Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2013 Mar 26;8(3):e56610. doi: 10.1371/journal.pone.0056610

Induction of Olfaction and Cancer-Related Genes in Mice Fed a High-Fat Diet as Assessed through the Mode-of-Action by Network Identification Analysis

Youngshim Choi 1, Cheol-Goo Hur 2, Taesun Park 1,*
Editor: Richard L Eckert3
PMCID: PMC3608641  PMID: 23555558

Abstract

The pathophysiological mechanisms underlying the development of obesity and metabolic diseases are not well understood. To gain more insight into the genetic mediators associated with the onset and progression of diet-induced obesity and metabolic diseases, we studied the molecular changes in response to a high-fat diet (HFD) by using a mode-of-action by network identification (MNI) analysis. Oligo DNA microarray analysis was performed on visceral and subcutaneous adipose tissues and muscles of male C57BL/6N mice fed a normal diet or HFD for 2, 4, 8, and 12 weeks. Each of these data was queried against the MNI algorithm, and the lists of top 5 highly ranked genes and gene ontology (GO)-annotated pathways that were significantly overrepresented among the 100 highest ranked genes at each time point in the 3 different tissues of mice fed the HFD were considered in the present study. The 40 highest ranked genes identified by MNI analysis at each time point in the different tissues of mice with diet-induced obesity were subjected to clustering based on their temporal patterns. On the basis of the above-mentioned results, we investigated the sequential induction of distinct olfactory receptors and the stimulation of cancer-related genes during the development of obesity in both adipose tissues and muscles. The top 5 genes recognized using the MNI analysis at each time point and gene cluster identified based on their temporal patterns in the peripheral tissues of mice provided novel and often surprising insights into the potential genetic mediators for obesity progression.

Introduction

Microarray analysis has enabled the use of whole-genome expression profiling to understand the mechanisms underlying obesity and metabolic complications and to identify key genetic mediators. Statistical approaches used to analyze microarray data can be classified into 2 major categories: methods that identify differentially expressed genes [1], [2] and those that classify genes according to the functional dependency (e.g., hierarchical clustering) [3]. Although microarray analysis has yielded some promising results, it is not a very practical method considering the fact that identification of genes directly affected by a condition is difficult from the hundreds to thousands of genes that exhibit changes in expression. To overcome this problem, Berneardo et al. developed a model-based approach that accurately distinguishes a compound's targets from the indirect responders [4]. This approach, namely, the mode-of-action by network identification (MNI), involves the reverse engineering of a network model of regulatory interactions in an organism of interest by using a training dataset of whole-genome expression profiles. The MNI algorithm has been applied successfully to identify disease mediators as well as drug targets by studying gene-expression data from yeast [4], humans (A. Ergun and J.J. Collins, unpublished data), bacteria, and other organisms (X.H., unpublished data).

Differential expression can be studied from a static or temporal viewpoint. In a static experiment, the arrays are obtained irrespective of time, essentially taking a snapshot of gene expression. On the other hand, in a temporal experiment, the arrays are collected over a time course, facilitating the study of the dynamic behavior of gene expression. Most previously obtained microarray datasets were static, that is, the results obtained on the basis of the measurement of gene expression at a single time point [5]. Since the regulation of gene expression is a dynamic process, it is important to identify and characterize the changes in gene expression over time. Therefore, numerous time-series microarray experiments have been performed to study such biological processes such as abiotic stress, disease progression, and drug responses [6][8].

Microarray analysis for studying the mechanisms underlying obesity was first reported by Soukas et al. in 2000 [9]. They used approximately 6,500 murine genes in pairs of adipose tissues in ob/ob mice and wild-type lean mice. Subsequently, many such studies were conducted: more than 30 microarray approaches have been exploited in assessing the changes in gene expression in the adipose tissues, liver, hypothalamus, skeletal muscles, small intestines, and kidneys of lean and obese animals or human subjects. A frequent limitation of these studies is that they are not time-resolved and do not necessarily provide information of an end-point or disease stage. Considerably less is known about the key genetic mediators of HFD-induced obesity and the dynamics of changes in metabolic processes related to this condition. To gain more insight into the genetic mediators associated with the onset and progression of diet-induced obesity and metabolic diseases, we studied the molecular changes in response to the HFD by using an integrative time-resolved approach.

Materials and Methods

Ethics statement

All animal experiments were performed in accordance with the Korean Food and Drug Administration (KFDA) guidelines. Protocols were reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) of the Yonsei Laboratory Animal Research Center (YLARC) (Permit #: 2011-0061). All mice were maintained in the specific pathogen-free facility of the YLARC.

Animals and diets

Five-week-old male C57BL/6N mice were obtained from Orient Bio (Gyeonggi-do, South Korea). All animals were housed in specific pathogen-free conditions, with 21±2.0°C temperature, 50±5% relative humidity, and a 12 h-light/12 h-dark cycle. From a week before the diet intervention was started, all animals were fed standard chow. At the beginning of the study, mice were divided into 2 groups: (1) control group fed the normal diet (ND, n = 40) and (2) a group fed the high-fat diet (HFD, n = 40). Mice were provided food and water ad libitum. The body weight and food intake were monitored throughout the study. At 2, 4, 8, and 12 weeks after the initiation of the study, 10 animals from each group were killed. Tissues were snap-frozen immediately in liquid nitrogen and stored at −80°C until further processing.

RNA extraction for microarray analysis

Total RNA was extracted from the epididymal and subcutaneous fat tissues and gastrocnemius muscle of each mouse by using Trizol (Invitrogen, CA, USA), according to the manufacturer's recommendations. Concentrations and purity of RNA samples were determined using a Nano Drop ND-1000 spectrophotometer (Nano Drop Technologies, Inc., Wilmington, DE, USA). RNA preparations were considered suitable for array hybridization only if samples showed intact 18S and 28S rRNA bands and displayed no chromosomal peaks or RNA degradation products. The integrity of the RNA samples was determined using a Bioanalyzer 2100 System (Agilent Technologies, Palo Alto, CA, USA).

Real-time quantitative PCR

Real-time PCR amplification was performed with the SYBR Premix Ex Taq kit (Takara, Kyoto, Japan) on a Light Cycler 2 (Roche Applied Science, Indianapolis, USA). The initial denaturation step was at 95°C for 10 s, followed by 40 cycles of amplification at 95°C for 3 s and 60°C for 40 s. mRNA expression was determined using the relative standard curve method and normalized to the housekeeping gene. The primers (sense and antisense, respectively) were as follows: Gli2, 5′- GCC AAC CAG AAC AAG CAG AA-3′, 5′- CGC TTA TGA ATG GTG ATG GG -3′; Gucy2c, 5′- GTG CGG TTA CTG CTC TTC CA -3′, 5′- TTG TCC ATC ATC AGG ACG CT -3′; Olfr1181, 5′- CCT GAC AGT CAT GGC CTT TG -3′, 5′- ACC CAG GAA GCC CAG ATA AA -3′; Atp8b3, 5′- GTT TGA GCA GGA TGT GAC CG -3′, 5′- GGC TTG CAT GAA AAT GCT GT -3′; Tmem46, 5′- TTT TCC AGC AGC AGG AGC TA -3′, 5′- GCT GAG GAG AAA AGG GAT GC -3′; Pthr2, 5′- ATG CAA GGG AGA AAC CCA TC -3′, 5′- TAG ATC CTC CCA CAC AGC CA -3′; Cdh7, 5′- TGG ACT GGG CAT TTT CAA GA -3′, 5′- GGG GAT CAG CAT CTC GAT TT -3′; Mep1b, 5′- GAT GGC CAC ATA CCA TTC CA -3′, 5′- TAA GGC GAT AGC GCT CAA AA -3′; Lamc3, 5′- GAC ATG GGC TCT TGC TAC GA -3′, 5′- CGT TCT CGA ACT CAG GCA GA -3′; GAPDH, 5′- GGA GAT TGT TGC CAT CAA CG -3′, 5′- TTT GCC GTG AGT GGA GTC AT -3′.

Microarray hybridization and data analysis

Equal amounts of total RNA were pooled from 10 mice in each experimental group and subjected to microarray experiments in triplicate. For analysis, 2 μg total RNA was labeled and amplified using the Universal Linkage System antisense RNA (aRNA) labeling kit (Kreatech Diagnostics, Amsterdam, The Netherlands). The Cy5-labeled aRNAs were resuspended in 10 μL of hybridization solution (GenoCheck, Korea). The labeled aRNAs were hybridized to the NimbleGen mouse whole genome 12-plex array (Roche NimbleGen, Inc., WI, USA) that contained 60-mer probes representing 42,576 genes (average 3 probes per target). The arrays were scanned using a GenePix 4000B microarray scanner. The data were extracted from the scanned images using NimbleScan software version 2.4 (Roche NimbleGen), and the Robust Multichip Average algorithm was used to generate gene expression values. The normalized and log-transformed intensity values were then analyzed using GeneSpring GX 10 (Agilent Technologies, Santa Clara, CA, USA) and GenePlex (Istech, Inc., Seoul, South Korea). The details of labeling, hybridization, scanning, and normalization of the data are provided on the NimbleGen website (http://www.nimblegen.com). Gene expression levels between the ND and HFD samples were assessed by comparing the average expression ratios of each group. Hierarchical clustering was performed in GeneSpring GX 7.3.1 software (Agilent Technologies, Santa Clara, CA), using average gene expression values under HFD condition divided by the median of ND gene expression, per time-point.

MNI algorithm

We constructed a compendium dataset consisting of hundreds of expression profiles in the organism of interest; that the expression profiles were downloaded from the Gene Expression Omnibus, a public repository of microarray studies. The MNI algorithm was applied, using the method developed by Xing et al. [10], and was configured to output the top 200 mediators for each sample and generate the associated Z-scores for those probe sets. The Z-score for probe sets that were not within the list of the top 100 probe sets identified as mediators for a given sample were set to zero. To identify a characteristic list of genes within each group, the Z-scores across samples and probe sets for corresponding genes were averaged and ranked. The top 100 genes within that list were selected to be reported as significant genetic mediators. A higher average Z-score is an indication of higher number of occurrences of a gene on the lists generated by the MNI algorithm in each group. The 100 highest ranked genes were classified according to the biological process in which they are involved as per the criteria established by the GO.

Results

Effect of HFD feeding on visceral adiposity

The body weight gains of mice fed the 2 diets over the 12-week period are shown in Figure 1A. The difference in body weight between the 2 groups continued to increase over the course of experimental feeding: the difference was about 45% by 12 weeks. The increase in body weight associated with the HFD was partially attributed to the expansion of visceral adipose tissues. The masses of the epididymal, perirenal, mesenteric, and retroperitoneal fat pads of the mice fed HFD for 12 weeks were 42%, 40%, 54%, and 42%, respectively; the difference in the masses was larger in the HFD-fed mice than in the ND-fed group (Figure 1B–F). Moreover, HFD-fed mice exhibited significant reductions in the wet weights of the gastrocnemius (−13%) and soleus (−16%) muscles at 12 weeks compared with those in the ND-fed mice (Figure 1G and H).

Figure 1. Changes in body weight, visceral fat-pad weights, and muscle masses over time.

Figure 1

(A) Body weight gain. (B) Total visceral fat-pad weight. (C) Epididymal fat-pad weight. (D) Perirenal fat-pad weight. (E) Mesenteric fat-pad weight. (F) Retroperitoneal fat-pad weight. (G) Gastrocnemius muscle mass. (H) Soleus muscle mass. Data are presented as means ± SEM. *P<0.05.

Transcription response of WAT and muscle to HFD during the 12-week time-course

Gene expression profiling in the WAT and muscle of mice was assessed through the oligonucleotide microarray analysis. Among 25,291 genes on the NimbleGen Mouse Whole Oligo 12-plex chip used in this study, 21,890 genes (86%) were identified as known genes. After determination of the temporal effects of the HFD across 12-week time-course, we focused on dissecting the HFD specific effects on the transcriptome of epididymal and subcutaneous fats and muscle. Microarray data were analyzed by hierarchical clustering of enriched functional groups of genes (based on Gene Ontology) and the major results are graphically illustrated in a heat map (Figure 2). The HFD elicited distinct changes in gene expression in epididymal and subcutaneous fats and muscle of mice over time, and most significant changes were shown in epididymal fat tissue. Specifically, prominent expression changes were observed at the early phase (week 2 to week 4) and the enrichment of lipid metabolism and inflammatory processes were significant among the up-regulated HFD-responsive genes, whereas G-protein coupled receptor protein signaling pathway and electron transport were most significant among the down-regulated HFD-responsive genes in the epididymal fat tissue.

Figure 2. Heatmap of differentially expressed transcript sets.

Figure 2

Values used for clustering are average HFD vs. ND per time-point expression ratio. The branches of the condition tree are colored so to discriminate three subclusters with the largest distance, corresponding to three tissues of the time-course: epididymal adipose tissue (red), subcutaneous adipose tissue (blue) and gastrocnemious muscle (green). This is summarized in the color bar underneath the cluster diagram.

MNI analysis of the time course treatment with the HFD

To elucidate the time course and metabolic processes underlying obesity progression induced by the HFD, we determined the gene expression profiles of the epididymal and subcutaneous fat tissues and gastrocnemius muscle of mice by using oligonucleotide microarray analysis. Each of these data was queried against the reconstructed network (MNI algorithm), and the resulting potential genetic mediators in each case were ranked according to the Z-score statistic. The lists of top 5 potential genetic mediators for obesity progression in the epididymal and subcutaneous fat tissues and gastrocnemius muscle of mice fed the HFD for 2, 4, 8, and 12 weeks are shown in Tables 1, 2, 3. The most characteristic genes across all tissues in the list were associated with cancer; the genes in this category included Nek11, Gli2, Tmem46, Mep1b, Ccdc109b, Rab23, Patz1, and Hdac9. The second representative functional theme was related to olfactory transduction, and these genes included Olfr1181, Olfr1173, Olfr855, Olfr1056, Olfr716, and Tmem16b. To validate the microarray results quantitatively, we analyzed the mRNA expression levels of top-ranked genes by real-time PCR. In all cases, a strong correspondence between the microarray data and the real-time PCR results was observed (Figure 3). We also measured the basal expression levels of selected genes including several olfactory receptors in the epididymal fat tissues of ND- or HFD-fed mice, using real-time PCR. The results indicated that the basal expression levels of highly ranked olfactory genes (Olfr1181, Olfr513, Olfr960, and Olfr1245) were comparable to those of top five genes (Gli2, Gucy2c, Atp8b3, and Tmem46) identified by MNI analysis at week 4 in the epididymal adipose tissue (Figure S1).

Table 1. List of top 5 genes identified by the MNI analysis at each time point in the epididymal fat tissue of HFD-induced obese mice.

Rank Gene accession No. Gene symbol Description Function Fold change
Epididymal fat tissue
2 week 1 NM_023173 Dusp12 Dual specificity phosphatase 12 Insulin resistance 2.52
2 NM_172461 Nek11 NIMA (never in mitosis gene a)-related expressed kinase 11 Cancer 0.43
3 NM_145489 AI661453 Expressed sequence AI661453 Unknown 0.23
4 NM_177078 Adrbk2 Adrenergic receptor kinase, beta 2 Bipolar disorder 2.73
5 NM_013679 Svs6 Seminal vesicle secretory protein 6 Unknown 0.77
4 week 1 XM_136212 Gli2 GLI-Kruppel family member GLI2 Cancer 2.73
2 NM_145067 Gucy2c Guanylate cyclase 2c Cancer 0.34
3 NM_001011816 Olfr1181 Olfactory receptor 1181 Olfactory transduction 5.47
4 NM_026094 Atp8b3 ATPase, Class I, type 8B, member 3 ATP binding 3.25
5 NM_145463 Tmem46 Transmembrane protein 46 Cancer 0.55
8 week 1 NM_199155 Tas2r110 Taste receptor, type 2, member 110 Sensory perception of taste 2.81
2 AK138164 Cntn5 Contactin 5 Cell adhesion 0.33
3 NM_152220 Stx3 Syntaxin 3 Arachidonic acid binding 1.81
4 NM_178924 Upk1b Uroplakin 1B Epithelial cell differentiation 1.79
5 NM_028622 Lce1c Late cornified envelope 1C Unknown 0.32
12 week 1 NM_139270 Pthr2 Parathyroid hormone receptor 2 Parathyroid hormone receptor activity 2.39
2 NM_172853 Cdh7 Cadherin 7, type 2 Calcium ion binding 2.42
3 NM_008586 Mep1b Meprin 1 beta Cancer 0.33
4 NM_011836 Lamc3 Laminin gamma 3 Cell adhesion 0.49
5 NM_145463 Tmem46 Transmembrane protein 46 Cancer 0.59

Table 2. List of top 5 genes identified by the MNI analysis at each time point in the subcutaneous fat tissue of HFD-induced obese mice.

Rank Gene accession No. Gene symbol Description Function Fold change
Subcutaneous fat tissue
2 week 1 NM_025779 Ccdc109b Coiled-coil domain containing 109B Cancer 0.54
2 NM_001025438 Camk2d Calcium/calmodulin-dependent protein kinase II, delta Calmodulin binding 0.65
3 AB211064 L1td1 LINE-1 type transposase domain containing 1 Unknown 2.61
4 NM_026345 Mansc1 MANSC domain containing 1 Unknown 3.03
5 NM_207566 Olfr1173 Olfactory receptor 1173 Olfactory transduction 1.97
4 week 1 NM_008529 Ly6e Lymphocyte antigen 6 complex, locus E Adrenal gland development 1.31
2 NM_146524 Olfr855 Olfactory receptor 855 Olfactory transduction 1.57
3 NM_018744 Sema6a Sema domain, transmembrane domain (TM), and cytoplasmic domain, (semaphorin) 6A Nervous system development 0.73
4 AB211064 L1td1 LINE-1 type transposase domain containing 1 Unknown 3.13
5 NM_183015 Ccnb3 Cyclin B3 Cell cycle 2.58
8 week 1 NM_153111 Fev FEV (ETS oncogene family) Nervous system development 0.39
2 AB211064 L1td1 LINE-1 type transposase domain containing 1 Unknown 2.8
3 NM_147018 Olfr1056 Olfactory receptor 1056 Olfactory transduction 0.73
4 NM_008999 Rab23 RAB23, member RAS oncogene family Cancer 0.56
5 NM_080644 Cacng5 Calcium channel, voltage-dependent, gamma subunit 5 Calcium ion transport 0.36
12 week 1 NM_001024852 Auts2 Autism susceptibility candidate 2 Mental retardation 0.53
2 NM_178046 Svil Supervillin Unknown 0.57
3 NM_018764 Pcdh7 Protocadherin 7 Cell adhesion 1.82
4 NM_146604 Olfr716 Olfactory receptor 716 Olfactory transduction 3.08
5 BC089489 4930474M22Rik RIKEN cDNA 4930474M22 gene Unknown 1.9

Table 3. List of top 5 genes identified by the MNI analysis at each time point in the gastrocnemius muscle of HFD-induced obese mice.

Rank Gene accession No. Gene symbol Description Function Fold change
Gastrocnemius muscle
2 week 1 NM_019574 Patz1 POZ (BTB) and AT hook containing zinc finger 1 Cancer 1.8
2 NM_020610 Nrip3 Nuclear receptor interacting protein 3 Inflammation 0.42
3 NM_024124 Hdac9 Histone deacetylase 9 Cancer 0.55
4 XM_975536 Armc4 Armadillo repeat containing 4 Unknown 0.54
5 NM_139226 Onecut3 One cut domain, family member 3 DNA binding 1.34
4 week 1 NM_153589 Tmem16b Transmembrane protein 16B Olfactory transduction 0.74
2 NM_175540 Eda2r Ectodysplasin A2 isoform receptor Alopecia 0.88
3 NM_008355 Il13 Interleukin 13 Inflammation 1.57
4 NM_010608 Kcnk3 Potassium channel, subfamily K, member 3 Ion transport 1.22
5 XM_129809 Ogfrl1 Opioid growth factor receptor-like 1 Unknown 0.7
8 week 1 NM_024124 Hdac9 Histone deacetylase 9 Cancer 0.6
2 NM_011990 Slc7a11 Solute carrier family 7 Amino acid transport 0.7
3 NM_175420 9330176C04Rik RIKEN cDNA 9330176C04 gene Unknown 2.17
4 NM_016961 Mapk9 Mitogen activated protein kinase 9 Insulin resistance 0.59
5 NM_027462 Wars2 Tryptophanyl tRNA synthetase 2 (mitochondrial) Vasculogenesis 0.56
12 week 1 NM_008114 Gfi1 Growth factor independent 1B Hematopoiesis 0.74
2 NM_145435 Pyy Peptide YY Insulin resistance 1.49
3 NM_138648 Olr1 Oxidized low density lipoprotein (lectin-like) receptor 1 Inflammation 0.33
4 NM_177861 Tmem67 Transmembrane protein 67 Mental retardation 0.54
5 XM_887155 Igsf10 Immunoglobulin superfamily, member 10 Unknown 0.42

Figure 3. Quantitative PCR.

Figure 3

Quantitative real-time PCR analysis of the mRNA expression on selected gene targets identified by MNI analysis in the epididymal fat tissues of mice. Results are presented as the average ± SEM of at least 3 separate experiments.

Functional analysis of the highly ranked genetic mediators

We next focused on the GO-annotated pathways that were significantly overrepresented among the highly ranked genetic mediators. For our analysis, we subjected the 100 highest ranked genes identified by MNI analysis in the epididymal and subcutaneous fat tissue and gastrocnemius muscle of mice with diet-induced obesity to pathway analysis based on the GO biological process annotations (Tables 4, 5, 6). We found that the olfactory transduction was highly enriched in the epididymal and subcutaneous fat tissue and gastrocnemius muscle of the HFD-fed mice compared to the ND-fed mice at all time points. Even the second representative functional theme of epididymal fat was related to cancer at all time points. The pathways thought to be associated with obesity progression in the epididymal fat as per the MNI analysis included Wnt signaling pathway, melanogenesis, chemokine signaling pathway, focal adhesion, MAPK signaling pathway, purine metabolism, regulation of actin cytoskeleton, neuroactive ligand-receptor interaction, and extracellular matrix (ECM)-receptor interaction. In the subcutaneous fat, other pathways identified by the MNI analysis for obesity progression included calcium signaling pathway, gonadotropin-releasing hormone (GnRH) signaling pathway, axon guidance, cell cycle, and tyrosine metabolism. In the gastrocnemius muscle, besides the olfactory transduction mentioned above, the over-represented groups identified according to GO biological processes for obesity progression were those involved in the various cellular processes such as neuroactive ligand-receptor interaction, cytokine-cytokine receptor interaction, pathways associated with cancer, insulin signaling pathway, pathways associated with colorectal cancer, adipocytokine signaling pathway, type II diabetes mellitus, and cell adhesion molecules.

Table 4. The enriched pathways among top 100 genetic mediators identified by the MNI analysis at each time point in the epididymal fat tissue of HFD-induced obese mice.

GO ontology Ranked pathway genes (rank)
Epididymal fat tissue
2 week Olfactory transduction Adrbk2 (4), Olfr513 (6), Olfr433 (15), Camk2g (28), Olfr1245 (29), Olfr1143 (36), Olfr996 (55), Olfr960 (57), Arrb2 (79)
Wnt signaling pathway Camk2g (28), Rhoa (31), Wnt10a (95)
Melanogenesis Camk2g (28), Adcy5 (80), Wnt10a (95)
Chemokine signaling pathway Rhoa (31), Arrb2 (79), Adcy5 (80)
Focal adhesion Rhoa (31), Lamc3 (44), Bcl2 (77)
Pathways in cancer Rhoa (31), Bcl2 (77), Wnt10a (95)
4 week Olfactory transduction Camk2g (17), Olfr513 (27), Olfr960 (31), Olfr1245 (43), Arrb2 (55)
Pathways in cancer Gli2 (1), Lamc3 (9), Bcl2 (58), Fgf5 (65)
MAPK signaling pathway Arrb2 (55), Fgf5 (65), Mapkapk5 (75)
Purine metabolism Gucy2c (2), Nme7 (47), Cant1 (85)
8 week Olfactory transduction Olfr536 (11), Olfr513 (31), Olfr654 (37), Camk2g (74), Olfr652 (99)
Focal adhesion Flnc(16), Bcl2 (64), Col6a2 (83), Rhoa (86), Mylk (96),
Regulation of actin cytoskeleton Fgf5 (26), Rhoa (86), Mylk (96)
Pathways in cancer Fgf5 (26), Bcl2 (64), Rhoa (86)
12 week Olfactory transduction Olfr16 (6), Camk2g (10), Olfr715 (24), Olfr1245 (32), Olfr536 (36), Olfr1143 (52)
Neuroactive ligand-receptor interaction Pth2r (1), Agtrl1 (16), Vipr2 (27), P2rx6 (74)
Focal adhesion Lamc3 (4), Bcl2 (75), Col6a2 (76), Rhoa (88)
ECM-receptor interaction Lamc3 (4), Cd44 (59), Col6a2 (76)
Pathways in cancer Lamc3 (4), Bcl2 (75), Rhoa (88)

Table 5. The enriched pathways among top 100 genetic mediators identified by the MNI analysis at each time point in the subcutaneous fat tissue of HFD-induced obese mice.

GO ontology Ranked pathway genes (rank)
Subcutaneous fat tissue
2 week Olfactory transduction Camk2b (2), Olfr1173 (5), Olfr823 (9), Guca1a (41), Olfr411 (49), Olfr1408 (51), Olfr875 (64)
Calcium signaling pathway Camk2b (2), Cacna1d (15), Htr4 (45), Ryr1 (96)
GnRH signaling pathway Camk2b (2), Cacna1d (15), Cga (48)
4 week Olfactory transduction Olfr855 (2), Olfr888 (13), Olfr305 (36), Olfr1173 (49), Olfr395 (52), Olfr411 (55), Olfr823 (83), Olfr1409 (89)
Axon guidance Sema6a (3), Abl1 (23)
Cell cycle Ccnb3 (5), Abl1 (23)
Regulation of actin cytoskeleton Vav3 (9), Ssh2 (25)
Tyrosine metabolism Fah (26), Aoc3 (27)
MAPK signaling pathway Cacna1d (28), Hspa1a (44)
8 week Olfactory transduction Olfr1056 (3), Olfr960 (15), Olfr1408 (26), Olfr855 (35), Olfr609 (42), Olfr1173 (49), Olfr411 (71), Olfr205 (91), Olfr1121 (94)
Focal adhesion Shc4 (63), Diap1 (77), Lamb2 (82), Vav3 (83)
12 week Olfactory transduction Olfr716 (4), Olfr45 (16), Olfr960 (19), Olfr1173 (32), Olfr411 (34), Olfr1408 (43), Olfr855 (45), Olfr875 (96)
GnRH signaling pathway Cacna1d (29), Cga (58)
Chemokine signaling pathway Vav3 (33), Gng3 (79), Shc4 (84)

Table 6. The enriched pathways among top 100 genetic mediators identified by the MNI analysis at each time point in the gastrocnemius muscle of HFD-induced obese mice.

GO ontology Ranked pathway genes (rank)
Gastrocnemius muscle
2 week Olfactory transduction Clca3 (6), Olfr739 (14), Olfr488 (40), Olfr474 (55)
4 week Olfactory transduction Olfr800 (28), Olfr978 (33), Olfr474 (49), Olfr488 (73)
Neuroactive ligand-receptor interaction Oprl1 (30), Mc1r (78), Tbxa2r (89), Tspo (97)
Cytokine-cytokine receptor interaction Eda2r (2), Il13 (3), Cx3cl1 (91)
Pathways in cancer Dcc (10), Amn (71), Mlh1 (85)
8 week Olfactory transduction Clca3 (9), Olfr739 (48), Olfr1305 (64), Olfr1395 (80), Olfr140 (81), Olfr63 (85), Olfr689 (86)
Pathways in cancer Mapk9 (4), Dcc (15), Mapk10 (78), Grb2 (87), Mitf (89)
Insulin signaling pathway Mapk9 (4), Irs3 (40), Mapk10 (78), Grb2 (87)
Colorectal cancer Mapk9 (4), Dcc (15), Mapk10 (78), Grb2 (87)
Adipocytokine signaling pathway Type II diabetes mellitus Mapk9 (4), Irs3 (40), Mapk10 (78)
12 week Olfactory transduction Olfr689 (22), Olfr488 (24), Olfr347 (38), Olfr1204 (69)
Cell adhesion molecules (CAMs) Cd22 (23), Nlgn3 (44), Ptprm (62)
Cytokine-cytokine receptor interaction Eda2r (43), Ppbp (53), Cx3cl1 (57)

Representative time-course profile clusters

We subjected the 40 highest ranked genes identified by MNI analysis at each time point in the epididymal and subcutaneous fat tissues and gastrocnemius muscle of mice with diet-induced obesity to clustering based on their temporal pattern. Figure 4 shows genes that were observed to have decreasing ranking across time point, with a peak at 2 week. Biological processes controlled by genes in this cluster included regulation of insulin resistance (EA: Dusp12), cancer (EA: Nek11, A4gnt; SA: Srp9), inflammation (EA: Siglece; M: Nrip3, Sez6l), and olfactory transduction (EA: Olfr 513, 433; SA: Olfr 823) (Tables 7, 8, 9). The genes shown in Figures 5 and 6 exhibited the highest rank at the intermediate time points of 4 and 8 weeks, respectively. For both clusters, the majority of genes in this category were associated with cancer (EA: Gli2, Gucy2c, Lsm1, Duoxa1, Lasp1; SA: Vav3, Kcnrg, Tle6, Rab23; M: Dcc, Rassf2, Perp, Pdgfr1), inflammation (SA: Btn2a2, Def6; M: ll13, Rap1gds1), insulin resistance (SA: Neurod4; M: Mapk9), and olfactory transduction (EA: Olfr 1181, 960, 536, 654, 527; SA: Olfr 855, 888, 305, 1056, 960, 685, 1048; M: Olfr 699, 800, 978, 232, 872) (Tables 10, 11, 12, 13, 14, 15). Figure 7 shows genes that exhibited increasing ranking across all time points, with a peak at 12 week. Biological processes for genes in this cluster included regulation of adipogenesis (EA: Smad7, Adhfe1), food intake (M: Pyy), inflammation (EA: Folr2, Pde7a, Vipr2; SA: Rfxdc2, Aqp5, Cpb2), cancer (M: Lin28, Gstm1, Safb2), and olfactory transduction (EA: Olfr 16, 1000; SA: Olfr 716, 45; M: Olfr 689, 347) (Tables 16, 17, 18). The genes shown in Figure 8 exhibited a constant high MNI ranking throughout the time course. This cluster contained genes related to cancer (EA: Tmem46; SA: Trim62; M: Hdac9), insulin resistance (EA: Camk2g), hepatic fibrosis (M: Tmem67), and olfactory transduction (SA: Olfr 1173, 411, 855) (Table 19).

Figure 4. Results of MNI analysis at week 2.

Figure 4

Genes that exhibited decreasing ranking across time points as revealed by the MNI analysis, with a peak at 2 week in the peripheral tissues of mice. (A) Epididymal adipose tissue. (B) Subcutaneous adipose tissue. (C) Muscle.

Table 7. List of genes that exhibited decreasing ranking across time point as revealed by the MNI analysis, with a peak at 2 week in the epididymal adipose tissue of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Epididymal adipose tissue
Insulin resistance
NM_023173 Dusp12 Dual specificity phosphatase 12 2.5 1.8 2.1 1.7
Inflammation
NM_031181 Siglece Sialic acid binding Ig-like lectin E 2.1 1.9 1.7 1.7
Cancer
NM_172461 Nek11 NIMA (never in mitosis gene a)-related expressed kinase 11 0.4 0.5 0.6 0.6
XM_286168 A4gnt Alpha-1,4-N-acetylglucosaminyltransferase 2.0 1.7 1.6 1.5
Olfactory transduction
NM_146723 Olfr513 Olfactory receptor 513 0.3 0.3 0.3 0.4
NM_146717 Olfr433 Olfactory receptor 433 2.1 2.0 1.9 1.5
Others
NM_022318 Popdc2 Popeye domain containing 2 2.6 2.0 1.8 1.9
XM_901428 EG629397 Predicted gene, EG629397 1.8 1.7 1.5 1.4
XM_899101 Camsap1 Calmodulin regulated spectrin-associated protein 1-like 1 3.4 2.2 2.3 2.6
NM_177769 Elmod1 ELMO domain containing 1 1.7 1.6 1.5 1.4
NM_010768 Matk Megakaryocyte-associated tyrosine kinase 2.3 1.8 1.7 1.8
NM_177078 Adrbk2 Adrenergic receptor kinase, beta 2 2.7 2.1 2.2 2.1
NM_013679 Svs6 Seminal vesicle secretory protein 6 0.8 0.7 0.7 0.7
XM_486653 EG434760 Predicted gene, EG434760 0.4 0.5 0.4 0.4
NM_001030297 Tmprss11c Transmembrane protease, serine 11c 1.9 1.5 2.0 1.7
NM_026338 4921517D21Rik RIKEN cDNA 4921517D21 gene 2.9 2.5 2.8 2.0
NM_133879 Zbtb4 Zinc finger and BTB domain containing 48 0.4 0.2 0.3 0.5
NM_001034875 Gm1006 Gene model 1006, (NCBI) 2.1 2.1 1.7 2.0
NM_016802 Rhoa Ras homolog gene family, member A 2.1 2.1 1.7 2.0
NM_177887 BC022651 cDNA sequence BC022651 1.8 1.4 1.6 1.5
NM_207670 Grasp GRIP1 associated protein 1.4 1.2 1.3 1.3
NM_183124 Defb41 Defensin beta 41 0.7 0.8 0.8 0.7

Table 8. List of genes that exhibited decreasing ranking across time point as revealed by the MNI analysis, with a peak at 2 week in the subcutaneous adipose tissue of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Subcutaneous adipose tissue
Cancer
NM_012058 Srp9 Signal recognition particle 9 0.5 0.6 0.7 0.5
Olfactory transduction
NM_146673 Olfr823 Olfactory receptor 823 0.6 0.6 0.7 0.7
Others
NM_025779 Ccdc109b Coiled-coil domain containing 109B 0.5 0.7 0.7 0.6
NM_001025438 Camk2d Calcium/calmodulin-dependent protein kinase II, delta 0.8 0.8 0.8 0.8
AB211064 L1td1 LINE-1 type transposase domain containing 1 3.1 2.8 1.9 1.9
NM_008748 Dusp8 Dual specificity phosphatase 8 0.6 0.6 0.8 0.8
XM_905633 Armcx4 Armadillo repeat containing, X-linked 4 1.9 2.0 2.4 2.4
NM_001034860 EG229571 Predicted gene, EG229571 1.6 1.5 1.4 1.4
XM_898433 AK129128 cDNA sequence AK129128 2.3 1.8 1.9 1.4
XM_355152 Gm967 Gene model 967, (NCBI) 0.5 0.6 0.6 0.7
NM_016869 Corin Corin 0.7 0.7 0.8 0.8
NM_153386 Clrn1 Clarin 1 1.3 1.2 1.3 1.3
NM_029911 Kcnk10 Potassium channel, subfamily K, member 10 0.7 0.7 0.8 0.8
NM_026943 Snrpd2 Small nuclear ribonucleoprotein D2 0.6 0.8 0.7 0.7
NM_134000 Traf3ip2 Traf3 interacting protein 2 1.5 1.3 1.2 1.2
NM_023790 Wdr5 WD repeat domain 54 1.8 1.3 1.3 0.9
XM_987216 1700072O05Rik RIKEN cDNA 1700072O05 gene 0.6 0.6 0.6 0.7
XM_146632 9030420J04Rik RIKEN cDNA 9030420J04 gene 2.5 2.1 2.0 1.5
NM_015756 Shroom3 Shroom family member 3 1.9 1.4 1.3 1.5

Table 9. List of genes that exhibited decreasing ranking across time point as revealed by the MNI analysis, with a peak at 2 week in the muscle of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Muscle
Inflammation
NM_020610 Nrip3 Nuclear receptor interacting protein 3 0.4 0.5 0.7 0.5
BC065117 Sez6l Seizure related 6 homolog like 1.7 1.7 1.7 1.3
Others
NM_019574 Patz1 POZ (BTB) and AT hook containing zinc finger 1 1.8 1.4 1.3 1.5
XM_975536 Armc4 Armadillo repeat containing 4 0.5 0.4 0.5 0.6
NM_139226 Onecut3 One cut domain, family member 3 1.3 1.4 1.3 1.2
NM_177596 EG210853 Predicted gene, EG210853 0.4 0.6 0.7 0.7
NM_175455 6430502M16RIk RIKEN cDNA 6430502M16 gene 2.3 1.6 1.7 1.7
NM_010243 Fut9 Fucosyltransferase 9 0.8 0.9 0.9 0.9
NM_024178 Alg14 Asparagine-linked glycosylation 14 homolog (yeast) 0.4 0.5 0.6 0.6
NM_138954 Rfpl4 Ret finger protein-like 4 0.6 0.8 0.7 0.7
XM_900215 0610010O12Rik RIKEN cDNA 0610010O12 gene 1.6 1.3 1.4 1.3
NM_025679 5730470L24Rik RIKEN cDNA 5730470L24 gene 0.8 0.8 0.8 0.8
NM_027588 Nt5c1b 5′-nucleotidase, cytosolic IB 0.7 0.7 0.7 0.6
NM_177239 Mysm1 Myb-like, SWIRM and MPN domains 1 0.5 0.6 0.5 0.7
XM_126537 6030468B19Rik RIKEN cDNA 6030468B19 gene 1.6 1.4 1.3 1.2
NM_028047 1500002O20Rik RIKEN cDNA 1500002O20 gene 1.3 1.2 1.3 1.2
U66058 Lig3 Ligase III, DNA, ATP-dependent 0.8 0.8 0.8 0.8

Figure 5. Results of MNI analysis at week 4.

Figure 5

Genes that exhibited the highest rank at the intermediate time point of 4 week as revealed by the MNI analysis in the peripheral tissues of mice. (A) Epididymal adipose tissue. (B) Subcutaneous adipose tissue. (C) Muscle.

Figure 6. Results of MNI analysis at week 8.

Figure 6

Genes that exhibited the highest rank at the intermediate time point of 8 week as revealed by the MNI analysis in the peripheral tissues of mice. (A) Epididymal adipose tissue. (B) Subcutaneous adipose tissue. (C) Muscle.

Table 10. List of genes that exhibited the highest ranking at the intermediate time point of 4 week as revealed by the MNI analysis in the epididymal adipose tissue of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Epididymal adipose tissue
cancer
XM_136212 Gli2 GLI-Kruppel family member GLI2 2.2 2.7 2.4 1.8
NM_145067 Gucy2c Guanylate cyclase 2c 0.4 0.3 0.5 0.5
NM_138721 Lsm1 U7 snRNP-specific Sm-like protein LSM10 0.3 0.9 0.8 0.6
NM_145395 Duoxa1 Dual oxidase maturation factor 1 1.7 1.7 1.7 1.5
NM_010688 Lasp1 LIM and SH3 protein 1 2.1 2.5 2.8 2.3
olfacroty transduction
NM_001011816 Olfr1181 Olfactory receptor 1181 3.1 5.5 2.9 3.9
NM_146279 Olfr960 Olfactory receptor 960 2.4 2.3 2.1 1.8
Others
NM_026094 Atp8b3 ATPase, Class I, type 8B, member 3 2.5 3.3 2.5 2.1
NM_029857 Tmco4 Transmembrane and coiled-coil domains 4 0.5 0.3 0.6 0.4
NM_181419 BC050092 cDNA sequence BC050092 2.2 3.4 2.8 2.6
NM_145514 Wdr26 WD repeat domain 26 0.4 0.3 0.4 0.4
XM_125673 Cxxc6 CXXC finger 6 2.4 2.9 2.2 1.8
BC019404 6030465E24Rik RIKEN cDNA 6030465E24 gene 1.4 1.7 1.6 1.6
NM_028015 Lass5 Longevity assurance homolog 5 (S. cerevisiae) 2.4 3.5 2.4 2.3
NM_010184 Fcer1a Fc receptor, IgE, high affinity I, alpha polypeptide 0.6 0.5 0.7 0.6
NM_007378 Abca4 ATP-binding cassette, sub-family A (ABC1), member 4 1.7 1.9 1.8 1.5
NM_029607 2310003C23Rik RIKEN cDNA 2310003C23 gene 0.4 0.3 0.4 0.5
NM_001033541 EG331493 Predicted gene, EG331493 1.3 1.5 1.4 1.4
NM_019656 Tspan6 Tetraspanin 6 2.0 2.8 2.4 2.2
NM_027366 Ly6g6e Lymphocyte antigen 6 complex, locus G6E 1.7 1.5 1.5 1.4
BC096543 B930095G15Rik RIKEN cDNA B930095G15 gene 3.6 3.8 2.7 2.7
NM_133199 Scn4a Sodium channel, voltage-gated, type IV, alpha 0.4 0.4 0.5 0.5
NM_027661 Hsfy2 Heat shock transcription factor, Y linked 2 1.4 1.4 1.5 1.3

Table 11. List of genes that exhibited the highest ranking at the intermediate time point of 4 week as revealed by the MNI analysis in the subcutaneous adipose tissue of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Subcutaneous adipose tissue
Inflammation
NM_175938 Btn2a2 Butyrophilin, subfamily 2, member A2 1.6 2.4 1.8 1.5
Cancer
NM_020505 Vav3 Vav 3 oncogene 1.3 1.6 1.4 1.5
NM_206974 Kcnrg Potassium channel regulator 1.3 1.7 1.5 1.3
NM_053254 Tle6 Transducin-like enhancer of split 6, homolog of drosophila E(spl) 0.7 0.6 0.8 0.7
olfactory transduction
NM_146524 Olfr855 Olfactory receptor 855 1.7 1.6 2.0 1.6
NM_146424 Olfr888 Olfactory receptor 888 0.6 0.5 0.6 0.7
NM_146616 Olfr305 Olfactory receptor 305 0.7 0.6 0.6 0.7
Others
NM_018744 Sema6a Sema domain, transmembrane domain (TM), and cytoplasmic domain, (semaphorin) 6A 0.8 0.7 0.7 0.8
NM_183015 Ccnb3 Cyclin B3 1.8 2.6 1.5 1.8
NM_134220 V1ri3 Vomeronasal 1 receptor, I3 1.6 2.3 1.6 1.7
NM_026082 Dock7 Dedicator of cytokinesis 7 1.6 2.2 1.5 1.4
NM_153761 Mill2 MHC I like leukocyte 2 0.7 0.6 0.7 0.8
NM_001031621 Abca17 ATP-binding cassette, sub-family A (ABC1), member 17 1.3 1.7 1.4 1.4
NM_175013 Pgm5 Phosphoglucomutase 5 1.5 1.5 1.5 1.3
NM_178712 Gpr64 G protein-coupled receptor 64 1.3 1.6 1.6 1.4
NM_177839 Tnn Tenascin N 0.8 0.7 0.8 0.9
NM_198644 Zfat1 ZFAT zinc finger 1 2.1 2.7 2.1 2.0
NM_145512 Sft2d2 SFT2 domain containing 2 1.2 1.5 1.4 1.3
NM_029012 Sppl3 Signal peptide peptidase 3 0.7 0.5 0.7 0.7
NM_009594 Abl1 v-abl Abelson murine leukemia oncogene 1 0.7 0.7 0.7 0.8
NM_177710 Ssh2 Slingshot homolog 2 (Drosophila) 0.7 0.6 0.8 0.7
NM_010176 Fah Fumarylacetoacetate hydrolase 0.7 0.7 0.8 0.8
NM_009675 Aoc3 Amine oxidase, copper containing 3 1.2 1.4 1.3 1.3
XM_149023 Gm553 Gene model 553, (NCBI) 2.0 1.9 1.8 1.6
XM_987216 1700072O05Rik RIKEN cDNA 1700072O05 gene 0.6 0.6 0.6 0.7
NM_173784 Ubtd2 Ubiquitin domain containing 2 1.6 2.0 1.6 1.4
NM_183131 4930451I11Rik RIKEN cDNA 4930451I11 gene 1.8 2.2 1.8 1.4
NM_027661 Hsfy2 Heat shock transcription factor, Y linked 2 1.9 3.0 0.5 0.9
XM_354998 Ccdc78 Coiled-coil domain containing 78 1.2 1.5 1.3 1.2
NM_001033407 Gm815 Gene model 815, (NCBI) 0.7 0.5 0.7 0.7

Table 12. List of genes that exhibited the highest ranking at the intermediate time point of 4 week as revealed by the MNI analysis in the muscle of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Muscle
Inflammation
NM_008355 Il13 Interleukin 13 1.3 1.6 1.3 1.3
NM_145544 Rap1gds1 RAP1, GTP-GDP dissociation stimulator 1 1.3 1.5 1.5 1.4
Cancer
NM_007831 Dcc Deleted in colorectal carcinoma 0.8 0.6 0.6 0.7
NM_175445 Rassf2 Ras association (RalGDS/AF-6) domain family 2 0.6 0.5 0.7 0.7
olfactory transduction
NM_153589 Tmem16b Transmembrane protein 16B 0.7 0.7 0.7 0.7
NM_001011862 Olfr699 Olfactory receptor 699 0.5 0.4 0.6 0.7
NM_146548 Olfr800 Olfactory receptor 800 0.7 0.5 0.6 0.6
NM_147105 Olfr978 Olfactory receptor 978 0.7 0.5 0.5 0.7
Others
NM_175540 Eda2r Ectodysplasin A2 isoform receptor 0.8 0.9 0.8 0.9
NM_010608 Kcnk3 Potassium channel, subfamily K, member 3 1.1 1.2 1.2 1.2
XM_129809 Ogfrl1 Opioid growth factor receptor-like 1 0.8 0.7 0.8 0.8
NM_011776 Zp3 Zona pellucida glycoprotein 3 1.8 2.0 1.5 1.4
NM_009130 Scg3 Secretogranin III 0.6 0.5 0.7 0.8
NM_173732 BC030440 cDNA sequence BC030440 1.3 1.2 1.2 1.2
XM_356935 EG383229 Predicted gene, EG383229 1.3 1.5 1.2 1.3
XM_992003 Thsd7a Thrombospondin, type I, domain containing 7A 1.5 1.3 1.4 1.4
NM_178005 Lrrtm2 Leucine rich repeat transmembrane neuronal 2 0.8 0.7 0.7 0.8
NM_011012 Oprl1 Opioid receptor-like 1 1.3 1.6 1.5 1.4
NM_028968 Ifitm7 Interferon induced transmembrane protein 7 0.6 0.4 0.6 0.6
NM_175204 5830406J20Rik RIKEN cDNA 5830406J20 gene 1.6 1.9 1.7 1.5
NM_013903 Mmp20 Matrix metallopeptidase 20 (enamelysin) 0.7 0.7 0.8 0.8
NM_177860 EG329763 Predicted gene, EG329763 0.8 0.7 0.7 0.7

Table 13. List of genes that exhibited the highest ranking at the intermediate time point of 8 week as revealed by the MNI analysis in the epididymal adipose tissue of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Epididymal adipose tissue
Olfactory transduction
NM_146520 Olfr536 Olfactory receptor 536 0.6 0.6 0.5 0.5
NM_146379 Olfr654 Olfactory receptor 654 1.9 2.4 2.8 1.9
NM_001011776 Olfr527 Olfactory receptor 527 1.7 1.5 1.8 1.8
Others
NM_199155 Tas2r110 Taste receptor, type 2, member 110 1.9 2.5 2.8 2.8
AK138164 Cntn5 Contactin 5 0.4 0.4 0.3 0.5
NM_152220 Stx3 Syntaxin 3 1.8 1.4 1.8 1.8
NM_178924 Upk1b Uroplakin 1B 1.4 1.8 1.8 1.7
NM_028622 Lce1c Late cornified envelope 1C 0.5 0.4 0.3 0.5
NM_027815 9030624J02Rik RIKEN cDNA 9030624J02 gene 0.7 0.7 0.8 0.7
NM_008633 Mtap4 Microtubule-associated protein 4 1.4 1.4 1.4 1.2
NM_020518 Vsig2 V-set and immunoglobulin domain containing 2 2.0 1.8 2.1 1.8
NM_138311 H1foo H1 histone family, member O, oocyte-specific 0.6 0.6 0.5 0.5
NM_153124 St8sia5 ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 5 0.8 0.7 0.7 0.7
XM_898823 Flnc Filamin C, gamma (actin binding protein 280) 0.5 0.5 0.4 0.4
NM_181419 BC050092 cDNA sequence BC050092 2.2 3.4 2.8 2.6
XM_001004783 EG544848 Predicted gene, EG544848 0.5 0.6 0.5 0.5
NM_009608 Actc1 Actin, alpha, cardiac 1.8 2.3 2.7 2.3
NM_010203 Fgf5 Fibroblast growth factor 5 0.5 0.5 0.5 0.5
NM_011827 Hcst Hematopoietic cell signal transducer 2.0 2.2 2.0 1.7
NM_026825 Lrrc16 Leucine rich repeat containing 16 1.4 1.5 1.7 1.5
NM_029415 Slc10a6 Solute carrier family 10 (sodium/bile acid cotransporter family), member 6 0.5 0.4 0.3 0.5
U10093 Klra7 Killer cell lectin-like receptor, subfamily A, member 7 1.2 1.2 1.2 1.2
NM_007379 Abca2 ATP-binding cassette, sub-family A (ABC1), member 2 0.5 0.6 0.5 0.4
NM_001039878 Strn Striatin, calmodulin binding protein 4 0.6 0.6 0.5 0.6

Table 14. List of genes that exhibited the highest ranking at the intermediate time point of 8 week as revealed by the MNI analysis in the subcutaneous adipose tissue of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Subcutaneous adipose tissue
Insulin resistance
NM_007501 Neurod4 Neurogenic differentiation 4 0.8 0.8 0.6 0.7
Inflammation
NM_027185 Def6 Differentially expressed in FDCP 6 1.9 2.0 3.0 1.7
Cancer
NM_008999 Rab23 RAB23, member RAS oncogene family 0.8 0.8 0.6 0.7
olfactory transduction
NM_147018 Olfr1056 Olfactory receptor 1056 0.8 0.9 0.7 0.8
NM_146279 Olfr960 Olfactory receptor 960 1.4 1.6 1.7 1.9
NM_001011857 Olfr685 Olfactory receptor 685 0.8 0.8 0.7 0.9
NM_146764 Olfr1408 Olfactory receptor 1408 0.6 0.6 0.7 0.7
Others
NM_153111 Fev FEV (ETS oncogene family) 0.7 0.5 0.4 0.6
NM_080644 Cacng5 Calcium channel, voltage-dependent, gamma subunit 5 0.6 0.6 0.4 0.5
NM_153592 Erlin2 ER lipid raft associated 2 1.3 1.3 1.6 1.3
NM_183036 Defb38 Defensin beta 38 1.6 2.1 2.8 2.0
NM_207022 Tas2r118 Taste receptor, type 2, member 118 2.0 1.6 2.9 2.1
XM_132900 LOC625758 Hypothetical LOC625758 2.0 2.4 2.6 1.5
NM_001008424 Cdsn Corneodesmosin 0.8 0.7 0.6 0.7
NM_025346 Rmnd5b Required for meiotic nuclear division 5 homolog B (S. cerevisiae) 1.6 1.9 2.8 2.0
NM_176846 Exph5 Exophilin 5 0.5 0.7 0.5 0.5
NM_146107 Actr1b ARP1 actin-related protein 1 homolog B (yeast) 0.5 0.6 0.3 0.6
XM_986681 Sytl2 Synaptotagmin-like 2 1.3 1.5 1.3 1.4
XM_618920 LOC544808 Hypothetical LOC544808 1.7 1.6 1.7 1.9
XM_110525 Cnksr1 Connector enhancer of kinase suppressor of Ras 1 1.3 1.5 1.4 1.3
NM_172263 Pde8b Phosphodiesterase 8B 0.8 0.7 0.6 0.8
NM_025638 Gdpd1 Glycerophosphodiester phosphodiesterase domain containing 1 0.3 0.4 0.5 0.4
NM_026520 2900009I07Rik RIKEN cDNA 2900009I07 gene 0.8 0.7 0.5 0.7

Table 15. List of genes that exhibited the highest ranking at the intermediate time point of 8 week as revealed by the MNI analysis in the muscle of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Muscle
Insulin resistance
NM_016961 Mapk9 Mitogen activated protein kinase 9 0.8 0.7 0.6 0.8
Cancer
NM_022032 Perp PERP, TP53 apoptosis effector 0.6 0.7 0.5 0.7
NM_026840 Pdgfrl Platelet-derived growth factor receptor-like 0.8 0.7 0.7 0.8
Olfactory transduction
NM_146686 Olfr232 Olfactory receptor 232 0.6 0.6 0.6 0.7
NM_146560 Olfr872 Olfactory receptor 872 1.3 1.3 1.3 1.2
Others
NM_027462 Wars2 Tryptophanyl tRNA synthetase 2 (mitochondrial) 0.7 0.8 0.6 0.7
XM_903020 Osbpl7 Oxysterol binding protein-like 7 1.2 1.4 1.6 1.4
XM_484317 EG432803 Predicted gene, EG432803 0.7 0.8 0.6 0.8
NM_001031772 Lin28 Lin-28 homolog B (C. elegans) 0.7 0.7 0.7 0.7
NM_001001602 Dab2ip Disabled homolog 2 (Drosophila) interacting protein 1.5 1.9 2.2 1.5
NM_145582 Atpbd3 ATP binding domain 3 0.7 0.6 0.6 0.6
AK014527 Cep192 Centrosomal protein 192 1.3 1.4 1.6 1.3
NM_028127 Frmd6 FERM domain containing 6 0.6 0.7 0.6 0.8
NM_172286 6430548M08Rik RIKEN cDNA 6430548M08 gene 1.2 1.2 1.2 1.1
NM_011105 Pkdrej Polycystic kidney disease (polycystin) and REJ (sperm receptor for egg jelly, sea urchin homolog)-like 1.5 1.9 2.0 2.2
XM_983096 LOC666331 Hypothetical protein LOC666331 1.2 1.3 1.6 1.4
NM_024196 Tbc1d2 TBC1 domain family, member 20 0.8 0.8 0.7 0.8
NM_198306 Galnt9 UDP-N-acetyl-alpha-D-galactosamine: polypeptide N-acetylgalactosaminyltransferase 9 0.8 0.6 1.0 0.7
AK014852 Glb1 Galactosidase, beta 1 like 3 1.4 1.4 1.4 1.5
NM_001004182 EG434008 Predicted gene, EG434008 0.8 0.7 0.7 0.7
NM_011284 Rpa2 Replication protein A2 1.6 1.5 1.9 1.4
NM_025381 Atp6v1f ATPase, H+ transporting, lysosomal V1 subunit F 1.4 1.3 1.3 1.4
NM_010571 Irs3 Insulin receptor substrate 3 0.6 0.7 0.6 0.8

Figure 7. Results of MNI analysis at week 12.

Figure 7

Genes that exhibited increasing ranking across time points as revealed by the MNI analysis, with a peak at 12 week in the peripheral tissues of mice. (A) Epididymal adipose tissue. (B) Subcutaneous adipose tissue. (C) Muscle.

Table 16. List of genes that exhibited increasing ranking across time points as revealed by the MNI analysis, with a peak at 12 week in the epididymal adipose tissue of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Epididymal adipose tissue
NM_175236 Adhfe1 Alcohol dehydrogenase, iron containing, 1 0.6 0.6 0.7 0.6
AF015260 Smad7 MAD homolog 7 (Drosophila) 0.7 0.6 0.7 0.5
Inflammation
NM_008035 Folr2 Folate receptor 2 (fetal) 0.7 0.7 0.7 0.6
NM_008802 Pde7a Phosphodiesterase 7A 2.1 1.9 2.2 2.7
NM_009511 Vipr2 Vasoactive intestinal peptide receptor 2 2.2 2.0 2.0 1.8
Cancer
NM_008586 Mep1b Meprin 1 beta 0.5 0.5 0.5 0.3
NM_181321 Tmc6 Transmembrane channel-like gene family 6 1.8 1.8 1.7 2.3
Olfactory transduction
NM_008763 Olfr16 Olfactory receptor 16 1.4 2.5 1.2 2.3
NM_001011695 Olfr1000 Olfactory receptor 1000 2.2 2.2 2.5 3.0
Others
NM_139270 Pthr2 Parathyroid hormone receptor 2 2.2 3.1 2.8 2.4
NM_172434 Tnrc4 Trinucleotide repeat containing 4 1.3 1.5 1.4 1.5
NM_173402 Rgs12 Regulator of G-protein signaling 12 0.7 0.7 0.7 0.5
XM_977897 B230217C12Rik RIKEN cDNA B230217C12 gene 1.8 1.4 1.8 1.8
EG433070 EG433070 Predicted gene, EG433070 1.7 1.8 2.0 2.5
NM_175259 Tmem58 Transmembrane protein 58 1.5 1.3 1.5 1.6
NM_026412 D2Ertd750e DNA segment, Chr 2, ERATO Doi 750, expressed 0.7 0.6 0.7 0.6
NM_181323 C130090K23Rik RIKEN cDNA C130090K23 gene 1.5 1.4 1.8 1.5
NM_175406 Atp6v0d2 ATPase, H+ transporting, lysosomal V0 subunit D2 1.4 1.4 1.5 1.3
NM_007725 Cnn2 Calponin 2 0.6 0.6 0.6 0.5
NM_001033274 Brd1 Bromodomain containing 1 1.7 2.2 2.2 2.3

Table 17. List of genes that exhibited increasing ranking across time points as revealed by the MNI analysis, with a peak at 12 week in the subcutaneous adipose tissue of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Subcutaneous adipose tissue
Inflammation
NM_001024852 Auts2 Autism susceptibility candidate 2 0.8 0.9 0.8 0.5
NM_001033536 Rfxdc2 Regulatory factor X domain containing 2 homolog (human) 1.2 1.4 1.3 1.4
NM_009701 Aqp5 Aquaporin 5 0.7 0.7 0.8 0.6
XM_285901 Gm792 Gene model 792, (NCBI) 0.7 0.6 0.5 0.4
NM_019775 Cpb2 Carboxypeptidase B2 (plasma) 1.7 1.6 1.5 2.5
Cancer
NM_009050 Ret Ret proto-oncogene 1.2 1.3 1.2 1.5
Olfactory transduction
NM_146604 Olfr716 Olfactory receptor 716 1.9 1.7 2.0 3.1
NM_146963 Olfr45 Olfactory receptor 45 1.6 1.5 1.9 2.2
Others
NM_178046 Svil Supervillin 0.7 0.8 0.7 0.6
NM_018764 Pcdh7 Protocadherin 7 1.5 1.4 1.3 1.8
NM_030061 Spink12 Serine peptidase inhibitor, Kazal type 12 1.4 1.4 1.3 1.7
NM_178741 Klhl8 Kelch-like 8 (Drosophila) 1.3 1.3 1.3 1.6
NM_175696 C530028O21Rik RIKEN cDNA C530028O21 gene 1.3 1.3 1.2 1.5
NM_177711 4932411G14Rik RIKEN cDNA 4932411G14 gene 0.7 0.6 0.7 0.5
NM_181579 Pof1b Premature ovarian failure 1B 1.7 1.8 1.6 2.2
XM_489612 4921511H13Rik RIKEN cDNA 4921511H13 gene 1.3 1.4 1.2 1.6
NM_172456 Endogl1 Endonuclease G-like 1 1.6 1.9 1.4 2.2
NM_001033488 Gm1964 Gene model 1964, (NCBI) 1.5 1.4 1.5 1.5
NM_181318 Rasgef1b RasGEF domain family, member 1B 0.7 0.8 0.8 0.6
NM_183017 Ttll12 Tubulin tyrosine ligase-like family, member 12 0.8 0.6 0.7 0.7
NM_010075 Dpp6 Dipeptidylpeptidase 6 1.1 1.2 1.2 1.3
NM_026175 Sf3a1 Splicing factor 3a, subunit 1 1.5 1.4 1.7 1.9

Table 18. List of genes that exhibited increasing ranking across time points as revealed by the MNI analysis, with a peak at 12 week in the muscle of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Muscle
Insulin resistance
NM_145435 Pyy Peptide YY 1.4 1.4 1.3 1.5
Inflammation
NM_138648 Olr1 Oxidized low density lipoprotein (lectin-like) receptor 1 0.5 0.6 0.5 0.3
NM_009132 Scin Scinderin 0.6 0.6 0.7 0.7
Cancer
NM_145833 Lin28 Lin-28 homolog (C. elegans) 0.7 0.7 0.7 0.7
NM_010358 Gstm1 Glutathione S-transferase, mu 1 1.3 1.4 1.4 1.3
NM_001029979 Safb2 Scaffold attachment factor B2 0.7 0.7 0.7 0.6
Olfactory transduction
NM_146750 Olfr689 Olfactory receptor 689 0.6 0.7 0.7 0.5
NM_146943 Olfr347 Olfactory receptor 347 1.2 1.3 1.2 1.5
Others
NM_009372 Tgif1 TG interacting factor 1 1.2 1.1 0.8 0.6
XM_887155 Igsf10 Immunoglobulin superfamily, member 10 0.7 0.5 0.6 0.4
NM_009418 Tpp2 Tripeptidyl peptidase II 1.2 1.4 1.2 1.5
NM_008118 Gif Gastric intrinsic factor 0.8 0.7 0.7 0.8
NM_026972 Cd209b CD209b antigen 0.9 0.8 0.6 0.7
NM_145358 Camkk2 Calcium/calmodulin-dependent protein kinase kinase 2, beta 1.2 1.2 1.3 1.2
BC051526 Cd22 CD226 antigen 0.8 0.9 0.9 0.7
NM_029946 4931407K02Rik RIKEN cDNA 4931407K02 gene 1.3 1.1 1.2 1.3
NM_172652 4632411B12Rik RIKEN cDNA 4632411B12 gene 1.7 1.4 1.4 1.9
NM_011598 Fabp9 Fatty acid binding protein 9, testis 0.8 0.8 0.9 0.7
NM_011023 Otx1 Orthodenticle homolog 1 (Drosophila) 0.7 0.7 0.7 0.5
NM_019579 Mpp5 Membrane protein, palmitoylated 5 0.5 0.5 0.6 0.3
NM_007555 Bmp5 Bone morphogenetic protein 5 0.9 0.9 0.8 0.8
NM_001039047 Trim58 Tripartite motif-containing 58 1.3 1.2 1.5 1.5
NM_020049 Slc6a14 Solute carrier family 6 (neurotransmitter transporter), member 14 0.8 0.6 0.7 0.6

Figure 8. Results of MNI analysis throughout the time course.

Figure 8

Genes that exhibited a constant high MNI ranking throughout the time course as revealed by the MNI analysis in the peripheral tissues of mice. (A) Epididymal adipose tissue. (B) Subcutaneous adipose tissue. (C) Muscle.

Table 19. List of genes that exhibited a constant high MNI ranking throughout the time course as revealed by the MNI analysis in the peripheral tissues of mice.

Gene accession No. Gene symbol Description Fold change (HFD/ND)
2wk 4wk 8wk 12wk
Epididymal adipose tissue
Insulin resistance
NM_001039138 Camk2g Calcium/calmodulin-dependent protein kinase II gamma 0.6 0.5 0.6 0.5
Others
NM_145463 Tmem46 Transmembrane protein 46 0.6 0.6 0.7 0.6
Subcutaneous adipose tissue
olfactory transduction
NM_207566 Olfr1173 Olfactory receptor 1173 2.0 2.3 1.9 2.7
NM_146709 Olfr411 Olfactory receptor 411 1.5 1.4 1.4 1.6
NM_146524 Olfr855 Olfactory receptor 855 1.7 1.6 2.0 1.6
Others
NM_178110 Trim62 Tripartite motif-containing 62 1.6 1.8 1.6 1.8
Muscle
Cancer
NM_024124 Hdac9 Histone deacetylase 9 0.6 0.5 0.6 0.6
Others
NM_177861 Tmem67 Transmembrane protein 67 0.7 0.6 0.7 0.5

Discussion

Animals use their olfactory system to monitor the chemical environment for molecules that reveal food sources or toxic substances and signal the presence of predators [11]. There are numerous olfactory receptors of different types, with as many as 1,000 in the mammalian genome that represent approximately 3% of the human genomeentire genetic information [12]. The information related to odor gathered by these olfactory receptors is funneled through a common signaling pathway. When an olfactory receptor binds to its odorant, it activates a single species of G protein, the olfactory trimeric G protein (Golf), which in turn activates the olfactory isoform of adenylate cyclase (AC3) [12]. Converging evidence has demonstrated that the olfactory system is a target for hormones related to metabolism and food-intake regulation; it adapts its function to nutritional needs by promoting or inhibiting food foraging [13]. Recent studies have found that obese patients display decreased olfactory acuity [14] and are significantly more likely to have absolute olfactory dysfunction or anosmia [15]. Furthermore, Simchen et al. showed that the abilities to detect and identify odors have been found to decrease as body mass index (BMI) increases in subjects less than 65 years old, independent of any linkage to food odor or gender [16]. Recently, the elements of olfactory-like chemosensory signaling have been found to also present in nonolfactory tissues such as testis [17], brain [18], and heart [19]. To our knowledge, this is the first study that shows a differential mRNA expression and high MNI ranking of olfactory receptors in the epididymal and subcutaneous fat tissues and muscles between ND and HFD-fed mice (Tables 1, 2, 3). These results imply that the olfactory receptors and the molecules involved in olfactory transduction might be the mediators of HFD-induced obesity progression in the peripheral tissues. This hypothesis is supported by the fact that the increased cAMP production by AC3 activates cAMP responsive element binding (CREB) protein, leading to increased adipogenesis in an obese mouse model. Furthermore, mice lacking AC3, which is a downstream regulator of olfactory receptors, exhibit obesity that is apparently caused by low locomotor activity, hyperphagia, and leptin insensitivity [20]. In future studies, it will be intriguing to further investigate the role of individual olfactory receptors in peripheral tissues, such as the pancreas, liver, muscle, and fat, to better understand the activation process of these signaling pathways and their physiological roles.

Cancer-related genes such as Nek11, A4gnt, Srp9, Gli2, Gucy2c, Lsm1, Duoxa1, Lasp1, Ret, Bex2, Vav3, Kcnrg, Tle6, Rab23, Dcc, Rassf2, Perp, Pdgfr1, Lin28, Gstm1, Safb2, Tmem46, and Hdac9 were remarkably overrepresented in time-course clusters identified by the MNI analysis in the epididymal and subcutaneous fat tissues and gastrocnemius muscle of mice with diet-induced obesity (Figs. 4, 5, 6, 7, 8). Of these 23 genes, 6 are known breast cancer-related genes (Lsm1, Duoxa1, Ret, Bex2, Rassf2, and Safb2). Lsm1 is a transforming oncogene that is amplified and overexpressed in breast cancer [21] and might affect either cell cycle progression or apoptosis [22]. Duoxa1, which was originally identified as a numb-interacting protein, was recently shown to function as a maturation factor in breast cancer [23]. Ret exhibits both estrogen- and retinoic acid-dependent transcriptional modulation in breast cancer [24]. Bex2 has a significant role in promoting cell survival and growth in breast cancer cells [25], [26], and Rassf2 might function as a tumor suppressor gene in in vitro cell migration and cell cycle progression [27]. The expression of Safb2 protein, which functions as estrogen receptor co-repressor and growth inhibitor, was lost in approximately 20% of breast cancers [28]. Many studies have attempted to determine the relationship between diet and breast cancer. Dietary fat is a source of endogenous estrogen and has been suggested as a possible risk factor for breast cancer [29]. To our knowledge, this is the first study showing an association between these 6 genes involved in breast cancer development and HFD-induced obesity in a rodent model.

Colon cancer-related genes such as Nek11, Gucy2c, Srp9, Tle6, and Pdgfrl were also overrepresented in the time-course clusters identified by the MNI analysis in the epididymal and subcutaneous fat tissues and gastrocnemius muscle of mice with diet-induced obesity (Figs. 4, 5, 6, 7, 8). Nek11, a member of the NIMA-related kinase family, phosphorylates Cdc25a and controls its degradation; Cdc25a phosphorylation is required for cell cycle progression in colorectal cancer cells [30]. Gucy2c and Srp9 have been shown to be overexpressed in colorectal cancer cells and were recently shown to function as a candidate biomarker for colon cancer [31], [32]. Tle6 is recurrently overexpressed in human colon cancer and enhances cell proliferation, colony formation, migration, and xenograft tumorigenicity [33]. Pdgfrl acts as a tumor suppressor and inhibits the growth of colorectal cancer cells [34]. Epidemiological studies indicate that both high body weight and high body mass index (BMI) were significantly associated with an increased colon cancer risk. Intra-abdominal visceral obesity, high plasma glucose levels, HbA1C, and C-peptide were also found to be associated with increased risk of colorectal cancer [35][38]. The current study showed that the above-mentioned genes that are involved in the regulation of colon cancer might play a genetic role in the development of obesity. No mechanistic insights have been reported to explain the relationship between the regulation of cancer-related genes in the adipose tissue or muscle and cancer susceptibility. It could be probable that the changes in the expression of cancer-related genes in the adipose tissue may accompany the regulation of same genes in epithelial tissues such as breast or colon.

Genes that were found to have the highest rank at the early phase and return to baseline after several weeks might be considered genetic mediators of acute-phase response in metabolic processes related to HFD-induced obesity. Dusp12 was one of the 58 genes that were observed to have decreasing ranking during the development of obesity, with a peak at 2 week. Previous studies identified several single nucleotide polymorphisms in this gene associated with type 2 diabetes in different populations, including Caucasians and Chinese [39]. Dusp12 is a glucokinase-associated protein that participates in glycolysis in the liver and dephosphorylation of cytoplasmic glucokinase in the pancreatic beta cells [40]. Therefore, Dusp12 might play a role in the regulation of glycolysis during the early stages of obesity. When glycolysis was decreased, whole-body glucose disposal was also reduced, indicating a decrease in glucose utilization in the peripheral tissues in response to the HFD. The latter likely results from an impaired glucose transport that precedes impaired insulin signaling.

Def6 and Mapk9 were one of the 145 genes that were found to have the highest rank at the intermediate time points of 4 or 8 weeks during the development of obesity. Def6, a novel type of activator for Rho GTPase, is expressed in myeloid cells, and disruption of Def6 expression leads to defects in toll-like receptor 4 (TLR4) signaling and innate immune responses [41]. Rho GTPases have been shown to be recruited to the cytosolic domain of TLR and the closely related interleukin 1 receptor (IL-1R) and to regulate the production of proinflammatory cytokines [42], [43]. In the present study, the high MNI ranking of Def6 in the subcutaneous adipose tissue of HFD-fed mice suggested that it might participate in the regulation of obesity-induced inflammation through TLR4 signaling. Mapk9, which is ubiquitously expressed, can invoke transcription factors such as c-Jun and many other apoptosis-related proteins [44]. Interestingly, recent studies have shown that the knockdown of Mapk9 leads to reduced serum levels of glucose, insulin, and homeostatic model assessment and therefore reverses insulin resistance in HFD-fed mice [45]. These findings provide supporting evidences to the high MNI ranking of Mapk9 associated with HFD-induced obesity observed in the present study. However, further studies are required to elucidate the precise function of Mapk9 in the development of HFD-induced type 2 diabetes.

Smad7, Adhfe1, and Pyy are one of the 65 genes that showed increasing ranking during the development of obesity, with a peak at 12 weeks. Smad7 was initially characterized as a factor induced by shear stress in vascular endothelial cells [46]. Only recently, new functions of Smad7 were elucidated: it inhibits transforming growth factor-β (TGF-β)-activated responses [46]. TGF-β is known to inhibit adipose differentiation of preadipocyte cell lines and primary cultures [47] and to block adipogenesis in vivo [48]. This suggests that Smad7 enhances adipogenesis through the inhibition of TGF-β signaling. Adhfe1 was characterized as a hydroxyacid-oxoacid transhydrogenase that catalyzes the conversion of γ-hydroxybutyrate to succinic semialdehyde [49]. Recently, Adhfe1 was suggested to play a role in adipocyte differentiation. The expression of Adhfe1 transcript is tightly linked to the phenotype of mature adipocytes both in vivo and in vitro, although the mechanisms underlying Adhfe1-mediated regulation of adipogenesis remain poorly understood [50]. Pyy, which is expressed and secreted in endocrine intestinal cells, plays a role in reducing appetite and caloric intake [51]. Recently, plasma Pyy concentrations were found to be decreased in both obese humans [52] and diet-induced obese mice [53]. These studies might suggest that Smad7 and Adhfe1 play a role in obesity by amplifying the aggressive effect of adipogenesis.

Camk2g and Tmem67 are one of the 8 genes that exhibited a constant high MNI ranking from 2 to 12 weeks. The increase of cytosolic Ca2+ in the beta cells is central to the initiation of insulin secretion under physiological conditions [54]. Recent findings suggest that Camk2g involved in the regulation of calcium in the islet beta cells is a candidate gene for type 2 diabetes [55]. The Tmem67 gene mediates a fundamental developmental stage of ciliary formation and epithelial morphogenesis [56]. In addition, defects in the Tmem67 gene resulted in Meckel syndrome type 3, Joubert syndrome type 6, and nephronophthisis 11, which show many clinical phenotypic similarities, including hepatic fibrosis [56], [57]. Consumption of fat-rich diets seems to play an important role in the pathogenesis of hepatic steatosis and its progression to fibrosis [58]. The constantly high MNI ranking of Tmem67 from 2 to 12 weeks associated with a HFD suggests that Tmem67 might participate in the development of hepatic fibrosis.

In summary, this study is the most comprehensive investigation of the gene expression patterns conducted using a time-resolved approach to gain insight into the development of HFD-induced obesity in a mouse model. A reverse-engineered gene network was used for the first time for the identification of key genetic mediators and pathways that have been implicated in the initiation and advancement of obesity. We highlighted the sequential induction of distinct olfactory receptors and stimulation of cancer-related genes during the development of obesity. To our knowledge, the proposed changes in the olfactory transduction machinery as per the MNI ranking have not been previously reported. These putative mechanisms clearly need further investigation. The top 5 genes recognized through the MNI analysis at each time points (2, 4, 8, and 12 weeks) and gene clusters identified based on their temporal patterns in the 3 different tissues (visceral and subcutaneous adipose tissues and muscle) of mice need special attention as potential genetic mediators for obesity progression.

Supporting Information

Figure S1

The basal expression levels of some target genes identified by MNI analysis. Quantitative real-time PCR analysis of the basal expression on highly ranked olfactory genes and top 5 genes at week 4 in the epididymal adipose tissues of (A) ND- or (B) HFD-fed mice. Results are presented as the average ± SEM of at least 3 separate experiments.

(TIF)

Funding Statement

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MEST) (No. 2012-0000643). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1. Jain N, Thatte J, Braciale T, Ley K, O'Connell M, et al. (2003) Local-pooled-error test for identifying differentially expressed genes with a small number of replicated microarrays. Bioinformatics 19: 1945–1951. [DOI] [PubMed] [Google Scholar]
  • 2. Storey JD, Tibshirani R (2003) Statistical methods for identifying differentially expressed genes in DNA microarrays. Methods in molecular biology 224: 149–157. [DOI] [PubMed] [Google Scholar]
  • 3. Eisen MB, Spellman PT, Brown PO, Botstein D (1998) Cluster analysis and display of genome-wide expression patterns. Proceedings of the National Academy of Sciences of the United States of America 95: 14863–14868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. di Bernardo D, Thompson MJ, Gardner TS, Chobot SE, Eastwood EL, et al. (2005) Chemogenomic profiling on a genome-wide scale using reverse-engineered gene networks. Nature biotechnology 23: 377–383. [DOI] [PubMed] [Google Scholar]
  • 5. Kerr MK, Martin M, Churchill GA (2000) Analysis of variance for gene expression microarray data. Journal of computational biology: a journal of computational molecular cell biology 7: 819–837. [DOI] [PubMed] [Google Scholar]
  • 6. Cobb JP, Mindrinos MN, Miller-Graziano C, Calvano SE, Baker HV, et al. (2005) Application of genome-wide expression analysis to human health and disease. Proceedings of the National Academy of Sciences of the United States of America 102: 4801–4806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Hsu KL, Pilobello KT, Mahal LK (2006) Analyzing the dynamic bacterial glycome with a lectin microarray approach. Nature chemical biology 2: 153–157. [DOI] [PubMed] [Google Scholar]
  • 8. McAdams HH, Shapiro L (2003) A bacterial cell-cycle regulatory network operating in time and space. Science 301: 1874–1877. [DOI] [PubMed] [Google Scholar]
  • 9. Soukas A, Cohen P, Socci ND, Friedman JM (2000) Leptin-specific patterns of gene expression in white adipose tissue. Genes & development 14: 963–980. [PMC free article] [PubMed] [Google Scholar]
  • 10. Xing H, Gardner TS (2006) The mode-of-action by network identification (MNI) algorithm: a network biology approach for molecular target identification. Nature protocols 1: 2551–2554. [DOI] [PubMed] [Google Scholar]
  • 11. Zhang X, Rogers M, Tian H, Zou DJ, Liu J, et al. (2004) High-throughput microarray detection of olfactory receptor gene expression in the mouse. Proceedings of the National Academy of Sciences of the United States of America 101: 14168–14173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Gilad Y, Lancet D (2003) Population differences in the human functional olfactory repertoire. Molecular biology and evolution 20: 307–314. [DOI] [PubMed] [Google Scholar]
  • 13.Palouzier-Paulignan B, Lacroix MC, Aime P, Baly C, Caillol M, et al.. (2012) Olfaction Under Metabolic Influences. Chem Senses. [DOI] [PMC free article] [PubMed]
  • 14. Richardson BE, Vander Woude EA, Sudan R, Thompson JS, Leopold DA (2004) Altered olfactory acuity in the morbidly obese. Obes Surg 14: 967–969. [DOI] [PubMed] [Google Scholar]
  • 15. Richardson BE, Vanderwoude EA, Sudan R, Leopold DA, Thompson JS (2012) Gastric bypass does not influence olfactory function in obese patients. Obes Surg 22: 283–286. [DOI] [PubMed] [Google Scholar]
  • 16. Simchen U, Koebnick C, Hoyer S, Issanchou S, Zunft HJ (2006) Odour and taste sensitivity is associated with body weight and extent of misreporting of body weight. Eur J Clin Nutr 60: 698–705. [DOI] [PubMed] [Google Scholar]
  • 17. Parmentier M, Libert F, Schurmans S, Schiffmann S, Lefort A, et al. (1992) Expression of members of the putative olfactory receptor gene family in mammalian germ cells. Nature 355: 453–455. [DOI] [PubMed] [Google Scholar]
  • 18. Mombaerts P (1999) Molecular biology of odorant receptors in vertebrates. Annual review of neuroscience 22: 487–509. [DOI] [PubMed] [Google Scholar]
  • 19. Young JM, Trask BJ (2002) The sense of smell: genomics of vertebrate odorant receptors. Human molecular genetics 11: 1153–1160. [DOI] [PubMed] [Google Scholar]
  • 20. Wang Z, Li V, Chan GC, Phan T, Nudelman AS, et al. (2009) Adult type 3 adenylyl cyclase-deficient mice are obese. PloS one 4: e6979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Kelley JR, Fraser MM, Hubbard JM, Watson DK, Cole DJ (2003) CaSm antisense gene therapy: a novel approach for the treatment of pancreatic cancer. Anticancer research 23: 2007–2013. [PubMed] [Google Scholar]
  • 22. Fraser MM, Watson PM, Fraig MM, Kelley JR, Nelson PS, et al. (2005) CaSm-mediated cellular transformation is associated with altered gene expression and messenger RNA stability. Cancer research 65: 6228–6236. [DOI] [PubMed] [Google Scholar]
  • 23. Ostrakhovitch EA, Li SS (2010) NIP1/DUOXA1 expression in epithelial breast cancer cells: regulation of cell adhesion and actin dynamics. Breast cancer research and treatment 119: 773–786. [DOI] [PubMed] [Google Scholar]
  • 24. Ahmad N, Kumar R (2011) Steroid hormone receptors in cancer development: a target for cancer therapeutics. Cancer letters 300: 1–9. [DOI] [PubMed] [Google Scholar]
  • 25. Naderi A, Teschendorff AE, Beigel J, Cariati M, Ellis IO, et al. (2007) BEX2 is overexpressed in a subset of primary breast cancers and mediates nerve growth factor/nuclear factor-kappaB inhibition of apoptosis in breast cancer cell lines. Cancer research 67: 6725–6736. [DOI] [PubMed] [Google Scholar]
  • 26. Naderi A, Liu J, Bennett IC (2010) BEX2 regulates mitochondrial apoptosis and G1 cell cycle in breast cancer. International journal of cancer Journal international du cancer 126: 1596–1610. [DOI] [PubMed] [Google Scholar]
  • 27. Vos MD, Ellis CA, Elam C, Ulku AS, Taylor BJ, et al. (2003) RASSF2 is a novel K-Ras-specific effector and potential tumor suppressor. The Journal of biological chemistry 278: 28045–28051. [DOI] [PubMed] [Google Scholar]
  • 28. Oesterreich S (2003) Scaffold attachment factors SAFB1 and SAFB2: Innocent bystanders or critical players in breast tumorigenesis? Journal of cellular biochemistry 90: 653–661. [DOI] [PubMed] [Google Scholar]
  • 29. Ruggeri BA, Klurfeld DM, Kritchevsky D, Furlanetto RW (1989) Caloric restriction and 7,12-dimethylbenz(a) anthracene-induced mammary tumor growth in rats: alterations in circulating insulin, insulin-like growth factors I and II, and epidermal growth factor. Cancer research 49: 4130–4134. [PubMed] [Google Scholar]
  • 30. Sorensen CS, Melixetian M, Klein DK, Helin K (2010) NEK11: linking CHK1 and CDC25A in DNA damage checkpoint signaling. Cell cycle 9: 450–455. [DOI] [PubMed] [Google Scholar]
  • 31. Mejia A, Schulz S, Hyslop T, Weinberg DS, Waldman SA (2009) GUCY2C reverse transcriptase PCR to stage pN0 colorectal cancer patients. Expert review of molecular diagnostics 9: 777–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Rho JH, Qin S, Wang JY, Roehrl MH (2008) Proteomic expression analysis of surgical human colorectal cancer tissues: up-regulation of PSB7, PRDX1, and SRP9 and hypoxic adaptation in cancer. Journal of proteome research 7: 2959–2972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Chen PC, Kuraguchi M, Velasquez J, Wang Y, Yang K, et al. (2008) Novel roles for MLH3 deficiency and TLE6-like amplification in DNA mismatch repair-deficient gastrointestinal tumorigenesis and progression. PLoS genetics 4: e1000092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Guo FJ, Zhang WJ, Li YL, Liu Y, Li YH, et al. (2010) Expression and functional characterization of platelet-derived growth factor receptor-like gene. World journal of gastroenterology: WJG 16: 1465–1472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Ahmed RL, Schmitz KH, Anderson KE, Rosamond WD, Folsom AR (2006) The metabolic syndrome and risk of incident colorectal cancer. Cancer 107: 28–36. [DOI] [PubMed] [Google Scholar]
  • 36. Pischon T, Lahmann PH, Boeing H, Friedenreich C, Norat T, et al. (2006) Body size and risk of colon and rectal cancer in the European Prospective Investigation Into Cancer and Nutrition (EPIC). Journal of the National Cancer Institute 98: 920–931. [DOI] [PubMed] [Google Scholar]
  • 37. Saydah SH, Platz EA, Rifai N, Pollak MN, Brancati FL, et al. (2003) Association of markers of insulin and glucose control with subsequent colorectal cancer risk. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology 12: 412–418. [PubMed] [Google Scholar]
  • 38. Ma J, Giovannucci E, Pollak M, Leavitt A, Tao Y, et al. (2004) A prospective study of plasma C-peptide and colorectal cancer risk in men. Journal of the National Cancer Institute 96: 546–553. [DOI] [PubMed] [Google Scholar]
  • 39. Hu C, Zhang R, Wang C, Ma X, Wang J, et al. (2011) Lack of association between genetic polymorphisms within DUSP12 – ATF6 locus and glucose metabolism related traits in a Chinese population. BMC medical genetics 12: 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Munoz-Alonso MJ, Guillemain G, Kassis N, Girard J, Burnol AF, et al. (2000) A novel cytosolic dual specificity phosphatase, interacting with glucokinase, increases glucose phosphorylation rate. The Journal of biological chemistry 275: 32406–32412. [DOI] [PubMed] [Google Scholar]
  • 41. Chen Q, Gupta S, Pernis AB (2009) Regulation of TLR4-mediated signaling by IBP/Def6, a novel activator of Rho GTPases. Journal of leukocyte biology 85: 539–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Arbibe L, Mira JP, Teusch N, Kline L, Guha M, et al. (2000) Toll-like receptor 2-mediated NF-kappa B activation requires a Rac1-dependent pathway. Nature immunology 1: 533–540. [DOI] [PubMed] [Google Scholar]
  • 43. Chen LY, Zuraw BL, Liu FT, Huang S, Pan ZK (2002) IL-1 receptor-associated kinase and low molecular weight GTPase RhoA signal molecules are required for bacterial lipopolysaccharide-induced cytokine gene transcription. Journal of immunology 169: 3934–3939. [DOI] [PubMed] [Google Scholar]
  • 44. Jaeschke A, Rincon M, Doran B, Reilly J, Neuberg D, et al. (2005) Disruption of the Jnk2 (Mapk9) gene reduces destructive insulitis and diabetes in a mouse model of type I diabetes. Proceedings of the National Academy of Sciences of the United States of America 102: 6931–6935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Singh R, Wang Y, Xiang Y, Tanaka KE, Gaarde WA, et al. (2009) Differential effects of JNK1 and JNK2 inhibition on murine steatohepatitis and insulin resistance. Hepatology 49: 87–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Topper JN, Cai J, Qiu Y, Anderson KR, Xu YY, et al. (1997) Vascular MADs: two novel MAD-related genes selectively inducible by flow in human vascular endothelium. Proceedings of the National Academy of Sciences of the United States of America 94: 9314–9319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Ignotz RA, Massague J (1985) Type beta transforming growth factor controls the adipogenic differentiation of 3T3 fibroblasts. Proceedings of the National Academy of Sciences of the United States of America 82: 8530–8534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Clouthier DE, Comerford SA, Hammer RE (1997) Hepatic fibrosis, glomerulosclerosis, and a lipodystrophy-like syndrome in PEPCK-TGF-beta1 transgenic mice. The Journal of clinical investigation 100: 2697–2713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Lyon RC, Johnston SM, Panopoulos A, Alzeer S, McGarvie G, et al. (2009) Enzymes involved in the metabolism of gamma-hydroxybutyrate in SH-SY5Y cells: identification of an iron-dependent alcohol dehydrogenase ADHFe1. Chemico-biological interactions 178: 283–287. [DOI] [PubMed] [Google Scholar]
  • 50. Kim JY, Tillison KS, Zhou S, Lee JH, Smas CM (2007) Differentiation-dependent expression of Adhfe1 in adipogenesis. Archives of biochemistry and biophysics 464: 100–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Xu J, McNearney TA, Chen JD (2011) Impaired postprandial releases/syntheses of ghrelin and PYY (3–36) and blunted responses to exogenous ghrelin and PYY (3–36) in a rodent model of diet-induced obesity. Journal of gastroenterology and hepatology 26: 700–705. [DOI] [PubMed] [Google Scholar]
  • 52. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, et al. (2003) Inhibition of food intake in obese subjects by peptide YY3-36. The New England journal of medicine 349: 941–948. [DOI] [PubMed] [Google Scholar]
  • 53. le Roux CW, Batterham RL, Aylwin SJ, Patterson M, Borg CM, et al. (2006) Attenuated peptide YY release in obese subjects is associated with reduced satiety. Endocrinology 147: 3–8. [DOI] [PubMed] [Google Scholar]
  • 54. Gloyn AL, Desai M, Clark A, Levy JC, Holman RR, et al. (2002) Human calcium/calmodulin-dependent protein kinase II gamma gene (CAMK2G): cloning, genomic structure and detection of variants in subjects with type II diabetes. Diabetologia 45: 580–583. [DOI] [PubMed] [Google Scholar]
  • 55. Easom RA (1999) CaM kinase II: a protein kinase with extraordinary talents germane to insulin exocytosis. Diabetes 48: 675–684. [DOI] [PubMed] [Google Scholar]
  • 56. Dawe HR, Smith UM, Cullinane AR, Gerrelli D, Cox P, et al. (2007) The Meckel-Gruber Syndrome proteins MKS1 and meckelin interact and are required for primary cilium formation. Human molecular genetics 16: 173–186. [DOI] [PubMed] [Google Scholar]
  • 57. Seeman T, Seemanova E, Nuernberg G, Nuernberg P, Janssen S, et al. (2010) Polycystic kidney and hepatic disease with mental retardation is nephronophthisis 11 caused by MKS3/TMEM67 mutations. Pediatric nephrology 25: 2375–2376. [DOI] [PubMed] [Google Scholar]
  • 58. Barbuio R, Milanski M, Bertolo MB, Saad MJ, Velloso LA (2007) Infliximab reverses steatosis and improves insulin signal transduction in liver of rats fed a high-fat diet. The Journal of endocrinology 194: 539–550. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Figure S1

The basal expression levels of some target genes identified by MNI analysis. Quantitative real-time PCR analysis of the basal expression on highly ranked olfactory genes and top 5 genes at week 4 in the epididymal adipose tissues of (A) ND- or (B) HFD-fed mice. Results are presented as the average ± SEM of at least 3 separate experiments.

(TIF)


Articles from PLoS ONE are provided here courtesy of PLOS

RESOURCES