Next Article in Journal
What Is Breast in the Bone?
Next Article in Special Issue
Metabolic Response to XD14 Treatment in Human Breast Cancer Cell Line MCF-7
Previous Article in Journal
Screening the Toxicity of Selected Personal Care Products Using Embryo Bioassays: 4-MBC, Propylparaben and Triclocarban
Previous Article in Special Issue
New Potential Biomarker for Methasterone Misuse in Human Urine by Liquid Chromatography Quadrupole Time of Flight Mass Spectrometry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Why Are Omics Technologies Important to Understanding the Role of Nutrition in Inflammatory Bowel Diseases?

by
Lynnette R. Ferguson
1,* and
Matthew P. G. Barnett
2
1
Discipline of Nutrition and Dietetics and Auckland Cancer Research Society, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
2
Food Nutrition & Health Team, Food & Bio-Based Products Group, AgResearch Limited, Palmerston North 4442, New Zealand
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2016, 17(10), 1763; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17101763
Submission received: 31 August 2016 / Revised: 29 September 2016 / Accepted: 10 October 2016 / Published: 21 October 2016
(This article belongs to the Special Issue Metabolomic Technologies in Medicine)

Abstract

:
For many years, there has been confusion about the role that nutrition plays in inflammatory bowel diseases (IBD). It is apparent that good dietary advice for one individual may prove inappropriate for another. As with many diseases, genome-wide association studies across large collaborative groups have been important in revealing the role of genetics in IBD, with more than 200 genes associated with susceptibility to the disease. These associations provide clues to explain the differences in nutrient requirements among individuals. In addition to genes directly involved in the control of inflammation, a number of the associated genes play roles in modulating the gut microbiota. Cell line models enable the generation of hypotheses as to how various bioactive dietary components might be especially beneficial for certain genetic groups. Animal models are necessary to mimic aspects of the complex aetiology of IBD, and provide an important link between tissue culture studies and human trials. Once we are sufficiently confident of our hypotheses, we can then take modified diets to an IBD population that is stratified according to genotype. Studies in IBD patients fed a Mediterranean-style diet have been important in validating our hypotheses and as a proof-of-principle for the application of these sensitive omics technologies to aiding in the control of IBD symptoms.

Graphical Abstract

1. Introduction

Inflammatory bowel diseases (IBD), including Crohn’s disease (CD) and ulcerative colitis (UC), are immune-mediated disorders characterised by chronic and relapsing inflammation of the gastrointestinal tract. Although IBD were rarely observed a century ago, their prevalence has increased in recent years [1]. For example, an Australia/New Zealand survey in 2014 showed high and increasing rates of IBD, especially CD [2]. While there is a clear genetic susceptibility to the diseases [3,4,5,6], diet [7], environment [8], and the resident intestinal microbial population (the microbiota) [9] are also thought to play an important role in the onset and progression of the disease.
Nutrigenomics New Zealand was a research programme established in 2004, with the aim of utilising the burgeoning field of omics technologies in identifying and developing bioactive compounds, functional foods, and/or diets for human consumption [10,11,12]. The initial target of this project was IBD, including both CD and UC. Although these diseases were initially seen merely as providing a proof of principle, they have proved to be extremely relevant to a range of inflammatory disorders, and provide an excellent example of the application of new developments in systems biology to nutrition and food science [13,14,15]. The insights provided are appropriate not only to the maintenance of good health, but also to protection against disease symptomology and progression.
The role of nutrition in IBD has been confused for many years, because it is clear that good dietary advice for one individual with the disease may not be the most appropriate advice for others [16,17,18]. Our hypothesis was that the differences in the impact of nutritional factors between individuals relate to genetic differences. As was already mentioned, there is strong evidence of a genetic basis to IBD, and this has largely been revealed by genome-wide association studies (GWAS), many of them done by the International IBD Genetics Consortium (IIBDGC) [3,19]. It has become increasingly clear that large numbers of subjects are important to reveal the genetic basis of disease, and IBD has almost become a gold standard for international collaborations. It is also important that diet is accurately assessed if gene-diet interactions are to be evaluated [20,21,22].
Where genes important to an individual can be identified, this opens the possibility of exploiting the potential value of gene-specific bioactive food components in regulating the disease, using high throughput (HTP) screens. Again, many of the sensitive technologies provided by systems biology can be valuable in validating the potential effects of these compounds in animal or human studies. The omics approaches to foods enables an order of magnitude step in the understanding of their complexities and interactions with the host, and is sometime termed “foodomics” [23,24]. Foodomics has also been described as part of the host-microbiota-expososome interplay [25]. It highlights health benefits from nutrients and enables a detailed understanding of the metabolic consequences not only on the human body, but also on the gut microbiome. Transcriptomics, proteomics, and metabolic profiling are used, not only for screening food components, foods or diets for novel activity, but also in validating their effects in human populations [10,26,27]. A potential sequence of in vitro to in vivo studies is illustrated in Figure 1.
The rhizome of turmeric (Curcuma longa L., Zingiberaceae) can be crushed to form a yellow-orange powder. Not only has this powder long been used as a spice for cooking, it has also been used in traditional medicines in various countries, including India and China, for several centuries [28,29]. It is claimed to have beneficial effects against several inflammatory diseases, including IBD and cancer. The colour of turmeric is due to curcuminoids, including curcumin, which interact with a number of molecular targets [30]. Curcumin is claimed to act therapeutically in IBD, and is currently being developed for this purpose by the pharmaceutical industry [31,32]. Since various omics technologies have been used in the studies that played an important part in the proof of curcumin’s efficacy, curcumin will be used as an exemplar throughout the text.

2. Defining the Role of Genes in Inflammatory Bowel Disease (IBD)

IBD is almost a classic example of the importance of genetics in disease development. It was long recognised that there was a familial basis to the disease, and twin studies provided important information [33]. Specifically, it was shown that monozygotic twins had a higher concordance of disease risk than dizygotic twins. However, it was also apparent that environmental factors, such as smoking, played a significant role [33]. Early studies to understand the nature of the genetic component used association studies, but progress was slow. These studies compare the frequency of a chromosome variant, or a set of such markers, in unrelated patients with the disease, in comparison with a group of healthy controls. This enables the detection of markers which associate with disease risk. Linkage analysis considers DNA variations between a pair of linked markers, which will be associated with both markers. More specifically, linkage disequilibrium (LD) considers the extent to which an allele of one SNP is association with an allele of another SNP within the same population [34]. As with many other complex diseases, the genetic basis of IBD appears to reflect a combination of small variations in the DNA, typically in the form of single nucleotide polymorphisms (SNPs) or copy number variants (CNVs) that increase the susceptibility to disease, rather than directly causing it [35].
Technologies evolved, and genome-wide scanning using microsatellite markers identified shared regions of the chromosomes that were over-represented in diseased individuals, thereby improving the understanding of the disease. By 1996, the chromosome regions associated with IBD were identified as being on chromosome 16 [37], as well as 3, 7, and 12 [38]. Other chromosome regions followed fairly rapidly, and it was suggested that 1–9 chromosomal regions were associated with disease susceptibility [39]. In 2001, the first gene was unequivocally identified as associated with IBD risk—nucleotide oligomerisation domain 2 (NOD2) [40,41]—which mapped to chromosome 16. These authors reported three independent associations for CD, including a frameshift variant and two missense variants of the NOD2 gene. This gene activates the nuclear transcription factor, nuclear factor NF-κB, and plays a role in the recognition of microbial pathogens.
Since this is a relatively rare complex of diseases, the importance of increased numbers of subjects for studies became apparent, leading to some important international collaborations. It had also become apparent that progress was relatively slow using the previous methods, and the field further progressed with the development of array technologies that scanned a number of genetic variants. A microarray involves the hybridisation of a target nucleic acid to a large set of probes attached to a solid support [42]. The original technologies used arrays of complementary DNA (cDNA), a double-stranded DNA, spotted onto glass microscope slides hybridised to a large set of oligonucleotide probes, to detect variations in a gene sequence. However, these methods rapidly evolved.
Association studies were increasingly being applied to population databases in GWAS [34]. Rather than considering individual candidate genes, GWAS uses a SNP chip that spans the genome, typically considering between 500,000 and 1,000,000 SNPs, and requiring far more complex statistical analyses. The Wellcome Trust Case Control Consortium used such an approach in a collaborative venture that included 17,000 subjects, both with and without diseases including IBD, and follow-up studies also examined high numbers of individuals from different populations [43]. While these studies initially identified approximately seven variants associated with CD, meta-analysis brought this number to 71 [35]. Such studies revealed hitherto unsuspected mechanisms, including autophagy, in disease pathogenesis. They also showed that a number of IBD loci are shared with other inflammatory diseases.
The Immunochip (more specifically, the Infinium® ImmunoArray BeadChip) was developed by Illumina, covering just under 200,000 genetic polymorphisms and designed to span loci that had previously been associated with specific regions associated with major autoimmune and inflammatory diseases [44]. The cost was lower than more standard GWAS chips, because it was produced in very large numbers. This cost and specificity was part of the rationale that led the expansion of the International IBD Genetics Consortium (IIBDGC), and led to the multi-authored paper by Jostins et al. [19]. These authors performed a meta-analysis of the then-current GWAS studies of CD and UC, followed by extensive validation of significant findings. They used the Immunochip method to add data from an extra 14,763 CD cases, 10,920 UC cases, and 15,977 controls, leading to a combined total of more than 75,000 cases and controls. They found 71 new associations, leading to a total of 163 IBD loci that met statistical significance. It was also apparent that many IBD loci are important in other immune-mediated disorders, especially ankylosing spondylitis and psoriasis. The loci also overlapped with those associated with susceptibility to mycobacterial infection.
Subsequent work using the Immunochip has revealed specific variants associated with the diseases in certain specific populations. For example, Huang and co-workers identified particular SNPs that associated with African Americans, while Yang et al. described six additional susceptibility loci occurring in Korean populations [45,46]. However, in general, there is considerable commonality across populations [47].
Genetic analysis has also been important in enabling better predictions of disease location and pathogenesis. The study by Cleynen et al. included patients from 49 centres across 16 European countries, as well as North America and Australasia [3]. They considered genotype-phenotype associations seen with current clinical sub-phenotypes in 34,819 patients overall (19,713 CD, 14,683 UC), all genotyped on the Immunochip array. They found that three loci (NOD2, major histocompatability complex (MHC), and macrophage stimulating 1 (MST1 3p21)) associated with IBD sub-phenotypes, especially disease location. They found that the genetic risk score strongly distinguished colonic from ileal CD, and suggested that there is a continuum of disorders within IBD, much better explained by these three groups than by CD and UC, according to the current definitions. The location and its genetic components were shown to have major implications for changes in disease pathogenesis over time.

3. Defining the Phenotype in IBD

While genetics plays a major role in IBD characteristics and progression, the desire of personalising nutrition in this disease is that strategic dietary regulation may be able to modify disease symptomology. Important technologies are summarised below, and an illustration as to how these technologies have been applied to animal studies (for example, of curcumin) in IBD is provided in Figure 1, with an example of the animal study design shown in Figure 2.

3.1. Transcriptomics

The transcriptome is defined by the set of all messenger RNA (mRNA) molecules produced in one or a population of cells [48]. Thus, it provides a measure of the genes that are actively being expressed at a given time. The field of transcriptomics has been dominated by microarray solutions provided by companies such as Affymetrix, Agilent Technologies, and Illumina. However, RNA-Seq (defined as transcriptome profiling using next-generation sequencing (NGS)) is increasingly being utilised for such work. The stated advantages of RNA-Seq over microarrays include providing better detection of low abundance transcripts, and having a greater detection of a wide range of transcripts as compared with microarrays [49]. However, the cost is considerably greater than for microarrays at present, and it is not being as widely used as the older array-based technology.
Preliminary data from such analyses are often presented in the form of a heat map that visually displays the genes that are differentially regulated before and after a treatment, and the strength of the changes involved [50]. Having obtained the levels of differentially expressed mRNA probes, it becomes important to understand the functions and/or biological pathways which they are involved in. A number of authors have utilized software packages such as Ingenuity Pathways Analysis (IPA) for such analyses [50,51]. However, the cost of this may argue against its use in many studies. Free software, such as PathVisio, that visualizes data and enables pathway statistics, or Cytoscape, for integrating complex networks, have been used by a number of groups [52].

3.2. Epigenetics Analyses

The genome reveals the potential nature of cellular proteins, while the transcriptome shows which proteins are currently being transcribed through the formation of mRNA. Epigenetic events alter the expression of genes without altering the nature of the DNA code. Various important mechanisms include DNA methylation, histone modifications, and non-coding RNAs (ncRNA). The latter group represents the majority of such events in mammals, ranging from short ncRNAs including microRNAs, to long stretches of RNA (long ncRNA or lncRNA) [53]. Interestingly, there is increasing evidence for interactions between these two groups. Epigenetic mechanisms to regulate gene expression are critical in the cellular response to environmental stimuli, including diet. While hypothesis-based studies dominated earlier work, genome-wide analyses, e.g., the methylation specific amplification microarray (MSAM) consider variations across the genome [54]. NGS technologies are also increasingly used to study epigenetic changes.

3.3. Proteomics

Proteomics has been defined as “the analysis of the protein complement present in a cell, organ, or organism at any given time [55]”.
While the genome describes the potential cellular proteins, and the transcriptome gives a measure of which genes are being transcribed at a given time, neither of these methods provides information on the actual protein composition (“the proteome”). Since the protein, in general, represents the functional unit derived from genes and mRNA, it ultimately determines the phenotype and accurate assessment of the proteome is, therefore, essential. Proteins and/or peptides are generally separated out through gel electrophoresis or liquid chromatography, both of which can be either one- or two-dimensional, and are typically coupled with mass spectrometry. A comprehensive description of current technologies for a proteomics approach to the assessment of biological samples has been published relatively recently [55].
A commonly employed method to assess the proteome is 2D gel electrophoresis using difference gel electrophoresis (DIGE) technology, by which (for example) proteins differentially expressed during intestinal inflammation can be recognized, and subsequently identified using mass spectrometry technology (liquid chromatography-mass spectrometry (LCMS) or LC-MS/MS). Although multidimensional liquid chromatography-mass spectrometry (MDLC-MS) approaches are increasingly being applied for qualitative and quantitative proteome analysis (as reviewed: [56,57]), a combined 2D DIGE/MS approach remains an important and widely used technique. This approach can also be used to identify changes in the proteome when inflammation is reduced in response to dietary compounds, such as curcumin [51], and to, therefore, better understand the mechanisms by which inflammation is ameliorated.

3.4. Metabolomics

Metabolomics is increasingly being used to study both the nature of diseases such as IBD, and the implication of variations of nutrition on disease characteristics. Formally, this field has been defined as the “systematic study of the unique chemical fingerprints that specific cellular processes leave behind” [58]. The ultimate goal is to understand the effects of dietary components on metabolic regulation and consequent implications for human health. This is complicated by the fact that the metabolites that make up the profile of a given sample may result from at least three different sets of signals: (1) dietary compounds (nutrients and bioactives); (2) xenobiotics from environmental sources which are absorbed and metabolized; and (3) metabolic signals produced by the large-bowel microflora.
Metabolomics data can be used to identify overall patterns or effects, for example by applying methods such as partial least squares discriminate analysis to assess how the metabolite profile of an individual compares to those of other individuals. Such an approach may be applied to better diagnose a health outcome, such as irritable bowel syndrome [59], or to distinguish between different dietary treatments, outcomes which cannot be achieved with conventional techniques. Metabolomics can also be used to identify specific metabolic changes in response to an intervention, such as a diet, although the success of this approach depends on libraries of small molecules being developed to facilitate metabolite identification. A major technical challenge faced by metabolomics is the wide variety of chemical and physical properties of the metabolites, ranging from highly polar through to non-polar compounds.
One critical aspect of metabolomics which has driven many of the recent advances is the potential for collection of relatively non-invasive samples, such as faecal, urine, blood, buccal swabs, or breath volatiles. Based on data derived from appropriate animal models, such samples can enable the accurate assessment of important outcomes within a target tissue (such as the gastrointestinal (GI) tract) based on their metabolite profile. Technologies for metabolomics research broadly fall into two categories, being either nuclear magnetic resonance (NMR)-spectroscopy or mass spectrometry-based. However, within these are a range of sub-categories, reflecting the varied nature of the metabolites within a sample. For example, MS-based technologies cover the spectrum from polar (hydrophilic interaction chromatography (HILIC) for compounds such as amino acids, organic acids, di/trisaccharides) through semi-polar (C18, e.g., flavonoids) to non-polar (C1, e.g., phospholipids, triacylglycerols and diacylglycerols).

3.5. Metagenomics

It has become increasingly clear that the intestinal microbiota (bacteria inhabiting the gastrointestinal tract) play an essential role in human health. Indeed, some authors have likened the microbiota to an organ in its own right [60]. The colonic microbiota plays an even more critical role in IBD pathogenesis. Metagenomics is the current technology which describes the microbial genomic composition of the GI tract, usually measured from a faecal sample [61]. Changes in the composition of these bacterial populations, and/or of their collective genome, can have important implications for disease susceptibility, including intestinal inflammation [62,63].
Early studies on these populations required culture-based techniques, using anaerobic conditions. More recently, culture-independent techniques including NGS have provided important advances in understanding the diversity of the microbiota. The species composition can be measured using 16s rRNA sequencing, while comprehensive sequencing of the collective genome enables an understanding of the functional capacity of the microbiota. In addition, RNA-seq approaches allow a measurement of the expression of microbial genes to be measured. Hobbs et al., [61] developed a statistical method to characterise transcriptional regulatory networks from metagenomics data. Such approaches allow an assessment of the complex interplay between host and microbial genes, and their impact on the health of the host.

4. In Vitro Approaches to Identifying Bioactive Food Components Likely to Benefit IBD

Once the nature of genetic variations related to disease susceptibility are unequivocally established, this information can be used to design a cell line or other high-throughput screen that reflects the changes in cellular function associated with the disease. Such an approach can mimic the genotype, thereby allowing either food fractions or known dietary components to be considered for their ability to overcome the effect of the defective gene [64,65].
The genotype-specific NOD2 assay developed by Philpott and co-workers [66] provides an example of a genotype-specific assay. As already indicated, a functional NOD2 gene activates the nuclear transcription factor NF-κB, and plays an important role in the recognition of microbial pathogens. The NOD2 gene is also known as caspase-activated recruitment domain 15 (CARD15), and it encodes an intracellular receptor for the bacterial component muramyl dipeptide (MDP) [67]. An important component in normal cellular signalling is the production of the inflammatory cytokine tumour necrosis factor-α (TNF-α), which has several functions in the inflammatory process that is part of a general immune response to tissue damage and infection. It is central to the pathogenesis of CD because it is produced in excess during chronic inflammation. Part of this process involves stimulation of a signal transduction pathway activating NF-κB [68]. Philpott et al. developed a screen to test for food extracts with the potential to reduce inflammation mediated by this event [66]. The NOD2 3020insC polymorphism is a relatively common and frequently studied genetic variant in CD [69]. It is a frameshift mutation that leads to the formation of a premature stop codon, which prevents the ability of the cell to respond to MDP and, subsequently, to activate the transcription factor NF-κB. This assay thus models the NOD2 3020insC genotype and considers whether any food compound or extract has the ability to restore the normal signalling activity of cells carrying this variant, in comparison to that of cells carrying the wild-type gene.
A pUNO-hNOD2a vector was used in transfections as the wild-type NOD2 reference vector. The 3020insC NOD2 variant (subsequently designated pUNO-hNOD2a 3020insC) was generated from this by site-directed mutagenesis, using the Gene Tailor Site-Directed Mutagenesis System. For the experiments, the human embryonic kidney cell line 293 T (HEK293T) was grown according to standard protocols. These cells were transfected using Lipofectamine 2000 with a galactosidase vector (transfection control), pNifty2-SEAP vector (as the NF-κB reporter) and either the wild-type (to give a NOD2 wild-type cell line) or constructed vector (to give the NOD2 mutant cell line), or no other vector (the control cell line). Cells were allowed to settle into multi-well dishes and grown for a time before the addition of standard compounds or food extracts over a serial dilution range. After a further 24 h incubation, cells were stimulated by the addition of MDP or had an equivalent volume of media added. After further incubation, an aliquot of the cell supernatant was transferred to fresh plates containing a Quanti-Blue reagent for the detection of secreted alkaline phosphatase (SEAP) production, or treated with WST-1 reagent to measure cell viability. Absorbances were read at 635 nm (Quanti-Blue) and 450 nm (WST-1), and SEAP production was normalised against transfection efficiency. To compensate for any possible toxicity of extracts, SEAP production was also normalized against cell viability.
An example more relevant to curcumin, but using a similar rationale, was studied by McCann and co-workers [30]. Among various molecular pathways involved in susceptibility to IBD are those involved in intestinal epithelial barrier function and immune response [70]. The solute carrier family 22 member 4 (SLC22A4) gene is important for barrier function, encoding an organic cation transporter protein involved in the movement of proteins across the membrane of epithelial cells. McCann et al. considered the risk variant (rs1050152 at position 503F) which has a higher transport activity compared with the wild-type gene, leading to inappropriate transport of organic cations across the intestinal epithelial barrier [71]. As their example of a mutant that affected immune response, they utilised a variant in the gene encoding interleukin-10 (IL10). This is an immune-suppressive cytokine that reduces excessive inflammatory response, thereby creating an imbalance between pro- and anti-inflammatory mechanisms [72]. The rs1800896 SNP in this gene at position −1082 (the −1082A variant) reduces IL10 transcription and cytokine production in IBD [73]. Thus, these authors considered the effects of an extract of turmeric and factions of this extract to overcome the functional effects of the two IBD-associated genetic variants—solute carrier protein 22 A4 (SLC22A4, rs1050152) and interleukin-10 (IL10, rs1800896). They developed an assay using HEK293 cells to examine the in vitro capacity of a turmeric extract, and fractions thereof, to affect the functionality of these two gene variants [30]. In parallel, they measured the curcumin content of the extract and fractions using high pressure liquid chromatography (HPLC). They found that an extract of turmeric beneficially affected the gene variants associated with IBD, by a reduction of inappropriate epithelial cell transport (SLC22A4, 503F) or through an increase in gene promoter activity of the IL10 anti-inflammatory cytokine (IL10, −1082A). The effect of turmeric on IL10 was linked with its curcumin content. A similar effect was seen for several chromatographically separated fractions of turmeric.
In the progressive development of curcumin as a pharmaceutical solution to IBD, the low bioavailability of the purified compound has been recognised as important. This is largely caused by its low solubility and stability in the digestive tract. Several different formulations have been developed to overcome this problem, including a solid lipid particle formulation (Curcumin SLCP, Longvida) [74]. These authors considered the anti-inflammatory effects of such a formulation in lipopolysaccharide (LPS)-stimulated RAW 264.7 cultured murine macrophages. They utilised transient transfection technologies with an NF-κB reporter construct to show that this derived compound could inhibit the transcriptional activity of nuclear factor NF-κB in this cell line, and that it was more efficient in this process than the original material.
If a tentative bioactive food compound has been previously identified and seems likely to have potential in IBD, then several of the omics technologies can be applied to test the range of potential mechanisms of action. While effective activation of the immune response is critical to maintaining human health by responding to invading pathogens or other sorts of potentially deleterious injury, over-activation of the immune response leads to inflammation. Activation of the inflammasome is mediated by the innate immune system, and plays an important role in these responses [75]. Several pattern recognition receptors (PRRs) are components of the inflammasome complex, including the nucleotide-binding domain, leucine-rich repeat containing proteins (NLRs or NOD-like receptors) in both mice and humans. The Human Inflammasomes RT Profiler™ PCR Array measures the expression of 84 of the genes associated with this process, which have been linked to various diseases, such as IBD. Thus, although Miller and co-workers had planned their inflammasome array studies on curcumin to provide a comment on its potential in malignant mesothelioma, the data are also relevant to IBD [75]. These authors used mouse and human malignant mesothelioma cell models, in in vitro studies. Curcumin significantly down-regulated levels of expression of various genes involved in inflammation, including NF-κB, toll-like receptor (TLR), and interleukin 1 (IL-1β).
Proteomics was applied to studying the molecular targets of curcumin on a range of gastric cancer cell lines [76]. As well as defining dose response curves and demonstrating that curcumin caused apoptosis, they used two-dimensional gel electrophoresis (2-DE) and matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF-TOF) mass spectrometry to study the target proteins. Of the proteins which showed more than a 1.5-fold change in curcumin-treated cells compared to untreated controls, 33 proteins were up-regulated and 42 proteins down-regulated by the treatment. Of relevance to IBD were 16% of proteins involved in metabolism and 2% in immune response. A similar study was carried out in the human breast cancer cell line MCF-7. Proteomic identification of differentially-expressed proteins in curcumin-treated MCF-7 cells identified 12 such proteins, although these genes did not seem to relate specifically to inflammatory diseases [77].
In vitro studies have also been informative on the likely effects of curcumin on the gut microbiome. Lou and coworkers [78] obtained faecal samples from a healthy male volunteer who had avoided alcohol or foods rich in curcumin for the previous 48 h. Following homogenisation of the sample, the supernatant was cultivated in an anaerobic chamber, divided into two and curcumin added to one of the subcultures, followed by overnight incubation. Ultra-performance liquid chromatography/quadrupole (UPLC-Q)-TOF MS enabled identification of the curcumin metabolites that were produced by the intestinal bacteria in the culture. They revealed novel metabolic pathways of curcumin in its interactions with the microbiome.

5. Animal Studies to Confirm the Role of Bioactive Components Likely to Benefit IBD

As described in the previous section, in vitro studies can provide highly relevant data via the application of omics techniques in a variety of contexts, including IBD. However, to date, there is no in vitro model which can adequately model the level of complexity of a biological system such as the GI tract. It is necessary to clearly establish safety and efficacy in a model before considering a human intervention, and animal models are currently the only viable option to achieve this. Thus, although human clinical outcomes are critical to establish the impact of any proposed nutritional intervention, there is still a place for appropriate animal models, for three key reasons.
As already mentioned, the GI tract is a highly complex environment in which food compounds, host cells, and the intestinal microbiota interact, and this cannot yet be adequately modelled in vitro. Animal models enable such modelling, which is important to better understand the mechanisms by which any impact of an intervention may occur. They also provide the ability to measure changes in the relevant tissues (in this case, intestinal tissue) and correlate these with peripheral changes (e.g., urine, blood) which have the potential to be used as biomarkers in a minimally-invasive manner for any subsequent human study; for example, to monitor symptoms and, therefore, assess the efficacy, or otherwise, of any intervention. This is of particular importance in the case of metabolomics, which is increasingly being applied to matrices, such as plasma and urine. It is also necessary to compare and correlate microbiota from the GI tract with those in faecal samples, so that subsequent analysis of faecal samples from human intervention studies can be used with a reasonable degree of confidence as a proxy for what is occurring in the GI tract.
Finally, it is important to establish robust use of omics techniques before applying them to human samples, and animal models provide the opportunity to do just that.
As an example, we describe some work of our own, and of others, on the potential beneficial effect of curcumin on intestinal inflammation. This includes what may be direct effects of curcumin on intestinal inflammation, as well as some which more likely occur through modulation of the intestinal microbiota.
Within the Nutrigenomics New Zealand programme, two mouse models of IBD were primarily utilized: the interleukin-10 gene-deficient (Il10−/−) mouse [79,80,81], and the multiple drug resistance (Mdr1a) mouse [82,83,84]. Both of these models exhibit symptoms similar to those seen in human IBD, resulting from the disruption of either the Il10 gene (which encodes the anti-inflammatory cytokine interleukin-10) or the Abcb1a gene (also known as Mdr1a, which encodes the membrane drug-efflux pump p-glycoprotein 3), and both have been used to investigate the effects of a range of foods or food components on inflammation.
We have used the Mdr1a model to assess the effects of polyphenols, including those from green tea [82] and, of particular relevance for the current discussion, curcumin [51,84]. Initially using a transcriptomics approach, we showed that curcumin’s beneficial effect on colon inflammation may occur through up-regulation of xenobiotic metabolism and down-regulation of immune response pathways, in particular those mediated by the pregnane X receptor (Pxr) and peroxisome proliferator-activated receptor α (Pparα) activation of retinoid X receptor (Rxr). We also provided evidence that curcumin may down-regulate genes involved in oxidative stress and fibrogenesis pathways [84]. Subsequent use of proteomics analysis, and integration of gene and protein data, demonstrated that curcumin may regulate the actin cytoskeleton and thereby enhance the integrity of the intestinal barrier (a potentially important factor in inflammation) via α-catenin [51].
Other research suggests that the beneficial effects of curcumin for colitis may be as a result of maintaining a more diverse colonic microbial ecology [85]. A reduction in the diversity of the intestinal microbiota has been associated with intestinal inflammation in animal studies, including mice [86] and pigs [87], and in human studies [88] and, therefore, the maintenance of diversity represents a plausible mechanism by which curcumin may exert its effect. Systems biology approaches on IBD have usefully begun to focus on gut microbial metabolism [89].

6. Human Studies

As we have described above, a wide range of in vitro and animal studies support the conclusion that curcumin is likely to be effective both in preventing inflammation, and in suppressing the negative effects of inflammation once the symptoms have begun [90]. Continuous oxidative damage can initiate the inflammatory signalling cascade in IBD. In this context, curcumin acts to scavenge free radicals and also up-regulate various antioxidant enzymes. It also alters a number of key signalling pathways, for example inhibiting cyclooxygenases 1 and 2 (COX-1 and COX-2), tumour necrosis factor-α (TNF-α), and the transcription factor NF-κB. In addition, it has been demonstrated as safe to be co-administered with conventional therapy, albeit in relatively small human trials to date, and is being actively developed as both a chemopreventive and pharmaceutical agent for IBD [91,92]. As Aggawal and co-workers point out, its TNF-α inhibition alone justifies the cost of this development, since current TNF-α inhibitors are used not only for IBD, but also for osteoarthritis, psoriasis, and ankylosis, at a cost of $15,000–$20,000 per person per year [31].
Despite the promise in other study approaches, curcumin has given variable results in human trials [93]. Indeed, to date, convincing data for curcumin alone was only available for two studies, comprising data for 99 patients. However, in combination with mainstream therapy, curcumin has significantly improved patient symptoms or allowed for a drop in dosage of corticosteroids. For example, Lang et al. showed that curcumin, in combination with mesalamine, induced remission of ulcerative colitis symptoms after four weeks of treatment [94]. Clinical remission was achieved by no patients in the placebo-alone group in this time, but by more than half of the patients also receiving curcumin. Most of the authors of these studies have concluded that larger clinical trials may need to be conducted. Additionally, we suggest that patient stratification according to genotype may be effective and could be considered for future work.
As with many other natural products, although curcumin shows some efficacy in vivo, it has a limited shelf-life and is not absorbed efficiently. A number of novel formulations have been developed to overcome these problems. Kitture and co-workers used another natural product, Aloe vera, to synthesise a template in which to immobilise the curcumin [95]. In antioxidant assays, the curcumin-loaded template showed superior properties to either the curcumin or Aloe vera alone. This loaded template also facilitated the efficient release of curcumin within five hours, quicker than could occur with curcumin alone. Closer to clinical applications is the use of nanoparticles to facilitate the colonic delivery of curcumin [32,96]. These particles have been found to accumulate preferentially in inflamed regions, and can, potentially, aid delivery to the areas that will most benefit from the compound. Various chemical structures have been compared in order to optimise the final material which goes into large-scale clinical trials.

7. Conclusions

The so-call omics approaches to studying disease are relatively recent, with increasingly sensitive and cost-effective technologies emerging over the past 10–15 years. IBD provide an excellent example where there have been order of magnitude changes in understanding the nature of the diseases, and the way in which natural (or other) therapies may interact in the natural course of the disease. Fundamental to the understanding of cellular mechanisms have been genomic studies, considering not only the human genes, but also the gut microbiome [89]. Transcriptomics, proteomics, and metabolomics are among many of the sensitive technologies that not only help to explain the nature of the disease, but enable short-term studies with highly sensitive endpoints to test new therapeutic strategies. We have used curcumin as an example for which several of these technologies have been applied to date. This example demonstrates that understanding the nature of the disease targets enables potential consideration of an increasing number of natural products, or more targeted therapies, that provide significant hope for better control of disease in future generations.

Acknowledgments

Nutrigenomics New Zealand was a collaboration between AgResearch Ltd., Plant & Food Research, and The University of Auckland, with funding through the New Zealand Ministry of Business, Innovation & Employment (MBIE) which ran from 2004–2014.

Author Contributions

Lynnette R. Ferguson and Matthew P. G. Barnett jointly conceived the idea, each wrote different manuscript sections and both edited the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ooi, C.J.; Makharia, G.K.; Hilmi, I.; Gibson, P.R.; Fock, K.M.; Ahuja, V.; Ling, K.L.; Lim, W.C.; Thia, K.T.; Wei, S.C.; et al. Asia Pacific Consensus Statements on Crohn’s disease. Part 1: Definition, diagnosis, and epidemiology: (Asia Pacific Crohn’s Disease Consensus—Part 1). J. Gastroenterol. Hepatol. 2016, 31, 45–55. [Google Scholar] [CrossRef] [PubMed]
  2. Day, A.S.; Lemberg, D.A.; Gearry, R.B. Inflammatory bowel disease in Australasian children and adolescents. Gastroenterol. Res. Pract. 2014, 2014, 703890–703897. [Google Scholar] [CrossRef] [PubMed]
  3. Cleynen, I.; Boucher, G.; Jostins, L.; Schumm, L.P.; Zeissig, S.; Ahmad, T.; Andersen, V.; Andrews, J.M.; Annese, V.; Brand, S.; et al. Inherited determinants of Crohn’s disease and ulcerative colitis phenotypes: A genetic association study. Lancet 2016, 387, 156–167. [Google Scholar] [CrossRef]
  4. Xavier, R.J.; Podolsky, D.K. Unravelling the pathogenesis of inflammatory bowel disease. Nature 2007, 448, 427–434. [Google Scholar] [CrossRef] [PubMed]
  5. Cho, J.H.; Brant, S.R. Recent insights into the genetics of inflammatory bowel disease. Gastroenterology 2011, 140, 1704–1712. [Google Scholar] [CrossRef] [PubMed]
  6. Kaser, A.; Zeissig, S.; Blumberg, R.S. Inflammatory bowel disease. Annu. Rev. Immunol. 2010, 28, 573–621. [Google Scholar] [CrossRef] [PubMed]
  7. Ruemmele, F.M. Role of diet in inflammatory bowel disease. Ann. Nutr. Metab. 2016, 68, 33–41. [Google Scholar] [CrossRef] [PubMed]
  8. Loftus, E.V., Jr. Clinical epidemiology of inflammatory bowel disease: Incidence, prevalence, and environmental influences. Gastroenterology 2004, 126, 1504–1517. [Google Scholar] [CrossRef] [PubMed]
  9. Chassaing, B.; Darfeuille-Michaud, A. The commensal microbiota and enteropathogens in the pathogenesis of inflammatory bowel diseases. Gastroenterology 2011, 140, 1720–1728. [Google Scholar] [CrossRef] [PubMed]
  10. Marlow, G.; Ellett, S.; Ferguson, I.R.; Zhu, S.; Karunasinghe, N.; Jesuthasan, A.C.; Han, D.Y.; Fraser, A.G.; Ferguson, L.R. Transcriptomics to study the effect of a Mediterranean-inspired diet on inflammation in Crohn’s disease patients. Hum. Genom. 2013, 7, 24–33. [Google Scholar] [CrossRef] [PubMed]
  11. Ferguson, L.R. Nutrigenetics, nutrigenomics and inflammatory bowel diseases. Expert Rev. Clin. Immunol. 2013, 9, 717–726. [Google Scholar] [CrossRef] [PubMed]
  12. Barnett, M.; Young, W.; Cooney, J.; Roy, N. Metabolomics and proteomics, and what to do with all these “omes”: Insights from nutrigenomic investigations in New Zealand. J. Nutrigenet. Nutrigenom. 2014, 7, 274–282. [Google Scholar] [CrossRef] [PubMed]
  13. Evelo, C.T.; van Bochove, K.; Saito, J.T. Answering biological questions: Querying a systems biology database for nutrigenomics. Genes Nutr. 2011, 6, 81–87. [Google Scholar] [CrossRef] [PubMed]
  14. Fan, S.; Pan, Z.; Geng, Q.; Li, X.; Wang, Y.; An, Y.; Xu, Y.; Tie, L.; Pan, Y.; Li, X. Layered signaling regulatory networks analysis of gene expression involved in malignant tumorigenesis of non-resolving ulcerative colitis via integration of cross-study microarray profiles. PLoS ONE 2013, 8, e67142. [Google Scholar] [CrossRef] [PubMed]
  15. Polytarchou, C.; Koukos, G.; Iliopoulos, D. Systems biology in inflammatory bowel diseases: Ready for prime time. Curr. Opin. Gastroenterol. 2014, 30, 339–346. [Google Scholar] [CrossRef] [PubMed]
  16. Marlow, G.; Han, D.Y.; Triggs, C.M.; Ferguson, L.R. Food intolerance: Associations with the rs12212067 polymorphism of FOXO3 in Crohn’s disease patients in New Zealand. J. Nutrigenet. Nutrigenom. 2015, 8, 70–80. [Google Scholar] [CrossRef] [PubMed]
  17. Petermann, I.; Triggs, C.M.; Huebner, C.; Han, D.Y.; Gearry, R.B.; Barclay, M.L.; Demmers, P.S.; McCulloch, A.; Ferguson, L.R. Mushroom intolerance: a novel diet-gene interaction in Crohn’s disease. Br. J. Nutr. 2009, 102, 506–508. [Google Scholar] [CrossRef] [PubMed]
  18. Triggs, C.M.; Munday, K.; Hu, R.; Fraser, A.G.; Gearry, R.B.; Barclay, M.L.; Ferguson, L.R. Dietary factors in chronic inflammation: food tolerances and intolerances of a New Zealand Caucasian Crohn’s disease population. Mutat. Res. 2010, 690, 123–138. [Google Scholar] [CrossRef] [PubMed]
  19. Jostins, L.; Ripke, S.; Weersma, R.K.; Duerr, R.H.; McGovern, D.P.; Hui, K.Y.; Lee, J.C.; Philip Schumm, L.; Sharma, Y.; Anderson, C.A.; et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012, 491, 119–124. [Google Scholar] [CrossRef] [PubMed]
  20. Boeing, H. Nutritional epidemiology: New perspectives for understanding the diet-disease relationship? Eur. J. Clin. Nutr. 2013, 67, 424–429. [Google Scholar] [CrossRef] [PubMed]
  21. Fallaize, R.; Forster, H.; Macready, A.L.; Walsh, M.C.; Mathers, J.C.; Brennan, L.; Gibney, E.R.; Gibney, M.J.; Lovegrove, J.A. Online dietary intake estimation: reproducibility and validity of the Food4Me food frequency questionnaire against a 4-day weighed food record. J. Med. Internet Res. 2014, 16, e190. [Google Scholar] [CrossRef] [PubMed]
  22. Forster, H.; Fallaize, R.; Gallagher, C.; O’Donovan, C.B.; Woolhead, C.; Walsh, M.C.; Macready, A.L.; Lovegrove, J.A.; Mathers, J.C.; Gibney, M.J.; et al. Online dietary intake estimation: The Food4Me food frequency questionnaire. J. Med. Internet Res. 2014, 16, e150. [Google Scholar] [CrossRef] [PubMed]
  23. Bordoni, A.; Capozzi, F. Foodomics for healthy nutrition. Curr. Opin. Clin. Nutr. Metab. Care 2014, 17, 418–424. [Google Scholar] [CrossRef] [PubMed]
  24. Capozzi, F.; Bordoni, A. Foodomics: A new comprehensive approach to food and nutrition. Genes Nutr. 2013, 8, 1–4. [Google Scholar] [CrossRef] [PubMed]
  25. Putignani, L.; Dallapiccola, B. Foodomics as part of the host-microbiota-exposome interplay. J. Proteom. 2016, 147, 3–20. [Google Scholar] [CrossRef] [PubMed]
  26. Herrero, M.; Simo, C.; Garcia-Canas, V.; Ibanez, E.; Cifuentes, A. Foodomics: MS-based strategies in modern food science and nutrition. Mass Spectrom. Rev. 2012, 31, 49–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Ibanez, C.; Valdes, A.; Garcia-Canas, V.; Simo, C.; Celebier, M.; Rocamora-Reverte, L.; Gomez-Martinez, A.; Herrero, M.; Castro-Puyana, M.; Segura-Carretero, A.; et al. Global Foodomics strategy to investigate the health benefits of dietary constituents. J. Chromatogr. A 2012, 1248, 139–153. [Google Scholar] [CrossRef] [PubMed]
  28. Witkin, J.M.; Li, X. Curcumin, an active constiuent of the ancient medicinal herb Curcuma longa L.: Some uses and the establishment and biological basis of medical efficacy. CNS Neurol. Disord. Drug Targets 2013, 12, 487–497. [Google Scholar] [CrossRef] [PubMed]
  29. Hatcher, H.; Planalp, R.; Cho, J.; Torti, F.M.; Torti, S.V. Curcumin: From ancient medicine to current clinical trials. Cell. Mol. Life Sci. 2008, 65, 1631–1652. [Google Scholar] [CrossRef] [PubMed]
  30. McCann, M.J.; Johnston, S.; Reilly, K.; Men, X.; Burgess, E.J.; Perry, N.B.; Roy, N.C. The effect of turmeric (Curcuma longa) extract on the functionality of the solute carrier protein 22 A4 (SLC22A4) and interleukin-10 (IL-10) variants associated with inflammatory bowel disease. Nutrients 2014, 6, 4178–4190. [Google Scholar] [CrossRef] [PubMed]
  31. Aggarwal, B.B.; Gupta, S.C.; Sung, B. Curcumin: An orally bioavailable blocker of TNF and other pro-inflammatory biomarkers. Br. J. Pharmacol. 2013, 169, 1672–1692. [Google Scholar] [CrossRef] [PubMed]
  32. Beloqui, A.; Coco, R.; Memvanga, P.B.; Ucakar, B.; des Rieux, A.; Preat, V. pH-sensitive nanoparticles for colonic delivery of curcumin in inflammatory bowel disease. Int. J. Pharm. 2014, 473, 203–212. [Google Scholar] [CrossRef] [PubMed]
  33. Tysk, C.; Lindberg, E.; Jarnerot, G.; Floderus-Myrhed, B. Ulcerative colitis and Crohn’s disease in an unselected population of monozygotic and dizygotic twins. A study of heritability and the influence of smoking. Gut 1988, 29, 990–996. [Google Scholar] [CrossRef] [PubMed]
  34. Bush, W.S.; Moore, J.H. Chapter 11: Genome-wide association studies. PLoS Comput. Biol. 2012, 8, e1002822. [Google Scholar] [CrossRef] [PubMed]
  35. Franke, A.; McGovern, D.P.; Barrett, J.C.; Wang, K.; Radford-Smith, G.L.; Ahmad, T.; Lees, C.W.; Balschun, T.; Lee, J.; Roberts, R.; et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat. Genet. 2010, 42, 1118–1125. [Google Scholar] [CrossRef] [PubMed]
  36. Ikram, M.K.; Sim, X.; Jensen, R.A.; Cotch, M.F.; Hewitt, A.W.; Ikram, M.A.; Wang, J.J.; Klein, R.; Klein, B.E.; Breteler, M.M.; et al. Four novel Loci (19q13, 6q24, 12q24, and 5q14) influence the microcirculation in vivo. PLoS Genet. 2010, 6, e1001184. [Google Scholar] [CrossRef]
  37. Hugot, J.P.; Laurent-Puig, P.; Gower-Rousseau, C.; Olson, J.M.; Lee, J.C.; Beaugerie, L.; Naom, I.; Dupas, J.L.; van Gossum, A.; Orholm, M.; et al. Mapping of a susceptibility locus for Crohn’s disease on chromosome 16. Nature 1996, 379, 821–823. [Google Scholar] [CrossRef] [PubMed]
  38. Satsangi, J.; Parkes, M.; Louis, E.; Hashimoto, L.; Kato, N.; Welsh, K.; Terwilliger, J.D.; Lathrop, G.M.; Bell, J.I.; Jewell, D.P. Two stage genome-wide search in inflammatory bowel disease provides evidence for susceptibility loci on chromosomes 3, 7 and 12. Nat. Genet. 1996, 14, 199–202. [Google Scholar] [CrossRef] [PubMed]
  39. Ek, W.E.; D’Amato, M.; Halfvarson, J. The history of genetics in inflammatory bowel disease. Ann. Gastroenterol. 2014, 27, 294–303. [Google Scholar] [PubMed]
  40. Hugot, J.P.; Chamaillard, M.; Zouali, H.; Lesage, S.; Cezard, J.P.; Belaiche, J.; Almer, S.; Tysk, C.; O’Morain, C.A.; Gassull, M.; et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature 2001, 411, 599–603. [Google Scholar] [CrossRef] [PubMed]
  41. Ogura, Y.; Bonen, D.K.; Inohara, N.; Nicolae, D.L.; Chen, F.F.; Ramos, R.; Britton, H.; Moran, T.; Karaliuskas, R.; Duerr, R.H.; et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature 2001, 411, 603–606. [Google Scholar] [CrossRef] [PubMed]
  42. Afman, L.; Muller, M. Nutrigenomics: From molecular nutrition to prevention of disease. J. Am. Diet. Assoc. 2006, 106, 569–576. [Google Scholar] [CrossRef] [PubMed]
  43. Burton, P.R.; Clayton, D.G.; Cardon, L.R.; Craddock, N.; Deloukas, P.; Duncanson, A.; Kwiatkowski, D.P.; McCarthy, M.I.; Ouwehand, W.H.; Samani, N.J.; et al. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 2007, 447, 661–678. [Google Scholar] [CrossRef] [PubMed]
  44. Cortes, A.; Brown, M.A. Promise and pitfalls of the immunochip. Arthritis Res. Ther. 2011, 13, 101–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Huang, C.; Haritunians, T.; Okou, D.T.; Cutler, D.J.; Zwick, M.E.; Taylor, K.D.; Datta, L.W.; Maranville, J.C.; Liu, Z.; Ellis, S.; et al. Characterization of genetic loci that affect susceptibility to inflammatory bowel diseases in African Americans. Gastroenterology 2015, 149, 1575–1586. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, S.K.; Hong, M.; Choi, H.; Zhao, W.; Jung, Y.; Haritunians, T.; Ye, B.D.; Kim, K.J.; Park, S.H.; Lee, I.; et al. Immunochip analysis identification of 6 additional susceptibility loci for Crohn’s disease in Koreans. Inflamm. Bowel Dis. 2015, 21, 1–7. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, J.Z.; van Sommeren, S.; Huang, H.; Ng, S.C.; Alberts, R.; Takahashi, A.; Ripke, S.; Lee, J.C.; Jostins, L.; Shah, T.; et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet. 2015, 47, 979–986. [Google Scholar] [CrossRef] [PubMed]
  48. Duffus, J.H.; Nordberg, M.; Templeton, D.M. IUPAC glossary of terms used in toxicology. Pure Appl. Chem. 2007, 79, 1153–1344. [Google Scholar] [CrossRef]
  49. 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] [PubMed]
  50. Fok, W.C.; Livi, C.; Bokov, A.; Yu, Z.; Chen, Y.; Richardson, A.; Perez, V.I. Short-term rapamycin treatment in mice has few effects on the transcriptome of white adipose tissue compared to dietary restriction. Mech. Ageing Dev. 2014, 140, 23–29. [Google Scholar] [CrossRef] [PubMed]
  51. Cooney, J.M.; Barnett, M.P.; Dommels, Y.E.; Brewster, D.; Butts, C.A.; McNabb, W.C.; Laing, W.A.; Roy, N.C. A combined omics approach to evaluate the effects of dietary curcumin on colon inflammation in the Mdr1a−/− mouse model of inflammatory bowel disease. J. Nutr. Biochem. 2016, 27, 181–192. [Google Scholar] [CrossRef] [PubMed]
  52. Shangguan, H.; Tan, S.Y.; Zhang, J.R. Bioinformatics analysis of gene expression profiles in hepatocellular carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 2054–2061. [Google Scholar] [PubMed]
  53. Yoon, J.H.; Abdelmohsen, K.; Gorospe, M. Functional interactions among microRNAs and long noncoding RNAs. Semin. Cell Dev. Biol. 2014, 34, 9–14. [Google Scholar] [CrossRef] [PubMed]
  54. Kellermayer, R.; Balasa, A.; Zhang, W.; Lee, S.; Mirza, S.; Chakravarty, A.; Szigeti, R.; Laritsky, E.; Tatevian, N.; Smith, C.W.; et al. Epigenetic maturation in colonic mucosa continues beyond infancy in mice. Hum. Mol. Genet. 2010, 19, 2168–2176. [Google Scholar] [CrossRef] [PubMed]
  55. Baynam, G.; Walters, M.; Claes, P.; Kung, S.; LeSouef, P.; Dawkins, H.; Bellgard, M.; Girdea, M.; Brudno, M.; Robinson, P.; et al. Phenotyping: Targeting genotype’s rich cousin for diagnosis. J. Paediatr. Child Health 2015, 51, 381–386. [Google Scholar] [CrossRef] [PubMed]
  56. Di Palma, S.; Hennrich, M.L.; Heck, A.J.; Mohammed, S. Recent advances in peptide separation by multidimensional liquid chromatography for proteome analysis. J. Proteom. 2012, 75, 3791–3813. [Google Scholar] [CrossRef] [PubMed]
  57. Wu, Q.; Yuan, H.; Zhang, L.; Zhang, Y. Recent advances on multidimensional liquid chromatography-mass spectrometry for proteomics: From qualitative to quantitative analysis—A review. Anal. Chim. Acta 2012, 731, 1–10. [Google Scholar] [CrossRef] [PubMed]
  58. Daviss, B. Growing pains for metabolomics. Scientist 2005, 19, 25–28. [Google Scholar]
  59. Shankar, V.; Reo, N.V.; Paliy, O. Simultaneous fecal microbial and metabolite profiling enables accurate classification of pediatric irritable bowel syndrome. Microbiome 2015, 3, 73–78. [Google Scholar] [CrossRef] [PubMed]
  60. Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.; Hirschfield, G.M.; Hold, G.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The gut microbiota and host health: A new clinical frontier. Gut 2015, 65, 330–339. [Google Scholar] [CrossRef] [PubMed]
  61. Hobbs, E.T.; Pereira, T.; O’Neill, P.K.; Erill, I. A Bayesian inference method for the analysis of transcriptional regulatory networks in metagenomic data. Algorithms Mol. Biol. 2016, 11, 19–30. [Google Scholar] [CrossRef] [PubMed]
  62. Druart, C.; Alligier, M.; Salazar, N.; Neyrinck, A.M.; Delzenne, N.M. Modulation of the gut microbiota by nutrients with prebiotic and probiotic properties. Adv. Nutr. 2014, 5, 624–633. [Google Scholar] [CrossRef] [PubMed]
  63. Peterson, C.T.; Sharma, V.; Elmen, L.; Peterson, S.N. Immune homeostasis, dysbiosis and therapeutic modulation of the gut microbiota. Clin. Exp. Immunol. 2015, 179, 363–377. [Google Scholar] [CrossRef] [PubMed]
  64. Martin, H.; Burgess, E.J.; Smith, W.A.; McGhie, T.K.; Cooney, J.M.; Lunken, R.C.; de Guzman, E.; Trower, T.; Perry, N.B. JAK2 and AMP-kinase inhibition in vitro by food extracts, fractions and purified phytochemicals. Food Funct. 2015, 6, 305–312. [Google Scholar] [CrossRef] [PubMed]
  65. Nasef, N.A.; Mehta, S.; Murray, P.; Marlow, G.; Ferguson, L.R. Anti-inflammatory activity of fruit fractions in vitro, mediated through toll-like receptor 4 and 2 in the context of inflammatory bowel disease. Nutrients 2014, 6, 5265–5279. [Google Scholar] [CrossRef] [PubMed]
  66. Philpott, M.; Mackay, L.; Ferguson, L.R.; Forbes, D.; Skinner, M. Cell culture models in developing nutrigenomics foods for inflammatory bowel disease. Mutat. Res. 2007, 622, 94–102. [Google Scholar] [CrossRef] [PubMed]
  67. Girardin, S.E.; Boneca, I.G.; Viala, J.; Chamaillard, M.; Labigne, A.; Thomas, G.; Philpott, D.J.; Sansonetti, P.J. Nod2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP) detection. J. Biol. Chem. 2003, 278, 8869–8872. [Google Scholar] [CrossRef] [PubMed]
  68. Murillo, L.S.; Morre, S.A.; Pena, A.S. Toll-like receptors and NOD/CARD proteins: Pattern recognition receptors are key elements in the regulation of immune response. Drugs Today (Barc) 2003, 39, 415–438. [Google Scholar] [CrossRef] [PubMed]
  69. Economou, M.; Trikalinos, T.A.; Loizou, K.T.; Tsianos, E.V.; Ioannidis, J.P. Differential effects of NOD2 variants on Crohn’s disease risk and phenotype in diverse populations: a metaanalysis. Am. J. Gastroenterol. 2004, 99, 2393–2404. [Google Scholar] [CrossRef] [PubMed]
  70. Vermeire, S. Towards a novel molecular classification of IBD. Dig. Dis. 2012, 30, 425–427. [Google Scholar] [CrossRef] [PubMed]
  71. Girardin, M.; Dionne, S.; Goyette, P.; Rioux, J.; Bitton, A.; Elimrani, I.; Charlebois, P.; Qureshi, I.; Levy, E.; Seidman, E.G. Expression and functional analysis of intestinal organic cation/L-carnitine transporter (OCTN) in Crohn’s disease. J. Crohns Colitis 2012, 6, 189–197. [Google Scholar] [CrossRef] [PubMed]
  72. Sanchez-Munoz, F.; Dominguez-Lopez, A.; Yamamoto-Furusho, J.K. Role of cytokines in inflammatory bowel disease. World J. Gastroenterol. 2008, 14, 4280–4288. [Google Scholar] [CrossRef] [PubMed]
  73. Zhu, H.; Lei, X.; Liu, Q.; Wang, Y. Interleukin-10–1082A/G polymorphism and inflammatory bowel disease susceptibility: A meta-analysis based on 17,585 subjects. Cytokine 2013, 61, 146–153. [Google Scholar] [CrossRef] [PubMed]
  74. Nahar, P.P.; Slitt, A.L.; Seeram, N.P. Anti-inflammatory effects of novel standardized solid lipid curcumin formulations. J. Med. Food 2015, 18, 786–792. [Google Scholar] [CrossRef] [PubMed]
  75. Miller, J.M.; Thompson, J.K.; MacPherson, M.B.; Beuschel, S.L.; Westbom, C.M.; Sayan, M.; Shukla, A. Curcumin: A double hit on malignant mesothelioma. Cancer Prev. Res. (Phila.) 2014, 7, 330–340. [Google Scholar] [CrossRef] [PubMed]
  76. Cai, X.Z.; Huang, W.Y.; Qiao, Y.; Du, S.Y.; Chen, Y.; Chen, D.; Yu, S.; Che, R.C.; Liu, N.; Jiang, Y. Inhibitory effects of curcumin on gastric cancer cells: a proteomic study of molecular targets. Phytomedicine 2013, 20, 495–505. [Google Scholar] [CrossRef] [PubMed]
  77. Fang, H.Y.; Chen, S.B.; Guo, D.J.; Pan, S.Y.; Yu, Z.L. Proteomic identification of differentially expressed proteins in curcumin-treated MCF-7 cells. Phytomedicine 2011, 18, 697–703. [Google Scholar] [CrossRef] [PubMed]
  78. Lou, Y.; Zheng, J.; Hu, H.; Lee, J.; Zeng, S. Application of ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry to identify curcumin metabolites produced by human intestinal bacteria. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2015, 985, 38–47. [Google Scholar] [CrossRef] [PubMed]
  79. Barnett, M.P.; McNabb, W.C.; Cookson, A.L.; Zhu, S.; Davy, M.; Knoch, B.; Nones, K.; Hodgkinson, A.J.; Roy, N.C. Changes in colon gene expression associated with increased colon inflammation in interleukin-10 gene-deficient mice inoculated with Enterococcus species. BMC Immunol. 2010, 11, 39–60. [Google Scholar] [CrossRef] [PubMed]
  80. Cooney, J.M.; Barnett, M.P.; Brewster, D.; Knoch, B.; McNabb, W.C.; Laing, W.A.; Roy, N.C. Proteomic analysis of colon tissue from interleukin-10 gene-deficient mice fed polyunsaturated fatty acids with comparison to transcriptomic analysis. J. Proteome Res. 2012, 11, 1065–1077. [Google Scholar] [CrossRef] [PubMed]
  81. Knoch, B.; Barnett, M.P.; Zhu, S.; Park, Z.A.; Nones, K.; Dommels, Y.E.; Knowles, S.O.; McNabb, W.C.; Roy, N.C. Genome-wide analysis of dietary eicosapentaenoic acid- and oleic acid-induced modulation of colon inflammation in interleukin-10 gene-deficient mice. J. Nutrigenet. Nutrigenom. 2009, 2, 9–28. [Google Scholar] [CrossRef] [PubMed]
  82. Barnett, M.P.; Cooney, J.M.; Dommels, Y.E.; Nones, K.; Brewster, D.T.; Park, Z.; Butts, C.A.; McNabb, W.C.; Laing, W.A.; Roy, N.C. Modulation of colonic inflammation in Mdr1a−/− mice by green tea polyphenols and their effects on the colon transcriptome and proteome. J. Nutr. Biochem. 2013, 24, 1678–1690. [Google Scholar] [CrossRef] [PubMed]
  83. Dommels, Y.E.M.; Butts, C.; Zhu, S.T.; Davy, M.; Martell, S.; Hedderly, D.; Barnett, M.P.G.; Broadley, K.; Roy, N.C. Characterization of inflammation in Mdr1a−/− mice and identification of related gene expression changes in colonic epithelial cells. Genes Nutr. 2007, 2, 209–223. [Google Scholar] [CrossRef] [PubMed]
  84. Nones, K.; Dommels, Y.E.; Martell, S.; Butts, C.; McNabb, W.C.; Park, Z.A.; Zhu, S.; Hedderley, D.; Barnett, M.P.; Roy, N.C. The effects of dietary curcumin and rutin on colonic inflammation and gene expression in multidrug resistance gene-deficient Mdr1a−/− mice, a model of inflammatory bowel diseases. Br. J. Nutr. 2009, 101, 169–181. [Google Scholar] [CrossRef] [PubMed]
  85. McFadden, R.M.; Larmonier, C.B.; Shehab, K.W.; Midura-Kiela, M.; Ramalingam, R.; Harrison, C.A.; Besselsen, D.G.; Chase, J.H.; Caporaso, J.G.; Jobin, C.; et al. The role of curcumin in modulating colonic microbiota during colitis and colon cancer prevention. Inflamm. Bowel Dis. 2015, 21, 2483–2494. [Google Scholar] [CrossRef] [PubMed]
  86. Bassett, S.A.; Young, W.; Barnett, M.P.; Cookson, A.L.; McNabb, W.C.; Roy, N.C. Changes in composition of caecal microbiota associated with increased colon inflammation in interleukin-10 gene-deficient mice inoculated with Enterococcus species. Nutrients 2015, 7, 1798–1816. [Google Scholar] [CrossRef] [PubMed]
  87. Lapthorne, S.; Pereira-Fantini, P.M.; Fouhy, F.; Wilson, G.; Thomas, S.L.; Dellios, N.L.; Scurr, M.; O’Sullivan, O.; Ross, R.P.; Stanton, C.; et al. Gut microbial diversity is reduced and is associated with colonic inflammation in a piglet model of short bowel syndrome. Gut Microbes 2013, 4, 212–221. [Google Scholar] [CrossRef] [PubMed]
  88. Andoh, A.; Kuzuoka, H.; Tsujikawa, T.; Nakamura, S.; Hirai, F.; Suzuki, Y.; Matsui, T.; Fujiyama, Y.; Matsumoto, T. Multicenter analysis of fecal microbiota profiles in Japanese patients with Crohn’s disease. J. Gastroenterol. 2012, 47, 1298–1307. [Google Scholar] [CrossRef] [PubMed]
  89. Moco, S.; Candela, M.; Chuang, E.; Draper, C.; Cominetti, O.; Montoliu, I.; Barron, D.; Kussmann, M.; Brigidi, P.; Gionchetti, P.; et al. Systems biology approaches for inflammatory bowel disease: Emphasis on gut microbial metabolism. Inflamm. Bowel Dis. 2014, 20, 2104–2114. [Google Scholar] [CrossRef] [PubMed]
  90. Baliga, M.S.; Joseph, N.; Venkataranganna, M.V.; Saxena, A.; Ponemone, V.; Fayad, R. Curcumin, an active component of turmeric in the prevention and treatment of ulcerative colitis: Preclinical and clinical observations. Food Funct. 2012, 3, 1109–1117. [Google Scholar] [CrossRef] [PubMed]
  91. Sreedhar, R.; Arumugam, S.; Thandavarayan, R.A.; Karuppagounder, V.; Watanabe, K. Curcumin as a therapeutic agent in the chemoprevention of inflammatory bowel disease. Drug Discov. Today 2016, 21, 843–849. [Google Scholar] [CrossRef] [PubMed]
  92. Ghosh, S.; Banerjee, S.; Sil, P.C. The beneficial role of curcumin on inflammation, diabetes and neurodegenerative disease: A recent update. Food Chem. Toxicol. 2015, 83, 111–124. [Google Scholar] [CrossRef] [PubMed]
  93. Taylor, R.A.; Leonard, M.C. Curcumin for inflammatory bowel disease: A review of human studies. Altern. Med. Rev. 2011, 16, 152–156. [Google Scholar] [PubMed]
  94. Lang, A.; Salomon, N.; Wu, J.C.; Kopylov, U.; Lahat, A.; Har-Noy, O.; Ching, J.Y.; Cheong, P.K.; Avidan, B.; Gamus, D.; et al. Curcumin in combination with mesalamine induces remission in patients with mild-to-moderate ulcerative colitis in a randomized controlled trial. Clin. Gastroenterol. Hepatol. 2015, 13, 1444–1449. [Google Scholar] [CrossRef] [PubMed]
  95. Kitture, R.; Ghosh, S.; More, P.A.; Date, K.; Gaware, S.; Datar, S.; Chopade, B.A.; Kale, S.N. Curcumin-loaded, self-assembled aloevera template for superior antioxidant activity and trans-membrane drug release. J. Nanosci. Nanotechnol. 2015, 15, 4039–4045. [Google Scholar] [CrossRef] [PubMed]
  96. Beloqui, A.; Memvanga, P.B.; Coco, R.; Reimondez-Troitino, S.; Alhouayek, M.; Muccioli, G.G.; Alonso, M.J.; Csaba, N.; de la Fuente, M.; Preat, V. A comparative study of curcumin-loaded lipid-based nanocarriers in the treatment of inflammatory bowel disease. Colloids Surf. B Biointerfaces 2016, 143, 327–335. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A potential sequence of in vitro to in vivo studies, including the application of omics technologies. This flow diagram shows a potential approach, working from in vitro through in vivo studies to human clinical trials, with the application of omics technologies at several stages. (1) There is increasing use of high-throughput techniques, such as single nucleotide polymorphism (SNP) chips, which enable genome-wide assessment to identify genetic factors which may be linked to a particular disease or health outcome. Relevant SNPs can then be incorporated into suitable in vitro assays (2) in which food compounds can be assessed for their abilities to interact with the SNP of interest and modulate its function. Effective food compounds can then be tested in appropriate animal models (3) which exhibit the relevant phenotype and/or have the SNP of interest (or one in a related gene). This can initially be used to establish if the food has an effect on phenotype and, subsequently a range of omics techniques can be applied, and data derived from them integrated, to better understand the mechanism by which a food may exert its effect. Food compounds which show efficacy both in vitro and in vivo may then be suitable as candidates for human studies, both pre-clinically (for example, to assess any possible biomarkers identified in the animal studies (4)) and, finally, clinically, to ascertain a clinically-relevant end-point, such as an improvement of IBD-associated symptoms (5). The Manhattan plot shown in (1) was originally published by Ikram, M.K.; et al. [36] and was obtained from Wikipedia (https://commons.wikimedia.org/wiki/File:Manhattan_Plot.png). This image file is licensed under the Creative Commons Attribution 2.5 Generic license.
Figure 1. A potential sequence of in vitro to in vivo studies, including the application of omics technologies. This flow diagram shows a potential approach, working from in vitro through in vivo studies to human clinical trials, with the application of omics technologies at several stages. (1) There is increasing use of high-throughput techniques, such as single nucleotide polymorphism (SNP) chips, which enable genome-wide assessment to identify genetic factors which may be linked to a particular disease or health outcome. Relevant SNPs can then be incorporated into suitable in vitro assays (2) in which food compounds can be assessed for their abilities to interact with the SNP of interest and modulate its function. Effective food compounds can then be tested in appropriate animal models (3) which exhibit the relevant phenotype and/or have the SNP of interest (or one in a related gene). This can initially be used to establish if the food has an effect on phenotype and, subsequently a range of omics techniques can be applied, and data derived from them integrated, to better understand the mechanism by which a food may exert its effect. Food compounds which show efficacy both in vitro and in vivo may then be suitable as candidates for human studies, both pre-clinically (for example, to assess any possible biomarkers identified in the animal studies (4)) and, finally, clinically, to ascertain a clinically-relevant end-point, such as an improvement of IBD-associated symptoms (5). The Manhattan plot shown in (1) was originally published by Ikram, M.K.; et al. [36] and was obtained from Wikipedia (https://commons.wikimedia.org/wiki/File:Manhattan_Plot.png). This image file is licensed under the Creative Commons Attribution 2.5 Generic license.
Ijms 17 01763 g001
Figure 2. Experimental design for an in vivo study. An example of the design of an animal study in which the effect of a food or food compound on a phenotype, and a range of associated omics outcomes, can be assessed. In this case, a relevant model of IBD (such as the multidrug resistance 1 (Mdr1a) or interleukin-10 gene-deficient (Il10−/−) mouse) and its appropriate control is used. Both control and IBD model are fed either a control diet (such as the AIN-76A rodent diet), or the control diet supplemented with a food compound that has shown potential in an in vitro model. Body weight and food intake can be assessed during the experimental period, and at the end of the study a range of samples is collected. This would include intestinal samples to assess any effects of the compound on intestinal inflammation, as well as a range of less invasive samples (blood, urine, faeces) in which potential biomarkers of the intestinal phenotype may be present, which could be applicable to future human studies.
Figure 2. Experimental design for an in vivo study. An example of the design of an animal study in which the effect of a food or food compound on a phenotype, and a range of associated omics outcomes, can be assessed. In this case, a relevant model of IBD (such as the multidrug resistance 1 (Mdr1a) or interleukin-10 gene-deficient (Il10−/−) mouse) and its appropriate control is used. Both control and IBD model are fed either a control diet (such as the AIN-76A rodent diet), or the control diet supplemented with a food compound that has shown potential in an in vitro model. Body weight and food intake can be assessed during the experimental period, and at the end of the study a range of samples is collected. This would include intestinal samples to assess any effects of the compound on intestinal inflammation, as well as a range of less invasive samples (blood, urine, faeces) in which potential biomarkers of the intestinal phenotype may be present, which could be applicable to future human studies.
Ijms 17 01763 g002

Share and Cite

MDPI and ACS Style

Ferguson, L.R.; Barnett, M.P.G. Why Are Omics Technologies Important to Understanding the Role of Nutrition in Inflammatory Bowel Diseases? Int. J. Mol. Sci. 2016, 17, 1763. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17101763

AMA Style

Ferguson LR, Barnett MPG. Why Are Omics Technologies Important to Understanding the Role of Nutrition in Inflammatory Bowel Diseases? International Journal of Molecular Sciences. 2016; 17(10):1763. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17101763

Chicago/Turabian Style

Ferguson, Lynnette R., and Matthew P. G. Barnett. 2016. "Why Are Omics Technologies Important to Understanding the Role of Nutrition in Inflammatory Bowel Diseases?" International Journal of Molecular Sciences 17, no. 10: 1763. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17101763

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