Accepted Manuscript Title: Nuclear receptors as regulators of stem cell and cancer stem cell metabolism Author: Zoltan Simandi Ixchelt Cuaranta-Monroy Laszlo Nagy PII: DOI: Reference:

S1084-9521(13)00107-9 http://dx.doi.org/doi:10.1016/j.semcdb.2013.10.002 YSCDB 1470

To appear in:

Seminars in Cell & Developmental Biology

Please cite this article as: Simandi Z, Cuaranta-Monroy I, Nagy L, Nuclear receptors as regulators of stem cell and cancer stem cell metabolism, Seminars in Cell and Developmental Biology (2013), http://dx.doi.org/10.1016/j.semcdb.2013.10.002 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Nuclear receptors as regulators of stem cell and cancer stem cell metabolism

ip t

Zoltan Simandi1, Ixchelt Cuaranta-Monroy1 and Laszlo Nagy1,2#

1

Department of Biochemistry and Molecular Biology, Research Center for Molecular Medicine, University of Debrecen, Medical and Health Science Center MTA-DE “Lendulet” Immunogenomics Research Group

cr

2

E-mail: [email protected] Phone: +36-52-416-432

Ac ce pt e

Fax: +36-52-314-989

M

Contact Information:

d

#

an

us

Egyetem tér 1., Debrecen, Hungary, H-4012

Highlights

Nuclear receptors (NRs) are key links between metabolism and cell fate decisions. ESRRB, DAX-1, LRH-1, TR4, NGF1-B, LXRb and RARs are NRs expressed in pluripotent embryonic stem cells. ESRRB, DAX-1 and LRH-1 are regulators of metabolism in embryonic stem cells. TR4, NGF1-B, LXRb and RARs are putative regulators of stemness. The discussed NRs play a role in tumor progression and might be metabolic regulators of cancer stem cells.

Page 1 of 25

Abstract

an

us

cr

ip t

Cellular metabolism is underpinning physiological processes in all cells. These include housekeeping functions as well as specific activities unique to a particular cell type. A growing number of studies in various experimental models indicate that metabolism is tightly connected to embryonic development as well. It is also emerging that metabolic processes have regulatory roles and by changing metabolism, cellular processes and even fates can be influenced. Nuclear receptors (NRs) are transcription factors, responding to changes in metabolites and are implicated in diverse biological processes such as embryonic development, differentiation, metabolism and cancer. Therefore, NRs are key links between metabolism and cell fate decisions. In this review, we introduce ESRR , DAX-1 and LRH-1 as putative regulators of metabolism in pluripotent embryonic stem cells. We also discuss the role of TR4, NGF1 , LXR and RARs in stemness. In addition, we summarize our current understanding of the potential roles of NRs in cancer stem cells.

Ac ce pt e

d

M

Key words: metabolism, nuclear receptors, pluripotency, embryonic stem cells, cancer stem cells

Inroduction

Unicellular organisms or cells within multicellular organisms need to make decisions throughout their lifespan. The constantly changing environment dictating whether a cell stays quiescent, proliferate or differentiate [1]. It is well established in yeast that nutrient levels are the major determinants of cell fate. Recent evidence suggests that cellular metabolism determines the cell fate specification of mammalian cells as well [2-4]. Murine embryonic stem cells (ESCs) provide a unique model system to understand the importance of metabolic regulation in cell fate decisions in vertebrates. ESCs are derived from the inner cell mass (ICM) of developing blastocysts and display an almost unlimited proliferation capacity in vitro [5]. Moreover, this cell type retains the ability to contribute to all cell lineages. The rate of cell division of mouse ESCs is very high; a cell doubling takes place in every 4-5 hours. This is even faster than any fast growing cancer cell. Therefore, ESCs have a high

2

Page 2 of 25

requirement for carbon, hydrogen and nitrogen to support biosynthesis of cell building blocks required for replication. Accordingly, metabolic features of ESCs are significantly different from that of terminally differentiated cell types (reviewed in [6]).

ip t

Metabolism in pluripotent stem cells

Ac ce pt e

d

M

an

us

cr

One of the major differences is the way these cells can produce sufficient energy. Oxidative consumption of glucose does not enable a fast rate of ATP production. In contrast, partial breakdown of glucose through glycolysis and shunting of intermediates through the pentose phosphate pathway provide sufficient NADPH and energy for rapid cell proliferation. Oxidative capacity is reduced and glycolysis-dependent anabolic pathways are enriched in stem cells (Figure 1). This underlines the not so critical role of mitochondria in these cells. In fact, ESCs have a low number of mitochondria and harbor a sparse mitochondrial infrastructure with immature cristae and limited perinuclear localization [2]. ESCs are routinely cultured under atmospheric (20%) oxygen tensions; however these cells are originally derived from embryos which reside in a 3-5% oxygen (hypoxic) environment. However the mechanism is just partly understood, emerging pieces of evidence suggest that such hypoxic conditions in cell culturing increase the quality of stemness [7, 8] and promotes the reprogramming of differentiated cells to induced pluripotent stem cells [9]. Under low oxygen tension hypoxia-inducible factors (HIFs), key transcription factors in hypoxia, are activated and directly regulate transcription of genes involved in self-renewal [10]. Different isoforms of the HIF-family are also known to regulate the transcription of glycolytic enzymes. For instance, HIF1 upregulates the expression of pyruvate dehydrogenase kinase (PDK), which is in turn, inhibits glucose oxidation [11]. Indeed, human ESCs cultured at 5% O2 consume significantly more glucose, less pyruvate and produce more lactate compared to those maintained at 20% O2 [12]. Comparison of ESCs and cells from early stages of differentiation has identified differences in lipid metabolism as well. Stem cells show a global enrichment of unsaturated fatty acid metabolites in the pluripotent state. While high levels of unsaturated fatty acids preserve pluripotency, addition of saturated metabolites to the ESCs is known to accelerate the differentiation program [13]. A recent study nicely demonstrates that lipid biosynthesis underlies the proliferative capacity of stem and progenitor cells in the brain [14]. Another recently discovered example of unique metabolic feature of stem cell is related to the distinct mode of amino acid catabolism. Threonine dehydrogenase (Tdh), an enzyme that catalyzes threonine oxidation (Thr), shows more than 200 times higher expression in ESCs. The restriction of Thr or inhibition of Tdh results in decreased accumulation of SAdenosylMethionine (SAM) that leads to decreased trimethylation of histone 3 lysine 4 (H3K4me3) and thus abolished stem cell growth [15].

3

Page 3 of 25

us

Are nuclear receptors regulating metabolism in stem cells?

cr

ip t

The above listed examples (summarized in Figure 1) strongly suggest that metabolic signals most likely play an essential role in regulation of stemness, pluripotency and cell fate decisions and the regulation of these signaling pathways are essential for proper embryonic development. A central question is how embryonic stem cells sense, regulate and respond to these environmental cues. Several lines of evidence suggest that a family of transcription factors, called nuclear receptors, might play important roles in recognizing different signals and convert it to transcriptional outcome and epigenetic changes. A goal of this review is to provide insight into the metabolic regulatory role of nuclear receptors in stem cell biology with an outlook to cancer stem cells.

an

The human and mouse genomes encode 48 and 49 nuclear receptors, respectively. Intense research on NRs in the last two decades has uncovered diverse roles of these receptors in developmental biology and metabolism [16].

Ac ce pt e

d

M

Nuclear receptors are ligand activated transcription factors. With a few exceptions, such as DAX1 and SHP1, all NRs expressed in vertebrates share a common domain structure. A highly conserved DNA-binding domain (DBD) ensure the binding to receptor-specific DNA sequences, while a less conserved ligand-binding domain (LBD) determines the signal specific action. Several small lipophilic molecule are known to function through NRs, however still a large fraction of nuclear receptors, often called orphan receptors, has no identified ligand. Based on their ligands and mechanistic action NRs are diveded into four groups: 1, steroid receptors 2, retinoid X receptor heterodimers 3, dimeric orphan receptors 4, monomeric orphan receptors (for detailes on grouping and nomenclature visit the website of NURSA (www.nursa.org)). DAX-1, LRH-1, SF1 and ESRR are linked directly to the maintenance of pluripotency in mESCs (reviewed in [17]) and recent genome-wide studies allow now a deeper insight into their regulatory role. A systematic profiling of NRs [18] and our independent gene expression characterization of mESCs (re-analysis of microarray data published by [19]) reveal in addition the high expression of TR4, NGF1 , LXR , RAR and RAR in undifferentiated ESCs (Figure 2). The importance of these nuclear receptors in stem cell biology is largely unknown. We will here summarize how DAX-1, LRH-1 and ESRR might regulate pluripotency and differentiation from a metabolic point of view. To further enhance the possible connection between these NRs, metabolism and pluripotency, we re-analyzed available genome-wide chromatin-immunoprecipitation data (ChIP-chip and ChIP-seq) and included several putative metabolic targets, that are likely to be regulated by these factors. Moreover, we sum up our recent knowledge on the TR4, NGF1 , LXR and RARs in the context of development and metabolism.

4

Page 4 of 25

DAX-1 (NR0B1) DAX-1 is a unique member of the NR family. Instead of binding to regulatory DNA sequences directly it appears to bind to other transcription factors and act mainly as a co-repressor [20].

us

cr

ip t

DAX-1 has a restricted expression pattern to the hypothalamic-pituitary-adrenal/gonadal-axis and was shown as an important regulator of steroidogenesis, gonadal development and sex determination [21]. Forming a pair with SF-1 (steroidogenic factor-1), an other NR family member, DAX-1 is playing a role in the transcriptional regulation of enzymes involved in conversion of cholesterol to steroid hormones [22]. The first indication of an essential role of DAX-1 in embryogenesis and pluripotency was revealed by the difficulty encountered in establishing DAX-1 knockout mice [23]. DAX-1 was identified as a highly expressed gene in ESCs and gene-silencing of DAX-1 was found to result loss of pluripotency of these cells [24].

Ac ce pt e

d

M

an

A recent analysis identified DAX-1 binding genomic regions in ESCs as part of the transcriptional network of pluripotency [25]. This data is valuable to get information how DAX-1 can regulate pluripotency. The list of putative DAX-1 regulated genes are available as a Supplement of the above manuscript. In agreement with the repressor function of DAX-1, several differentiation-related genes can be find on the list, such as members of the Hox-genes (Hoxa2, Hoxa9, Hoxa11, Hoxb4, Hoxc10 and Hoxc12), Wnt-signal (Wnt11, Wnt16, Wnt8a) and early lineage-specific markers (Pax6, Stra8, Foxa1, Foxd3, Lefty1, T, Rest). However, promoter region of stem cell marker Oct3/4 and Nanog are also bound by DAX-1, indicating its potential contextdependent activator function as well. The list contains several genes which are known to be involved in metabolic processes, such as Apoa2, Apoe, Atp5a1, Pgm2, Pgam1, Fabp3, etc. Importantly, members of stem cell related metabolic pathways are also represented in force. Glucose transporter (Slc2a3), much of glycolytic enzymes (Hk3, Gpi1, Pfkp, Gapdh, Pgk1, Ldhb, Pdhb), enzyme in pentose phosphate pathway (Pgls), nucleotide synthesis (Prps1), metabolism of threonine (Tdh, Gcat) and fatty acid synthesis (Hmgcr, Dhcr7) are all putative DAX-1 targets (Figure 1, Table 1). Based on these findings it is secure to say that DAX-1 has a largely uncharacterized metabolic regulatory function in stem cells.

LRH-1 (NR5A2)

Liver receptor homolog-1 (LRH-1) has been implicated in diverse biological processes, including bile acid metabolism, steroidogenesis and cell proliferation [26]. In adult, LRH-1 is known to be highly expressed in liver and promotes expression of genes involved in hepatic cholesterol uptake and efflux, HDL formation and fatty acid synthesis [27]. LRH-1 also controls the first step of hepatic glucose uptake through direct transcriptional regulation of the glucokinase gene. This suggests that LRH-1, at least in hepatocytes, acts as a critical regulatory component of the glucose-sensing system [28].

5

Page 5 of 25

ESRR

(NR3B2)

M

an

us

cr

ip t

Importantly, LRH-1 is also expressed during early mouse embryogenesis. LRH-1-/- embryos display growth retardation, epiblast disorganization, a mild embryonic-extraembryonic constriction, as well as abnormal thickening of the proximo-posterior epiblast [29]. LRH-1 has been identified as a key transcription factor in cellular reprogramming [30]. Similar to DAX-1, there is available genome-wide study for LRH-1, in which the LRH-1 occupied regions have been identified [31]. We re-analyzed this dataset and annotated the LRH-1 binding sites to the closest genes. Considering the limitations of this type of analysis, we performed a prediction of enriched pathways (Table 2). Several genes, involved in various metabolic processes, seem to be regulated directly by LRH-1. Genes encoding enzymes of glycolysis and gluconeogenesis (Hk2, Pfkm, Pgam2, Ldha, Ldhc, Aldh3a2, Aldh9a1), pentose phosphate pathway (H6pd, Rpia), fatty acid biosynthesis (Acacb, Acaca) or fatty acid metabolism (Aldh3a2, Aldh9a1, Acsl1, Cyp4a31, Hadh) are all possess LRH-1 occupied region in the close proximity (Figure 1, Table 1). Geneexpression studies combined with LRH-1 ChIP data will complete our understanding on the LRH-1-dependent regulatory mechanisms in ESCs. Inverse agonists that have been recently described for the LRH-1 [32], might also accelerate discoveries related to biological function of this nuclear receptor.

Ac ce pt e

d

There are three isoforms of the ESRR genes in mouse and human cells: ESRR , and . ESRRs share a homology with estrogen receptors (ERs), but ESRRs do not bind estrogen or other known physiological ligands [33]. ESRRs are typically expressed in tissues with high metabolic demands and they have a predominant role in orchestrating mitochondrial biogenesis and cellular energy metabolism such as oxidative phosphorylation (OxPhos), tricarboxylic acid (TCA) cycle, fatty acid oxidation and ATP synthesis. ESRRs directly acivate transcription of genes involved in these metabolic processes [34, 35]. In ESCs only one isoform, ESRR shows high expression (Figure 2). However, this one seems to be indispensable for pluripotency [36]. Expression of ESRR is regulated by Nanog, a key transcription factor of ES cells [37]. Loss of ESRR results spontaneous differentiation of ESCs while the overexpression of ESRR has been shown to promote reprogramming of mouse embryonic fibroblasts to induced pluripotent stem cells [38]. Only limited information is available why ESRR is so important in the maintenance of pluripotency. Recent genome-wide studies identified the binding sites of ESRR in mouse embryonic stem cells. The analysis revealed that ESRR -binding sites partly overlap with the Nanog-Oct3/4-Sox2 transcription factor binding sites [39], however large fraction of binding sites are independent from the core complex. ESRR was previously suggested to bind as a monomer [40]. In contrast, a detailed analysis identified three equally enriched full-site motifs, DR0, DR5, and DR8, but no half-site enrichment, suggesting that ESRR is likely to bind in a form of dimer to DNA [41]. So far, ESRR ChIP-seq data has not been analyzed in a deeper biological context. To get a preliminary

6

Page 6 of 25

cr

ip t

insight into the ESRR -mediated functions in ESCs, we re-analyzed an available ChIP-seq data [39] and in Table 3 summarized the top biological functions identified for the putative ESRR regulated genes. This clearly indicates that ESRR beside the regulation of pluripotency, plays a role in the regulation of metabolic processes. Indeed, much of the glycolytic, lipid- and amino acid-related enzymes that are important in stemness, are seems to be regulated by ESRR (Figure 1, Table 1). So far, this is the first indication that ESRR regulate the metabolic state of ESCs and raise the possibility of an ESRR -dependent metabolic regulation of stem cell pluripotency. However, these preliminary indications of ESRR mediated regulation of stem cell metabolism will requires additional analysis and validation.

us

TR4 (NR2C2)

d

M

an

Our genome-wide gene expressional data suggest that TR4 is expressed at a higher level in stem cells (Figure 2) and a recent study implicated also its functional importance in pluripotency [42]. Importantly, TR4 -/- mice display significant growth retardation [43]. In addition, gene disruption results in defects of spermatogenesis, in testis and in cerebellar development. Importantly, TR4 is expressed in metabolic tissues, including liver, muscle, brown and white adipose tissues and regulate metabolic genes, such as apolipoprotein E, enoyl-CoA hydratase, acyl-CoA oxidase [44, 45]. Moreover, TR4 also regulates phosphoenolpyruvate carboxykinase (PEPCK), a key enzyme controlling the rate of gluconeogenesis [46]. Thus, its high expression in stem cell might be essential in the regulation of metabolism via controlling mainly carbohydrate metabolism.

NGF1

Ac ce pt e

A recent study has determined the TR4 binding regions in four different cell types [47]. The majority of TR4 targets are shared between the different cell types and responsible for RNA metabolism and protein translation. However, cell-type specific TR4 binding could be also observed and these results further confirmed that TR4 is regulate genes involved in cellular metabolism. Genome-wide studies of TR4 binding in ESCs are not available yet. Loss of function experiments will be also required to identify the TR4 regulated genes in ESCs and in early embryonic differentiation. This might be helpful to understand the phenotypic alterations of the TR4-/- animals as well.

(NR4A1)

The NR4A family includes three highly homologous isoforms, NR4A1, NR4A2 and NR4A1, also known as Nur77, Nurr1 and Nor1, respectively. X-ray crystallography studies suggest that the ligand-binding pocket of these receptors is unable to accommodate ligands [48]. Previous studies have pointed to NR4A1 as a transcriptional regulator of glucose metabolism in liver and skeletal muscle [49]. Moreover, later studies demonstrated that Nur77 also regulates hepatic cholesterol metabolism [50]. Nur77 modulates lipid metabolism in hepatocytes by 7

Page 7 of 25

decreasing the expression of SREBP1c, which is a master regulator of triglyceride metabolism. In the context of developmental biology, Nur77 has been shown to be involved in cellular differentiation, apoptosis, proliferation, T-cell function.

cr

ip t

Higher expression level of NGF1 in ESCs is also indicated by our data (Figure 2). However, it is still very tentative to suggest NR4A1 as a metabolic regulator of stemness. ChIP-seq experiments in combination with transcriptome analyses will establish how Nur77 modulates the expression profile of a specific cell-type under defined conditions.

RAR alpha and gamma (NR1B1 and NR1B3)

LXR

Ac ce pt e

d

M

an

us

Three genes code for receptors that functionally bind retinoic acid (RA): RAR , and . Retinoic acid (RA) acts as an important signaling molecule during embryonic development and is commonly used for induction of in vitro differentiation of ESCs [51]. Ligands for RAR activation are being synthetized upon LIF removal and induction of differentiation [52]. These formally suggest that RARs, present in ESCs, should fulfill rather an inhibitory role than activator function in transcription. Indeed, a well-accepted model is that in the absence of ligand RAR and RXR dimers bind to DNA and interact with co-repressors to repress transcriptional events. Active repression by unliganded RARs has been proposed in development [53]. To understand the function of unliganded RARs is stem cells, genome-wide characterization of RAR genomic binding has been carried out [54]. Analysis of the RAR-bound sites shows that among the enzymes summarized in Figure 1 and Table 1, only Ldhb and Pdk1 seem to have RAR occupied promoter region. Validation of these targets will be required to confirm the putative role of RARs in the metabolic regulation of undifferentiated ESCs.

(NR1H2)

The liver X receptors form permissive heterodimers with retinoid X receptors (RXRs) and are important regulators of lipid metabolism [55, 56]. The LXR family consists of the two subtypes: LXR and LXR . LXR and LXR are detected in the liver starting at 11.5 days postcoitum [57]. Later, LXR expression remains high in organs involved in lipid homeostasis, such as liver, intestine, and brown adipose tissue, whereas LXR is more ubiquitously expressed and enriched in tissues of neuronal and endocrine origin. LXR -deficient mice are viable and show minor metabolic differences as compared to the wild-type [58]. As a remarkable difference, reduced fertility has been shown [59]. Oxysterols, oxidative derivatives of cholesterol has been identified as physiological ligands for LXRs, thus LXRs seems to act as a cholesterol sensors. Treatment of rodents with synthetic LXR ligands results in decreased hepatic gluconeogenesis and increased lipogenesis, indicating that

8

Page 8 of 25

LXR serves as a transcription factor that integrates liver carbohydrate and lipid metabolism [60]. A recent genome-wide study has identified LXR and RXR binding regions in mouse liver [61].

ip t

Taken together these results suggest that however Lxr is expressed in ESCs and might regulate metabolic pathways, it is unlikely that would be essential for pluripotency. In the same time, experimental evidence is required to confirm this statement.

Cancer stem cells and metabolism

M

an

us

cr

Numerous studies have revealed the existence of crosstalk between various nuclear receptor signaling pathways. One interesting example in the context of metabolism and differentiation is the potential link between LXR-dependent cholesterol metabolism and retinoic acid biosynthesis. Cholesterol metabolites, predominantly the oxysterols were identified that induces the expression of retinal dehydrogenases (RALDHs) via upregulation of sterol regulatory element binding protein-1c (SREBP1c) [62]. RALDHs synthesize retinoic acid (RA) from retinal [63]. RA is known to critically important for embryogenesis and also involved in proliferation, differentiation and apoptosis. However this crosstalk was mainly characterized in liver cells results in the same study also raise a similar mechanism in embryonic carcinoma cells. Existence of such a crosstalk between LXR and RAR signaling might play important roles in the metabolic switch when stem cell differentiation starts.

Ac ce pt e

d

The pluripotent nature of various cancers has been recognized early on [64]. Overall, there are two major cancer development theories as far as stem cell involvement is concerned: origin from cancer stem cells (CSCs) and de-differentiation of mature cells. The first one has been published by Furth et al., suggesting that a single cell from a tumor can initiate a new tumor in a recipient mouse [64]. Since then several studies have been performed to investigate the theory, which in principle states that the different tumor populations arise from a delimitated number of cells which conserves the same characteristics than stem cells in adult tissues [65]. CSCs can occur in diverse solid tumors, such as glioblastomas and breast cancer, but their existence has been reported in acute myeloid leukemias (AML) as well [65, 66]. A number of markers has been used for the isolation of CSCs, such as CD133 or SSEA-1, however difficulties of isolating pure populations of CSCs are still limiting our understanding of these cell populations. Elucidation of the pathways that regulate the maintenance and survival of CSCs is important for the development of novel therapies. Metabolic pathways have recently been implicated in governing the function of CSCs. In general, cancer cells have very similar metabolic features compared to ESCs [67]. Glucose metabolism is typically characterized by a shift from aerobic, oxidative metabolism to a glycolytic pathway. This aerobic glycolysis phenotype is known as the Warburg effect [68]. Metabolic switch to aerobic glycolysis is driven primarily by oncogenic signals and transcription factors. HIF1 is one of the most studied one, either the intratumoral hypoxic condition or genetic defects results in its stabilization and downstream regulatory

9

Page 9 of 25

function [69]. Identification of additional factors that play a role in metabolic switch of cancer cell is one of the hot topics in research.

Nuclear receptors as potential targets in cancer (stem) cell metabolism

cr

ip t

NRs have been extensively studied and linked directly to pathogenesis and treatment of various cancers; however their expression profile and putative role in cancer stem cells remained completely unknown. Importantly, CSCs have ESC-like transcriptional programs [70], thus detection of the above discussed pluripotency-related NRs, such as ESRR , LRH-1, DAX-1 are not unexpected in CSCs.

Ac ce pt e

d

M

an

us

Interestingly, many of these NRs have been already linked to poor prognosis of cancer. LRH-1 was shown to promote motility and invasiveness in breast cancer cells [71] and regulate steroidogenic gene transcription in endometrial cancer cells [72]. LRH-1 transcription is also activated in human pancreatic cancer cells [73]. The other stem cell specific nuclear receptor DAX-1 has been also recognized as a putative target in cancer treatment [74]. It is a direct target of the EWS/FLI1 oncoprotein, and found to be highly expressed in Ewing’s tumors. Genes implicated in cell cycle progression are regulated by DAX-1. ESRR , and are identified coactivating factors of HIF and thus might directly modulate HIF-mediated gene transcription of cancer cells. Further studies are required to helpus understand which pathways are regulated by NRs in cancer cells and CSCs. A recent study applied ChIP-seq to map the in vivo genome-wide binding (cistrome) of NRs in both normal and cancer cells [41]. Such studies combined with gene expression data will bring us closer to understand how NRs mechanistically contribute to cancer development. The above stated important role of NRs in cancer biology and their well established role in metabolism pose many interesting questions: most importantly whether NRs play any role in cancer cells to sense and regulate the level of nutrients to facilitate tumor development and progression. There are growing number of pieces of evidence indicating a direct relationship between NRs, metabolism and cancer. Anti-estrogen therapy for breast cancer was the first targeted therapy used in any type of cancer. Since its discovery, several metabolic pathways affected by anti- estrogens in cancer cells has been identified [75]. Activation of LXR signaling by oxysterols is an other example of how NR ligands can be used to target metabolic pathways in the treatment of various cancers [76]. Oxysterols are known to interfere with proliferation and initiate cell death of many cancer cells. Oxysterols and LXRsignaling might also help to treat “incurable” conditions, such as glioblastoma multiforme (GBM). GBM, a WHO grade IV astrocytoma, is the most aggressive and common primary brain tumor in adults. The median survival of GBM patients is approximately 1 year. Importantly, LXR

10

Page 10 of 25

agonists have been shown to inhibit GBM growth and promote tumor cell death, and LXR was initiated as a novel antitumor agents in glioblastoma [77, 78]. Role of NR agonists has been also studied and their roles confirmed in the metabolism and gene expression regulation of breast cancer stem cells [79, 80].

an

us

cr

ip t

A proof of principle for NR-mediated cancer therapy is the use of all-trans retinoic acid (atRA) treatment in AML. In AML the presence of leukemia stem cells has been demonstrated. It has been shown that they are hierarchically organized due to the heterogeneity in their self-renewal capacity and in longevity of the produced clone [81-83]. In this context, Alcalay et al., found that expressing some fusion proteins in myeloid precursors, previously observed in murine models of AML which includes the transcription factor RAR , the gene expression pattern was congruent with the induction of genes related to the maintenance of the stem cell phenotype and repression of DNA repair genes. Importantly, metabolism was one of the functional classifications of AMLfusion protein target genes and some of the identified genes could be validated in patient samples [84].

Conclusions

M

These examples provide evidence that targeting metabolic pathways through the activation of NRs may represent effective strategies for eradicating CSCs.

Ac ce pt e

d

Recent studies are just beginning to show the importance of metabolism in cell fate determination, including cancer development. Role of nuclear receptors are still poorly characterized in the metabolic regulation of pluripotency. The recent development of highthroughput sequencing methodologies such as RNA-seq and ChIP-seq combined with metabolomics analyses holds the promise to provide more accurate gene expression measurements and deeper mechanistic insights into the regulatory role of nuclear receptors in cellular metabolism. This knowledge can be potentially turned into novel therapeautic approaches later.

Acknowledgement

The authors would like to acknowledge Erik Czipa and Dr. Endre Barta for the contibution in data analyzis, and the Nagy laboratory for discussions and comments on the manuscript. L.N is supported by a grant from the Hungarian Scientific Research Fund (OTKA K100196), and TÁMOP422_2012_0023 VÉD-ELEM implemented through the New Hungary Development Plan co-financed by the European Social Fund and the European Regional Development Fund. Z.S is a recipient of TÁMOP-422/B10/1_2010_0024 grant.

11

Page 11 of 25

References

[1] Lu C, Thompson CB. Metabolic regulation of epigenetics. Cell Metab 2012;16:9-17.

ip t

[2] Chung S, Dzeja PP, Faustino RS, Perez-Terzic C, Behfar A, Terzic A. Mitochondrial oxidative metabolism is required for the cardiac differentiation of stem cells. Nat Clin Pract Cardiovasc Med 2007;4 Suppl 1:S60-7.

us

cr

[3] Cho YM, Kwon S, Pak YK, Seol HW, Choi YM, Park do J, et al. Dynamic changes in mitochondrial biogenesis and antioxidant enzymes during the spontaneous differentiation of human embryonic stem cells. Biochem Biophys Res Commun 2006;348:1472-8.

[4] Wang J, Alexander P, Wu L, Hammer R, Cleaver O, McKnight SL. Dependence of mouse embryonic stem cells on threonine catabolism. Science 2009;325:435-9.

an

[5] Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature 1981;292:154-6.

M

[6] Folmes CD, Dzeja PP, Nelson TJ, Terzic A. Metabolic plasticity in stem cell homeostasis and differentiation. Cell Stem Cell 2012;11:596-606.

d

[7] Ezashi T, Das P, Roberts RM. Low O2 tensions and the prevention of differentiation of hES cells. Proc Natl Acad Sci U S A 2005;102:4783-8.

Ac ce pt e

[8] Westfall SD, Sachdev S, Das P, Hearne LB, Hannink M, Roberts RM, et al. Identification of oxygen-sensitive transcriptional programs in human embryonic stem cells. Stem Cells Dev 2008;17:869-81. [9] Yoshida Y, Takahashi K, Okita K, Ichisaka T, Yamanaka S. Hypoxia enhances the generation of induced pluripotent stem cells. Cell Stem Cell 2009;5:237-41. [10] Covello KL, Kehler J, Yu H, Gordan JD, Arsham AM, Hu CJ, et al. HIF-2alpha regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth. Genes Dev 2006;20:557-70. [11] Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab 2006;3:177-85. [12] Forristal CE, Christensen DR, Chinnery FE, Petruzzelli R, Parry KL, Sanchez-Elsner T, et al. Environmental oxygen tension regulates the energy metabolism and self-renewal of human embryonic stem cells. PLoS One 2013;8:e62507.

12

Page 12 of 25

[13] Yanes O, Clark J, Wong DM, Patti GJ, Sanchez-Ruiz A, Benton HP, et al. Metabolic oxidation regulates embryonic stem cell differentiation. Nat Chem Biol 2010;6:411-7.

ip t

[14] Knobloch M, Braun SM, Zurkirchen L, von Schoultz C, Zamboni N, Arauzo-Bravo MJ, et al. Metabolic control of adult neural stem cell activity by Fasn-dependent lipogenesis. Nature 2013;493:226-30.

cr

[15] Shyh-Chang N, Locasale JW, Lyssiotis CA, Zheng Y, Teo RY, Ratanasirintrawoot S, et al. Influence of threonine metabolism on S-adenosylmethionine and histone methylation. Science 2013;339:222-6.

us

[16] Sonoda J, Pei L, Evans RM. Nuclear receptors: decoding metabolic disease. FEBS Lett 2008;582:2-9.

an

[17] Wagner RT, Cooney AJ. Minireview: the diverse roles of nuclear receptors in the regulation of embryonic stem cell pluripotency. Mol Endocrinol 2013;27:864-78. [18] Xie CQ, Jeong Y, Fu M, Bookout AL, Garcia-Barrio MT, Sun T, et al. Expression profiling of nuclear receptors in human and mouse embryonic stem cells. Mol Endocrinol 2009;23:724-33.

M

[19] Simandi Z, Balint BL, Poliska S, Ruhl R, Nagy L. Activation of retinoic acid receptor signaling coordinates lineage commitment of spontaneously differentiating mouse embryonic stem cells in embryoid bodies. FEBS Lett 2010;584:3123-30.

Ac ce pt e

d

[20] Sablin EP, Woods A, Krylova IN, Hwang P, Ingraham HA, Fletterick RJ. The structure of corepressor Dax-1 bound to its target nuclear receptor LRH-1. Proc Natl Acad Sci U S A 2008;105:18390-5. [21] Guo W, Burris TP, McCabe ER. Expression of DAX-1, the gene responsible for X-linked adrenal hypoplasia congenita and hypogonadotropic hypogonadism, in the hypothalamicpituitary-adrenal/gonadal axis. Biochem Mol Med 1995;56:8-13. [22] Iyer AK, McCabe ER. Molecular mechanisms of DAX1 action. Mol Genet Metab 2004;83:60-73. [23] Achermann JC, Meeks JJ, Jameson JL. Phenotypic spectrum of mutations in DAX-1 and SF1. Mol Cell Endocrinol 2001;185:17-25. [24] Khalfallah O, Rouleau M, Barbry P, Bardoni B, Lalli E. Dax-1 knockdown in mouse embryonic stem cells induces loss of pluripotency and multilineage differentiation. Stem Cells 2009;27:1529-37. [25] Kim J, Chu J, Shen X, Wang J, Orkin SH. An extended transcriptional network for pluripotency of embryonic stem cells. Cell 2008;132:1049-61.

13

Page 13 of 25

[26] Pare JF, Malenfant D, Courtemanche C, Jacob-Wagner M, Roy S, Allard D, et al. The fetoprotein transcription factor (FTF) gene is essential to embryogenesis and cholesterol homeostasis and is regulated by a DR4 element. J Biol Chem 2004;279:21206-16.

ip t

[27] Fayard E, Auwerx J, Schoonjans K. LRH-1: an orphan nuclear receptor involved in development, metabolism and steroidogenesis. Trends Cell Biol 2004;14:250-60.

cr

[28] Oosterveer MH, Mataki C, Yamamoto H, Harach T, Moullan N, van Dijk TH, et al. LRH-1dependent glucose sensing determines intermediary metabolism in liver. J Clin Invest 2012;122:2817-26.

us

[29] Labelle-Dumais C, Jacob-Wagner M, Pare JF, Belanger L, Dufort D. Nuclear receptor NR5A2 is required for proper primitive streak morphogenesis. Dev Dyn 2006;235:3359-69.

an

[30] Wang W, Yang J, Liu H, Lu D, Chen X, Zenonos Z, et al. Rapid and efficient reprogramming of somatic cells to induced pluripotent stem cells by retinoic acid receptor gamma and liver receptor homolog 1. Proc Natl Acad Sci U S A 2011;108:18283-8.

M

[31] Heng JC, Feng B, Han J, Jiang J, Kraus P, Ng JH, et al. The nuclear receptor Nr5a2 can replace Oct4 in the reprogramming of murine somatic cells to pluripotent cells. Cell Stem Cell 2009;6:167-74.

d

[32] Busby S, Nuhant P, Cameron M, Mercer BA, Hodder P, Roush WR, et al. Discovery of Inverse Agonists for the Liver Receptor Homologue-1 (LRH1; NR5A2). 2010.

Ac ce pt e

[33] Giguere V, Yang N, Segui P, Evans RM. Identification of a new class of steroid hormone receptors. Nature 1988;331:91-4. [34] Poidatz D, Dos Santos E, Brule A, De Mazancourt P, Dieudonne MN. Estrogen-related receptor gamma modulates energy metabolism target genes in human trophoblast. Placenta 2012;33:688-95. [35] Kim DK, Ryu D, Koh M, Lee MW, Lim D, Kim MJ, et al. Orphan nuclear receptor estrogen-related receptor gamma (ERRgamma) is key regulator of hepatic gluconeogenesis. J Biol Chem 2012;287:21628-39. [36] Martello G, Sugimoto T, Diamanti E, Joshi A, Hannah R, Ohtsuka S, et al. Esrrb is a pivotal target of the Gsk3/Tcf3 axis regulating embryonic stem cell self-renewal. Cell Stem Cell 2012;11:491-504. [37] Festuccia N, Osorno R, Halbritter F, Karwacki-Neisius V, Navarro P, Colby D, et al. Esrrb is a direct Nanog target gene that can substitute for Nanog function in pluripotent cells. Cell Stem Cell 2012;11:477-90.

14

Page 14 of 25

[38] Feng B, Jiang J, Kraus P, Ng JH, Heng JC, Chan YS, et al. Reprogramming of fibroblasts into induced pluripotent stem cells with orphan nuclear receptor Esrrb. Nat Cell Biol 2009;11:197-203.

ip t

[39] Chen X, Xu H, Yuan P, Fang F, Huss M, Vega VB, et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 2008;133:1106-17.

cr

[40] Sem DS, Casimiro DR, Kliewer SA, Provencal J, Evans RM, Wright PE. NMR spectroscopic studies of the DNA-binding domain of the monomer-binding nuclear orphan receptor, human estrogen related receptor-2. The carboxyl-terminal extension to the zinc-finger region is unstructured in the free form of the protein. J Biol Chem 1997;272:18038-43.

us

[41] Tang Q, Chen Y, Meyer C, Geistlinger T, Lupien M, Wang Q, et al. A comprehensive view of nuclear receptor cancer cistromes. Cancer Res 2011;71:6940-7.

an

[42] Shyr CR, Kang HY, Tsai MY, Liu NC, Ku PY, Huang KE, et al. Roles of testicular orphan nuclear receptors 2 and 4 in early embryonic development and embryonic stem cells. Endocrinology 2009;150:2454-62.

M

[43] Collins LL, Lee YF, Heinlein CA, Liu NC, Chen YT, Shyr CR, et al. Growth retardation and abnormal maternal behavior in mice lacking testicular orphan nuclear receptor 4. Proc Natl Acad Sci U S A 2004;101:15058-63.

Ac ce pt e

d

[44] Kim E, Xie S, Yeh SD, Lee YF, Collins LL, Hu YC, et al. Disruption of TR4 orphan nuclear receptor reduces the expression of liver apolipoprotein E/C-I/C-II gene cluster. J Biol Chem 2003;278:46919-26. [45] Yan ZH, Karam WG, Staudinger JL, Medvedev A, Ghanayem BI, Jetten AM. Regulation of peroxisome proliferator-activated receptor alpha-induced transactivation by the nuclear orphan receptor TAK1/TR4. J Biol Chem 1998;273:10948-57. [46] Liu NC, Lin WJ, Kim E, Collins LL, Lin HY, Yu IC, et al. Loss of TR4 orphan nuclear receptor reduces phosphoenolpyruvate carboxykinase-mediated gluconeogenesis. Diabetes 2007;56:2901-9. [47] O'Geen H, Lin YH, Xu X, Echipare L, Komashko VM, He D, et al. Genome-wide binding of the orphan nuclear receptor TR4 suggests its general role in fundamental biological processes. BMC Genomics 2010;11:689. [48] Flaig R, Greschik H, Peluso-Iltis C, Moras D. Structural basis for the cell-specific activities of the NGFI-B and the Nurr1 ligand-binding domain. J Biol Chem 2005;280:19250-8. [49] Chao LC, Wroblewski K, Zhang Z, Pei L, Vergnes L, Ilkayeva OR, et al. Insulin resistance and altered systemic glucose metabolism in mice lacking Nur77. Diabetes 2009;58:2788-96.

15

Page 15 of 25

[50] Zhang P, Hu Y, Yang J, Zheng L, Wang Q. The orphan nuclear receptor Nur77 regulates hepatic cholesterol metabolism through the suppression of LDLR and HMGCR expression. Mol Med Rep 2012;5:1541-7.

ip t

[51] Soprano DR, Teets BW, Soprano KJ. Role of retinoic acid in the differentiation of embryonal carcinoma and embryonic stem cells. Vitam Horm 2007;75:69-95.

cr

[52] Lane MA, Chen AC, Roman SD, Derguini F, Gudas LJ. Removal of LIF (leukemia inhibitory factor) results in increased vitamin A (retinol) metabolism to 4-oxoretinol in embryonic stem cells. Proc Natl Acad Sci U S A 1999;96:13524-9.

us

[53] Weston AD, Blumberg B, Underhill TM. Active repression by unliganded retinoid receptors in development: less is sometimes more. J Cell Biol 2003;161:223-8.

an

[54] Delacroix L, Moutier E, Altobelli G, Legras S, Poch O, Choukrallah MA, et al. Cell-specific interaction of retinoic acid receptors with target genes in mouse embryonic fibroblasts and embryonic stem cells. Mol Cell Biol 2010;30:231-44.

M

[55] Willy PJ, Umesono K, Ong ES, Evans RM, Heyman RA, Mangelsdorf DJ. LXR, a nuclear receptor that defines a distinct retinoid response pathway. Genes Dev 1995;9:1033-45. [56] Calkin AC, Tontonoz P. Transcriptional integration of metabolism by the nuclear sterolactivated receptors LXR and FXR. Nat Rev Mol Cell Biol 2012;13:213-24.

Ac ce pt e

d

[57] Annicotte JS, Schoonjans K, Auwerx J. Expression of the liver X receptor alpha and beta in embryonic and adult mice. Anat Rec A Discov Mol Cell Evol Biol 2004;277:312-6. [58] Alberti S, Schuster G, Parini P, Feltkamp D, Diczfalusy U, Rudling M, et al. Hepatic cholesterol metabolism and resistance to dietary cholesterol in LXRbeta-deficient mice. J Clin Invest 2001;107:565-73. [59] Steffensen KR, Robertson K, Gustafsson JA, Andersen CY. Reduced fertility and inability of oocytes to resume meiosis in mice deficient of the Lxr genes. Mol Cell Endocrinol 2006;256:9-16. [60] Laffitte BA, Chao LC, Li J, Walczak R, Hummasti S, Joseph SB, et al. Activation of liver X receptor improves glucose tolerance through coordinate regulation of glucose metabolism in liver and adipose tissue. Proc Natl Acad Sci U S A 2003;100:5419-24. [61] Boergesen M, Pedersen TA, Gross B, van Heeringen SJ, Hagenbeek D, Bindesboll C, et al. Genome-wide profiling of liver X receptor, retinoid X receptor, and peroxisome proliferatoractivated receptor alpha in mouse liver reveals extensive sharing of binding sites. Mol Cell Biol 2011;32:852-67.

16

Page 16 of 25

[62] Huq MD, Tsai NP, Gupta P, Wei LN. Regulation of retinal dehydrogenases and retinoic acid synthesis by cholesterol metabolites. EMBO J 2006;25:3203-13.

ip t

[63] Wang X, Penzes P, Napoli JL. Cloning of a cDNA encoding an aldehyde dehydrogenase and its expression in Escherichia coli. Recognition of retinal as substrate. J Biol Chem 1996;271:16288-93. [64] Clevers H. The cancer stem cell: premises, promises and challenges. Nature medicine 2011;17:313-9.

cr

[65] Jordan CT, Guzman ML, Noble M. Cancer stem cells. The New England journal of medicine 2006;355:1253-61.

us

[66] Magee JA, Piskounova E, Morrison SJ. Cancer stem cells: impact, heterogeneity, and uncertainty. Cancer Cell 2012;21:283-96.

an

[67] Quail DF, Taylor MJ, Postovit LM. Microenvironmental regulation of cancer stem cell phenotypes. Curr Stem Cell Res Ther 2012;7:197-216.

M

[68] Warburg O. On the origin of cancer cells. Science 1956;123:309-14.

d

[69] Gonzalez IR, Moreno-Manzano V, Rodriguez-Jimenez FJ, Sepulveda P, Sanchez-Puelles JM. The biology of HIFalpha proteins in cell differentiation and disease. Vitam Horm 2011;87:367-79.

Ac ce pt e

[70] Visvader JE, Lindeman GJ. Cancer stem cells: current status and evolving complexities. Cell Stem Cell 2012;10:717-28. [71] Chand AL, Herridge KA, Thompson EW, Clyne CD. The orphan nuclear receptor LRH-1 promotes breast cancer motility and invasion. Endocr Relat Cancer 2010;17:965-75. [72] Dube C, Bergeron F, Vaillant MJ, Robert NM, Brousseau C, Tremblay JJ. The nuclear receptors SF1 and LRH1 are expressed in endometrial cancer cells and regulate steroidogenic gene transcription by cooperating with AP-1 factors. Cancer Lett 2009;275:127-38. [73] Benod C, Vinogradova MV, Jouravel N, Kim GE, Fletterick RJ, Sablin EP. Nuclear receptor liver receptor homologue 1 (LRH-1) regulates pancreatic cancer cell growth and proliferation. Proc Natl Acad Sci U S A 2011;108:16927-31. [74] Lalli E, Alonso J. Targeting DAX-1 in embryonic stem cells and cancer. Expert Opin Ther Targets 2010;14:169-77. [75] Hah N, Kraus WL. Hormone-regulated transcriptomes: Lessons learned from estrogen signaling pathways in breast cancer cells. Mol Cell Endocrinol 2013.

17

Page 17 of 25

[76] de Weille J, Fabre C, Bakalara N. Oxysterols in cancer cell proliferation and death. Biochem Pharmacol 2013;86:154-60.

ip t

[77] Guo D, Reinitz F, Youssef M, Hong C, Nathanson D, Akhavan D, et al. An LXR agonist promotes glioblastoma cell death through inhibition of an EGFR/AKT/SREBP-1/LDLRdependent pathway. Cancer Discov 2011;1:442-56. [78] Moschetta A. Nuclear receptor LXR as a novel therapeutic antitumoral target in glioblastoma. Cancer Discov 2011;1:381-2.

us

cr

[79] Papi A, Guarnieri T, Storci G, Santini D, Ceccarelli C, Taffurelli M, et al. Nuclear receptors agonists exert opposing effects on the inflammation dependent survival of breast cancer stem cells. Cell Death Differ 2012;19:1208-19.

an

[80] Nguyen-Vu T, Vedin LL, Liu K, Jonsson P, Lin JZ, Candelaria NR, et al. Liver X receptor ligands disrupt breast cancer cell proliferation through an E2F-mediated mechanism. Breast Cancer Res 2013;15:R51.

M

[81] Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994;367:645-8.

d

[82] Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997;3:730-7.

Ac ce pt e

[83] Hope KJ, Jin L, Dick JE. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nature immunology 2004;5:738-43. [84] Alcalay M, Meani N, Gelmetti V, Fantozzi A, Fagioli M, Orleth A, et al. Acute myeloid leukemia fusion proteins deregulate genes involved in stem cell maintenance and DNA repair. The Journal of clinical investigation 2003;112:1751-61.

Figure 1.

18

Page 18 of 25

ip t cr us an

Ac ce pt e

d

M

Figure 1. Metabolism in pluripotent embryonic stem cells (ESCs). Rapidly proliferating pluripotent stem cells has unique metabolism. I. ATP production: ATP synthesis is decoupled from O2 consumption and depends on glycolysis. Gylcolytic enzymes: hexokinase (HK), glucose6-phosphate isomerase (GPI), phosphofructokinase (PFK), aldolase (ALDO), triosephosphate isomerase (TPI), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), phosphoglycerate kinase (PGK), pyruvate dehydrogenase (PDH), lactate dehydrogenase (LDH). II. Nucleotide synthesis: Pentose phosphate pathway (Pentose PP) is active in ESCs and by converting glucose6-phospate (G6P) into ribose-5-phosphate for nucleotide synthesis, it also produces NADPH. Pentose PP enzymes among others: hexose-6-phosphate dehydrogenase (H6PD), 6phosphogluconolactonase (PGLS), ribose 5-phosphate isomerase A (RPIA), transketolase (TKT) Phosphoribosyl pyrophosphate synthetase 1 (PRPS1) catalyzes the phosphoribosylation of ribose 5-phosphate and necessary for purine metabolism. CAD encodes a trifunctional protein that is involved in pyrimidine nucleotide synthesis. III. Amino acid metabolism: ESCs requires the catabolism of L-threonine (Thr). The degradation of L-threonine to glycine consists of a two-step biochemical pathway involving the enzymes L-threonine dehydrogenase (Tdh) and 2-amino-3ketobutyrate coenzyme A ligase (GCAT). Glycine dehydrogenase (GLDC) is involved in the degradation of glycin. IV. Lipid metabolism: Unsaturated fatty acids preserve the pluripotency. 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) and 7-dehydrocholesterol reductase (DHCR7) are enzymes for cholesterol synthesis. V. Epigenetic regulation: Acetyl-CoA (AcCoA) and S-AdenosylMethionine (SAM) are substrates for histone acetylation and methylation, respectively VI. Hypoxia: hypoxic culture conditions results the stabilization of hypoxia inducible factor 1 alpha (HIF1 ) and enhance the transcription of pyruvate dehydrogenase kinase (Pdk). Pdk inhibits glucose oxidation. Out of all the shown enzymes, only the marked (*) ones are not regulated by DAX-1, LRH-1 and/or ESRR (see Table 1).

19

Page 19 of 25

M

an

us

cr

ip t

Figure 2.

Ac ce pt e

d

Figure 2. Per chip normalized signal intensity data of nuclear receptors in undifferentiated ESCs. Values of all entities and some known embryonic stem cell markers are also shown. Red line indicates the median value of all entities. One dot represents one gene. Gene symbols on the top of the figure indicate the dominantly expressed genes from each group and are considered to be expressed in stem cells. Nuclear receptors shown in red are discussed in the text.

Table 1. Summary of DAX-1, LRH-1 and ESRR Gene symbol

GLUT1/3

HK GPI

regulated metabolic genes DAX-1

LRH-1

ESRR

Yes

Yes

(Slc2a1)

(Slc2a1, a3)

Yes

Yes

Yes

(Hk3)

(Hk2)

(Hk1, 2, 3)

Facilitated glucose transporter, member Yes 1 (Slc2a3) Hexokinase glucose-6-phosphate isomerase

Yes

Yes

20

Page 20 of 25

(Gpi1)

(Gpi1)

Yes

Yes

Yes

(Pfkp)

(Pfkl, m, p)

(Pfkl, m, p)

phosphofructokinase

PFK

Yes

ip t

aldolase

ALDO

(Aldoa) Yes

PGK

phosphoglycerate kinase

Yes

us

glyceraldehyde-3-phosphate dehydrogenase

an

GAPDH

cr

triosephosphate isomerase 1

TPI1

LDH

M

Yes

lactate dehydrogenase

PGLS RPIA* TKT PRPS1

CAD*

hexose-6-phosphate dehydrogenase

Ac ce pt e

H6PD

d

(Ldh )

6-phosphogluconolactonase

Yes

(Tpi1)

Yes Yes (Pgk1)

Yes

Yes

(Ldh , Ldh )

(Ldh , Ldh )

Yes

Yes Yes

ribose 5-phosphate isomerase A transketolase

Yes

phosphoribosyl pyrophosphate synthetase 1

Yes

carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase Yes

PDH

pyruvate dehydrogenase complex (Pdhb)

PDK

pyruvate dehydrogenase kinase

Yes

Yes

21

Page 21 of 25

(Pdk1)

(Pdk1, 2, 4)

Yes

Yes

GLDC*

glycine dehydrogenase

TDH

L-threonine dehydrogenase

Yes

HMGCR

3-hydroxy-3-methylglutaryl-CoA reductase

Yes

DHCR7

7-dehydrocholesterol reductase

Yes

HIF

hypoxia inducible factor

Yes

Yes

ip t

glycine C-acetyltransferase

Yes

cr

GCAT

Yes

us

Yes

(HIF1 )

an

(HIF1 )

Yes

M

Table 2. Top 20 Biological Process GO Terms for genes annotated to LRH-1 (NR5A2) binding site(s). Enrichme nt

d

Term

Ac ce pt e

TermID

Genes in Term

Target Genes in Term

Fraction of Targets in Term

GO:003132 3

regulation of cellular metabolic 4.43E-34 process

3995

832

0.27

GO:001921 9

regulation of nucleobasecontaining compound metabolic process

1.91E-33

3072

673

0.22

GO:005117 1

regulation of nitrogen compound metabolic process

3.68E-33

3113

679

0.22

GO:000727 5

multicellular organismal development

5.78E-33

3492

743

0.24

GO:006025 5

regulation of macromolecule metabolic process

6.76E-33

3904

812

0.26

GO:008009 0

regulation of primary metabolic process

9.81E-33

3956

820

0.27

22

Page 22 of 25

positive regulation of cellular process

3.78E-32

3118

676

0.22

GO:001046 8

regulation of gene expression

6.55E-32

3135

678

0.22

GO:200011 2

regulation of cellular macromolecule biosynthetic process

1.69E-31

2820

622

GO:005125 2

regulation of RNA metabolic process

3.69E-31

2644

GO:003132 6

regulation of cellular biosynthetic process

8.12E-31

GO:003250 2

developmental process

1.42E-30

GO:001055 6

regulation of macromolecule biosynthetic process

1.56E-30

GO:000988 9

regulation of biosynthetic process

GO:004851 8

positive regulation of biological process

GO:000635 5

regulation of transcription, DNA-dependent

cr

ip t

GO:004852 2

us

590

0.20

0.19

654

0.21

3854

794

0.26

2893

631

0.20

3058

659

0.21

2.04E-30

3452

726

0.24

2.07E-30

2580

576

0.19

GO:001922 2

regulation of metabolic process 3.02E-30

4667

926

0.30

GO:200114 1

regulation of RNA biosynthetic 3.20E-30 process

2584

576

0.19

GO:004885 6

anatomical structure development

5.91E-30

3404

716

0.23

GO:001604 3

cellular component organization

4.43E-28

3172

669

0.22

M

an

3020

Ac ce pt e

d

2.00E-30

23

Page 23 of 25

Table 3. Top 20 Biological Process GO Terms for genes annotated to ESRR site(s).

Term

P-value

GO:004423 7

cellular metabolic process

1.21E-141

6331

GO:000815 2

metabolic process

3.70E-129

7476

GO:004423 8

primary metabolic process

1.17E-120

GO:003132 3

regulation of cellular metabolic 1.24E-112 process

GO:003250 2

developmental process

GO:004426 0

cellular macromolecule metabolic process

GO:004885 6

anatomical structure development

GO:000727 5

multicellular organismal development

Number in common

5120

Fraction of List

0.36

5912

0.42

us

cr

ip t

TermID

Numbe r in Term

(NR3B2) binding

5162

0.36

3813

3184

0.22

3670

3062

0.22

4637

3768

0.27

1.22E-99

3222

2707

0.19

5.24E-99

3323

2782

0.20

GO:001922 2

regulation of metabolic process 6.62E-99

4482

3648

0.26

GO:008009 0

regulation of primary metabolic process

1.19E-98

3772

3119

0.22

GO:004851 8

positive regulation of biological process

1.41E-89

3258

2710

0.19

GO:005117 1

regulation of nitrogen compound metabolic process

2.49E-88

2971

2490

0.18

GO:001921 9

regulation of nucleobasecontaining compound t b li

3.05E-88

2937

2464

0.17

an

6468

M

1.31E-106

Ac ce pt e

d

6.93E-101

24

Page 24 of 25

metabolic process cellular component organization

1.10E-87

2977

2493

0.18

GO:004317 0

macromolecule metabolic process

1.93E-86

5167

4114

0.29

GO:004873 1

system development

1.15E-85

2770

2331

GO:004852 2

positive regulation of cellular process

3.13E-85

2936

GO:005117 9

localization

3.16E-84

GO:007184 0

cellular component organization or biogenesis

2.48E-83

GO:006025 5

regulation of macromolecule metabolic process

5.49E-82

cr 2456

0.17

us

0.16

2767

0.19

3109

2582

0.18

3762

3067

0.22

an

3352

Ac ce pt e

d

M

ip t

GO:001604 3

25

Page 25 of 25

Nuclear receptors as regulators of stem cell and cancer stem cell metabolism.

Cellular metabolism is underpinning physiological processes in all cells. These include housekeeping functions as well as specific activities unique t...
396KB Sizes 0 Downloads 0 Views