Next Article in Journal
β-Carotene Status Is Associated with Inflammation and Two Components of Metabolic Syndrome in Patients with and without Osteoarthritis
Previous Article in Journal
Changes in HMO Concentrations throughout Lactation: Influencing Factors, Health Effects and Opportunities
Previous Article in Special Issue
Fasting: How to Guide
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Comparison of Gene Expression Profiles of Rat Tissues after Mild and Short-Term Calorie Restrictions

1
Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
2
Biomedical Gerontology Laboratory, Faculty of Human Sciences, Waseda University, 2-579-1 Mikajima, Tokorozawa, Saitama 359-1164, Japan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2021, 13(7), 2277; https://doi.org/10.3390/nu13072277
Submission received: 30 April 2021 / Revised: 22 June 2021 / Accepted: 24 June 2021 / Published: 30 June 2021

Abstract

:
Many studies have shown the beneficial effects of calorie restriction (CR) on rodents’ aging; however, the molecular mechanism explaining these beneficial effects is still not fully understood. Previously, we conducted transcriptomic analysis on rat liver with short-term and mild-to-moderate CR to elucidate its early response to such diet. Here, we expanded transcriptome analysis to muscle, adipose tissue, intestine, and brain and compared the gene expression profiles of these multiple organs and of our previous dataset. Several altered gene expressions were found, some of which known to be related to CR. Notably, the commonly regulated genes by CR include nicotinamide phosphoribosyltransferase and heat shock protein 90, which are involved in declining the aging process and thus potential therapeutic targets for aging-related diseases. The data obtained here provide information on early response markers and key mediators of the CR-induced delay in aging as well as on age-associated pathological changes in mammals.

1. Introduction

Many studies have shown that calorie restriction (CR) has beneficial health effects on rodents and human [1,2]. Several molecular mechanisms, including the oxidative stress stimulation and nutrient-sensing pathways, have been proposed to explain the role of CR for improving health and extending life [3]. Recent advances in science and technology have allowed omics-based (e.g., transcriptomics, proteomics, metabolomics) deep investigation of the molecular mechanism of CR [4,5]. However, it is still unclear how CR delays the aging process and the surge of age-related diseases.
The liver, muscle, adipose tissue, intestine, and brain are main organs with pivotal roles in the metabolism and absorption of food and nutrients. These organs, which are known to communicate with each other [6,7], are significant for the regulation of homeostasis by nutrient sensing. In addition, all organs and tissues are affected by CR, a major protective factor for most age-related diseases in humans. Therefore, investigating the commonalities and differences among multiple organs in response to CR is an important insight that can help disclosing the molecular mechanisms underlying the effects of CR.
In general, any experiment related to CR is performed in a longevity and aging study that requires a long-term observation. Reported animal studies were conducted under relatively long-term and severe CR conditions such as 30% dietary restriction for several months, which is difficult to achieve in humans. However, transcriptomic alterations usually occurs much earlier than phenotypic alterations thereby enabling us to find early response markers of CR.
Previously, we examined the liver transcriptome of rats with short-term (one week and one month) and mild-to-moderate (5%, 10%, 20%, and 30%) CR [8]. In the present study we expanded transcriptome analysis to other target organs, such as muscle, adipose tissue, intestine, and brain, and compared the gene expression profiles of these organs among them and to our previous dataset to find early response markers and key mediators of the CR-induced beneficial effects related to age.

2. Materials and Methods

2.1. Animal Experiment

The methods used in the animal experiment have been described previously [8]. Male Wistar rats purchased at 5 weeks of age from Japan SLC (Tokyo, Japan) were kept individually and fed the American Institute of Nutrition-93G powdered diet ad libitum for 1 week or 1 month. The daily intakes of these two groups were recorded, and then 100%, 95%, 90%, 80%, and 70% of their daily intakes were provided to five groups (control, 5, 10, 20, and 30% CR, respectively) of five rats each for 1 week or 1 month. (Figure 1). The rats were anesthetized with diethyl ether on the last day of the experiment, after an overnight (16 h) fast and their white adipose tissue (visceral fat), skeletal muscle (gastrocnemius), brain (hypothalamus area), small intestine (collected only after 1 month), and liver (data reported previously) were collected. All procedures were done according to the Animal Usage Committee of the Faculty of Agriculture at the University of Tokyo’s regulations, and the committee’s consent was acquired (permission number 1818T0011).

2.2. Microarray Experiments

Total RNA was extracted from adipose tissue, the hypothalamus, muscle, and the intestine for the microarray experiment. As previously stated, total RNA was extracted from these tissues using the RNeasy mini kit (Qiagen, Valencia, CA, USA) [8]. The DNA microarray analysis was performed with the Affymetrix GeneChip according to the standard Affymetrix (Thermo Fisher Scientific, Santa Clara, CA, USA) protocols. For the adipose tissue analysis, a pool of complementary RNA was divided in half and used separately for the hybridization to the Affymetrix GeneChip Rat Expression Set 230 Array. A pool of complementary RNA was employed for hybridization to the Affymetrix GeneChip Rat Genome 230 2.0 Array for muscle and hypothalamic investigations. A pool of complementary RNA was used to hybridize to the Affymetrix GeneChip Rat Expression Set 230 Array for the small intestine analysis.

2.3. Differentially Expressed Gene Probes

As previously described [8], we used the default criteria of the Affymetrix GeneChip Operating Software MAS5.0, which was used as follows: ‘detection p value’—present, p < 0.04; marginal, 0.04 ≤ p < 0.06; and absent, p ≥ 0.06; and ‘change p value’—increase, p ≤ 0.0025; marginal increase, 0.0025 < p ≤ 0.003; decrease, p ≥ 0.998; marginal decrease, 0.997 ≤ p < 0.998; and no change, 0.003 < p < 0.997. An algorithm was used to generate signal log ratio, which is a quantitative estimate of the change in gene expression. We did not use a fold-change cut-off to get dynamic expression changes generated by mild-to-moderate CR. A conservative approach in the analysis with a combination of stringent filtering methods was used to reduce false positives. Probe sets that were ‘absent’ in at least one of each hybridization pair were excluded. Comparisons with a ‘no change’ and a ‘marginal increase’ and a ‘marginal decrease’ call were eliminated. For the adipose tissue analysis, we used duplicate GeneChips were used on each group sample, and the expression change was taken as informative if the change call of both chips was either ‘increase’ or ‘decrease’, and in the same direction. We used one GeneChip on each group sample for the muscle, hypothalamus, and small intestine studies, and the expression change was considered informative if the change call was either ‘increase’ or ‘decrease’. The probe sets that showed changes in the same direction across all levels of CR were regarded to be “CR responsive” genes. Microsoft Excel (Microsoft Corp., Redmond, WA, USA) was used to filter the data and identify probe sets that overlapped. The GEO site (http://www.ncbi.nlm.nih.gov/geo/, accessed on 27 June 2021) contains raw data set for all tissues (accession number GSE18297 and GSE176300). The change of the informative gene expression was further validated using randomly selected gene probes in the liver of rats, as previously described [8].

2.4. Gene Ontology Analysis

The functional annotation tool of the Database for Annotation, Visualization, and Integrated Discovery (DAVID) 6.8 was used to undertake gene ontology (GO) analysis on the 30% CR group, which received the most robust CR intervention [9]. This web-based functional annotation tool picks up enrichment in gene groups corresponding to biological functions or categories. For the analysis of CR responsive genes, all informative genes defined were used. For the comparison between each CR treatments and its respective controls, Gene Ontology: Biological Process categories (BP_Direct) were significantly over-represented, as determined by Fisher’s exact test (Adjusted p-value < 1 × 10−4 by the Benjamini and Hochberg method). We also re-analyzed the gene expression data previously obtained for the liver using this version of DAVID.

3. Results

3.1. The Number of CR Responsive Genes

The number of gene probes that were changed by CR was different among the examined tissues (Table 1). There are fewer gene probes in the 5% and 10% CR groups than in the 20% and 30% CR groups for the one week (1 w) liver and adipose tissues and one month (1 m) adipose tissue and intestine experiments. Additional details regarding the number of gene probes that were altered are shown in Supplementary Table S1.

3.2. CR Responsive Genes for Each Tissue

The top five gene probes that were consistently up- or down-regulated across all CR levels (defined as CR responsive genes) for each tissue are displayed in Table 2 and Table 3. The values for the change in expression of each gene are shown on a log scale and are sorted according to the values found for the 30% CR groups after one month of experiment, as this was the most robust intervention in the present study.
Our previous study demonstrated a significant up-regulation of metallothionein genes (namely Mt2a and Mt1a), involved in metal detoxification and oxidative stress, and down-regulation of fatty acid synthase (FAS) genes, which code for key enzymes tangled in fatty acid synthesis in rat liver of rats with mild-to-moderate CR [8]. These major findings were confirmed here by the re-analysis of the GO data obtained for the 30% CR group using DAVID. Both the up-regulation of the ‘oxidation-reduction process [GO:0055114] (p = 8.65 × 10−13)’ and ‘fatty acid beta-oxidation [GO:0006635] (p = 1.39 × 10−6)’ categories (Supplementary Table S2) and the down-regulation of ‘lipid metabolic process [GO:0006629] (p = 4.03 × 10−5)’ were demonstrated (Supplementary Table S3).
Table 2 and Table 3 demonstrate that 45 and 14 gene probes were up- and down-regulated in adipose tissue, respectively, as compared to the control group. The D site of the albumin promoter (albumin D-box) binding protein (Dbp) gene, which is involved in circadian rhythm regulation, was shown to be the most up-regulated. The steroidogenic acute regulatory protein (Star) gene, which codes for the rate-limiting enzyme in the production of steroid hormones from cholesterol [10], was also significantly up-regulated. Most significantly down-regulated genes included the secretory leukocyte peptidase inhibitor (Slpi) and the adipose-derived inflammatory factor that is reported to be increased with obesity [11]. These genes code for important lipid-metabolizing enzymes. Correspondingly, GO analysis identified the up-regulation of the ‘tricarboxylic acid cycle’ [GO:0006099] (p = 1.63 × 10−13) and ‘fatty acid beta-oxidation‘ [GO:0006635] (p = 1.1 × 10−11) in the top-ranked categories (Supplementary Table S2).
Fifty genes were up-regulated, and 51 genes were down-regulated in muscle. Flavin containing monooxygenase 1 (FMO1), a novel regulator of energy balance that promotes metabolic efficiency, was one of the top-ranked genes that were up-regulated. The G0/G1 switch 2 (G0s2), which controls lipid metabolism in muscle [12], was significantly down-regulated. GO analysis indicated that the ‘response to hypoxia‘ [GO:0001666] (p = 1.29 × 10−4), ‘muscle contraction‘ [GO:0006936] (p = 3.5 × 10−4), and ‘actin cytoskeleton organization‘ [GO:0030036] (p = 5.90 × 10−4) were significantly over-represented in the down-regulated category (Supplementary Table S3).
The number of altered gene probes at the 1 m-hypothalamus sample was small; hence, only two known genes were identified as down-regulated and as CR responsive genes in this tissue. One of these genes, the nuclear receptor subfamily 4, group A, member 3 (Nr4a3), may promote food intake as well as body weight gain via its actions in the brain [13]. Another gene, the early growth response 1 (Egr1), is known as a neurogenic transcription factor and it is associated with appetite signaling [14].
Forty-seven genes were up-regulated and 23 genes were down-regulated in the small intestine. The top five up-regulated genes included sulfotransferase family 1A member 1 (Sult1a1), which is highly expressed in the small intestine [15] and is important in xenobiotic metabolism. The down-regulated genes included the gut peptide neuromedin U (Nmu) that has been reported to decrease food intake and body weight [16]. Interestingly, ‘aging‘ [GO:0007568] (p = 3.93 × 10−4) category was up-regulated in the intestine (Supplementary Table S2).

3.3. Up-Regulated Genes across All Tissues Studied

As we could not identify all CR responsive genes across all tissues, the genes that exhibited the greatest changes across multiple tissues were screened. The top ten up-regulated genes, including at least 20 out of 36 CR conditions across multiple organs, are listed in Table 4. The most commonly up-regulated gene was Nr1d2, an orphan nuclear receptor known as a circadian regulator. This gene was up-regulated in almost all CR conditions except in the 1w-hypothalamus. Furthermore, the aldo-keto reductase family 1, member C14 (Akr1c14) and the nicotinamide phosphoribosyltransferase (Nampt) genes were up-regulated within 21 CR conditions. Other up-regulated genes across 20 CR conditions included the glutamate-ammonia ligase (Glul), sulfotransferase family 1A member 1 (Sult1a1), CD36 molecule (thrombospondin receptor) (Cd36), flavin containing monooxygenase 1 (Fmo1), epoxide hydrolase 1, microsomal (xenobiotic) (Ephx1), and Dbp.
The expression signatures differed among tissues. For example, no up-regulated genes were observed in the brain, except for Nr1d2 and Sult1a1. Genes Sult1a1, Ephx1, and Dbp were not found in the liver, and no changes in Fmo1 expressions were observed in the brain and intestine.
A complete list of the up-regulated genes across the multiple tissues is displayed in Supplementary Table S4.

3.4. Down-Regulated Genes across All Tissues Studied

Genes down-regulated in response to CR in at least 19 of 36 CR conditions among the multiple tissues are also listed in Table 4. The most commonly down-regulated gene was CKLF-like MARVEL transmembrane domain containing 6 (Cmtm6), which is tangled in immune response and inflammatory activities. Cytochrome P450, family 51 (Cyp51), known to be involved in cholesterol biosynthesis, was ranked second. The heat shock protein 90, alpha (cytosolic), class A member 1 (Hsp90aa1) was ranked the third most commonly down-regulated gene, although no change was detected in the liver tissue. Genes coding for structural proteins such as actin, gamma 1 (Actg1), tubulin, beta 4B class IVb (Tubb4b), and tubulin, beta 2A class IIa (Tubb2a) were down-regulated as expected. Additionally, fatty acid desaturase 1 (Fads1), tropomyosin 4 (Tpm4), and sphingosine-1-phosphate receptor 1 (S1pr1) were down-regulated.
No changes in Cmtm6 and Actg1 expressions were detected in the intestine. No changes in the expressions of Tubb4b, Tpm4, S1pr1, and Tubb2a were detected in the hypothalamus and intestine.

3.5. Comparison with a Meta-Analysis of the CR Effect

Two meta-analyses have been conducted for 33 and 61 CR studies [17,18], as summarized in Supplementary Table S5. We compared our 36 CR responsive genes found in each tissue with the findings reported for these CR meta-analyses datasets. The overlapped genes are shown in Supplementary Table S6. Eighteen of our 36 CR responsive genes overlapped with those of the previous meta-analyses, and all gene expressions except that of Dbp were changed in the same direction as our data.
The 18 most commonly regulated genes across all tissues were also compared using the CR meta-analyses. Seven of these 18 genes overlapped with the CR meta-analyses data (Table 5). More specifically, genes Nampt, Glul, Sult1a1, and Fmo1 were up-regulated, while Cmtm6, Cyp51, Hsp90aa1, Actg1, and Tubb2a were down-regulated in both our study and in the meta-analyses. The Sult1a1 and Actg1 genes overlapped in both meta-analyses datasets. Overall, nearly half of the genes found in our study were overlapped with those in the previous CR meta-analyses.

4. Discussion

The gene expression profiles of numerous tissues of young growing rats with mild-to-moderate and short-term CR were studied in this work. First, we presented the transcriptomic characteristics induced by CR in each tissue (Table 2 and Table 3). Notably, by comparing the CR responsive genes in each tissue found in our study with the dataset resulting from two previous meta-analyses, which contained severe CR conditions in multiple organs and tissues, nearly half of the genes were overlapped in the same direction (Supplementary Table S6). The remaining genes did not overlap or conflicted (only one gene) with those in the meta-analyses dataset, and this discrepancy may explain the differential responses to CR in the setting of each study (e.g., duration and strength of CR, dietary regimes, gender, species and developmental stage of animals used).
Second, we looked for a common element that could cause the favorable effect of CR by looking at the commonality of genes reacting to CR across diverse tissues. The top 18 genes that were consistently altered across the five examined tissues were therefore further evaluated (Table 4). Among them, seven genes (Nampt, Glul, Sult1a1, Fmo1, Cmtm6, Hsp90aa1, and Actg1) were overlapped with the CR meta-analyses dataset in the same direction (Table 5). These genes might be essential for CR beneficial effects and thus might be used as sensitive biomarkers of CR, as they responded to relatively mild CR conditions and in short-term in multiple tissues.
In a wide range of species, including rats and primates, it is commonly acknowledged that CR extends lifespan. We found several aging-related genes that changed across the multiple tissues examined and are represented by Nampt. This gene codes for the major rate-limiting enzyme in NAD+ production, which has been shown to decline with age in multiple tissues such as the liver, adipose, and brain [19]. The pathogenesis of age-related metabolic disorders is aided by the lowering of Nampt and NAD+ levels in many tissues [20,21]. A study performed using an eight-week CR also demonstrated the up-regulation of Nampt mRNA in rat organs [22], consistent with our data.
Another key biological process and target underlying aging is cellular senescence. We found the down-regulation of Hsp90aa1, a member of the Hsp90 superfamily. This gene codes for an abundant protein that functions as a molecular chaperone [23]. The inhibitors of Hsp90 have been shown recently to be a novel class of senolytics [24]. Multi-tissue dysregulation of Hsp90 members, as seen in our study, may explain the anti-aging effect of CR through the clearance of senescent cells. Moreover, the up-regulation of Glul, known to code for the glutamine synthetase that catalyzes the de novo synthesis of glutamine from glutamate and ammonia, was observed. Glul is ubiquitously expressed and particularly highly expressed in the muscle, liver, and brain [25]. Recently, Johomura et. al. showed that activation of glutaminolysis induced the production of ammonia, which neutralized the lower pH to improve the survival of the senescent cells [26]. Therefore, the up-regulation of Glul induced by CR may inversely repress this process, thus promoting the death of senescent cells as well as a decline of aging effects.
One of the most prominent health benefits of CR is the prevention of malignancies. We found that cancer-related genes such as Cmtm6 were down-regulated in response to CR. CMTM6 is a ubiquitously expressed protein that is known to be a critical regulator of PD-L1 [27,28], a target of immune checkpoint inhibitor therapy [29]. CMTM6 depletion in multiple tissues may decrease PD-L1 expression and cancer incidence through a CR-related mechanism. Moreover, S1pr1, a novel promising target in cancer therapy [30], was broadly down-regulated across the five tissues examined in our study. Thus, CR-induced metabolic changes may not only reduce the incidence of cancer, but also increase the efficacy of cancer therapies as proposed previously [31].
According to a current hypothesis of aging, aging is caused by a loss in detoxifying capacity, and CR affects the expressions of genes involved in xenobiotic metabolism [32]. In our study, the xenobiotic-related gene Fmo1 was up-regulated and overlapped with the meta-analyses dataset. FMOs are enzymes originally implicated in the oxidation of xenobiotics but have been recently implicated to promote longevity and health span [33,34]. EPHX1 is an enzyme that aids in the detoxification of cigarette-related chemicals [35], which is a significant risk factor for the development of certain cancers [36]. SULT1A1 is a phase II xenobiotic metabolizing enzyme that is extensively expressed in the liver and facilitates carcinogen sulfonation [37]. It is therefore no surprise that these detoxication enzymes are transcriptionally activated in response to CR in multiple tissues, and that they may contribute to the extension of the human lifespan.
Even minor calorie restriction, such as a 15% dietary restriction for 16 weeks, has been shown to enhance lipid metabolism [38]. We also observed a marked decrease of white adipose tissue weight under a 20% CR diet for one week [8]. However, many genes related to lipid metabolism were identified that did not overlap the meta-analyses dataset. Cd36, which is well known to be tangled in the regulating of lipogenesis in human adipose tissue [39] was up-regulated. CYP51, known to have a role in cholesterol biosynthesis in mammalian cells [40], and FADS1, a rate-limiting enzyme that generates long-chain polyunsaturated fatty acids [41], were down-regulated. As expected, the expression of these genes was mostly changed toward improving lipid metabolism in target tissues. Moreover, Nr1d2 (also referred to Rev-erbbeta), first discovered to be a gene regulator involved in circadian rhythm and fat accumulation [42], was most ubiquitously up-regulated in this study. It also controls lipid and energy homeostasis in skeletal muscle [43]. Interestingly, circadian clock gene was recently proposed to mediate the beneficial effect of CR and that may contribute to longevity [44].
Our present findings demonstrate that the expressions of key genes involved in the CR-induced beneficial effects were regulated even by very mild and short-term interventional CR conditions, such as 5%–10% CR for one week, which are applicable in humans. These expression profiles can also be used as reference data for removing the transcripts responding to the reduction of food intake often observed in in vivo nutrition research. Indeed, CR profiles have been successfully applied to identify sensitive transcriptomic biomarkers of selenium status [45]. In summary, the approach and data obtained in the present study will not only help providing insight on novel mechanisms associated with CR-induced health benefits but may also identify targets for functional and safety assessment of food and nutrients.
One limitation of our investigation is the absence of biological replicates, as we used pooled samples for DNA microarray analysis. However, we applied conservative criteria to screen the genes in the data filtering process and the changes in expression were partly validated by qPCR using randomly selected samples in a previous study [8]. Another limitation is that we used relatively young rats for the CR study since one of the purposes of this investigation was to obtain the reference gene expression data for in vivo nutrition research where the young growing rats are often used.

5. Conclusions

Our findings give essential knowledge on the molecular mechanisms underlying CR’s positive consequences. The present study also provides a way to identify dietary or therapeutic targets for aging-related diseases. Further studies are required to determine if the genes found in this study are involved in the essential mechanism of CR-induced beneficial effects in different species, including human.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/nu13072277/s1, Table S1: Number of genes altered by short-term and mild-to-moderate CR, Table S2: GO analysis of the genes up-regulated by 30%CR, Table S3: GO analysis of the genes down-regulated by 30%CR, Table S4: Genes commonly altered by CR, Table S5: Reported meta-analysis of CR transcriptome experiment, Table S6: The comparison of the top five genes consistently up- or down- regulated for each organ with meta-analysis data.

Author Contributions

K.S. wrote the manuscript, and M.I., H.J. and T.C. revised the manuscript. K.S. performed the data analyses. H.K. supervised the work. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Grants-in-Aid from the Japan Society for the Promotion of Science (KAKENHI, 18380077).

Institutional Review Board Statement

The study was approved by Animal Usage Committee of the Faculty of Agriculture at the University of Tokyo (permission number 1818T0011).

Informed Consent Statement

Not applicable.

Data Availability Statement

The microarray data have been deposited in the GEO database (https://www.ncbi.nlm.nih.gov/geo/, accessed on 27 June 2021) under accession number GSE176300.

Acknowledgments

Not applicable.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Most, J.; Tosti, V.; Redman, L.M.; Fontana, L. Calorie restriction in humans: An update. Ageing Res. Rev. 2017, 39, 36–45. [Google Scholar] [CrossRef]
  2. Austad, S.N.; Hoffman, J.M. Beyond calorie restriction: Aging as a biological target for nutrient therapies. Curr. Opin. Biotechnol. 2020, 70, 56–60. [Google Scholar] [CrossRef] [PubMed]
  3. Fontana, L.; Partridge, L. Promoting health and longevity through diet: From model organisms to humans. Cell 2015, 161, 106–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Amer, B.; Baidoo, E.E.K. Omics-driven biotechnology for industrial applications. Front. Bioeng. Biotechnol. 2021, 9, 613307. [Google Scholar] [CrossRef]
  5. Aon, M.A.; Bernier, M.; Mitchell, S.J.; Di Germanio, C.; Mattison, J.A.; Ehrlich, M.R.; Colman, R.J.; Anderson, R.M.; de Cabo, R. Untangling determinants of enhanced health and lifespan through a multi-omics approach in mice. Cell Metab. 2020, 32, 100–116.e4. [Google Scholar] [CrossRef] [PubMed]
  6. Castillo-Armengol, J.; Fajas, L.; Lopez-Mejia, I.C. Inter-organ communication: A gatekeeper for metabolic health. EMBO Rep. 2019, 20, e47903. [Google Scholar] [CrossRef]
  7. Wang, F.; So, K.-F.; Xiao, J.; Wang, H. Organ-organ communication: The liver’s perspective. Theranostics 2021, 11, 3317–3330. [Google Scholar] [CrossRef]
  8. Saito, K.; Ohta, Y.; Sami, M.; Kanda, T.; Kato, H. Effect of mild restriction of food intake on gene expression profile in the liver of young rats: Reference data for in vivo nutrigenomics study. Br. J. Nutr. 2010, 104, 941–950. [Google Scholar] [CrossRef] [Green Version]
  9. Huang, D.W.; Sherman, B.T.; Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009, 4, 44–57. [Google Scholar] [CrossRef]
  10. Manna, P.R.; Stetson, C.L.; Slominski, A.T.; Pruitt, K. Role of the steroidogenic acute regulatory protein in health and disease. Endocrine 2016, 51, 7–21. [Google Scholar] [CrossRef] [Green Version]
  11. Hoggard, N. Increase in circulating and adipose tissue expression of secretory leukocyte peptidase inhibitor (SLPI) with obesity and diabetes. Open Nutr. J. 2012, 6, 108–115. [Google Scholar] [CrossRef] [Green Version]
  12. Laurens, C.; Badin, P.-M.; Louche, K.; Mairal, A.; Tavernier, G.; Marette, A.; Tremblay, A.; Weisnagel, S.J.; Joanisse, D.R.; Langin, D.; et al. G0/G1 Switch Gene 2 controls adipose triglyceride lipase activity and lipid metabolism in skeletal muscle. Mol. Metab. 2016, 5, 527–537. [Google Scholar] [CrossRef]
  13. Xu, Y.; O’Malley, B.W.; Elmquist, J.K. Brain nuclear receptors and body weight regulation. J. Clin. Investig. 2017, 127, 1172–1180. [Google Scholar] [CrossRef] [Green Version]
  14. Cyr, N.E.; Toorie, A.M.; Steger, J.S.; Sochat, M.M.; Hyner, S.; Perello, M.; Stuart, R.; Nillni, E.A. Mechanisms by which the orexigen NPY regulates anorexigenic α-MSH and TRH. Am. J. Physiol. Endocrinol. Metab. 2013, 304, E640–E650. [Google Scholar] [CrossRef] [Green Version]
  15. Alnouti, Y.; Klaassen, C.D. Tissue distribution and ontogeny of sulfotransferase enzymes in mice. Toxicol. Sci. 2006, 93, 242–255. [Google Scholar] [CrossRef] [PubMed]
  16. Jarry, A.-C.; Merah, N.; Cisse, F.; Cayetanot, F.; Fiamma, M.-N.; Willemetz, A.; Gueddouri, D.; Barka, B.; Valet, P.; Guilmeau, S.; et al. Neuromedin U is a gut peptide that alters oral glucose tolerance by delaying gastric emptying via direct contraction of the pylorus and vagal-dependent mechanisms. FASEB J. 2019, 33, 5377–5388. [Google Scholar] [CrossRef]
  17. Swindell, W.R. Genes and gene expression modules associated with caloric restriction and aging in the laboratory mouse. BMC Genomics 2009, 10, 585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Plank, M.; Wuttke, D.; van Dam, S.; Clarke, S.A.; de Magalhães, J.P. A meta-analysis of caloric restriction gene expression profiles to infer common signatures and regulatory mechanisms. Mol. Biosyst. 2012, 8, 1339–1349. [Google Scholar] [CrossRef]
  19. Yaku, K.; Okabe, K.; Nakagawa, T. NAD metabolism: Implications in aging and longevity. Ageing Res. Rev. 2018, 47, 1–17. [Google Scholar] [CrossRef]
  20. Imai, S.; Yoshino, J. The importance of NAMPT/NAD/SIRT1 in the systemic regulation of metabolism and ageing. Diabetes Obes. Metab. 2013, 15 (Suppl. 3), 26–33. [Google Scholar] [CrossRef] [Green Version]
  21. Stein, L.R.; Imai, S.-I. Specific ablation of Nampt in adult neural stem cells recapitulates their functional defects during aging. EMBO J. 2014, 33, 1321–1340. [Google Scholar] [CrossRef] [Green Version]
  22. Song, J.; Ke, S.-F.; Zhou, C.-C.; Zhang, S.-L.; Guan, Y.-F.; Xu, T.-Y.; Sheng, C.-Q.; Wang, P.; Miao, C.-Y. Nicotinamide phosphoribosyltransferase is required for the calorie restriction-mediated improvements in oxidative stress, mitochondrial biogenesis, and metabolic adaptation. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 44–57. [Google Scholar] [CrossRef] [Green Version]
  23. Panaretou, B.; Prodromou, C.; Roe, S.M.; O’Brien, R.; Ladbury, J.E.; Piper, P.W.; Pearl, L.H. ATP binding and hydrolysis are essential to the function of the Hsp90 molecular chaperone in vivo. EMBO J. 1998, 17, 4829–4836. [Google Scholar] [CrossRef] [Green Version]
  24. Fuhrmann-Stroissnigg, H.; Niedernhofer, L.J.; Robbins, P.D. Hsp90 inhibitors as senolytic drugs to extend healthy aging. Cell Cycle 2018, 17, 1048–1055. [Google Scholar] [CrossRef] [Green Version]
  25. Zhang, J.; Pavlova, N.N.; Thompson, C.B. Cancer cell metabolism: The essential role of the nonessential amino acid, glutamine. EMBO J. 2017, 36, 1302–1315. [Google Scholar] [CrossRef] [Green Version]
  26. Johmura, Y.; Yamanaka, T.; Omori, S.; Wang, T.-W.; Sugiura, Y.; Matsumoto, M.; Suzuki, N.; Kumamoto, S.; Yamaguchi, K.; Hatakeyama, S.; et al. Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science 2021, 371, 265–270. [Google Scholar] [CrossRef] [PubMed]
  27. Burr, M.L.; Sparbier, C.E.; Chan, Y.-C.; Williamson, J.C.; Woods, K.; Beavis, P.A.; Lam, E.Y.N.; Henderson, M.A.; Bell, C.C.; Stolzenburg, S.; et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature 2017, 549, 101–105. [Google Scholar] [CrossRef] [Green Version]
  28. Mezzadra, R.; Sun, C.; Jae, L.T.; Gomez-Eerland, R.; de Vries, E.; Wu, W.; Logtenberg, M.E.W.; Slagter, M.; Rozeman, E.A.; Hofland, I.; et al. Identification of CMTM6 and CMTM4 as PD-L1 protein regulators. Nature 2017, 549, 106–110. [Google Scholar] [CrossRef] [PubMed]
  29. Salmaninejad, A.; Valilou, S.F.; Shabgah, A.G.; Aslani, S.; Alimardani, M.; Pasdar, A.; Sahebkar, A. PD-1/PD-L1 pathway: Basic biology and role in cancer immunotherapy. J. Cell. Physiol. 2019, 234, 16824–16837. [Google Scholar] [CrossRef] [PubMed]
  30. Rostami, N.; Nikkhoo, A.; Ajjoolabady, A.; Azizi, G.; Hojjat-Farsangi, M.; Ghalamfarsa, G.; Yousefi, B.; Yousefi, M.; Jadidi-Niaragh, F. S1PR1 as a novel promising therapeutic target in cancer therapy. Mol. Diagn. Ther. 2019, 23, 467–487. [Google Scholar] [CrossRef]
  31. Brandhorst, S.; Longo, V.D. Fasting and caloric restriction in cancer prevention and treatment. Recent Results Cancer Res. 2016, 207, 241–266. [Google Scholar]
  32. Fu, Z.D.; Klaassen, C.D. Short-term calorie restriction feminizes the mRNA profiles of drug metabolizing enzymes and transporters in livers of mice. Toxicol. Appl. Pharmacol. 2014, 274, 137–146. [Google Scholar] [CrossRef] [Green Version]
  33. Guo, D.; Shen, Y.; Li, W.; Li, Q.; Miao, Y.; Zhong, Y. Upregulation of flavin-containing monooxygenase 3 mimics calorie restriction to retard liver aging by inducing autophagy. Aging 2020, 12, 931–944. [Google Scholar] [CrossRef]
  34. Rossner, R.; Kaeberlein, M.; Leiser, S.F. Flavin-containing monooxygenases in aging and disease: Emerging roles for ancient enzymes. J. Biol. Chem. 2017, 292, 11138–11146. [Google Scholar] [CrossRef] [Green Version]
  35. Oesch, F.; Glatt, H.; Schmassmann, H. The apparent ubiquity of epoxide hydratase in rat organs. Biochem. Pharmacol. 1977, 26, 603–607. [Google Scholar] [CrossRef]
  36. Jacob, L.; Freyn, M.; Kalder, M.; Dinas, K.; Kostev, K. Impact of tobacco smoking on the risk of developing 25 different cancers in the UK: A retrospective study of 422,010 patients followed for up to 30 years. Oncotarget 2018, 9, 17420–17429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Hempel, N.; Wang, H.; LeCluyse, E.L.; McManus, M.E.; Negishi, M. The human sulfotransferase SULT1A1 gene is regulated in a synergistic manner by Sp1 and GA binding protein. Mol. Pharmacol. 2004, 66, 1690–1701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Park, C.Y.; Park, S.; Kim, M.S.; Kim, H.-K.; Han, S.N. Effects of mild calorie restriction on lipid metabolism and inflammation in liver and adipose tissue. Biochem. Biophys. Res. Commun. 2017, 490, 636–642. [Google Scholar] [CrossRef]
  39. Okamoto, F.; Tanaka, T.; Sohmiya, K.; Kawamura, K. CD36 abnormality and impaired myocardial long-chain fatty acid uptake in patients with hypertrophic cardiomyopathy. Jpn. Circ. J. 1998, 62, 499–504. [Google Scholar] [CrossRef] [Green Version]
  40. Tsuchiya, T.; Dhahbi, J.M.; Cui, X.; Mote, P.L.; Bartke, A.; Spindler, S.R. Additive regulation of hepatic gene expression by dwarfism and caloric restriction. Physiol. Genom. 2004, 17, 307–315. [Google Scholar] [CrossRef]
  41. Athinarayanan, S.; Fan, Y.-Y.; Wang, X.; Callaway, E.; Cai, D.; Chalasani, N.; Chapkin, R.S.; Liu, W. Fatty acid desaturase 1 influences hepatic lipid homeostasis by modulating the PPARα-FGF21 axis. Hepatol. Commun. 2020, 5, 461–477. [Google Scholar] [CrossRef] [PubMed]
  42. Burris, T.P. Nuclear hormone receptors for heme: REV-ERBalpha and REV-ERBbeta are ligand-regulated components of the mammalian clock. Mol. Endocrinol. 2008, 22, 1509–1520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ramakrishnan, S.N.; Lau, P.; Burke, L.J.; Muscat, G.E.O. REV-ERBbeta regulates the expression of genes involved in lipid absorption in skeletal muscle cells: Evidence for cross-talk between orphan nuclear receptors and myokines. J. Biol. Chem. 2005, 280, 8651–8659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chaudhari, A.; Gupta, R.; Makwana, K.; Kondratov, R. Circadian clocks, diets and aging. Nutr. Healthy Aging 2017, 4, 101–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Raines, A.M.; Sunde, R.A. Selenium toxicity but not deficient or super-nutritional selenium status vastly alters the transcriptome in rodents. BMC Genom. 2011, 12, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Overview of the animal experiment.
Figure 1. Overview of the animal experiment.
Nutrients 13 02277 g001
Table 1. Number of genes altered by short-term and mild-to-moderate CR.
Table 1. Number of genes altered by short-term and mild-to-moderate CR.
OrgansDuration5%10%20%30%Overlap
liver1 week18358890273490
1 month495454572570
adipose1 week3879811084167459
1 month304123216241255
muscle1 week68466312291517101
1 month1041109710741422
brain
(hypothalamus)
1 week13461413142913453
1 month7658119148
intestine-----70
1 month370503530558
The number of gene probes overlapped in the same direction throughout all the CR groups. The values were determined according to the criteria described in Materials and methods. 5%: 5% calorie restriction. 10%: 10% calorie restriction. 20%: 20% calorie restriction. 30%: 30% calorie restriction.
Table 2. Top five gene probes consistently up-regulated across all CR levels for each tissue.
Table 2. Top five gene probes consistently up-regulated across all CR levels for each tissue.
1 Week1 MonthGene TitleGene Symbol
Probe ID5%10%20%30%5%10%20%30%
liver
(40 gene probes)
1388271_at2.23.54.54.52.32.93.44.0metallothionein 2AMt2A
1371237_a_at2.43.64.14.02.93.43.53.9metallothionein 1a///transthyretinMt1a///Ttr
1387336_at1.02.43.73.72.02.63.23.5N-acetyltransferase 8Nat8
1387156_at3.94.44.44.13.94.13.83.3hydroxysteroid (17-beta) dehydrogenase 2Hsd17b2
1368213_at0.81.52.53.11.52.02.52.8P450 (cytochrome) oxidoreductasePor
adipose
(45 gene probes)
1387874_at2.52.72.92.92.82.93.02.5D site of albumin promoter (albumin D-box) binding proteinDbp
1388039_a_at1.01.72.02.41.62.32.32.4gamma-aminobutyric acid (GABA) B receptor 1Gabbr1
1368406_at1.21.62.32.41.41.72.22.3steroidogenic acute regulatory proteinStar
1372536_at0.81.21.92.30.91.41.92.2aarF domain containing kinase 3Adck3
1387174_a_at1.01.51.92.01.51.82.22.2steroidogenic acute regulatory proteinStar
muscle
(50 gene probes)
1378927_at0.81.01.61.61.41.51.81.8--
1387053_at0.61.00.91.50.90.91.11.6flavin containing monooxygenase 1Fmo1
1368971_a_at0.70.81.01.11.21.11.31.5synaptojanin 2Synj2
1369150_at0.30.90.61.20.70.60.81.4pyruvate dehydrogenase kinase, isozyme 4Pdk4
1370019_at0.70.91.21.41.01.11.21.4sulfotransferase family 1A member 1Sult1a1
brainnot identifed
intestine
(47 gene probes)
1370019_at 1.21.31.31.8sulfotransferase family 1A member 1Sult1a1
1371076_at 1.31.12.01.8cytochrome P450, family 2, subfamily b, polypeptide 1///cytochrome P450, family 2, subfamily b, polypeptide 2Cyp2b1///Cyp2b2
1368303_at 1.31.31.61.4period circadian clock 2Per2
1367774_at 0.51.00.71.3glutathione S-transferase alpha 1///glutathione S-transferase alpha-3-likeGsta1///LOC102550391
1369455_at 1.30.91.91.2ATP-binding cassette, subfamily G (WHITE), member 5Abcg5
Values are shown as log ratio vs control.
Table 3. Top five gene probes consistently down-regulate across all CR levels for each tissue.
Table 3. Top five gene probes consistently down-regulate across all CR levels for each tissue.
1 Week1 MonthGene TitleGene Symbol
Probe ID5%10%20%30%5%10%20%30%
liver
(50 gene probes)
1367707_at−1.55−2.45−4.3−4.4−1.7−2.85−3.6−4.4fatty acid synthaseFasn
1367708_a_at−1.2−2.15−4−4.3−1.6−2.75−3.5−3.9fatty acid synthaseFasn
1373718_at−1.6−2.9−3.75−4.05−2.55−3.15−3.45−3.4tubulin, beta 2A class IIaTubb2a
1370870_at−1.5−1.9−2.95−2.95−1.75−2.25−2.55−2.85malic enzyme 1, NADP(+)-dependent, cytosolicMe1
1367854_at−1.1−1.85−2.55−2.8−1.4−2−2.3−2.55ATP citrate lyaseAcly
adipose
(14 gene probes)
1367998_at−1.15−1.4−1.35−2.85−0.7−2.5−2.45−1.95secretory leukocyte peptidase inhibitorSlpi
1368294_at−1.1−0.5−1.4−2.3−0.9−1.45−1.8−1.8deoxyribonuclease 1-like 3Dnase1l3
1389006_at−0.75−0.7−1.2−2−0.7−1.2−1.6−1.6macrophage expressed 1Mpeg1
1368189_at−0.9−0.65−1.45−1.7−0.9−0.85−1.3−1.47-dehydrocholesterol reductaseDhcr7
1373718_at−0.45−0.8−0.8−0.95−0.75−0.95−0.8−1.15tubulin, beta 2A class IIaTubb2a
muscle
(51 gene probes)
1388395_at−0.7−1.7−2.5−2.9−2.5−3.5−3−2.2G0/G1switch 2G0s2
1378423_at−0.3−0.9−1.3−1.6−1.8−1.7−1.8−2.1nicotinamide riboside kinase 2Nmrk2
1379416_at−1.1−1.4−2−2.2−1.8−1.5−2.5−2autism susceptibility candidate 2-likeLOC100362819
1378586_at−2.5−2.2−2.2−2.2−2−1.7−0.6−1.9cytokine inducible SH2-containing proteinCish
1374204_at−1.4−1.1−1.8−1.9−2.5−2.6−2.5−1.4WD repeat and SOCS box-containing 1Wsb1
brain
(3 gene probes)
1375043_at−2.3−2.4−2−2.1−1.3−1.4−1.2−1.4----
1369067_at−1−1−0.9−1.1−0.9−1.2−1.2−1.1nuclear receptor subfamily 4, group A, member 3Nr4a3
1368321_at−0.7−0.7−0.4−0.7−1.2−1.4−1.2−0.9early growth response 1Egr1
intestine
(23 gene probes)
1369717_at −1−1.1−1.1−1.7neuromedin UNmu
1387758_at −0.5−1−0.2−1.7alkaline phosphatase 3, intestine, not Mn requiringAkp3
1378658_at −0.9−1.6−1−1.4chloride channel accessory 4Clca4
1368247_at −1.4−1.1−1.1−1.2heat shock 70kD protein 1A///heat shock 70kD protein 1B (mapped)Hspa1a///Hspa1b
1389986_at −0.7−0.5−1.1−1.1----
Values are shown as log ratio vs control.
Table 4. Top-ranked genes commonly regulated by CR across all tissues studied.
Table 4. Top-ranked genes commonly regulated by CR across all tissues studied.
Probe IDGene
Symbol
Gene Title# of Overlapped
Gene Probes
LiverAdiposeMuscleBrain(hypothalamus)Intestine
1 Week1 Month1 Week1 Month1 Week1 Month1 Week1 Month1 Month
5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%
1390430_atNr1d2nuclear receptor subfamily 1, group D, member 229
1370708_a_atAkr1c14aldo-keto reductase family 1, member C1421
1389014_atNamptnicotinamide phosphoribosyltransferase21
1367633_atGlulglutamate-ammonia ligase20
1370019_atSult1a1sulfotransferase family 1A member 120
1386870_atGlulglutamate-ammonia ligase20
1386901_atCd36CD36 molecule (thrombospondin receptor)20
1387053_atFmo1flavin containing monooxygenase 120
1387669_a_atEphx1epoxide hydrolase 1, microsomal (xenobiotic)20
1387874_atDbpD site of albumin promoter (albumin D-box) binding protein20
Probe IDGene
Symbol
Gene Title# of Overlapped
Gene Probes
LiverAdiposeMuscleBrain(hypothalamus)Intestine
1 Week1 Month1 weEk1 Month1 Week1 Month1 Week1 Month1 Month
5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%5%10%20%30%
1372056_atCmtm6CKLF-like MARVEL transmembrane domain containing 625
1367979_s_atCyp51cytochrome P450, family 5123
1388850_atHsp90aa1heat shock protein 90, alpha (cytosolic), class A member 121
1371327_a_atActg1actin, gamma 120
1367857_atFads1fatty acid desaturase 119
1371390_atTubb4btubulin, beta 4B class IVb19
1371653_atTpm4tropomyosin 419
1371840_atS1pr1sphingosine-1-phosphate receptor 119
1372727_at--19
1373718_atTubb2atubulin, beta 2A class IIa19
Red arrows represent the genes up-regulated and blue arrows represent the genes down-regulated by mild CR. Complete list is available at Supplementary Table S4.
Table 5. The overlap of top-ranked genes commonly regulated by CR across all tissues with previous meta-analyses data.
Table 5. The overlap of top-ranked genes commonly regulated by CR across all tissues with previous meta-analyses data.
Gene SymbolFunctionOur StudyRef #1Ref #2
NamptNAD metabolismup up
Glulglutamate metabolismup up
Sult1a1xenobiotic metabolismupupup
Fmo1xenobiotic metabolismup up
Dbpcircadian rhythmupupdown
Cmtm6immune systemdown down
Hsp90aa1chaperone proteindown down
Actg1structural proteindowndowndown
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Saito, K.; Ito, M.; Chiba, T.; Jia, H.; Kato, H. A Comparison of Gene Expression Profiles of Rat Tissues after Mild and Short-Term Calorie Restrictions. Nutrients 2021, 13, 2277. https://doi.org/10.3390/nu13072277

AMA Style

Saito K, Ito M, Chiba T, Jia H, Kato H. A Comparison of Gene Expression Profiles of Rat Tissues after Mild and Short-Term Calorie Restrictions. Nutrients. 2021; 13(7):2277. https://doi.org/10.3390/nu13072277

Chicago/Turabian Style

Saito, Kenji, Maiko Ito, Takuya Chiba, Huijuan Jia, and Hisanori Kato. 2021. "A Comparison of Gene Expression Profiles of Rat Tissues after Mild and Short-Term Calorie Restrictions" Nutrients 13, no. 7: 2277. https://doi.org/10.3390/nu13072277

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