Next Article in Journal
LncRNA H19 Regulates Breast Cancer DNA Damage Response and Sensitivity to PARP Inhibitors via Binding to ILF2
Previous Article in Journal
New 5-Hydroxycoumarin-Based Tyrosyl-DNA Phosphodiesterase I Inhibitors Sensitize Tumor Cell Line to Topotecan
Previous Article in Special Issue
Fibrates Affect Levels of Phosphorylated p38 in Intestinal Cells in a Differentiation-Dependent Manner
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Nuclear Receptors in Health and Diseases

1
Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
2
Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
Int. J. Mol. Sci. 2023, 24(11), 9153; https://doi.org/10.3390/ijms24119153
Submission received: 29 April 2023 / Revised: 11 May 2023 / Accepted: 18 May 2023 / Published: 23 May 2023
(This article belongs to the Special Issue Nuclear Receptors in Health and Diseases)
Nuclear receptors (NRs) are a vital superfamily of transcription factors that play crucial roles in physiology and pharmacology [1]. The human genome comprises 48 NRs, 24 of which are ligand-dependent transcription factors [2]. The NRs in humans are categorized into six subgroups based on their sequence homology and structure [1]: thyroid-hormone-receptor-like, retinoid-x-receptor-like, estrogen-receptor-like, nerve-growth-factor-IB-like, steroidogenic-factor-like, and germ-cell-nuclear-factor-like, which are presented in Table 1. These NRs play significant roles in various processes, including development, reproduction, aging, and cancer [3]. They act as key mediators in maintaining everyday health by regulating metabolism and circadian rhythms. NRs can directly bind to specific promoter elements on DNA to regulate the expression of target genes, which, in turn, control metabolism, homeostasis, development, and reproduction.
Most NRs consist of six functional domains [4], including the variable N-terminal regulatory domain (A–B), retained DNA-binding domain (DBD) (C), hinge region (D), conserved ligand-binding domain (LBD) (E), and variable C-terminal domain (F), as illustrated in Figure 1A. NRs are transcription factors that are activated by ligands, small lipid-based molecules, to regulate gene expression and cellular processes. This family comprises receptors (class I) for various steroid hormones and their derivatives, including estrogen, progesterone, glucocorticoids, vitamin D, oxysterols, and bile acids, as well as receptors for retinoic acids, thyroid hormones, fatty acids, and their derivatives [5]. Some NRs are cytoplasmic in the absence of ligands and form complexes with heat shock proteins (HSP) that regulate their cellular location, protein stability, steroid-hormone-binding capacity, and transcriptional activity [6]. Upon ligand binding, these receptors undergo dimerization and translocate to the nucleus, as depicted in Figure 1B. Optionally, class II NRs are retained in the nucleus and their chromatin-modifying activities are modulated by ligand binding to form heterodimers (usually with retinoid X receptors (RXR)) [7], as depicted in Figure 1C. NRs function primarily by transcriptionally regulating the expression of related target genes through the recruitment of coactivators or corepressors when ligands bind to the receptors [8]. NRs are highly druggable targets due to their ability to bind and be activated by a large number of small molecules, with 10–20% of FDA-approved medications currently targeting NRs [9]. These medications have an estimated market value of 30 billion dollars per year. Consequently, a variety of opinions exist for repurposing these approved medications and developing new therapies targeting NRs for numerous diseases.
The interaction between NRs and other biological processes presents an exciting area of research. DNA damage repair pathways are intimately connected to steroid signaling through transcriptional processes, which has implications for drug combinations in a variety of diseases. The previously unknown roles of NRs in epigenetic regulation and their involvement in essential signaling pathways and metabolic mechanisms indicate that this superfamily has the potential not only to affect development and normal functioning, but also to contribute to disease development. This Special Issue conveys several prominent researchers in the field of nuclear receptor research from around the world to examine the emerging roles of NRs (mainly including farnesoid X receptors (FXR), peroxisome proliferator-activated receptors (PPARs), constitutive activated receptors (CAR), estrogen-related receptor γ (ERRγ), and RXR, etc.) and their impact on health maintenance and disease pathogenesis, such as cancers, metabolic disorders, growth and development, inflammation, Parkinson’s disease, and kidney disease.
FXR, also referred to as NR1H4, is a nuclear receptor that responds to bile acid and plays a role in regulating the metabolisms of bile acid, lipid, and glucose to maintain homeostasis [10]. Several studies have suggested the role of FXR in regulating genes that manipulate intestinal glucose [11,12,13]. Jiufang et al. investigated the role of gut-specific FXR in glucose absorption using a unique double-tagged glucose kinetic approach in FXR△gut mice [14]. Although the FXR△gut mice exhibited a reduced expression of hexokinase 1 (Hk1) in the duodenum when subjected to an obesogenic challenge, the analysis of the glucose fluxes in these mice did not indicate that gut FXR plays a role in glucose absorption. These findings suggest that the delayed glucose uptake observed in FXR-deficient mice is not due to a lack of FXR in the intestine [14]. However, intestinal FXR is involved in regulating the surface area of the small intestine. Jae Man et al. reported that the gene encoding the transient receptor potential melastatin 6 (TRPM6), which is an Mg2+ channel, could be a direct target of the FXR in the intestinal epithelial cells of mice. The maintenance of the basal expression of intestinal TRPM6 in the presence of GW4064, an artificial FXR agonist, is dependent on the FXR expression in the intestinal epithelial cells [15]. Additionally, they proposed a gut FXR–TRPM6 axis that could link bile acid signaling to Mg2+ homeostasis. Yongdong Niu’s lab found that HBx C40, a C-terminal truncated X protein and known FXR-binding protein, contributes to tumor cell proliferation and migration by modifying the distribution of the cell cycle and causing apoptosis, independent of FXR. HBx C40 enhances the growth of FXR deficiency tumors in vivo by modifying the cellular cycle distribution and disrupting the glucose metabolism to promote the development of hepatocellular carcinoma [16]. FXR is also known to play a critical role in renal water reabsorption and is believed to provide a protective effect in both acute and chronic renal diseases, including those related to diabetes. Xiaoyan Zhang’s laboratory has summarized the recent developments in understanding the physiological and pathophysiological roles of FXR in the kidney [17].
Non-steroidal anti-inflammatory drugs (NSAIDs) are considered to be cancer prevention therapeutics due to their ability to restrict cyclooxygenases (COX), which are commonly overexpressed in various types of cancer [18]. Adam et al. demonstrated that NSAIDs can induce anti-neoplastic activity by activating PPARγ, which, in turn, induces proline dehydrogenase/proline oxidase (PRODH/POX)-dependent apoptosis [19]. Therefore, targeting proline metabolism and the PRODH/POX–PPARγ axis could be a potential new approach for treating breast cancer [19]. Lsaac et al. investigated the therapeutic effects of PPARs, particularly the gamma isoform (PPARγ), in preclinical animal models of Parkinson’s disease (PD) and clinical trials for PD [20]. They suggested the possible anti-α-synucleinopathy mechanisms downstream from these NRs [20]. Understanding the neuroprotective mechanisms of PPARs, using preclinical models that mimic PD as closely as possible, can facilitate better clinical trials for disease-modifying medications in PD [20]. Fenofibrate, a widely used cholesterol-lowering agent, acts as a ligand of PPARα [21]. Katerina et al. investigated the impact of fenofibrate on the activated p38 (p-p38) levels in colorectal cancer cell lines with differentiating statuses. They found that fibrates upregulated the p-p38 in undifferentiated HT-29 cells, whereas, in other cases, the p-p38 expression was reduced. The authors also observed that PPAR ligands affected the expression of the p-p38 in the cytoplasm of HT-29 cells, whereas, in Caco2 cells, fibrates affected the p-p38 expression in both the cytoplasm and nucleus [22].
The xenobiotic receptor CAR is activated by a variety of environmental chemicals and participates in xenobiotic metabolism [23,24]. The Yonggong Zhai lab investigated the effects of maternal exposure to the CAR-specific ligand 1,4-bis [2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP) on offspring health outcomes [25]. Maternal TCPOBOP exposure led to a greater inhibition of body weight in first-generation (F1) female mice compared to the control group. Additionally, maternal exposure to TCPOBOP resulted in a significant increase in the hepatic-drug-metabolizing enzyme expression in F1 generation [25]. Mechanistically, maternal TCPOBOP exposure triggered the intestinal inflammatory response and disrupted the antioxidant system in the offspring female mice, which hindered the absorption of nutrients in the intestine and posed a serious threat to the health of their offspring [25].
ERRγ, an orphan nuclear receptor, is a crucial transcription factor that plays a significant role in mitochondrial metabolism and energy homeostasis. Soon-Young and colleagues conducted cell-based experiments and demonstrated that GSK5182, a specific reverse agonist of ERRγ, can impede the ubiquitination of ERRγ, thereby stabilizing the protein [26]. The researchers also found that GSK5182’s stabilization of ERRγ is dependent on the presence of Y326, as a mutation of this amino acid to alanine (Y326A) prevented a ligand-induced stabilization of the protein. Furthermore, GSK5182 hindered the ubiquitination of ERRγ via the E3 Parkin ligase, which prevented its subsequent degradation [26].
Megumi et al. found that the RXR ligand, all-trans retinoic acid (ATRA), induced synergistic and beneficial effects on TGF-β2-treated human trabecular meshwork (HTM) cells cultured in both 2D and 3D environments. Notably, the effects varied considerably between the two cultures [27].
Valentina and colleagues provided a comprehensive summary of the role of nuclear receptors (NRs) in erythropoiesis, covering their involvement in both homeostatic and stressful conditions [28]. This knowledge is essential for regulating the production of red blood cells, particularly in disease conditions such as anemia. Additionally, this understanding is crucial for expanding erythroid cells in culture, enabling the long-term objective of cultured blood for transfusions.
The current Special Issue serves as an open-access platform for a collection of reviews and original research articles that discuss the advanced features of NRs in physiological and pathological states. These articles provide a fascinating resource on the various aspects of NRs, ranging from fundamental science to applied therapeutic approaches.

Funding

This research was supported by the start-up funding from Wuhan University (Nos.600460042).

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Auwerx, J.; Baulieu, E.; Beato, M.; Becker-Andre, M.; Burbach, P.H.; Camerino, G.; Chambon, P.; Cooney, A.; Dejean, A.; Dreyer, C.; et al. A unified nomenclature system for the nuclear receptor superfamily. Cell 1999, 97, 161–163. [Google Scholar]
  2. Mangelsdorf, D.J.; Thummel, C.; Beato, M.; Herrlich, P.; Schütz, G.; Umesono, K.; Blumberg, B.; Kastner, P.; Mark, M.; Chambon, P.; et al. The nuclear receptor superfamily: The second decade. Cell 1995, 83, 835–839. [Google Scholar] [CrossRef] [PubMed]
  3. Gronemeyer, H.; Gustafsson, J.Å.; Laudet, V. Principles for modulation of the nuclear receptor superfamily. Nat. Rev. Drug Discov. 2004, 3, 950–964. [Google Scholar] [CrossRef] [PubMed]
  4. Xu, P.; Zhai, Y.; Wang, J. The Role of PPAR and Its Cross-Talk with CAR and LXR in Obesity and Atherosclerosis. Int. J. Mol. Sci. 2018, 19, 1260. [Google Scholar] [CrossRef] [PubMed]
  5. Bain, D.L. Nuclear receptor structure: Implications for function. Annu. Rev. Physiol. 2007, 69, 201–220. [Google Scholar] [CrossRef]
  6. Echeverria, P.C.; Picard, D. Molecular chaperones, essential partners of steroid hormone receptors for activity and mobility. Biochim. Biophys. Acta 2010, 1803, 641–649. [Google Scholar] [CrossRef]
  7. Lazar, M.A. Maturing of the nuclear receptor family. J. Clin. Investig. 2017, 127, 1123–1125. [Google Scholar] [CrossRef]
  8. Hong, F.; Pan, S.; Guo, Y.; Xu, P.; Zhai, Y. PPARs as Nuclear Receptors for Nutrient and Energy Metabolism. Molecules 2019, 24, 2545. [Google Scholar] [CrossRef]
  9. Kinch, M.S.; Hoyer, D.; Patridge, E.; Plummer, M. Target selection for FDA-approved medicines. Drug Discov. Today 2015, 20, 784–789. [Google Scholar] [CrossRef]
  10. Sun, L.; Cai, J.; Gonzalez, F.J. The role of farnesoid X receptor in metabolic diseases, and gastrointestinal and liver cancer. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 335–347. [Google Scholar] [CrossRef]
  11. Zheng, X.; Chen, T.; Jiang, R.; Zhao, A.; Wu, Q.; Kuang, J.; Sun, D.; Ren, Z.; Li, M.; Zhao, M.; et al. Hyocholic acid species improve glucose homeostasis through a distinct TGR5 and FXR signaling mechanism. Cell Metab. 2021, 33, 791–803.e7. [Google Scholar] [CrossRef] [PubMed]
  12. Sun, L.; Xie, C.; Wang, G.; Wu, Y.; Wu, Q.; Wang, X.; Liu, J.; Deng, Y.; Xia, J.; Chen, B.; et al. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat. Med. 2018, 24, 1919–1929. [Google Scholar] [CrossRef] [PubMed]
  13. Chávez-Talavera, O.; Tailleux, A.; Lefebvre, P.; Staels, B. Bile Acid Control of Metabolism and Inflammation in Obesity, Type 2 Diabetes, Dyslipidemia, and Nonalcoholic Fatty Liver Disease. Gastroenterology 2017, 152, 1679–1694.e3. [Google Scholar] [CrossRef]
  14. Yang, J.; van Dijk, T.H.; Koehorst, M.; Havinga, R.; de Boer, J.F.; Kuipers, F.; van Zutphen, T. Intestinal Farnesoid X Receptor Modulates Duodenal Surface Area but Does Not Control Glucose Absorption in Mice. Int. J. Mol. Sci. 2023, 24, 4132. [Google Scholar] [CrossRef]
  15. Kim, E.Y.; Lee, J.M. Transcriptional Control of Trpm6 by the Nuclear Receptor FXR. Int. J. Mol. Sci. 2022, 23, 1980. [Google Scholar] [CrossRef] [PubMed]
  16. Wu, X.; Ni, Z.; Song, T.; Lv, W.; Chen, Y.; Huang, D.; Xie, Y.; Huang, W.; Niu, Y. C-Terminal Truncated HBx Facilitates Oncogenesis by Modulating Cell Cycle and Glucose Metabolism in FXR-Deficient Hepatocellular Carcinoma. Int. J. Mol. Sci. 2023, 24, 5174. [Google Scholar] [CrossRef]
  17. Guo, Y.; Xie, G.; Zhang, X. Role of FXR in Renal Physiology and Kidney Diseases. Int. J. Mol. Sci. 2023, 24, 2408. [Google Scholar] [CrossRef] [PubMed]
  18. Zappavigna, S.; Cossu, A.M.; Grimaldi, A.; Bocchetti, M.; Ferraro, G.A.; Nicoletti, G.F.; Filosa, R.; Caraglia, M. Anti-Inflammatory Drugs as Anticancer Agents. Int. J. Mol. Sci. 2020, 21, 2605. [Google Scholar] [CrossRef]
  19. Kazberuk, A.; Chalecka, M.; Palka, J.; Surazynski, A. Nonsteroidal Anti-Inflammatory Drugs as PPARgamma Agonists Can Induce PRODH/POX-Dependent Apoptosis in Breast Cancer Cells: New Alternative Pathway in NSAID-Induced Apoptosis. Int. J. Mol. Sci. 2022, 23, 1510. [Google Scholar] [CrossRef]
  20. Pérez-Segura, I.; Santiago-Balmaseda, A.; Rodríguez-Hernández, L.D.; Morales-Martínez, A.; Martínez-Becerril, H.A.; Martínez-Gómez, P.A.; Delgado-Minjares, K.M.; Salinas-Lara, C.; Martínez-Dávila, I.A.; Guerra-Crespo, M.; et al. PPARs and Their Neuroprotective Effects in Parkinson’s Disease: A Novel Therapeutic Approach in alpha-Synucleinopathy? Int. J. Mol. Sci. 2023, 24, 3264. [Google Scholar] [CrossRef]
  21. Hong, F.; Xu, P.; Zhai, Y. The Opportunities and Challenges of Peroxisome Proliferator-Activated Receptors Ligands in Clinical Drug Discovery and Development. Int. J. Mol. Sci. 2018, 19, 2189. [Google Scholar] [CrossRef] [PubMed]
  22. Cizkova, K.; Tauber, Z. Fibrates Affect Levels of Phosphorylated p38 in Intestinal Cells in a Differentiation-Dependent Manner. Int. J. Mol. Sci. 2023, 24, 7695. [Google Scholar] [CrossRef] [PubMed]
  23. Xu, P.; Hong, F.; Wang, J.; Dai, S.; Wang, J.; Zhai, Y. The CAR agonist TCPOBOP inhibits lipogenesis and promotes fibrosis in the mammary gland of adolescent female mice. Toxicol. Lett. 2018, 290, 29–35. [Google Scholar] [CrossRef] [PubMed]
  24. Pan, S. Environmental chemical TCPOBOP disrupts milk lipid homeostasis during pregnancy and lactation. Ecotoxicol. Environ. Saf. 2023, 249, 114463. [Google Scholar] [CrossRef]
  25. Pan, S.; Guo, Y.; Yu, W.; Zhang, J.; Qiao, X.; Li, L.; Xu, P.; Zhai, Y. Constitutive Androstane Receptor Agonist, TCPOBOP: Maternal Exposure Impairs the Growth and Development of Female Offspring in Mice. Int. J. Mol. Sci. 2023, 24, 2602. [Google Scholar] [CrossRef] [PubMed]
  26. Na, S.Y.; Kim, K.S.; Jung, Y.S.; Kim, D.K.; Kim, J.; Cho, S.J.; Lee, I.K.; Chung, J.; Kim, J.S.; Choi, H.S. An Inverse Agonist GSK5182 Increases Protein Stability of the Orphan Nuclear Receptor ERRgamma via Inhibition of Ubiquitination. Int. J. Mol. Sci. 2022, 24, 96. [Google Scholar] [CrossRef]
  27. Watanabe, M.; Sato, T.; Tsugeno, Y.; Higashide, M.; Furuhashi, M.; Umetsu, A.; Suzuki, S.; Ida, Y.; Hikage, F.; Ohguro, H. All-trans Retinoic Acids Synergistically and Beneficially Affect In Vitro Glaucomatous Trabecular Meshwork (TM) Models Using 2D and 3D Cell Cultures of Human TM Cells. Int. J. Mol. Sci. 2022, 23, 9912. [Google Scholar] [CrossRef]
  28. Pastori, V.; Pozzi, S.; Labedz, A.; Ahmed, S.; Ronchi, A.E. Role of Nuclear Receptors in Controlling Erythropoiesis. Int. J. Mol. Sci. 2022, 23, 2800. [Google Scholar] [CrossRef]
Figure 1. Schematic structure of NRs (nuclear receptors) and model of NR signaling. (A) General domain structure of NRs; (B) mechanism of class I NR action; and (C) mechanism of class II NR action.
Figure 1. Schematic structure of NRs (nuclear receptors) and model of NR signaling. (A) General domain structure of NRs; (B) mechanism of class I NR action; and (C) mechanism of class II NR action.
Ijms 24 09153 g001
Table 1. A summary of different NRs categorized according to their structure and sequence homology.
Table 1. A summary of different NRs categorized according to their structure and sequence homology.
SubfamilyNumberGroupNRNC SymbolAbbreviationName
1. Thyroid Hormone Receptor-like1.1Thyroid hormone receptorNR1A1TRαThyroid hormone receptor-α
NR1A2TRβThyroid hormone receptor-β
1.2Retinoic acid receptorNR1B1RARαRetinoic acid receptor-α
NR1B2RARβRetinoic acid receptor-β
NR1B3RARγRetinoic acid receptor-γ
1.3Peroxisome proliferator-activated receptorNR1C1PPARαPeroxisome proliferator-activated receptor-α
NR1C2PPAR-β/δPeroxisome proliferator-activated receptor-β/δ
NR1C3PPARγPeroxisome proliferator-activated receptor-γ
1.4Rev-ErbANR1D1Rev-ErbAαRev-ErbAα
NR1D2Rev-ErbAβRev-ErbAβ
1.5RAR-related orphan receptorNR1F1RORαRAR-related orphan receptor-α
NR1F2RORβRAR-related orphan receptor-β
NR1F3RORγRAR-related orphan receptor-γ
1.6Liver X receptor-likeNR1H2LXRβLiver X receptor-β
NR1H3LXRαLiver X receptor-α
NR1H4FXRFarnesoid X receptor
1.7Vitamin D receptor-likeNR1I1VDRVitamin D receptor
NR1I2PXRPregnane X receptor
NR1I3CARConstitutive androstane receptor
2. Retinoid X Receptor-like2.1Hepatocyte nuclear factor-4NR2A1HNF4αHepatocyte nuclear factor-4-α
NR2A2HNF4γHepatocyte nuclear factor-4-γ
2.2Retinoid X receptorNR2B1RXRαRetinoid X receptor-α
NR2B2RXRβRetinoid X receptor-β
NR2B3RXRγRetinoid X receptor-γ
2.3Testicular receptorNR2C1TR2Testicular receptor 2
NR2C2TR4Testicular receptor 4
2.4TLX/PNRNR2E1TLXHomologue of the Drosophila tailless gene
NR2E3PNRPhotoreceptor cell-specific nuclear receptor
2.5COUP/EARNR2F6EAR-2V-erbA-related
3. Estrogen Receptor-like3.1Estrogen receptorNR3A1ERαEstrogen receptor-α
NR3A2ERβEstrogen receptor-β
3.2Estrogen related receptorNR3B1ERRαEstrogen-related receptor-α
NR3B2ERRβEstrogen-related receptor-β
NR3B3ERRγEstrogen-related receptor-γ
3.33-Ketosteroid receptorsNR3C1GRGlucocorticoid receptor
NR3C2MRMineralocorticoid receptor
NR3C3PRProgesterone receptor
NR3C4ARAndrogen receptor
4. Nerve Growth Factor IB-like4.1NGFIB/NURR1/NOR1NR4A1NGFIBNerve Growth factor IB
NR4A2NURR1Nuclear receptor related 1
NR4A3NOR1Neuron-derived orphan receptor 1
5. Steroidogenic Factor-like5.1SF1/LRH1NR5A1SF1Steroidogenic factor 1
NR5A2LRH-1Liver receptor homolog-1
6. Germ Cell Nuclear Factor-like6.1GCNFNR6A1GCNFGerm cell nuclear factor
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xu, P. Nuclear Receptors in Health and Diseases. Int. J. Mol. Sci. 2023, 24, 9153. https://doi.org/10.3390/ijms24119153

AMA Style

Xu P. Nuclear Receptors in Health and Diseases. International Journal of Molecular Sciences. 2023; 24(11):9153. https://doi.org/10.3390/ijms24119153

Chicago/Turabian Style

Xu, Pengfei. 2023. "Nuclear Receptors in Health and Diseases" International Journal of Molecular Sciences 24, no. 11: 9153. https://doi.org/10.3390/ijms24119153

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop