aparicio, S.; Crespo, J.; Lopez, L.D.; Manna, P.P.; Giampieri, F.; Battino, M. Nutritional patterns associated with the maintenance of neurocognitive functions and the risk of dementia and Alzheimer’s disease: A focus on human studies. Pharmacol. Res. 2018, 131, 32–43. [Google Scholar] [CrossRef] [PubMed]
  36. Seidl, S.E.; Santiago, J.A.; Bilyk, H.; Potashkin, J.A. The emerging role of nutrition in Parkinson’s disease. Front. Aging Neurosci. 2014, 6, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Johansson, K.; Askling, J.; Alfredsson, L.; Di Giuseppe, D.; on behalf of the EIRA Study Group. Mediterranean diet and risk of rheumatoid arthritis: A population-based case-control study. Arthritis Res. Ther. 2018, 20, 175. [Google Scholar] [CrossRef]
  38. Skoczyńska, M.; Świerkot, J. The role of diet in rheumatoid arthritis. Reumatologia 2018, 56, 259–267. [Google Scholar] [CrossRef] [PubMed]
  39. Valdés, A.; Cifuentes, A.; León, C. Foodomics evaluation of bioactive compounds in foods. TrAC Trends Anal. Chem. 2017, 96, 2–13. [Google Scholar] [CrossRef]
  40. Aruoma, O.I.; Hausman-Cohen, S.; Pizano, J.; Schmidt, M.A.; Minich, D.M.; Joffe, Y.; Brandhorst, S.; Evans, S.J.; Brady, D.M. Personalized Nutrition: Translating the Science of Nutrigenomics into Practice: Proceedings from the 2018 American College of Nutrition Meeting. J. Am. Coll. Nutr. 2019, 38, 287–301. [Google Scholar] [CrossRef] [PubMed]
  41. Trujillo, E.; Davis, C.; Milner, J. Nutrigenomics, proteomics, metabolomics, and the practice of dietetics. J. Am. Diet. Assoc. 2006, 106, 403–413. [Google Scholar] [CrossRef]
  42. El-Sohemy, A. Nutrigenetics. Forum Nutr. 2007, 60, 25–30. [Google Scholar] [CrossRef] [PubMed]
  43. Gomez-Delgado, F.; Alcala-Diaz, J.F.; Garcia-Rios, A.; Delgado-Lista, J.; Ortiz-Morales, A.; Rangel-Zuñiga, O.; Tinahones, F.J.; Gonzalez-Guardia, L.; Malagon, M.M.; Bellido-Muñoz, E.; et al. Polymorphism at the TNF-alpha gene interacts with Mediterranean diet to influence triglyceride metabolism and inflammation status in metabolic syndrome patients: From the CORDIOPREV clinical trial. Mol. Nutr. Food Res. 2014, 58, 1519–1527. [Google Scholar] [CrossRef]
  44. Cornelis, M.C.; El-Sohemy, A.; Kabagambe, E.K.; Campos, H. Coffee, CYP1A2 genotype, and risk of myocardial infarction. JAMA 2006, 295, 1135–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ordovas, J.M.; Corella, D. Nutritional genomics. Annu. Rev. Genom. Hum. Genet. 2004, 5, 71–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Cornelis, M.C.; El-Sohemy, A. Coffee, caffeine, and coronary heart disease. Curr. Opin. Lipidol. 2007, 18, 13–19. [Google Scholar] [CrossRef] [PubMed]
  47. Tachibana, C. Transcriptomics today: Microarrays, RNA-seq, and more. Science 2015, 349, 544. [Google Scholar] [CrossRef]
  48. Zhao, S.; Fung-Leung, W.-P.; Bittner, A.; Ngo, K.; Liu, X. Comparison of RNA-Seq and Microarray in Transcriptome Profiling of Activated T Cells. PLoS ONE 2014, 9, e78644. [Google Scholar] [CrossRef]
  49. Liu, B.; Qian, S.B. Translational regulation in nutrigenomics. Adv. Nutr. 2011, 2, 511–519. [Google Scholar] [CrossRef] [PubMed]
  50. Tsuda, T.; Ueno, Y.; Yoshikawa, T.; Kojo, H.; Osawa, T. Microarray profiling of gene expression in human adipocytes in response to anthocyanins. Biochem. Pharmacol. 2006, 71, 1184–1197. [Google Scholar] [CrossRef] [PubMed]
  51. Van Erk, M.J.; Blom, W.A.M.; van Ommen, B.; Hendriks, H.F.J. High-protein and high-carbohydrate breakfasts differentially change the transcriptome of human blood cells. Am. J. Clin. Nutr. 2006, 84, 1233–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Qu, Z.; Li, W.; Fu, B. MicroRNAs in Autoimmune Diseases. BioMed Res. Int. 2014, 2014, 527895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Evangelatos, G.; Fragoulis, G.E.; Koulouri, V.; Lambrou, G.I. MicroRNAs in rheumatoid arthritis: From pathogenesis to clinical impact. Autoimmun. Rev. 2019, 18, 102391. [Google Scholar] [CrossRef]
  54. Kura, B.; Parikh, M.; Slezak, J.; Pierce, G.N. The Influence of Diet on MicroRNAs that Impact Cardiovascular Disease. Molecules 2019, 24, 1509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Cui, J.; Zhou, B.; Ross, S.A.; Zempleni, J. Nutrition, microRNAs, and Human Health. Adv. Nutr. 2017, 8, 105–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Zhang, L.; Chen, T.; Yin, Y.; Zhang, C.-Y.; Zhang, Y.-L. Dietary microRNA—A Novel Functional Component of Food. Adv. Nutr. 2019, 10, 711–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Chiang, K.; Shu, J.; Zempleni, J.; Cui, J. Dietary MicroRNA Database (DMD): An Archive Database and Analytic Tool for Food-Borne microRNAs. PLoS ONE 2015, 10, e0128089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Sarda, S.; Hannenhalli, S. Next-generation sequencing and epigenomics research: A hammer in search of nails. Genom. Inform. 2014, 12, 2–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Giacconi, R.; Malavolta, M.; Bürkle, A.; Moreno-Villanueva, M.; Franceschi, C.; Capri, M.; Slagboom, P.E.; Jansen, E.; Dollé, M.E.T.; Grune, T.; et al. Nutritional Factors Modulating Alu Methylation in an Italian Sample from The Mark-Age Study Including Offspring of Healthy Nonagenarians. Nutrients 2019, 11, 2986. [Google Scholar] [CrossRef] [Green Version]
  60. Brandt, B.; Rashidiani, S.; Bán, Á.; Rauch, T.A. DNA Methylation-Governed Gene Expression in Autoimmune Arthritis. Int. J. Mol. Sci. 2019, 20, 5646. [Google Scholar] [CrossRef] [Green Version]
  61. Carlos-Reyes, Á.; López-González, J.S.; Meneses-Flores, M.; Gallardo-Rincón, D.; Ruíz-García, E.; Marchat, L.A.; Astudillo-de la Vega, H.; Hernández de la Cruz, O.N.; López-Camarillo, C. Dietary Compounds as Epigenetic Modulating Agents in Cancer. Front. Genet. 2019, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Gay, S.; Wilson, A.G. The emerging role of epigenetics in rheumatic diseases. Rheumatology 2014, 53, 406–414. [Google Scholar] [CrossRef] [Green Version]
  63. Kalea, A.Z.; Drosatos, K.; Buxton, J.L. Nutriepigenetics and cardiovascular disease. Curr. Opin. Clin. Nutr. Metab. Care 2018, 21, 252–259. [Google Scholar] [CrossRef]
  64. Burdge, G.C.; Hoile, S.P.; Lillycrop, K.A. Epigenetics: Are there implications for personalised nutrition? Curr. Opin. Clin. Nutr. Metab. Care 2012, 15, 442–447. [Google Scholar] [CrossRef]
  65. Van Dijk, E.L.; Jaszczyszyn, Y.; Naquin, D.; Thermes, C. The Third Revolution in Sequencing Technology. Trends Genet. TIG 2018, 34, 666–681. [Google Scholar] [CrossRef] [PubMed]
  66. Midha, M.K.; Wu, M.; Chiu, K.P. Long-read sequencing in deciphering human genetics to a greater depth. Hum. Genet. 2019, 138, 1201–1215. [Google Scholar] [CrossRef]
  67. Gong, L.; Wong, C.H.; Idol, J.; Ngan, C.Y.; Wei, C.L. Ultra-long Read Sequencing for Whole Genomic DNA Analysis. J. Vis. Exp. JOVE 2019. [Google Scholar] [CrossRef] [Green Version]
  68. Kraft, F.; Kurth, I. Long-read sequencing in human genetics. Med. Genet. 2019, 31, 198–204. [Google Scholar] [CrossRef] [Green Version]
  69. Oikonomopoulos, S.; Bayega, A.; Fahiminiya, S.; Djambazian, H.; Berube, P.; Ragoussis, J. Methodologies for Transcript Profiling Using Long-Read Technologies. Front. Genet. 2020, 11. [Google Scholar] [CrossRef] [PubMed]
  70. Quick, J. Ultra-Long Read Nanopore Sequencing Methods for Metagenomics. J. Biomol. Tech. 2019, 30, S63. [Google Scholar]
  71. Li, J.; Xu, H.; Sun, Z.; Hou, Q.; Kwok, L.-Y.; Laga, W.; Wang, Y.; Ma, H.; Yu, Z.; Menghe, B.; et al. Effect of dietary interventions on the intestinal microbiota of Mongolian hosts. Sci. Bull. 2016, 61, 1605–1614. [Google Scholar] [CrossRef] [Green Version]
  72. Singh, A. Tools for metabolomics. Nat. Methods 2020, 17, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. McGee, E.E.; Kiblawi, R.; Playdon, M.C.; Eliassen, A.H. Nutritional Metabolomics in Cancer Epidemiology: Current Trends, Challenges, and Future Directions. Curr. Nutr. Rep. 2019, 8, 187–201. [Google Scholar] [CrossRef]
  74. Johansson-Persson, A.; Barri, T.; Ulmius, M.; Onning, G.; Dragsted, L.O. LC-QTOF/MS metabolomic profiles in human plasma after a 5-week high dietary fiber intake. Anal. Bioanal. Chem. 2013, 405, 4799–4809. [Google Scholar] [CrossRef] [PubMed]
  75. McCombie, G.; Browning, L.M.; Titman, C.M.; Song, M.; Shockcor, J.; Jebb, S.A.; Griffin, J.L. Omega-3 oil intake during weight loss in obese women results in remodelling of plasma triglyceride and fatty acids. Metab. Off. J. Metab. Soc. 2009, 5, 363–374. [Google Scholar] [CrossRef] [Green Version]
  76. Van Velzen, E.J.; Westerhuis, J.A.; van Duynhoven, J.P.; van Dorsten, F.A.; Grün, C.H.; Jacobs, D.M.; Duchateau, G.S.; Vis, D.J.; Smilde, A.K. Phenotyping tea consumers by nutrikinetic analysis of polyphenolic end-metabolites. J. Proteome Res. 2009, 8, 3317–3330. [Google Scholar] [CrossRef] [PubMed]
  77. Redeuil, K.; Smarrito-Menozzi, C.; Guy, P.; Rezzi, S.; Dionisi, F.; Williamson, G.; Nagy, K.; Renouf, M. Identification of novel circulating coffee metabolites in human plasma by liquid chromatography-mass spectrometry. J. Chromatogr. A 2011, 1218, 4678–4688. [Google Scholar] [CrossRef]
  78. Collins, C.; McNamara, A.E.; Brennan, L. Role of metabolomics in identification of biomarkers related to food intake. Proc. Nutr. Soc. 2019, 78, 189–196. [Google Scholar] [CrossRef]
  79. Parker, S.J.; Raedschelders, K.; Van Eyk, J.E. Emerging proteomic technologies for elucidating context-dependent cellular signaling events: A big challenge of tiny proportions. Proteomics 2015, 15, 1486–1502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Levy, E.; Slavov, N. Single cell protein analysis for systems biology. Essays Biochem. 2018, 62, 595–605. [Google Scholar] [CrossRef] [PubMed]
  81. Minakshi, P.; Kumar, R.; Ghosh, M.; Saini, H.M.; Ranjan, K.; Brar, B.; Prasad, G. Chapter 14—Single-Cell Proteomics: Technology and Applications. In Single-Cell Omics; Barh, D., Azevedo, V., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 283–318. [Google Scholar] [CrossRef]
  82. Schweigert, F.J. Nutritional Proteomics: Methods and Concepts for Research in Nutritional Science. Ann. Nutr. Metab. 2007, 51, 99–107. [Google Scholar] [CrossRef]
  83. Ganesh, V.; Hettiarachchy, N.S. Nutriproteomics: A promising tool to link diet and diseases in nutritional research. Biochim. Biophys. Acta BBA Proteins Proteom. 2012, 1824, 1107–1117. [Google Scholar] [CrossRef]
  84. Vojdani, A.; Gushgari, L.R.; Vojdani, E. Interaction between food antigens and the immune system: Association with autoimmune disorders. Autoimmun. Rev. 2020, 19, 102459. [Google Scholar] [CrossRef] [PubMed]
  85. Tsuda, R.; Ozawa, T.; Kobayashi, E.; Hamana, H.; Taki, H.; Tobe, K.; Sugiyama, E.; Iwamoto, M.; Imura, J.; Kishi, H.; et al. Monoclonal antibody against citrullinated peptides obtained from rheumatoid arthritis patients reacts with numerous citrullinated microbial and food proteins. Arthritis Rheumatol. 2015, 67, 2020–2031. [Google Scholar] [CrossRef] [Green Version]
  86. Manasson, J.; Blank, R.B.; Scher, J.U. The microbiome in rheumatology: Where are we and where should we go? Ann. Rheum. Dis. 2020, 79, 727. [Google Scholar] [CrossRef] [Green Version]
  87. Segal, J.P.; Mullish, B.H.; Quraishi, M.N.; Acharjee, A.; Williams, H.R.T.; Iqbal, T.; Hart, A.L.; Marchesi, J.R. The application of omics techniques to understand the role of the gut microbiota in inflammatory bowel disease. Ther. Adv. Gastroenterol. 2019, 12. [Google Scholar] [CrossRef] [Green Version]
  88. De Filippis, F.; Pasolli, E.; Tett, A.; Tarallo, S.; Naccarati, A.; De Angelis, M.; Neviani, E.; Cocolin, L.; Gobbetti, M.; Segata, N.; et al. Distinct Genetic and Functional Traits of Human Intestinal Prevotella copri Strains Are Associated with Different Habitual Diets. Cell Host Microbe 2019, 25, 444–453.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Wu, G.; Zhang, C.; Wu, H.; Wang, R.; Shen, J.; Wang, L.; Zhao, Y.; Pang, X.; Zhang, X.; Zhao, L.; et al. Genomic Microdiversity of Bifidobacterium pseudocatenulatum Underlying Differential Strain-Level Responses to Dietary Carbohydrate Intervention. MBio 2017, 8, e02348-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Malla, M.A.; Dubey, A.; Kumar, A.; Yadav, S.; Hashem, A.; Abd_Allah, E.F. Exploring the Human Microbiome: The Potential Future Role of Next-Generation Sequencing in Disease Diagnosis and Treatment. Front. Immunol. 2019, 9. [Google Scholar] [CrossRef] [PubMed]
  91. Tang, Z.-Z.; Chen, G.; Hong, Q.; Huang, S.; Smith, H.M.; Shah, R.D.; Scholz, M.; Ferguson, J.F. Multi-Omic Analysis of the Microbiome and Metabolome in Healthy Subjects Reveals Microbiome-Dependent Relationships Between Diet and Metabolites. Front. Genet. 2019, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Tang, W.H.W.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal Microbial Metabolism of Phosphatidylcholine and Cardiovascular Risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Mills, S.; Stanton, C.; Lane, J.A.; Smith, G.J.; Ross, R.P. Precision Nutrition and the Microbiome, Part I: Current State of the Science. Nutrients 2019, 11, 923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Slatko, B.E.; Gardner, A.F.; Ausubel, F.M. Overview of Next-Generation Sequencing Technologies. Curr. Protoc. Mol. Biol. 2018, 122, e59. [Google Scholar] [CrossRef] [PubMed]
  95. Hirst, M.; Marra, M.A. Next generation sequencing based approaches to epigenomics. Brief. Funct. Genom. 2010, 9, 455–465. [Google Scholar] [CrossRef]
  96. Verma, M.; Kumar, V. Chapter 11—Single-Cell Epigenomics: Technology and Applications. In Single-Cell Omics; Barh, D., Azevedo, V., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 215–229. [Google Scholar] [CrossRef]
  97. Hussey, B.; Lindley, M.R.; Mastana, S.S. Omega 3 fatty acids, inflammation and DNA methylation: An overview. Clin. Lipidol. 2017, 12, 24–32. [Google Scholar] [CrossRef]
  98. Kocic, H.; Damiani, G.; Stamenkovic, B.; Tirant, M.; Jovic, A.; Tiodorovic, D.; Peris, K. Dietary compounds as potential modulators of microRNA expression in psoriasis. Adv. Chronic Dis. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  99. Tili, E.; Michaille, J.-J.; Adair, B.; Alder, H.; Limagne, E.; Taccioli, C.; Ferracin, M.; Delmas, D.; Latruffe, N.; Croce, C.M. Resveratrol decreases the levels of miR-155 by upregulating miR-663, a microRNA targeting JunB and JunD. Carcinogenesis 2010, 31, 1561–1566. [Google Scholar] [CrossRef] [PubMed]
  100. Marques-Rocha, J.L.; Milagro, F.I.; Mansego, M.L.; Zulet, M.A.; Bressan, J.; Martínez, J.A. Expression of inflammation-related miRNAs in white blood cells from subjects with metabolic syndrome after 8 wk of following a Mediterranean diet–based weight loss program. Nutrition 2016, 32, 48–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Rao, V.S.; Srinivas, K.; Sujini, G.N.; Kumar, G.N.S. Protein-Protein Interaction Detection: Methods and Analysis. Int. J. Proteom. 2014, 2014, 147648. [Google Scholar] [CrossRef] [Green Version]
  102. Dürholz, K.; Hofmann, J.; Iljazovic, A.; Häger, J.; Lucas, S.; Sarter, K.; Strowig, T.; Bang, H.; Rech, J.; Schett, G.; et al. Dietary Short-Term Fiber Interventions in Arthritis Patients Increase Systemic SCFA Levels and Regulate Inflammation. Nutrients 2020, 12, 3207. [Google Scholar] [CrossRef]
  103. Murtaza, N.; Burke, L.M.; Vlahovich, N.; Charlesson, B.; O’Neill, H.M.; Ross, M.L.; Campbell, K.L.; Krause, L.; Morrison, M. Analysis of the Effects of Dietary Pattern on the Oral Microbiome of Elite Endurance Athletes. Nutrients 2019, 11, 614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Gibofsky, A. Epidemiology, pathophysiology, and diagnosis of rheumatoid arthritis: A Synopsis. Am. J. Manag. Care 2014, 20, S128–S135. [Google Scholar]
  105. Kurkó, J.; Besenyei, T.; Laki, J.; Glant, T.T.; Mikecz, K.; Szekanecz, Z. Genetics of rheumatoid arthritis—A comprehensive review. Clin. Rev. Allergy Immunol. 2013, 45, 170–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. MacGregor, A.J.; Snieder, H.; Rigby, A.S.; Koskenvuo, M.; Kaprio, J.; Aho, K.; Silman, A.J. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum. 2000, 43, 30–37. [Google Scholar] [CrossRef]
  107. Deane, K.D.; Demoruelle, M.K.; Kelmenson, L.B.; Kuhn, K.A.; Norris, J.M.; Holers, V.M. Genetic and environmental risk factors for rheumatoid arthritis. Best Pr. Res. Clin. Rheumatol. 2017, 31, 3–18. [Google Scholar] [CrossRef] [PubMed]
  108. Cutolo, M.; Nikiphorou, E. Don’t neglect nutrition in rheumatoid arthritis! RMD Open 2018, 4, e000591. [Google Scholar] [CrossRef] [Green Version]
  109. Kim, H.Y.; Kim, H.R.; Lee, S.H. Advances in systems biology approaches for autoimmune diseases. Immune Netw. 2014, 14, 73–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Zhang, F.; Wei, K.; Slowikowski, K.; Fonseka, C.Y.; Rao, D.A.; Kelly, S.; Goodman, S.M.; Tabechian, D.; Hughes, L.B.; Salomon-Escoto, K.; et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat. Immunol. 2019, 20, 928–942. [Google Scholar] [CrossRef]
  111. Chen, Y.-J.; Chang, W.-A.; Wu, L.-Y.; Hsu, Y.-L.; Chen, C.-H.; Kuo, P.-L. Systematic Analysis of Differential Expression Profile in Rheumatoid Arthritis Chondrocytes Using Next-Generation Sequencing and Bioinformatics Approaches. Int. J. Med. Sci. 2018, 15, 1129–1142. [Google Scholar] [CrossRef] [Green Version]
  112. Platzer, A.; Nussbaumer, T.; Karonitsch, T.; Smolen, J.S.; Aletaha, D. Analysis of gene expression in rheumatoid arthritis and related conditions offers insights into sex-bias, gene biotypes and co-expression patterns. PLoS ONE 2019, 14, e0219698. [Google Scholar] [CrossRef] [Green Version]
  113. Stephenson, W.; Donlin, L.T.; Butler, A.; Rozo, C.; Bracken, B.; Rashidfarrokhi, A.; Goodman, S.M.; Ivashkiv, L.B.; Bykerk, V.P.; Orange, D.E.; et al. Single-cell RNA-seq of rheumatoid arthritis synovial tissue using low-cost microfluidic instrumentation. Nat. Commun. 2018, 9, 791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Wang, X.; Xia, S.; Fu, B. RNA-seq analysis of synovial fibroblasts in human rheumatoid arthritis. Mol. Med. Rep. 2014, 10, 241–247. [Google Scholar] [CrossRef]
  115. Suzuki, A.; Terao, C.; Yamamoto, K. Linking of genetic risk variants to disease-specific gene expression via multi-omics studies in rheumatoid arthritis. Semin. Arthritis Rheum. 2019, 49, S49–S53. [Google Scholar] [CrossRef] [PubMed]
  116. Giannopoulou, E.G.; Elemento, O.; Ivashkiv, L.B. Use of RNA sequencing to evaluate rheumatic disease patients. Arthritis Res. Ther. 2015, 17, 167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Goulielmos, G.N.; Zervou, M.I.; Myrthianou, E.; Burska, A.; Niewold, T.B.; Ponchel, F. Genetic data: The new challenge of personalized medicine, insights for rheumatoid arthritis patients. Gene 2016, 583, 90–101. [Google Scholar] [CrossRef] [PubMed]
  118. Soukup, T.; Hloch, K.; Doseděl, M.; Tebbens, J.D.; Nekvindová, J.; Šembera, Š.; Veleta, T.; Pávek, P.; Barvík, I. The influence of coffee intake and genetics on adenosine pathway in rheumatoid arthritis. Pharmacogenomics 2020, 21, 735–749. [Google Scholar] [CrossRef]
  119. Gan, R.W.; Young, K.A.; Zerbe, G.O.; Demoruelle, M.K.; Weisman, M.H.; Buckner, J.H.; Gregersen, P.K.; Mikuls, T.R.; O’Dell, J.R.; Keating, R.M.; et al. Lower omega-3 fatty acids are associated with the presence of anti-cyclic citrullinated peptide autoantibodies in a population at risk for future rheumatoid arthritis: A nested case-control study. Rheumatology 2016, 55, 367–376. [Google Scholar] [CrossRef] [Green Version]
  120. Tasaki, S.; Suzuki, K.; Kassai, Y.; Takeshita, M.; Murota, A.; Kondo, Y.; Ando, T.; Nakayama, Y.; Okuzono, Y.; Takiguchi, M.; et al. Multi-omics monitoring of drug response in rheumatoid arthritis in pursuit of molecular remission. Nat. Commun. 2018, 9, 2755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Okada, Y.; Wu, D.; Trynka, G.; Raj, T.; Terao, C.; Ikari, K.; Kochi, Y.; Ohmura, K.; Suzuki, A.; Yoshida, S.; et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 2014, 506, 376–381. [Google Scholar] [CrossRef] [PubMed]
  122. Deane, K.D.; Norris, J.M.; Holers, V.M. Preclinical rheumatoid arthritis: Identification, evaluation, and future directions for investigation. Rheum. Dis. Clin. N. Am. 2010, 36, 213–241. [Google Scholar] [CrossRef] [Green Version]
  123. Lucchino, B.; Spinelli, F.R.; Iannuccelli, C.; Guzzo, M.P.; Conti, F.; Di Franco, M. Mucosa-Environment Interactions in the Pathogenesis of Rheumatoid Arthritis. Cells 2019, 8, 700. [Google Scholar] [CrossRef] [Green Version]
  124. Deane, K.D.; El-Gabalawy, H. Pathogenesis and prevention of rheumatic disease: Focus on preclinical RA and SLE. Nat. Rev. Rheumatol. 2014, 10, 212–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Korczowska, I. Rheumatoid arthritis susceptibility genes: An overview. World J. Orthop. 2014, 5, 544–549. [Google Scholar] [CrossRef] [PubMed]
  126. Saad, M.N.; Mabrouk, M.S.; Eldeib, A.M.; Shaker, O.G. Identification of rheumatoid arthritis biomarkers based on single nucleotide polymorphisms and haplotype blocks: A systematic review and meta-analysis. J. Adv. Res. 2016, 7, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Walsh, A.M.; Whitaker, J.W.; Huang, C.C.; Cherkas, Y.; Lamberth, S.L.; Brodmerkel, C.; Curran, M.E.; Dobrin, R. Integrative genomic deconvolution of rheumatoid arthritis GWAS loci into gene and cell type associations. Genome Biol. 2016, 17, 79. [Google Scholar] [CrossRef] [Green Version]
  128. Li, P.; Wang, X.; Guo, X.; Wen, Y.; Liu, L.; Liang, X.; Du, Y.; Wu, C.; Wang, S.; Zhang, F. Integrative analysis of genome-wide association study and expression quantitative trait loci datasets identified various immune cell-related pathways for rheumatoid arthritis. Ann. Hum. Genet. 2020, 84, 72–79. [Google Scholar] [CrossRef] [PubMed]
  129. Liu, T.; Lin, X.; Yu, H. Identifying genes related with rheumatoid arthritis via system biology analysis. Gene 2015, 571, 97–106. [Google Scholar] [CrossRef]
  130. O’Shea, J.J.; Plenge, R. JAK and STAT signaling molecules in immunoregulation and immune-mediated disease. Immunity 2012, 36, 542–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Diogo, D.; Okada, Y.; Plenge, R.M. Genome-wide association studies to advance our understanding of critical cell types and pathways in rheumatoid arthritis: Recent findings and challenges. Curr. Opin. Rheumatol. 2014, 26, 85–92. [Google Scholar] [CrossRef] [PubMed]
  132. Noack, M.; Miossec, P. Selected cytokine pathways in rheumatoid arthritis. Semin. Immunopathol. 2017, 39, 365–383. [Google Scholar] [CrossRef]
  133. Yap, H.-Y.; Tee, S.Z.-Y.; Wong, M.M.-T.; Chow, S.-K.; Peh, S.-C.; Teow, S.-Y. Pathogenic Role of Immune Cells in Rheumatoid Arthritis: Implications in Clinical Treatment and Biomarker Development. Cells 2018, 7, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Crofford, L.J. The Role of COX-2 in Rheumatoid Arthritis Synovial Tissues. Arthritis Res. 2000, 1, S30. [Google Scholar] [CrossRef] [Green Version]
  135. Hansildaar, R.; Vedder, D.; Baniaamam, M.; Tausche, A.K.; Gerritsen, M.; Nurmohamed, M.T. Cardiovascular risk in inflammatory arthritis: Rheumatoid arthritis and gout. Lancet Rheumatol. 2020. [Google Scholar] [CrossRef]
  136. Cunnane, G.; Whitehead, A.S. Amyloid precursors and amyloidosis in rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 1999, 13, 615–628. [Google Scholar] [CrossRef]
  137. DeMizio, D.J.; Geraldino-Pardilla, L.B. Autoimmunity and Inflammation Link to Cardiovascular Disease Risk in Rheumatoid Arthritis. Rheumatol. Ther. 2020, 7, 19–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Giles, J.T.; Danielides, S.; Szklo, M.; Post, W.S.; Blumenthal, R.S.; Petri, M.; Schreiner, P.J.; Budoff, M.; Detrano, R.; Bathon, J.M. Insulin resistance in rheumatoid arthritis: Disease-related indicators and associations with the presence and progression of subclinical atherosclerosis. Arthritis Rheumatol. 2015, 67, 626–636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Nakano, K.; Whitaker, J.W.; Boyle, D.L.; Wang, W.; Firestein, G.S. DNA methylome signature in rheumatoid arthritis. Ann. Rheum. Dis. 2013, 72, 110–117. [Google Scholar] [CrossRef] [PubMed]
  140. Karami, J.; Aslani, S.; Tahmasebi, M.N.; Mousavi, M.J.; Sharafat Vaziri, A.; Jamshidi, A.; Farhadi, E.; Mahmoudi, M. Epigenetics in rheumatoid arthritis; fibroblast-like synoviocytes as an emerging paradigm in the pathogenesis of the disease. Immunol. Cell Biol. 2020, 98, 171–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Glant, T.T.; Mikecz, K.; Rauch, T.A. Epigenetics in the pathogenesis of rheumatoid arthritis. BMC Med. 2014, 12, 35. [Google Scholar] [CrossRef] [Green Version]
  142. Mahendran, S.M.; Keystone, E.C.; Krawetz, R.J.; Liang, K.; Diamandis, E.P.; Chandran, V. Elucidating the endogenous synovial fluid proteome and peptidome of inflammatory arthritis using label-free mass spectrometry. Clin. Proteom. 2019, 16, 23. [Google Scholar] [CrossRef] [Green Version]
  143. Burska, A.; Boissinot, M.; Ponchel, F. Cytokines as biomarkers in rheumatoid arthritis. Mediat. Inflamm. 2014, 2014, 545493. [Google Scholar] [CrossRef]
  144. Aterido, A.; Tornero, J.; Blanco, F.; Fernández-Gutiérrez, B.; González, A.; Cañete, J.D.; Maymó, J.; Alperi-López, M.; Olivé, À.; Corominas, H.; et al. FRI0100 Multi-omics analysis identifies a gene signature associated with the clinical response to anti-tnf therapy in rheumatoid arthritis. J. Ann. Rheum. Dis. 2018, 77, 594. [Google Scholar] [CrossRef]
  145. Romão, V.C.; Vital, E.M.; Fonseca, J.E.; Buch, M.H. Right drug, right patient, right time: Aspiration or future promise for biologics in rheumatoid arthritis? Arthritis Res. Ther. 2017, 19, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Lindstrom, T.M.; Robinson, W.H. Biomarkers for rheumatoid arthritis: Making it personal. Scand. J. Clin. Lab. Investig. 2010, 242, 79–84. [Google Scholar] [CrossRef] [PubMed]
  147. Tao, W.; Concepcion, A.N.; Vianen, M.; Marijnissen, A.C.A.; Lafeber, F.P.G.J.; Radstake, T.R.D.J.; Pandit, A. Multi-omics and machine learning accurately predicts clinical response to Adalimumab and Etanercept therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2021, 73, 212–222. [Google Scholar] [CrossRef]
  148. Singh, V.; Kalliolias, G.D.; Ostaszewski, M.; Veyssiere, M.; Pilalis, E.; Gawron, P.; Mazein, A.; Bonnet, E.; Petit-Teixeira, E.; Niarakis, A. RA-map: Building a state-of-the-art interactive knowledge base for rheumatoid arthritis. Database J. Biol. Databases Curation 2020, 2020. [Google Scholar] [CrossRef] [PubMed]
  149. Teodoro, A.J. Bioactive Compounds of Food: Their Role in the Prevention and Treatment of Diseases. Oxidative Med. Cell. Longev. 2019, 2019, 3765986. [Google Scholar] [CrossRef] [Green Version]
  150. Christensen, J.J.; Ulven, S.M.; Thoresen, M.; Westerman, K.; Holven, K.B.; Andersen, L.F. Associations between dietary patterns and gene expression pattern in peripheral blood mononuclear cells: A cross-sectional study. Nutr. Metab. Cardiovasc. Dis. 2020, 30, 2111–2122. [Google Scholar] [CrossRef]
  151. Nguyen, Y.; Salliot, C.; Gelot, A.; Gambaretti, J.; Mariette, X.; Boutron-Ruault, M.-C.; Seror, R. Mediterranean diet and risk of rheumatoid arthritis: Findings from the French E3N-EPIC cohort study. Arthritis Rheumatol. 2021, 73, 69–77. [Google Scholar] [CrossRef] [PubMed]
  152. Konstantinidou, V.; Covas, M.I.; Sola, R.; Fitó, M. Up-to date knowledge on the in vivo transcriptomic effect of the Mediterranean diet in humans. Mol. Nutr. Food Res. 2013, 57, 772–783. [Google Scholar] [CrossRef] [PubMed]
  153. Camargo, A.; Delgado-Lista, J.; Garcia-Rios, A.; Cruz-Teno, C.; Yubero-Serrano, E.M.; Perez-Martinez, P.; Gutierrez-Mariscal, F.M.; Lora-Aguilar, P.; Rodriguez-Cantalejo, F.; Fuentes-Jimenez, F.; et al. Expression of proinflammatory, proatherogenic genes is reduced by the Mediterranean diet in elderly people. Br. J. Nutr. 2012, 108, 500–508. [Google Scholar] [CrossRef] [PubMed]
  154. Yubero-Serrano, E.M.; Gonzalez-Guardia, L.; Rangel-Zuñiga, O.; Delgado-Lista, J.; Gutierrez-Mariscal, F.M.; Perez-Martinez, P.; Delgado-Casado, N.; Cruz-Teno, C.; Tinahones, F.J.; Villalba, J.M.; et al. Mediterranean diet supplemented with coenzyme Q10 modifies the expression of proinflammatory and endoplasmic reticulum stress-related genes in elderly men and women. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2012, 67, 3–10. [Google Scholar] [CrossRef] [Green Version]
  155. Arpón, A.; Riezu-Boj, J.I.; Milagro, F.I.; Marti, A.; Razquin, C.; Martínez-González, M.A.; Corella, D.; Estruch, R.; Casas, R.; Fitó, M.; et al. Adherence to Mediterranean diet is associated with methylation changes in inflammation-related genes in peripheral blood cells. J. Physiol. Biochem. 2016, 73, 445–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Davis, C.; Bryan, J.; Hodgson, J.; Murphy, K. Definition of the Mediterranean Diet; a Literature Review. Nutrients 2015, 7, 9139–9153. [Google Scholar] [CrossRef]
  157. Oliviero, F.; Spinella, P.; Fiocco, U.; Ramonda, R.; Sfriso, P.; Punzi, L. How the Mediterranean diet and some of its components modulate inflammatory pathways in arthritis. Swiss Med. Wkly. 2015, 145, w14190. [Google Scholar] [CrossRef] [PubMed]
  158. Barbosa, D.S.; Simão, A.C.; Dichi, I. Chapter 114—Use of Olive Oil in Patients with Rheumatoid Arthritis. In Olives and Olive Oil in Health and Disease Prevention; Preedy, V.R., Watson, R.R., Eds.; Academic Press: San Diego, CA, USA, 2010; pp. 1057–1064. [Google Scholar] [CrossRef]
  159. Aparicio-Soto, M.; Sánchéz-Hidalgo, M.; Cárdeno, A.; Lucena, J.M.; Gonzáléz-Escribano, F.; Castillo, M.J.; Alarcón-de-la-Lastra, C. The phenolic fraction of extra virgin olive oil modulates the activation and the inflammatory response of T cells from patients with systemic lupus erythematosus and healthy donors. Mol. Nutr. Food Res. 2017, 61. [Google Scholar] [CrossRef]
  160. Castañer, O.; Covas, M.I.; Khymenets, O.; Nyyssonen, K.; Konstantinidou, V.; Zunft, H.F.; de la Torre, R.; Muñoz-Aguayo, D.; Vila, J.; Fitó, M. Protection of LDL from oxidation by olive oil polyphenols is associated with a downregulation of CD40-ligand expression and its downstream products in vivo in humans. Am. J. Clin. Nutr. 2012, 95, 1238–1244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Guo, Y.; Walsh, A.M.; Fearon, U.; Smith, M.D.; Wechalekar, M.D.; Yin, X.; Cole, S.; Orr, C.; McGarry, T.; Canavan, M.; et al. CD40L-Dependent Pathway Is Active at Various Stages of Rheumatoid Arthritis Disease Progression. J. Immunol. 2017, 198, 4490. [Google Scholar] [CrossRef] [Green Version]
  162. Melguizo-Rodríguez, L.; Manzano-Moreno, F.J.; Illescas-Montes, R.; Ramos-Torrecillas, J.; Luna-Bertos, E.d.; Ruiz, C.; García-Martínez, O. Bone Protective Effect of Extra-Virgin Olive Oil Phenolic Compounds by Modulating Osteoblast Gene Expression. Nutrients 2019, 11, 1722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Kostoglou-Athanassiou, I.; Athanassiou, L.; Athanassiou, P. The Effect of Omega-3 Fatty Acids on Rheumatoid Arthritis. Mediterr. J. Rheumatol. 2020, 31, 190–194. [Google Scholar] [CrossRef] [PubMed]
  164. Lourdudoss, C.; Di Giuseppe, D.; Wolk, A.; Westerlind, H.; Klareskog, L.; Alfredsson, L.; van Vollenhoven, R.F.; Lampa, J. Dietary Intake of Polyunsaturated Fatty Acids and Pain in Spite of Inflammatory Control Among Methotrexate-Treated Early Rheumatoid Arthritis Patients. Arthritis Care Res. 2018, 70, 205–212. [Google Scholar] [CrossRef] [PubMed]
  165. Di Giuseppe, D.; Wallin, A.; Bottai, M.; Askling, J.; Wolk, A. Long-term intake of dietary long-chain n-3 polyunsaturated fatty acids and risk of rheumatoid arthritis: A prospective cohort study of women. Ann. Rheum. Dis. 2014, 73, 1949–1953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Shapiro, J.A.; Koepsell, T.D.; Voigt, L.F.; Dugowson, C.E.; Kestin, M.; Nelson, J.L. Diet and rheumatoid arthritis in women: A possible protective effect of fish consumption. Epidemiology 1996, 7, 256–263. [Google Scholar] [CrossRef]
  167. Gioxari, A.; Kaliora, A.C.; Marantidou, F.; Panagiotakos, D.P. Intake of ω-3 polyunsaturated fatty acids in patients with rheumatoid arthritis: A systematic review and meta-analysis. Nutrition 2018, 45, 114–124.e4. [Google Scholar] [CrossRef]
  168. Lee, A.L.; Park, Y. The association between n-3 polyunsaturated fatty acid levels in erythrocytes and the risk of rheumatoid arthritis in Korean women. Ann. Nutr. Metab. 2013, 63, 88–95. [Google Scholar] [CrossRef] [PubMed]
  169. Gan, R.W.; Demoruelle, M.K.; Deane, K.D.; Weisman, M.H.; Buckner, J.H.; Gregersen, P.K.; Mikuls, T.R.; O’Dell, J.R.; Keating, R.M.; Fingerlin, T.E.; et al. Omega-3 fatty acids are associated with a lower prevalence of autoantibodies in shared epitope-positive subjects at risk for rheumatoid arthritis. Ann. Rheum. Dis. 2017, 76, 147–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Basu, A.; Schell, J.; Scofield, R.H. Dietary fruits and arthritis. Food Funct. 2018, 9, 70–77. [Google Scholar] [CrossRef]
  171. Murakami, I.; Murakami, K.; Hashimoto, M.; Tanaka, M.; Ito, H.; Fujii, T.; Torii, M.; Ikeda, K.; Kuwabara, A.; Tanaka, K.; et al. Intake frequency of vegetables or seafoods negatively correlates with disease activity of rheumatoid arthritis. PLoS ONE 2020, 15, e0228852. [Google Scholar] [CrossRef] [PubMed]
  172. Alwarith, J.; Kahleova, H.; Rembert, E.; Yonas, W.; Dort, S.; Calcagno, M.; Burgess, N.; Crosby, L.; Barnard, N.D. Nutrition Interventions in Rheumatoid Arthritis: The Potential Use of Plant-Based Diets. A Review. Front. Nutr. 2019, 6, 141. [Google Scholar] [CrossRef]
  173. Comee, L.; Taylor, C.A.; Nahikian-Nelms, M.; Ganesan, L.P.; Krok-Schoen, J.L. Dietary patterns and nutrient intake of individuals with rheumatoid arthritis and osteoarthritis in the United States. Nutrition 2019, 67–68, 110533. [Google Scholar] [CrossRef] [Green Version]
  174. Bärebring, L.; Winkvist, A.; Gjertsson, I.; Lindqvist, H.M. Poor Dietary Quality Is Associated with Increased Inflammation in Swedish Patients with Rheumatoid Arthritis. Nutrients 2018, 10, 1535. [Google Scholar] [CrossRef] [Green Version]
  175. Hu, Y.; Sparks, J.A.; Malspeis, S.; Costenbader, K.H.; Hu, F.B.; Karlson, E.W.; Lu, B. Long-term dietary quality and risk of developing rheumatoid arthritis in women. Ann. Rheum. Dis. 2017, 76, 1357–1364. [Google Scholar] [CrossRef]
  176. Yang, C.M.; Chen, Y.W.; Chi, P.L.; Lin, C.C.; Hsiao, L.D. Resveratrol inhibits BK-induced COX-2 transcription by suppressing acetylation of AP-1 and NF-κB in human rheumatoid arthritis synovial fibroblasts. Biochem. Pharmacol. 2017, 132, 77–91. [Google Scholar] [CrossRef] [PubMed]
  177. Oh, Y.-C.; Kang, O.-H.; Choi, J.-G.; Chae, H.-S.; Lee, Y.-S.; Brice, O.-O.; Jung, H.J.; Hong, S.-H.; Lee, Y.-M.; Kwon, D.-Y. Anti-Inflammatory Effect of Resveratrol by Inhibition of IL-8 Production in LPS-Induced THP-1. Cells 2009, 37, 1203–1214. [Google Scholar] [CrossRef]
  178. Rajasekhar, M.; Olsson, A.M.; Steel, K.J.A.; Georgouli, M.; Ranasinghe, U.; Brender Read, C.; Frederiksen, K.S.; Taams, L.S. MicroRNA-155 contributes to enhanced resistance to apoptosis in monocytes from patients with rheumatoid arthritis. J. Autoimmun. 2017, 79, 53–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Xiong, G.; Huang, Z.; Jiang, H.; Pan, Z.; Xie, J.; Wang, S. Inhibition of microRNA-21 decreases the invasiveness of fibroblast-like synoviocytes in rheumatoid arthritis via TGFβ/Smads signaling pathway. Iran. J. Basic Med. Sci. 2016, 19, 787–793. [Google Scholar] [PubMed]
  180. Li, X.; Tian, F.; Wang, F. Rheumatoid arthritis-associated microRNA-155 targets SOCS1 and upregulates TNF-α and IL-1β in PBMCs. Int. J. Mol. Sci. 2013, 14, 23910–23921. [Google Scholar] [CrossRef] [Green Version]
  181. Källberg, H.; Jacobsen, S.; Bengtsson, C.; Pedersen, M.; Padyukov, L.; Garred, P.; Frisch, M.; Karlson, E.W.; Klareskog, L.; Alfredsson, L. Alcohol consumption is associated with decreased risk of rheumatoid arthritis: Results from two Scandinavian case-control studies. Ann. Rheum. Dis. 2009, 68, 222–227. [Google Scholar] [CrossRef] [PubMed]
  182. Sundström, B.; Johansson, I.; Rantapää-Dahlqvist, S. Interaction between dietary sodium and smoking increases the risk for rheumatoid arthritis: Results from a nested case-control study. Rheumatology 2015, 54, 487–493. [Google Scholar] [CrossRef] [Green Version]
  183. Iris, M.; Tsou, P.-S.; Sawalha, A.H. Caffeine downregulates inflammatory pathways involved in autoimmunity. bioRxiv 2017, 241539. [Google Scholar] [CrossRef] [Green Version]
  184. Baghel, S.S.; Thakran, R.D.; Messi, C.; Kapoor, S.; Garg, S.; Kashyap, V.; Zaheer, Q.; Malaviya, A.N. SAT0746-HPR Coffee decreases methotrexate intolerance and increases its compliance in rheumatoid arthritis (RA): A study by rheumatology nurse counsellors. J. Ann. Rheum. Dis. 2018, 77, 1833–1834. [Google Scholar] [CrossRef]
  185. Van Bussel, I.P.G.; Jolink-Stoppelenburg, A.; De Groot, C.P.G.M.; Müller, M.R.; Afman, L.A. Differences in genome-wide gene expression response in peripheral blood mononuclear cells between young and old men upon caloric restriction. Genes Nutr. 2016, 11, 13. [Google Scholar] [CrossRef] [Green Version]
  186. Dao, M.C.; Sokolovska, N.; Brazeilles, R.; Affeldt, S.; Pelloux, V.; Prifti, E.; Chilloux, J.; Verger, E.O.; Kayser, B.D.; Aron-Wisnewsky, J.; et al. A Data Integration Multi-Omics Approach to Study Calorie Restriction-Induced Changes in Insulin Sensitivity. Front. Physiol. 2019, 9. [Google Scholar] [CrossRef]
  187. Roytblat, L.; Rachinsky, M.; Fisher, A.; Greemberg, L.; Shapira, Y.; Douvdevani, A.; Gelman, S. Raised interleukin-6 levels in obese patients. Obes. Res. 2000, 8, 673–675. [Google Scholar] [CrossRef] [PubMed]
  188. Hotamisligil, G.S.; Arner, P.; Caro, J.F.; Atkinson, R.L.; Spiegelman, B.M. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J. Clin. Investig. 1995, 95, 2409–2415. [Google Scholar] [CrossRef] [PubMed]
  189. Gallagher, L.; Cregan, S.; Biniecka, M.; Cunningham, C.; Veale, D.J.; Kane, D.J.; Fearon, U.; Mullan, R.H. Insulin-Resistant Pathways Are Associated with Disease Activity in Rheumatoid Arthritis and Are Subject to Disease Modification Through Metabolic Reprogramming: A Potential Novel Therapeutic Approach. Arthritis Rheumatol. 2020, 72, 896–902. [Google Scholar] [CrossRef] [PubMed]
  190. Brenner, M.; Laragione, T.; Gulko, P.S. Short-term low-magnesium diet reduces autoimmune arthritis severity and synovial tissue gene expression. Physiol. Genom. 2017, 49, 238–242. [Google Scholar] [CrossRef] [PubMed]
  191. Li, M.; Zhai, L.; Wei, W. High-Methionine Diet Attenuates Severity of Arthritis and Modulates IGF-I Related Gene Expressions in an Adjuvant Arthritis Rats Model. Mediat. Inflamm. 2016, 2016, 9280529. [Google Scholar] [CrossRef] [PubMed]
  192. Imada, K.; Lin, N.; Liu, C.; Lu, A.; Chen, W.; Yano, M.; Sato, T.; Ito, A. Nobiletin, a citrus polymethoxy flavonoid, suppresses gene expression and production of aggrecanases-1 and -2 in collagen-induced arthritic mice. Biochem. Biophys. Res. Commun. 2008, 373, 181–185. [Google Scholar] [CrossRef] [PubMed]
  193. Norling, L.V.; Headland, S.E.; Dalli, J.; Arnardottir, H.H.; Haworth, O.; Jones, H.R.; Irimia, D.; Serhan, C.N.; Perretti, M. Proresolving and cartilage-protective actions of resolvin D1 in inflammatory arthritis. JCI Insight 2016, 1, e85922. [Google Scholar] [CrossRef] [PubMed]
  194. Lin, I.C.; Yamashita, S.; Murata, M.; Kumazoe, M.; Tachibana, H. Equol suppresses inflammatory response and bone erosion due to rheumatoid arthritis in mice. J. Nutr. Biochem. 2016, 32, 101–106. [Google Scholar] [CrossRef] [PubMed]
  195. Ahmed, S. Green tea polyphenol epigallocatechin 3-gallate in arthritis: Progress and promise. Arthritis Res. 2010, 12, 208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Cassotta, M.; Pistollato, F.; Battino, M. Rheumatoid arthritis research in the 21st century: Limitations of traditional models, new technologies, and opportunities for a human biology-based approach. Altex 2020, 37, 223–242. [Google Scholar] [CrossRef] [PubMed]
  197. Steves, C.J.; Bird, S.; Williams, F.M.; Spector, T.D. The Microbiome and Musculoskeletal Conditions of Aging: A Review of Evidence for Impact and Potential Therapeutics. J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 2016, 31, 261–269. [Google Scholar] [CrossRef] [PubMed]
  198. Xu, H.; Zhao, H.; Fan, D.; Liu, M.; Cao, J.; Xia, Y.; Ju, D.; Xiao, C.; Guan, Q. Interactions between Gut Microbiota and Immunomodulatory Cells in Rheumatoid Arthritis. Mediat. Inflamm. 2020, 2020, 1430605. [Google Scholar] [CrossRef]
  199. Van de Wiele, T.; Van Praet, J.T.; Marzorati, M.; Drennan, M.B.; Elewaut, D. How the microbiota shapes rheumatic diseases. Nat. Rev. Rheumatol. 2016, 12, 398–411. [Google Scholar] [CrossRef]
  200. Zhang, X.; Zhang, D.; Jia, H.; Feng, Q.; Wang, D.; Liang, D.; Wu, X.; Li, J.; Tang, L.; Li, Y.; et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat. Med. 2015, 21, 895–905. [Google Scholar] [CrossRef] [PubMed]
  201. Zhong, D.; Wu, C.; Zeng, X.; Wang, Q. The role of gut microbiota in the pathogenesis of rheumatic diseases. Clin. Rheumatol. 2018, 37, 25–34. [Google Scholar] [CrossRef]
  202. Vaahtovuo, J.; Munukka, E.; Korkeamäki, M.; Luukkainen, R.; Toivanen, P. Fecal Microbiota in Early Rheumatoid Arthritis. J. Rheumatol. 2008, 35, 1500–1505. [Google Scholar]
  203. Chen, J.; Wright, K.; Davis, J.M.; Jeraldo, P.; Marietta, E.V.; Murray, J.; Nelson, H.; Matteson, E.L.; Taneja, V. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med. 2016, 8, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Wells, P.M.; Adebayo, A.S.; Bowyer, R.C.E.; Freidin, M.B.; Finckh, A.; Strowig, T.; Lesker, T.R.; Alpizar-Rodriguez, D.; Gilbert, B.; Kirkham, B.; et al. Associations between gut microbiota and genetic risk for rheumatoid arthritis in the absence of disease. Lancet Rheumatol. 2020, 2, e418–e427. [Google Scholar] [CrossRef]
  205. Singh, R.K.; Chang, H.-W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [Green Version]
  206. Paolino, S.; Pacini, G.; Patanè, M.; Alessandri, E.; Cattelan, F.; Goegan, F.; Pizzorni, C.; Gotelli, E.; Cutolo, M. Interactions between microbiota, diet/nutrients and immune/inflammatory response in rheumatic diseases: Focus on rheumatoid arthritis. Reumatologia 2019, 57, 151–157. [Google Scholar] [CrossRef]
  207. Zhenyukh, O.; Civantos, E.; Ruiz-Ortega, M.; Sánchez, M.S.; Vázquez, C.; Peiró, C.; Egido, J.; Mas, S. High concentration of branched-chain amino acids promotes oxidative stress, inflammation and migration of human peripheral blood mononuclear cells via mTORC1 activation. Free Radic. Biol. Med. 2017, 104, 165–177. [Google Scholar] [CrossRef] [PubMed]
  208. Coras, R.; Murillo-Saich, J.D.; Guma, M. Circulating Pro- and Anti-Inflammatory Metabolites and Its Potential Role in Rheumatoid Arthritis Pathogenesis. Cells 2020, 9, 827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Wammers, M.; Schupp, A.-K.; Bode, J.G.; Ehlting, C.; Wolf, S.; Deenen, R.; Köhrer, K.; Häussinger, D.; Graf, D. Reprogramming of pro-inflammatory human macrophages to an anti-inflammatory phenotype by bile acids. Sci. Rep. 2018, 8, 255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Ratajczak, W.; Rył, A.; Mizerski, A.; Walczakiewicz, K.; Sipak, O.; Laszczyńska, M. Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim. Pol. 2019, 66, 1–12. [Google Scholar] [CrossRef] [Green Version]
  211. Vinolo, M.A.R.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of inflammation by short chain fatty acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [Green Version]
  213. Li, M.; van Esch, B.C.A.M.; Henricks, P.A.J.; Folkerts, G.; Garssen, J. The Anti-inflammatory Effects of Short Chain Fatty Acids on Lipopolysaccharide- or Tumor Necrosis Factor α-Stimulated Endothelial Cells via Activation of GPR41/43 and Inhibition of HDACs. Front. Pharmacol. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
  214. Chan, M.M.; Yang, X.; Wang, H.; Saaoud, F.; Sun, Y.; Fong, D. The Microbial Metabolite Trimethylamine N-Oxide Links Vascular Dysfunctions and the Autoimmune Disease Rheumatoid Arthritis. Nutrients 2019, 11, 1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Pattison, D.J.; Symmons, D.P.; Lunt, M.; Welch, A.; Luben, R.; Bingham, S.A.; Khaw, K.T.; Day, N.E.; Silman, A.J. Dietary risk factors for the development of inflammatory polyarthritis: Evidence for a role of high level of red meat consumption. Arthritis Rheum. 2004, 50, 3804–3812. [Google Scholar] [CrossRef]
  216. Wang, Z.; Bergeron, N.; Levison, B.S.; Li, X.S.; Chiu, S.; Jia, X.; Koeth, R.A.; Li, L.; Wu, Y.; Tang, W.H.W.; et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 2019, 40, 583–594. [Google Scholar] [CrossRef]
  217. Chen, B.; Zhao, Y.; Li, S.; Yang, L.; Wang, H.; Wang, T.; Bin, S.; Gai, Z.; Heng, X.; Zhang, C.; et al. Variations in oral microbiome profiles in rheumatoid arthritis and osteoarthritis with potential biomarkers for arthritis screening. Sci. Rep. 2018, 8, 17126. [Google Scholar] [CrossRef] [PubMed]
  218. Tong, Y.; Zheng, L.; Qing, P.; Zhao, H.; Li, Y.; Su, L.; Zhang, Q.; Zhao, Y.; Luo, Y.; Liu, Y. Oral Microbiota Perturbations Are Linked to High Risk for Rheumatoid Arthritis. Front. Cell. Infect. Microbiol. 2020, 9. [Google Scholar] [CrossRef] [PubMed]
  219. Lerner, A.; Aminov, R.; Matthias, T. Dysbiosis May Trigger Autoimmune Diseases via Inappropriate Post-Translational Modification of Host Proteins. Front. Microbial. 2016, 7. [Google Scholar] [CrossRef]
  220. Tomar, N.; Gupta, C.; Kaushik, M.; Wadhawan, A. Nutrigenomics: A perio-nutrition interrelationship. J. Oral Res. Rev. 2017, 9, 32–36. [Google Scholar]
  221. Singh, S.; Kalra, P. Nutrigenomics in periodontics—An overview. Int. J. Oral Health Dent. 2017, 3, 74–76. [Google Scholar] [CrossRef]
  222. Badsha, H. Role of Diet in Influencing Rheumatoid Arthritis Disease Activity. Open Rheumatol. J. 2018, 12, 19–28. [Google Scholar] [CrossRef] [Green Version]
  223. Forsyth, C.; Kouvari, M.; D’Cunha, N.M.; Georgousopoulou, E.N.; Panagiotakos, D.B.; Mellor, D.D.; Kellett, J.; Naumovski, N. The effects of the Mediterranean diet on rheumatoid arthritis prevention and treatment: A systematic review of human prospective studies. Rheumatol. Int. 2018, 38, 737–747. [Google Scholar] [CrossRef]
  224. Young, S.P.; Kapoor, S.R.; Viant, M.R.; Byrne, J.J.; Filer, A.; Buckley, C.D.; Kitas, G.D.; Raza, K. The impact of inflammation on metabolomic profiles in patients with arthritis. Arthritis Rheum. 2013, 65, 2015–2023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Wang, M.; Huang, J.; Fan, H.; He, D.; Zhao, S.; Shu, Y.; Li, H.; Liu, L.; Lu, S.; Xiao, C.; et al. Treatment of Rheumatoid Arthritis Using Combination of Methotrexate and Tripterygium Glycosides Tablets-A Quantitative Plasma Pharmacochemical and Pseudotargeted Metabolomic Approach. Front. Pharm. 2018, 9, 1051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Hedrick, V.E.; Dietrich, A.M.; Estabrooks, P.A.; Savla, J.; Serrano, E.; Davy, B.M. Dietary biomarkers: Advances, limitations and future directions. Nutr. J. 2012, 11, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Seok, J.; Warren, H.S.; Cuenca, A.G.; Mindrinos, M.N.; Baker, H.V.; Xu, W.; Richards, D.R.; McDonald-Smith, G.P.; Gao, H.; Hennessy, L.; et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc. Natl. Acad. Sci. USA 2013, 110, 3507–3512. [Google Scholar] [CrossRef] [Green Version]
  228. Fritsche, K.L. The science of fatty acids and inflammation. Adv. Nutr. 2015, 6, 293S–301S. [Google Scholar] [CrossRef] [PubMed]
  229. Cassotta, M.; Forbes-Hernández, T.Y.; Calderón Iglesias, R.; Ruiz, R.; Elexpuru Zabaleta, M.; Giampieri, F.; Battino, M. Links between Nutrition, Infectious Diseases, and Microbiota: Emerging Technologies and Opportunities for Human-Focused Research. Nutrients 2020, 12, 1827. [Google Scholar] [CrossRef] [PubMed]
  230. Karczewski, K.J.; Snyder, M.P. Integrative omics for health and disease. Nat. Rev. Genet. 2018, 19, 299–310. [Google Scholar] [CrossRef] [PubMed]
  231. Paul, B.J.; Kandy, H.I.; Krishnan, V. Pre-rheumatoid arthritis and its prevention. Eur. J. Rheumatol. 2017, 4, 161–165. [Google Scholar] [CrossRef]
  232. Pinu, F.R.; Beale, D.J.; Paten, A.M.; Kouremenos, K.; Swarup, S.; Schirra, H.J.; Wishart, D. Systems Biology and Multi-Omics Integration: Viewpoints from the Metabolomics Research Community. Metabolites 2019, 9, 76. [Google Scholar] [CrossRef] [Green Version]
  233. Fondi, M.; Liò, P. Multi -omics and metabolic modelling pipelines: Challenges and tools for systems microbiology. Microbiol. Res. 2015, 171, 52–64. [Google Scholar] [CrossRef] [PubMed]
  234. Dihazi, H.; Asif, A.R.; Beissbarth, T.; Bohrer, R.; Feussner, K.; Feussner, I.; Jahn, O.; Lenz, C.; Majcherczyk, A.; Schmidt, B.; et al. Integrative omics—From data to biology. Expert Rev. Proteom. 2018, 15, 463–466. [Google Scholar] [CrossRef] [PubMed]
Figure 1. An integrative ‘omics’ approach will lead to a more human-focused research, precision nutrition, eventually resulting in better prevention strategies, as well as better management of rheumatoid arthritis.
Figure 1. An integrative ‘omics’ approach will lead to a more human-focused research, precision nutrition, eventually resulting in better prevention strategies, as well as better management of rheumatoid arthritis.
Nutrients 13 00763 g001
Table 1. Examples of ‘omics’ approaches and emerging technologies employed in nutritional research. Five major categories and their subcategories are shown.
Table 1. Examples of ‘omics’ approaches and emerging technologies employed in nutritional research. Five major categories and their subcategories are shown.
ApproachSubcategoryTargetsTechniquesReferences
GenomicsGenomicsGenes (DNA sequence)Second generation: Illumina; SOLID; Ion Torrent.
Third generation: PacBio; SMRT-seq; Illumina Tru-seq Synthetic Long-Read technology; Oxford Nanopore Technologies sequencing platform
[65,94]
EpigenomicsModification of DNA and DNA- binding proteinsWhole-genome bisulfite sequencing (for DNA Methylation Analysis); ChIP-seq (for DNA–Protein Interaction Analysis); ATAC-Seq (for Chromatin Accessibility Analysis)[95,96,97]
TranscriptomicsTranscriptomicsmRNARNA-microarrays; RNA-seq: Illumina, SOLID, Ion Torrent (second generation);
PacBio, SMRT-seq, Illumina TruSeq Synthetic Long-Read technology, Oxford Nanopore Technologies sequencing platform (third generation)
[47,48,49,50]
ncRNA-omicsnon-coding RNA (including microRNA) [57,98,99,100]
ProteomicsProteomicsProteinsProtein-microarrays; NMR Spectroscopy; MS; single-cell and ultrasensitive protein analyses[79,80,81]
InteractomicsProtein-protein interaction, protein-small molecules interactionTAP; Affinity Chromatography; Coimmunoprecipitation; Protein arrays; PFC; Phage display, NMR spectroscopy[84,85,101]
MetabolomicsMetabolomicsMetabolitesNMR; 1H NMR; MS; MALDI-TOF; SIMS; FTICR-MS[72,73,74,75,76,77,78]
LipidomicsLipids
AminomicsAminoacids
MicrobiomicsMicrobiomicsHuman Microbiota (including bacteria,
fungi, protozoa, and viruses)
[87,88,89,90,91,102,103]
Meta-genomicsMicrobiota DNA
Meta-transcriptomicsMicrobiota RNAs
Meta-proteomicsMicrobiota proteins
Meta-bolomicsMicrobiota metabolites
Abbreviations: ATAC-Seq, Assay for transposase-accessible chromatin-Squencing; ChIP-seq, Chromatin immunoprecipitation-Sequencing; FTICR-MS, Fourier transform ion cyclotron resonance-Mass Spectrometry. 1H NMR, Proton Nuclear Magnetic Resonance spectroscopy; lncRNAs, long non-coding RNAs; MALDI-TOF, Matrix-Assisted Laser Desorption Ionization Time of Flight; MS, Mass Spectrometry; NMR, Nuclear Magnetic Resonance; PacBio, Pacific Biosciences; PFC, Protein fragment Complementation; SIMS, Secondary Ion Mass Spectrometry; SMRT-seq, Single Molecule Real Time sequencing; SOLID: Sequencing by Oligonucleotide Ligation and Detection; TAP, Tandem Affinity Purification.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cassotta, M.; Forbes-Hernandez, T.Y.; Cianciosi, D.; Elexpuru Zabaleta, M.; Sumalla Cano, S.; Dominguez, I.; Bullon, B.; Regolo, L.; Alvarez-Suarez, J.M.; Giampieri, F.; et al. Nutrition and Rheumatoid Arthritis in the ‘Omics’ Era. Nutrients 2021, 13, 763. https://doi.org/10.3390/nu13030763

AMA Style

Cassotta M, Forbes-Hernandez TY, Cianciosi D, Elexpuru Zabaleta M, Sumalla Cano S, Dominguez I, Bullon B, Regolo L, Alvarez-Suarez JM, Giampieri F, et al. Nutrition and Rheumatoid Arthritis in the ‘Omics’ Era. Nutrients. 2021; 13(3):763. https://doi.org/10.3390/nu13030763

Chicago/Turabian Style

Cassotta, Manuela, Tamara Y. Forbes-Hernandez, Danila Cianciosi, Maria Elexpuru Zabaleta, Sandra Sumalla Cano, Irma Dominguez, Beatriz Bullon, Lucia Regolo, Josè Miguel Alvarez-Suarez, Francesca Giampieri, and et al. 2021. "Nutrition and Rheumatoid Arthritis in the ‘Omics’ Era" Nutrients 13, no. 3: 763. https://doi.org/10.3390/nu13030763

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