Estrogen receptor-associated receptor α and peroxisome proliferator-activated receptor γ in metabolism and disease (Review)

  • Authors:
    • Wei-Yi Huang
    • Peng-Ming Sun
  • View Affiliations

  • Published online on: December 22, 2020     https://doi.org/10.3892/mmr.2020.11795
  • Article Number: 156
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Estrogen receptor‑associated receptor α (ERRα) is an orphan nuclear receptor that lacks corresponding ligands. ERRα recruits co‑regulators to regulate gene transcription and plays an important role in human physiological functions. Peroxisome proliferator‑activated receptor γ (PPARγ) is also a nuclear receptor that regulates the expression of target genes via a ligand‑dependent mechanism, thereby participating in a series of physiological processes. Both ERRα and PPARγ are involved in the process of energy metabolism and tumorigenesis. In the present review, a concise overview of the important roles governed by ERRα and PPARγ in metabolism and their association with various disease are provided.

Introduction

Estrogen receptor-associated receptor (ERR) is an orphan nuclear receptor that exerts its biological function without binding to a ligand. In 1988, Giguère et al (1) identified a nuclear receptor that was highly homologous with ERα in nucleotide and amino acid sequences using cDNA for the DNA-binding domain of estrogen receptor α (ERα) as the probe. Both ERR and ER are type III nuclear receptors. To date, the following three subtypes have been found, ERRα (NR3B1), ERRβ (NR3B2) and ERRγ (NR3B3), in which ERRα is widely distributed in various adult tissues and participates in a variety of physiological processes, including mitochondrial biogenesis (2), gluconeogenesis, oxidative phosphorylation (3), fatty acid metabolism (4) and brown adipose tissue thermogenesis (5). It was also identified as an important regulator of the mammalian circadian clock, and its output pathways at both transcriptional and physiological levels regulated the expression of transcription factors involved in metabolic homeostasis (6). The ERRα-encoding gene is located at site 11q13 of the human chromosome and primarily consists of the following three functional domains: N terminal domain (NTD), DNA-binding domain (DBD) and ligand binding domain (LBD). Activation function 1 (AF1) is located at the NTD, while AF2 is located at the LBD (7). The DBD of ERRα contains two zinc fingers, which are used for identification and binding of special sequences at the regulatory region in the DNA of the target gene (8). AF2 regulates the transcriptional activity of nuclear receptors, primarily through functional interactions with coactivators, such as peroxisome proliferator-activated receptor γ coactivator-1 (PGC-1), or corepressors, such as nuclear factor RIP140 (8).

Peroxisome proliferator-activated receptor (PPAR) is a novel steroid hormone receptor discovered by Issemann and Green (9) in 1990, which can be activated by fatty acid-like peroxisome proliferator. PPARs are nuclear transcription factors activated by ligands and members of the type II nuclear hormone receptor superfamily. There are three subtypes of PPARs: PPARα, β/δ and γ (10). Typically, PPARs and retinoid X receptors (RXR) form a heterodimer and recruit a co-inhibitory protein complex to inhibit the transcription of target genes (10). When PPARs are combined with ligands and activated, this heterodimer may release co-inhibitor proteins and bind to coactivator proteins, and subsequently combine with the promoter of the target gene, upstream peroxisome proliferator response element (PPRE), to regulate its transcription and activate its biological function (10). The PPARγ gene is located in the p25 region of chromosome 3 and contains six regions known as regions A-F, which are divided into four functional domains: Amino terminal domain, DNA binding domain, transcriptional activity regulatory domain and ligand binding domain (11). PPARγ regulates gene transcription through binding of the DNA binding domain to PPRE, and a number of nuclear factors, such as protein kinase C, protein kinase A and 5′AMP-activated protein kinase can affect the activity of PPARγ after binding to this domain (12,13).

Distribution and function of ERRα and PPARγ

ERRα is expressed in a variety of tissues from embryonic development to adulthood. The expression of ERRα can be detected in the heart, brain, kidney, brown adipose tissue (BAT), intestines, bones and uterus (Table I) (14). The expression of ERRα is higher in metabolically active tissues, including the heart, white adipose tissue, BAT and macrophages, while it is relatively lower in the liver, lung and vagina (15,16). Studies have demonstrated that ERRs play an important role in the regulation of eukaryotic gene expression, embryonic development, cell proliferation, bone cell production and angiogenesis (1719). ERRα is an orphan nuclear receptor that does not have corresponding ligands, but may interact with and have a bypass effect on the classical oestrogen signalling pathway through competitive binding to the same target genes, transcription factors and coactivator proteins with ERα (7,20,21). Earlier studies reported the important role of ERRα in energy metabolism of the body via the regulation of its target genes. The metabolic processes that ERRα plays a role in include glucose metabolism (22,23), lipid metabolism (24) and mitochondrial oxidation metabolism (2527). ERRα regulates the process of glucose metabolism mainly by affecting the gluconeogenic pathway and the derivatization of mitochondria (28,29). ERRα influences the lipid metabolism process through targeting and regulating genes of the fatty acid β oxidation pathway, such as acetyl-coenzyme A dehydrogenase and malonyl coenzyme A decarboxylase (30). ERRα regulates mitochondrial oxidation metabolism by upregulating gene expression related to oxidative phosphorylation through combined action with PGC-1α as the coactivator (31). When the body is affected by changes in the external environment, such as hunger and cold temperatures, the upregulation of ERRα expression may promote energy generation and the utilization of body energy, achieving an optimal adaptive state (32).

Table I.

Expression levels of ERRα and PPARγ in various tissues.

Table I.

Expression levels of ERRα and PPARγ in various tissues.

Gene Top ten tissues of gene expression in C57/Bl6J mouse, displayed from high to low (14)
ERRαJejunumIleumOlfactory bulbKidneyHeartGall bladderMusclePreputial glandBATDuodenum
PPARγWATBATColonStomachPreputial glandThyroidAortaSkinOvaryEye

[i] ERRα, estrogen receptor-associated receptor α; PPARγ, peroxisome proliferator-activated receptor γ; WAT, white adipose tissue; BAT, brown adipose tissue.

The mRNA of PPARγ is made up of ~4,000 nucleotides. A total of four subtypes of mRNA can be produced by different promoters and alternative splicing: PPARγl, PPARγ2, PPARγ3 and PPARγ4 (33). The isomers of these four mRNA subtypes have different promoters, expression modes, ligand affinity and tissue distribution. PPARγ1 is the main subtype of PPARγ and is relatively widely distributed (34). It is primarily distributed in adipose tissue, liver, heart, pancreas, intestines, kidney and skeletal muscle. The expression levels of PPARγ2 are the highest in adipose tissue, and lowest in skeletal muscle (35). PPARγ3 is expressed only in macrophages and the large intestine (36). However, little is known concerning PPARγ4 expression. PPARγ is differently expressed in a variety of tissues (Table I) (14). PPARγ regulates the expression of target genes through ligand-dependent mechanisms, thereby participating in a series of physiological processes. There are two types of PPARγ ligands: Endogenous and exogenous (37). The exogenous ligands contain insulin sensitizers used in the treatment of clinical diabetes, tyrosine-containing drugs, such as GW1929, and phenylacetic acid derivatives, such as ibuprofen (38). The endogenous ligands are mainly prostaglandin-derived metabolites (39). PPARγ forms a heterodimer with RXRα, and then binds to a specific DNA sequence of the PPRE to activate target genes (40). Based on previous studies, PPARγ exerts various biological effects and plays important roles in lipid metabolism (41), glucose metabolism (42), atherosclerosis formation (43) and inflammatory response (44). In addition, as a nuclear hormone receptor, PPARγ can affect the function of fatty acids and its derivatives at the transcriptional level to regulate cell survival and control the occurrence and development of cancer in different tissues (45).

Both ERRα and PPARγ are members of the nuclear receptor superfamily, and as ligand-dependent transcription factors, they need to bind to co-factors to form heterodimers and participate in the regulation of their target genes. A genome-wide analysis of ERRα and ERRγ has confirmed their direct and overlapping binding at the promoter regions of a large number of mitochondrial genes, a number of which are PGC-1α targets (46). These genes cover various aspects of mitochondrial oxidative metabolism, ranging from glucose utilization, fatty acid oxidation, the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) (46). Using laser capture techniques, Teng et al (47) demonstrated that the expression of the selected ERRα target gene isocitrate dehydrogenase (IDH) was involved in the TCA cycle. PPARγ is a master regulator of macrophage polarization. Angajala et al (48) showed that macrophages control the first break of the TCA cycle that occurs in the enzymatic step involving IDH. Wei et al (49) demonstrated that rosiglitazone-activated PPARγ can induce ERRα expression. PGC-1α can target ERRα and transactivate nuclear factor erythroid 2-related factor (NRF)1/NRF2 target genes, which are the nuclear respiratory factors (50). In addition, research has revealed that the induction of NRF1 transcription factors is a prerequisite for the transcriptional activation of cytochrome c (cyt c), which is an important electron transporter in OXPHOS (51). ERRα was previously implicated in regulating the gene encoding medium-chain acyl-CoA dehydrogenase (MCAD), which catalyses the initial step in mitochondrial fatty acid oxidation (52). Additionally, MCAD was previously reported to be a target gene of PPARγ (53). Gandhi et al (54) demonstrated that increased PPARγ levels can regulate insulin-mediated glucose uptake through the translocation and activation of glucose transporter type 4 in the PI3K/phosphorylated-Akt signalling cascade. Therefore, both ERRα and PPARγ can regulate the amount of acetyl-CoA that will enter the TCA cycle by affecting fatty acid metabolism. The aforementioned findings indicated that PPARγ can also affect the production of pyruvates associated with the TCA cycle by affecting the glycolysis pathway. It was also suggested that ERRα expression can influence cyt c expression, which is closely associated with the OXPHOS process. Glycolysis, fatty acid metabolism, OXPHOS and the TCA cycle are all ubiquitous metabolic pathways in the body that provide the most direct energy source, ATP (Fig. 1).

Association of ERRα and PPARγ with disease

ERRα recruits co-regulators, is activated in a constitutive manner, regulates gene transcription, and serves an important role in cell physiological functions, as well as participates in the pathological processes of some diseases, such as diabetes, fatty liver and hepatocellular carcinoma (55). Research has demonstrated that the expression levels of OXPHOS-associated genes are downregulated early in the development of insulin resistance in human diabetes (56). ERRα is a target gene of PGC-1, and hence can regulate the expression of OXPHOS and fatty acid oxidation genes. Studies have reported that the expression levels of ERRα-regulated genes are decreased in patients with insulin resistance (57), and there is an association between insulin sensitivity and the expression of ERRα mRNA in human adipose tissue (58). Overaccumulation of triglycerides in liver cells leads to non-alcoholic fatty liver disease (NAFLD). Decreased expression of ERRα affects the intake of dietary fat, thus inhibiting NAFLD development (59). In addition, a previous study indicated that the absence of ERRα activity promoted the development of rapamycin-induced NAFLD (60). Furthermore, in a mouse model of pressure overload-induced left ventricular hypertrophy, ERRα expression was found to be significantly downregulated, which resulted in faster development of heart failure (61). In addition, several studies found that in rodent models of heart failure, including models of decompensated right ventricular hypertrophy and myocardial infarction, and genetic models that show accelerated heart failure, the expression of ERRα and its coactivator are reduced (6264).

A number of studies have demonstrated the close association between ERRα and the occurrence, development and clinical prognosis of various tumours. In hormone-dependent tumours, such as endometrial (65), ovarian (20), breast (66) and prostate cancer (67), ERRα may regulate tumour development through its effect on the ERα signalling pathway. In non-hormone-dependent tumours, including colorectal cancer, non-small cell lung cancer, nasopharyngeal carcinoma and glioma, ERRα may play a role by indirectly affecting gene transcription or proliferation of tumour cells. In endometrial cancer, a previous study revealed that upregulated expression of ERRα was significantly associated with tumour cell proliferation (68). Based on the findings of previous studies, it has been proposed that ERRs and ERs are co-expressed in ovarian cancer, and the interaction between these two families may be the molecular basis for the complex endocrine biological behaviour of ovarian cancer. Sun et al (20) showed that the ERRα was associated with the occurrence of ovarian cancer and the survival rate of patients, and could be used as a factor for poor prognosis of ovarian cancer. In addition, breast cancer is also a hormone-dependent tumour. Kraus et al (69) pointed out that ERRα could compete with ERα to bind to the oestrogen response element to regulate the transcription of target genes. Recent in vitro studies demonstrated that ERRα promoted triple-negative breast cancer (TNBC) cell migration and invasion, which was regulated by STAT3, providing a potential therapeutic option against TNBC metastasis (70). Previous studies on prostate cancer revealed that ERR protein was highly expressed in prostatic epithelial cells, whereas in prostate cancer cells expression was lower, and the increase of ERRα expression levels was significantly associated with prostate cancer development, disease prognosis and the survival rate of patients (67,71). ERRα-associated diseases and related tissues are shown in Table II (8).

Table II.

ERRα-associated diseases and related tissues.

Table II.

ERRα-associated diseases and related tissues.

TissueDiseases (8)
HeartVentricular hypertrophy, myocardial infarction and heart failure
WATObesity
Liver and muscleDiabetes and non-alcoholic fatty liver disease
BoneOsteoporosis
Human reproductive organsCancer

[i] ERRα, estrogen receptor-associated receptor α; WAT, white adipose tissue.

The biological functions of PPARγ are complex and diverse, and studies have provided a number of novel approaches for the clinical prevention and treatment of diabetes (72), atherosclerosis, hypertension, NFLAD (73) and kidney disease (74). For the treatment of diabetes, thiazolidinedione (TZD) drugs can promote glucose utilization in skeletal muscle and inhibit glucose synthesis in the liver (75). When activated by TZD, PPARγ can promote the expression of the PI3K subunit p85, promote c-Cbl associated protein (CAP) transcription, promote insulin signalling and improve insulin resistance (76). In islet α cells, activated PPARγ improved insulin resistance by suppressing the activity of the transcription factor Pax6 and suppressing the expression of glucagon at the transcription level (77). Studies have reported that PPARγ ligands can induce CD36 expression, promote the phagocytosis of oxidized low-density lipoprotein by macrophages and cause intracellular lipid accumulation (78,79). In addition to enabling lipids to be taken up by macrophages, PPARγ can also transfer excess intracellular cholesterol to the extracellular space via ATP-binding cassette transporter A1 protein (80). Intimal macrophages engulf cholesterol and form foam cells during the progression of atherosclerosis. PPARγ is expressed in the vascular endothelium, and PPARγ agonists can lower blood pressure (81). In vitro endothelial cell culture experiments found that TZD-like ligands can significantly promote the secretion of vasomotor factor C-type natriuretic peptide in bovine carotid artery endothelial cells and inhibit the secretion of the vasoconstrictor factor endothelin (82).

PPARγ is a nuclear hormone receptor and its transcriptional level may affect the oxidation of fatty acids and the mitochondrial biogenesis of BAT (83). Therefore, PPARγ is most likely involved in the development of cancer in different tissues by regulating cell proliferation and differentiation. The expression of PPARγ has been reported in various types of tumour cells, including breast (84), prostate (85) and lung cancer cells (86), and it has been found that the binding of PPARγ to its ligand could inhibit the growth of tumour cells (87). However, other studies found that the expression levels of PPARγ was significantly increased in endometrial (88) and epithelial ovarian cancer (89, 90). Dong (84) found that efatutazone, a PPARγ agonist, could promote the differentiation of tumour cells in breast cancer in a specific stage, and thus interfere with tumour occurrence and development. In a study on ovarian cancer, Luo et al (91) found that PPARγ could upregulate the expression levels of microRNA-125, and thereby inhibit the proliferation of ovarian cancer cells. In colon cancer, studies demonstrated that patients with high PPARγ expression were more likely to survive than those with low PPARγ expression (92). In lung cancer, PPAR activation may inhibit the metastasis of tumour cells by inhibiting the epithelium-mesenchymal transition (93). In pancreatic cancer, it was revealed that PPARγ was highly expressed in pancreatic cancer cells, and activation of PPARγ may inhibit the growth of PANC-1 cells (94). In gastric cancer, He et al (95) reported that rosiglitazone, a PPARγ agonist, could induce cell apoptosis, and thus inhibit the growth and invasion of tumour cells, and this effect could be reversed by GW9662, a PPARγ antagonist. PPARγ-associated diseases and related tissues are shown in Table III (96).

Table III.

PPARγ-associated diseases and related tissues.

Table III.

PPARγ-associated diseases and related tissues.

TissueDiseases (96)
WATDiabetes and atherosclerosis
CNSParkinsons disease, Alzheimers disease, brain injury and ALS
Heart Cardiomyopathies
KidneyKidney disease
BreastBreast cancer

[i] PPARγ, peroxisome proliferator-activated receptor γ; WAT, white adipose tissue; CNS, central nervous system; ALS, amyotrophic lateral sclerosis.

As aforementioned, both ERRα and PPARγ are involved in tumour development. Specifically, they were reported in studies on hormone-dependent tumours (endometrial, ovarian, breast and prostate cancer) and hormone-independent tumours (lung and colon cancer) (Fig. 2). Using R programming language (version 3.6.3; http://www.r-project.org/), based on The Cancer Genome Atlas database (https://portal.gdc.cancer.gov/), Pearson's correlation analysis was performed. It was found that ERRα expression was weakly positively correlated with PPARγ expression (correlation, r=0.16, P<0.01; Fig. 3). Using bioinformatics analysis, based on the Search Tool for the Retrieval of Interacting Genes database (97), the co-expression analysis revealed that ERRα and PPARγ have a co-expression relationship (Fig. 4), suggesting that the two genes may have several similar functions. The protein-protein interaction network (http://string-db.org/cgi/input.pl) between ERRα and PPARγ showed that ERRα and PPARγ proteins interacted with nuclear receptor coactivator 1, histone acetyltransferase p300, CREB-binding protein, leptin, adiponectin receptor protein 1, CCAAT/enhancer-binding protein b and fatty acid-binding protein adipocyte. Searching UniProt database (https://www.uniprot.org/) and GeneCards database (https://www.genecards.org/), it was found that these interacting proteins are involved in the activation of gene transcription, the modification of transcription factors and cellular energy metabolism (Fig. 5).

Conclusions and perspectives

To date, there are very few studies involving both ERRα and PPARγ. A previous study demonstrated that ERRα knockout with small interfering RNA resulted in decreased PPARγ expression levels in 3T3-L1 pre-adipocytes (98). Studies have also reported that PPREs are present at the ERRα promoter, and PPRE was the PPAR response element (49). A previous study revealed that rosiglitazone, as a PPARγ agonist, could induce the expression of ERRα after activating the expression of PPARγ, thus enhancing mitochondrial biogenesis and osteoclast function (49). Therefore, it can be hypothesized that there is an association between ERRα and PPARγ expression. However, further studies are required to verify and clarify this association.

In previous years, studies on ERRα, PPARγ and tumorigenesis were gradually applied to clinical diagnosis and treatment. In diseases that have been extensively studied, such as ovarian and breast cancer, ERRα is generally considered to be a factor closely related to the poor prognosis of tumours, and hence is also considered to be a potential target for tumour therapy. Meanwhile, PPARγ expression in tumours varies, and the relationship between PPARγ and tumour prognosis is yet to be determined. In metabolic diseases that have been comprehensively studied, such as diabetes, PPARγ has become an important therapeutic target (99). ERRα is also closely related to numerous metabolic diseases. Currently, thiazolidinediones, as PPARγ agonists, have been used in the clinical treatment of metabolic syndromes, and they are expected to play an important role in the treatment of inflammation and tumours (100102).

However, there are few reports concerning the association between ERRα and PPARγ, the underlying mechanism of their interaction and their combined role in diseases. ERRα and PPARγ are related to a number of diseases, and both act as transcription factors that regulate cellular metabolic functions. Studying the relationship between ERRα and PPARγ could help to further understand the progress of certain diseases and will be useful for drug research. In addition, researches on new drugs for the ERRs have been reported (103), and thus it may be possible to develop ERRα and PPARγ dual-targeted drugs to provide further insight into the treatment of diseases.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets analyzed during the current study are available in the TCGA database (https://portal.gdc.cancer.gov/) and STRING: functional protein association networks (http://string-db.org/cgi/input.pl).

Authors' contributions

WYH and PMS designed the study. WYH was the major contributor in writing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Giguère V, Yang N, Segui P and Evans RM: Identification of a new class of steroid hormone receptors. Nature. 331:91–94. 1988. View Article : Google Scholar : PubMed/NCBI

2 

Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, Troy A, Cinti S, Lowell B, Scarpulla RC, et al: Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell. 98:115–124. 1999. View Article : Google Scholar : PubMed/NCBI

3 

Mootha VK, Handschin C, Arlow D, Xie X, St Pierre J, Sihag S, Yang W, Altshuler D, Puigserver P, Patterson N, et al: Erralpha and Gabpa/b specify PGC-1alpha-dependent oxidative phosphorylation gene expression that is altered in diabetic muscle. Proc Natl Acad Sci USA. 101:6570–6575. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Huss JM, Torra IP, Staels B, Giguère V and Kelly DP: Estrogen-related receptor alpha directs peroxisome proliferator-activated receptor alpha signaling in the transcriptional control of energy metabolism in cardiac and skeletal muscle. Mol Cell Biol. 24:9079–9091. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Emmett MJ, Lim HW, Jager J, Richter HJ, Adlanmerini M, Peed LC, Briggs ER, Steger DJ, Ma T, Sims CA, et al: Histone deacetylase 3 prepares brown adipose tissue for acute thermogenic challenge. Nature. 546:544–548. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Dufour CR, Levasseur MP, Pham NH, Eichner LJ, Wilson BJ, Charest-Marcotte A, Duguay D, Poirier-Héon JF, Cermakian N and Giguère V: Genomic convergence among ERRα, PROX1, and BMAL1 in the control of metabolic clock outputs. PLoS Genet. 7:e10021432011. View Article : Google Scholar : PubMed/NCBI

7 

Giguère V: To ERR in the estrogen pathway. Trends Endocrinol Metab. 13:220–225. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Ranhotra HS: The estrogen-related receptor alpha: The oldest, yet an energetic orphan with robust biological functions. J Recept Signal Transduct Res. 30:193–205. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Issemann I and Green S: Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature. 347:645–650. 1990. View Article : Google Scholar : PubMed/NCBI

10 

Juge-Aubry C, Pernin A, Favez T, Burger AG, Wahli W, Meier CA and Desvergne B: DNA binding properties of peroxisome proliferator-activated receptor subtypes on various natural peroxisome proliferator response elements. Importance of the 5-flanking region. J Biol Chem. 272:25252–25259. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Vamecq J and Latruffe N: Medical significance of peroxisome proliferator-activated receptors. Lancet. 354:141–148. 1999. View Article : Google Scholar : PubMed/NCBI

12 

Chandra V, Huang P, Hamuro Y, Raghuram S, Wang Y, Burris TP and Rastinejad F: Structure of the intact PPAR-gamma-RXR- nuclear receptor complex on DNA. Nature. 456:350–356. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Burns KA and Vanden Heuvel JP: Modulation of PPAR activity via phosphorylation. Biochim Biophys Acta. 1771:952–960. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM and Mangelsdorf DJ: Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell. 126:789–799. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Bonnelye E, Vanacker JM, Dittmar T, Begue A, Desbiens X, Denhardt DT, Aubin JE, Laudet V and Fournier B: The ERR-1 orphan receptor is a transcriptional activator expressed during bone development. Mol Endocrinol. 11:905–916. 1997. View Article : Google Scholar : PubMed/NCBI

16 

Heard DJ, Norby PL, Holloway J and Vissing H: Human ERRgamma, a third member of the estrogen receptor-related receptor (ERR) subfamily of orphan nuclear receptors: Tissue-specific isoforms are expressed during development and in the adult. Mol Endocrinol. 14:382–392. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Bonnelye E, Vanacker JM, Spruyt N, Alric S, Fournier B, Desbiens X and Laudet V: Expression of the estrogen-related receptor 1 (ERR-1) orphan receptor during mouse development. Mech Dev. 65:71–85. 1997. View Article : Google Scholar : PubMed/NCBI

18 

Carnesecchi J and Vanacker JM: Estrogen-related receptors and the control of bone cell fate. Mol Cell Endocrinol. 432:37–43. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Likhite N, Yadav V, Milliman EJ, Sopariwala DH, Lorca S, Narayana NP, Sheth M, Reineke EL, Giguère V and Narkar V: Loss of estrogen-related receptor alpha facilitates angiogenesis in endothelial cells. Mol Cell Biol. 39:392019. View Article : Google Scholar

20 

Sun P, Sehouli J, Denkert C, Mustea A, Könsgen D, Koch I, Wei L and Lichtenegger W: Expression of estrogen receptor-related receptors, a subfamily of orphan nuclear receptors, as new tumor biomarkers in ovarian cancer cells. J Mol Med (Berl). 83:457–467. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Johnston SD, Liu X, Zuo F, Eisenbraun TL, Wiley SR, Kraus RJ and Mertz JE: Estrogen-related receptor alpha 1 functionally binds as a monomer to extended half-site sequences including ones contained within estrogen-response elements. Mol Endocrinol. 11:342–352. 1997. View Article : Google Scholar : PubMed/NCBI

22 

Wende AR, Huss JM, Schaeffer PJ, Giguère V and Kelly DP: PGC-1alpha coactivates PDK4 gene expression via the orphan nuclear receptor ERRalpha: A mechanism for transcriptional control of muscle glucose metabolism. Mol Cell Biol. 25:10684–10694. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, Rhee J, Adelmant G, Stafford J, Kahn CR, Granner DK, et al: Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature. 413:131–138. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Kitamura K, Erlangga JS, Tsukamoto S, Sakamoto Y, Mabashi-Asazuma H and Iida K: Daidzein promotes the expression of oxidative phosphorylation- and fatty acid oxidation-related genes via an estrogen-related receptor α pathway to decrease lipid accumulation in muscle cells. J Nutr Biochem. 77:1083152020. View Article : Google Scholar : PubMed/NCBI

25 

Cartoni R, Léger B, Hock MB, Praz M, Crettenand A, Pich S, Ziltener JL, Luthi F, Dériaz O, Zorzano A, et al: Mitofusins 1/2 and ERRalpha expression are increased in human skeletal muscle after physical exercise. J Physiol. 567:349–358. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Soriano FX, Liesa M, Bach D, Chan DC, Palacín M and Zorzano A: Evidence for a mitochondrial regulatory pathway defined by peroxisome proliferator-activated receptor-gamma coactivator-1 alpha, estrogen-related receptor-alpha, and mitofusin 2. Diabetes. 55:1783–1791. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Rangwala SM, Li X, Lindsley L, Wang X, Shaughnessy S, Daniels TG, Szustakowski J, Nirmala NR, Wu Z and Stevenson SC: Estrogen-related receptor alpha is essential for the expression of antioxidant protection genes and mitochondrial function. Biochem Biophys Res Commun. 357:231–236. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Herzog B, Cardenas J, Hall RK, Villena JA, Budge PJ, Giguère V, Granner DK and Kralli A: Estrogen-related receptor alpha is a repressor of phosphoenolpyruvate carboxykinase gene transcription. J Biol Chem. 281:99–106. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Ranhotra HS: Estrogen-related receptor alpha and mitochondria: Tale of the titans. J Recept Signal Transduct Res. 35:386–390. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Vega RB and Kelly DP: A role for estrogen-related receptor alpha in the control of mitochondrial fatty acid beta-oxidation during brown adipocyte differentiation. J Biol Chem. 272:31693–31699. 1997. View Article : Google Scholar : PubMed/NCBI

31 

LeBleu VS, O'Connell JT, Gonzalez Herrera KN, Wikman H, Pantel K, Haigis MC, Machado de Carvalho F, Damascena A, Domingos Chinen LT, Rocha RM, et al: PGC-1α mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat Cell Biol. 16:992–1003, 1001-1015. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Villena JA and Kralli A: ERRalpha: A metabolic function for the oldest orphan. Trends Endocrinol Metab. 19:269–276. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Aprile M, Ambrosio MR, D'Esposito V, Beguinot F, Formisano P, Costa V and Ciccodicola A: PPARG in human adipogenesis: Differential contribution of canonical transcripts and dominant negative isoforms. PPAR Res. 2014:5378652014. View Article : Google Scholar : PubMed/NCBI

34 

Salgia MM, Elix CC, Pal SK and Jones JO: Different roles of peroxisome proliferator-activated receptor gamma isoforms in prostate cancer. Am J Clin Exp Urol. 7:98–109. 2019.PubMed/NCBI

35 

Braissant O, Foufelle F, Scotto C, Dauça M and Wahli W: Differential expression of peroxisome proliferator-activated receptors (PPARs): Tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. Endocrinology. 137:354–366. 1996. View Article : Google Scholar : PubMed/NCBI

36 

Meirhaeghe A, Fajas L, Gouilleux F, Cottel D, Helbecque N, Auwerx J and Amouyel P: A functional polymorphism in a STAT5B site of the human PPAR gamma 3 gene promoter affects height and lipid metabolism in a French population. Arterioscler Thromb Vasc Biol. 23:289–294. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Martin H: Role of PPAR-gamma in inflammation. Prospects for therapeutic intervention by food components. Mutat Res. 669:1–7. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Tanis SP, Colca JR, Parker TT, Artman GD III, Larsen SD, McDonald WG, Gadwood RC, Kletzien RF, Zeller JB, Lee PH, et al: PPARγ-sparing thiazolidinediones as insulin sensitizers. Design, synthesis and selection of compounds for clinical development. Bioorg Med Chem. 26:5870–5884. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Finch ER, Tukaramrao DB, Goodfield LL, Quickel MD, Paulson RF and Prabhu KS: Activation of PPARγ by endogenous prostaglandin J2 mediates the antileukemic effect of selenium in murine leukemia. Blood. 129:1802–1810. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Michalik L, Desvergne B, Dreyer C, Gavillet M, Laurini RN and Wahli W: PPAR expression and function during vertebrate development. Int J Dev Biol. 46:105–114. 2002.PubMed/NCBI

41 

Lapsys NM, Kriketos AD, Lim-Fraser M, Poynten AM, Lowy A, Furler SM, Chisholm DJ and Cooney GJ: Expression of genes involved in lipid metabolism correlate with peroxisome proliferator-activated receptor gamma expression in human skeletal muscle. J Clin Endocrinol Metab. 85:4293–4297. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Iwata M, Haruta T, Usui I, Takata Y, Takano A, Uno T, Kawahara J, Ueno E, Sasaoka T, Ishibashi O, et al: Pioglitazone ameliorates tumor necrosis factor-alpha-induced insulin resistance by a mechanism independent of adipogenic activity of peroxisome proliferator - activated receptor-gamma. Diabetes. 50:1083–1092. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Ricote M, Huang J, Fajas L, Li A, Welch J, Najib J, Witztum JL, Auwerx J, Palinski W and Glass CK: Expression of the peroxisome proliferator-activated receptor gamma (PPARgamma) in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized low density lipoprotein. Proc Natl Acad Sci USA. 95:7614–7619. 1998. View Article : Google Scholar : PubMed/NCBI

44 

Jiang C, Ting AT and Seed B: PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature. 391:82–86. 1998. View Article : Google Scholar : PubMed/NCBI

45 

Yamashita H, Kawasawa YI, Shuman L, Zheng Z, Tran T, Walter V, Warrick JI, Chen G, Al-Ahmadie H, Kaag M, et al: Repression of transcription factor AP-2 alpha by PPARγ reveals a novel transcriptional circuit in basal-squamous bladder cancer. Oncogenesis. 8:692019. View Article : Google Scholar : PubMed/NCBI

46 

Fan W and Evans R: PPARs and ERRs: Molecular mediators of mitochondrial metabolism. Curr Opin Cell Biol. 33:49–54. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Teng CT, Li Y, Stockton P and Foley J: Fasting induces the expression of PGC-1α and ERR isoforms in the outer stripe of the outer medulla (OSOM) of the mouse kidney. PLoS One. 6:e269612011. View Article : Google Scholar : PubMed/NCBI

48 

Angajala A, Lim S, Phillips JB, Kim JH, Yates C, You Z and Tan M: Diverse roles of mitochondria in immune responses: novel insights into immuno-metabolism. Front Immunol. 9:16052018. View Article : Google Scholar : PubMed/NCBI

49 

Wei W, Wang X, Yang M, Smith LC, Dechow PC, Sonoda J, Evans RM and Wan Y: PGC1beta mediates PPARgamma activation of osteoclastogenesis and rosiglitazone-induced bone loss. Cell Metab. 11:503–516. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Scarpulla RC: Metabolic control of mitochondrial biogenesis through the PGC-1 family regulatory network. Biochim Biophys Acta. 1813:1269–1278. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Xia Y, Buja LM, Scarpulla RC and McMillin JB: Electrical stimulation of neonatal cardiomyocytes results in the sequential activation of nuclear genes governing mitochondrial proliferation and differentiation. Proc Natl Acad Sci USA. 94:11399–11404. 1997. View Article : Google Scholar : PubMed/NCBI

52 

Huss JM, Kopp RP and Kelly DP: Peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched nuclear receptors estrogen-related receptor-alpha and -gamma. Identification of novel leucine-rich interaction motif within PGC-1alpha. J Biol Chem. 277:40265–40274. 2002. View Article : Google Scholar : PubMed/NCBI

53 

Fliegner D, Westermann D, Riad A, Schubert C, Becher E, Fielitz J, Tschöpe C and Regitz-Zagrosek V: Up-regulation of PPARgamma in myocardial infarction. Eur J Heart Fail. 10:30–38. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Gandhi GR, Stalin A, Balakrishna K, Ignacimuthu S, Paulraj MG and Vishal R: Insulin sensitization via partial agonism of PPARγ and glucose uptake through translocation and activation of GLUT4 in PI3K/p-Akt signaling pathway by embelin in type 2 diabetic rats. Biochim Biophys Acta. 1830:2243–2255. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Xia H, Dufour CR and Giguère V: ERRα as a bridge between transcription and function: Role in liver metabolism and disease. Front Endocrinol (Lausanne). 10:2062019. View Article : Google Scholar : PubMed/NCBI

56 

Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, Laurila E, et al: PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 34:267–273. 2003. View Article : Google Scholar : PubMed/NCBI

57 

Schreiber SN, Emter R, Hock MB, Knutti D, Cardenas J, Podvinec M, Oakeley EJ and Kralli A: The estrogen-related receptor alpha (ERRalpha) functions in PPARgamma coactivator 1alpha (PGC-1alpha)-induced mitochondrial biogenesis. Proc Natl Acad Sci USA. 101:6472–6477. 2004. View Article : Google Scholar : PubMed/NCBI

58 

Rutanen J, Yaluri N, Modi S, Pihlajamäki J, Vänttinen M, Itkonen P, Kainulainen S, Yamamoto H, Lagouge M, Sinclair DA, et al: SIRT1 mRNA expression may be associated with energy expenditure and insulin sensitivity. Diabetes. 59:829–835. 2010. View Article : Google Scholar : PubMed/NCBI

59 

B'chir W, Dufour CR, Ouellet C, Yan M, Tam IS, Andrzejewski S, Xia H, Nabata K, St-Pierre J and Giguère V: Divergent role of estrogen-related receptor α in lipid- and fasting-induced hepatic steatosis in mice. Endocrinology. 159:2153–2164. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Chaveroux C, Eichner LJ, Dufour CR, Shatnawi A, Khoutorsky A, Bourque G, Sonenberg N and Giguère V: Molecular and genetic crosstalks between mTOR and ERRα are key determinants of rapamycin-induced nonalcoholic fatty liver. Cell Metab. 17:586–598. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Sakamoto T, Matsuura TR, Wan S, Ryba DM, Kim JU, Won KJ, Lai L, Petucci C, Petrenko N, Musunuru K, et al: A Critical Role for Estrogen-Related Receptor Signaling in Cardiac Maturation. Circ Res. 126:1685–1702. 2020. View Article : Google Scholar : PubMed/NCBI

62 

Mitra MS, Schilling JD, Wang X, Jay PY, Huss JM, Su X and Finck BN: Cardiac lipin 1 expression is regulated by the peroxisome proliferator activated receptor γ coactivator 1α/estrogen related receptor axis. J Mol Cell Cardiol. 51:120–128. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Gomez-Arroyo J, Mizuno S, Szczepanek K, Van Tassell B, Natarajan R, dos Remedios CG, Drake JI, Farkas L, Kraskauskas D, Wijesinghe DS, et al: Metabolic gene remodeling and mitochondrial dysfunction in failing right ventricular hypertrophy secondary to pulmonary arterial hypertension. Circ Heart Fail. 6:136–144. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Watson PA, Birdsey N, Huggins GS, Svensson E, Heppe D and Knaub L: Cardiac-specific overexpression of dominant-negative CREB leads to increased mortality and mitochondrial dysfunction in female mice. Am J Physiol Heart Circ Physiol. 299:H2056–H2068. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Sun PM, Gao M, Wei LH, Mustea A, Wang JL, Könsgen D, Lichtenegger W and Sehouli J: An estrogen receptor alpha-dependent regulation of estrogen receptor-related receptor alpha in the proliferation of endometrial carcinoma cells. Int J Gynecol Cancer. 16 (Suppl 2):564–568. 2006. View Article : Google Scholar : PubMed/NCBI

66 

Stein RA, Gaillard S and McDonnell DP: Estrogen-related receptor alpha induces the expression of vascular endothelial growth factor in breast cancer cells. J Steroid Biochem Mol Biol. 114:106–112. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Cheung CP, Yu S, Wong KB, Chan LW, Lai FM, Wang X, Suetsugi M, Chen S and Chan FL: Expression and functional study of estrogen receptor-related receptors in human prostatic cells and tissues. J Clin Endocrinol Metab. 90:1830–1844. 2005. View Article : Google Scholar : PubMed/NCBI

68 

Fujimoto J and Sato E: Clinical implication of estrogen-related receptor (ERR) expression in uterine endometrial cancers. J Steroid Biochem Mol Biol. 116:71–75. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Kraus RJ, Ariazi EA, Farrell ML and Mertz JE: Estrogen-related receptor alpha 1 actively antagonizes estrogen receptor-regulated transcription in MCF-7 mammary cells. J Biol Chem. 277:24826–24834. 2002. View Article : Google Scholar : PubMed/NCBI

70 

Ma JH, Qi J, Lin SQ, Zhang CY, Liu FY, Xie WD and Li X: STAT3 targets ERR-α to promote epithelial-mesenchymal transition, migration, and invasion in triple-negative breast cancer cells. Mol Cancer Res. 17:2184–2195. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Fujimura T, Takahashi S, Urano T, Kumagai J, Ogushi T, Horie-Inoue K, Ouchi Y, Kitamura T, Muramatsu M and Inoue S: Increased expression of estrogen-related receptor alpha (ERRalpha) is a negative prognostic predictor in human prostate cancer. Int J Cancer. 120:2325–2330. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Hong F, Pan S, Guo Y, Xu P and Zhai Y: PPARs as nuclear receptors for nutrient and energy metabolism. Molecules. 24:25452019. View Article : Google Scholar

73 

Fougerat A, Montagner A, Loiseau N, Guillou H and Wahli W: Peroxisome proliferator-activated receptors and their novel ligands as candidates for the treatment of non-alcoholic fatty liver disease. Cells. 9:92020. View Article : Google Scholar

74 

Shi Y, Zou Y, Shen Z, Xiong Y, Zhang W, Liu C and Chen S: Trace elements, PPARs, and metabolic syndrome. Int J Mol Sci. 21:212020. View Article : Google Scholar

75 

Thangavel N, Al Bratty M, Akhtar Javed S, Ahsan W and Alhazmi HA: Targeting peroxisome proliferator-activated receptors using thiazolidinediones: Strategy for design of novel antidiabetic drugs. Int J Med Chem. 2017:10697182017.PubMed/NCBI

76 

Ribon V, Johnson JH, Camp HS and Saltiel AR: Thiazolidinediones and insulin resistance: Peroxisome proliferatoractivated receptor gamma activation stimulates expression of the CAP gene. Proc Natl Acad Sci USA. 95:14751–14756. 1998. View Article : Google Scholar : PubMed/NCBI

77 

Schinner S, Dellas C, Schroder M, Heinlein CA, Chang C, Fischer J and Knepel W: Repression of glucagon gene transcription by peroxisome proliferator-activated receptor gamma through inhibition of Pax6 transcriptional activity. J Biol Chem. 277:1941–1948. 2002. View Article : Google Scholar : PubMed/NCBI

78 

Tontonoz P, Nagy L, Alvarez JG, Thomazy VA and Evans RM: PPARgamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell. 93:241–252. 1998. View Article : Google Scholar : PubMed/NCBI

79 

Maréchal L, Laviolette M, Rodrigue-Way A, Sow B, Brochu M, Caron V and Tremblay A: The CD36-PPARγ pathway in metabolic disorders. Int J Mol Sci. 19:192018. View Article : Google Scholar

80 

Chawla A, Barak Y, Nagy L, Liao D, Tontonoz P and Evans RM: PPAR-gamma dependent and independent effects on macrophage-gene expression in lipid metabolism and inflammation. Nat Med. 7:48–52. 2001. View Article : Google Scholar : PubMed/NCBI

81 

Chandra M, Miriyala S and Panchatcharam M: PPARγ and its role in cardiovascular diseases. PPAR Res. 2017:64046382017. View Article : Google Scholar : PubMed/NCBI

82 

Fukunaga Y, Itoh H, Doi K, Tanaka T, Yamashita J, Chun TH, Inoue M, Masatsugu K, Sawada N, Saito T, et al: Thiazolidinediones, peroxisome proliferator-activated receptor gamma agonists, regulate endothelial cell growth and secretion of vasoactive peptides. Atherosclerosis. 158:113–119. 2001. View Article : Google Scholar : PubMed/NCBI

83 

Chang JS and Ha K: A truncated PPAR gamma 2 localizes to mitochondria and regulates mitochondrial respiration in brown adipocytes. PLoS One. 13:e01950072018. View Article : Google Scholar : PubMed/NCBI

84 

Dong JT: Anticancer activities of PPARγ in breast cancer are context-dependent. Am J Pathol. 182:1972–1975. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Lin SJ, Lin CY, Yang DR, Izumi K, Yan E, Niu X, Chang HC, Miyamoto H, Wang N, Li G, et al: The differential effects of anti-diabetic thiazolidinedione on prostate cancer progression are linked to the TR4 nuclear receptor expression status. Neoplasia. 17:339–347. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Lakshmi SP, Reddy AT, Banno A and Reddy RC: PPAR agonists for the prevention and treatment of lung cancer. PPAR Res. 2017:82527962017. View Article : Google Scholar : PubMed/NCBI

87 

Yousefnia S, Momenzadeh S, Seyed Forootan F, Ghaedi K and Nasr Esfahani MH: The influence of peroxisome proliferator-activated receptor γ (PPARγ) ligands on cancer cell tumorigenicity. Gene. 649:14–22. 2018. View Article : Google Scholar : PubMed/NCBI

88 

Holland CM, Saidi SA, Evans AL, Sharkey AM, Latimer JA, Crawford RA, Charnock-Jones DS, Print CG and Smith SK: Transcriptome analysis of endometrial cancer identifies peroxisome proliferator-activated receptors as potential therapeutic targets. Mol Cancer Ther. 3:993–1001. 2004.PubMed/NCBI

89 

Zhang GY, Ahmed N, Riley C, Oliva K, Barker G, Quinn MA and Rice GE: Enhanced expression of peroxisome proliferator-activated receptor gamma in epithelial ovarian carcinoma. Br J Cancer. 92:113–119. 2005. View Article : Google Scholar : PubMed/NCBI

90 

Davidson B, Hadar R, Stavnes HT, Trope' CG and Reich R: Expression of the peroxisome proliferator-activated receptors-alpha, -beta, and -gamma in ovarian carcinoma effusions is associated with poor chemoresponse and shorter survival. Hum Pathol. 40:705–713. 2009. View Article : Google Scholar : PubMed/NCBI

91 

Luo S, Wang J, Ma Y, Yao Z and Pan H: PPARγ inhibits ovarian cancer cells proliferation through upregulation of miR-125b. Biochem Biophys Res Commun. 462:85–90. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Ogino S, Shima K, Baba Y, Nosho K, Irahara N, Kure S, Chen L, Toyoda S, Kirkner GJ, Wang YL, et al: Colorectal cancer expression of peroxisome proliferator-activated receptor gamma (PPARG, PPARgamma) is associated with good prognosis. Gastroenterology. 136:1242–1250. 2009. View Article : Google Scholar : PubMed/NCBI

93 

Reka AK, Kurapati H, Narala VR, Bommer G, Chen J, Standiford TJ and Keshamouni VG: Peroxisome proliferator-activated receptor-gamma activation inhibits tumor metastasis by antagonizing Smad3-mediated epithelial-mesenchymal transition. Mol Cancer Ther. 9:3221–3232. 2010. View Article : Google Scholar : PubMed/NCBI

94 

Dong YW, Wang XP and Wu K: Suppression of pancreatic carcinoma growth by activating peroxisome proliferator-activated receptor gamma involves angiogenesis inhibition. World J Gastroenterol. 15:441–448. 2009. View Article : Google Scholar : PubMed/NCBI

95 

He Q, Pang R, Song X, Chen J, Chen H, Chen B, Hu P and Chen M: Rosiglitazone suppresses the growth and invasiveness of SGC-7901 gastric cancer cells and angiogenesis in vitro via PPARgamma dependent and independent mechanisms. PPAR Res. 2008:6498082008. View Article : Google Scholar : PubMed/NCBI

96 

Wang YX: PPARs: Diverse regulators in energy metabolism and metabolic diseases. Cell Res. 20:124–137. 2010. View Article : Google Scholar : PubMed/NCBI

97 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47:D607–D613. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Ijichi N, Ikeda K, Horie-Inoue K, Yagi K, Okazaki Y and Inoue S: Estrogen-related receptor alpha modulates the expression of adipogenesis-related genes during adipocyte differentiation. Biochem Biophys Res Commun. 358:813–818. 2007. View Article : Google Scholar : PubMed/NCBI

99 

Prabhu DS and Rajeswari VD: PPAR-Gamma as putative gene target involved in Butein mediated anti-diabetic effect. Mol Biol Rep. 47:5273–5283. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Lebovitz HE: Thiazolidinediones: The forgotten diabetes medications. Curr Diab Rep. 19:1512019. View Article : Google Scholar : PubMed/NCBI

101 

Heneka MT, Sastre M, Dumitrescu-Ozimek L, Hanke A, Dewachter I, Kuiperi C, O'Banion K, Klockgether T, Van Leuven F and Landreth GE: Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice. Brain. 128:1442–1453. 2005. View Article : Google Scholar : PubMed/NCBI

102 

Wuertz BR, Darrah L, Wudel J and Ondrey FG: Thiazolidinediones abrogate cervical cancer growth. Exp Cell Res. 353:63–71. 2017. View Article : Google Scholar : PubMed/NCBI

103 

Sanghvi M, Moaddel R, Frazier C, Gandhari M and Wainer IW: Abstract 5500: Biochromatography for the screening of new drugs for the estrogen related receptors. Cancer Res. 70:55002010.

Related Articles

Journal Cover

February-2021
Volume 23 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang W and Huang W: Estrogen receptor-associated receptor α and peroxisome proliferator-activated receptor γ in metabolism and disease (Review). Mol Med Rep 23: 156, 2021
APA
Huang, W., & Huang, W. (2021). Estrogen receptor-associated receptor α and peroxisome proliferator-activated receptor γ in metabolism and disease (Review). Molecular Medicine Reports, 23, 156. https://doi.org/10.3892/mmr.2020.11795
MLA
Huang, W., Sun, P."Estrogen receptor-associated receptor α and peroxisome proliferator-activated receptor γ in metabolism and disease (Review)". Molecular Medicine Reports 23.2 (2021): 156.
Chicago
Huang, W., Sun, P."Estrogen receptor-associated receptor α and peroxisome proliferator-activated receptor γ in metabolism and disease (Review)". Molecular Medicine Reports 23, no. 2 (2021): 156. https://doi.org/10.3892/mmr.2020.11795