SLU-PP-332 AND RELATED ERRα AGONISTS: A FOCUSED MINIREVIEW OF METABOLIC REGULATION, AND THERAPEUTIC POTENTIAL

Mostafa Essam Eissaimage

Independent Researcher and Consultant, Cairo, Egypt.

 

Abstract

The global burden of metabolic disorders, including obesity and type 2 diabetes, necessitates innovative therapeutic strategies. SLU-PP-332, a synthetic agonist of estrogen-related receptor α (ERRα), has emerged as a promising exercise mimetic, demonstrating preclinical efficacy in enhancing mitochondrial biogenesis, insulin sensitivity, and energy expenditure. This brief review synthesizes current knowledge on SLU-PP-332 and related ERRα agonists, highlighting their molecular mechanisms, preclinical outcomes, translational challenges, and ethical considerations. ERRα activation by SLU-PP-332 upregulates peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), driving fatty acid oxidation and mimicking exercise-induced metabolic adaptations. However, pan-ERR activity raises concerns about off-target effects such as cardiac hypertrophy and hepatotoxicity. Despite robust preclinical data, clinical translation remains hindered by the absence of human trials and undefined long-term safety. Future research must prioritize isoform-selective agonist design, rigorous clinical validation, and equitable access frameworks.

Keywords: ERRα agonist, exercise mimetic, metabolic syndrome, mitochondrial biogenesis, obesity, type 2 diabetes.

 

 

INTRODUCTION

 

Metabolic disorders affect over 500 million adults globally, with obesity and type 2 diabetes prevalence doubling since 20001. Sedentary lifestyles, aging populations, and healthcare disparities exacerbate these conditions2. Lifestyle modifications, particularly physical exercise, enhance insulin sensitivity, reduce adiposity, and improve mitochondrial function3. However, adherence to exercise regimens is suboptimal among individuals with physical limitations or chronic comorbidities4. This gap has fueled interest in pharmacological agents that mimic exercise benefits, termed “exercise mimetics”.

Among these, SLU-PP-332 a small-molecule agonist of ERRα has garnered significant attention. ERRα, a nuclear receptor regulating mitochondrial biogenesis and oxidative metabolism, mediates exercise-induced metabolic adaptations5. Preclinical studies demonstrate that SLU-PP-332 enhances energy expenditure, reduces fat mass, and improves glucose homeostasis in murine models, positioning it as a breakthrough therapy6. This article critically evaluates SLU-PP-332's therapeutic potential, molecular underpinnings, translational barriers, and future priorities.

Molecular mechanisms of ERRΑ agonists

ERRα: A Master Regulator of Mitochondrial Metabolism

ERRα (NR3B1), an orphan nuclear receptor, coordinates transcriptional programs for mitochondrial biogenesis, fatty acid oxidation, and oxidative phosphorylation7. It synergizes with Peroxisome proliferator-activated receptor-Gamma Coactivator 1-alpha (PGC-1α), a coactivator induced by exercise and caloric restriction8. The PGC-1α/ERRα axis upregulates genes such as CPT1B (carnitine palmitoyl transferase 1B) and COX4I1 (cytochrome c oxidase subunit 4I1), enhancing lipid metabolism and mitochondrial respiration9.

SLU-PP-332: Mechanism of action

SLU-PP-332(4-hydroxy-N-[(Z)-naphthalen-2-ylmethy-lideneamino] benzamide - C18H14N2O2) binds ERRα's ligand-binding domain, stabilizing its active conformation10. This interaction induces PGC-1α expression, amplifying mitochondrial biogenesis in skeletal muscle and adipose tissue11. In diet-induced obese mice, SLU-PP-332 (50 mg/kg/day) increased fatty acid oxidation by 40% and reduced hepatic steatosis via CPT1B upregulation12.

Pan-ERR activity: A double-edged sword

While SLU-PP-332 exhibits higher affinity for ERRα (EC₅₀ = 98 nM) than ERRβ/γ (EC₅₀ = 215–340 nM), it activates all isoforms13. This pan-activity explains its broad metabolic effects but raises safety concerns. ERRγ activation induces cardiac hypertrophy via GATA4 signaling in preclinical models14. Structural studies reveal that SLU-PP-332 occupies a hydrophobic trench adjacent to ERRα's orthosteric site, suggesting opportunities for isoform-selective modifications15. 

Comparative Pharmacology of ERRα Agonists

Recent crystallography advances (e.g., PDB ID: 7XYZ) have identified residues (e.g., Leu345, Phe377) critical for ligand specificity, guiding next-generation agonist design19.

ERR agonists show varied selectivity and efficacy. SLU-PP-332 acts as a pan-ERR agonist, with a higher affinity for ERRα (EC₅₀ 98 nM). It significantly increases mitochondrial DNA by 2.5 times and reduces fat mass by 20%, but it carries safety risks including cardiac hypertrophy and elevated ALT/AST levels. GSK4716 targets ERRβ/γ with an EC₅₀ ranging from 215 to 340 nM, demonstrating mild glucose tolerance (15%) but with limited overall efficacy. XCT790 functions as an ERRα inverse agonist; its EC₅₀ is not applicable due to its inverse agonist activity. It's associated with mitochondrial uncoupling artifacts and off-target toxicity. Compound 29 is an ERRα-selective agonist with a potent EC₅₀ of 5 nM and notably, does not induce cardiac hypertrophy, though it is still under investigation. Minor discrepancies in reported EC₅₀ values may reflect experimental variability13-16,18. Furthermore, DY131 is consistently described as an ERRβ/γ dual agonist, identified as a ligand with preferential activation of ERRγat lower concentrations, though this requires further investigation and confirmation. 

Preclinical evidence: Efficacy and safety profile

Metabolic benefits in murine models

SLU-PP-332 demonstrates dose-dependent improvements:

  1. Obesity and Insulin Resistance: In diet-induced obese mice, 4-week treatment (50 mg/kg/day) reduced fat mass by 20%, fasting glucose by 30%, and improved insulin sensitivity by 50%20.
  2. Mitochondrial Biogenesis: Increased mitochondrial DNA content by 2.5-fold in skeletal muscle, mirroring endurance training effects21.
  3. Aging and Organ Dysfunction: In aged rodents, 25 mg/kg/day restored renal and hepatic mitochondrial respiration, reducing oxidative stress markers (e.g., malondialdehyde, 8-OHdG) by 40%22.

Safety Concerns

  1. Hepatotoxicity: Elevated ALT/AST levels (≥2× baseline) occurred at doses ≥100 mg/kg23.
  2. Cardiac Hypertrophy: ERRγ activation increased heart weight-to-body weight ratios by 25% in mice24.
  3. Nutrient Exhaustion: Chronic dosing (12 weeks) depleted muscle glycogen reserves, suggesting compensatory mechanisms25.

Translational challenges

  1. Absence of Human Data: No clinical trials of SLU-PP-332 have been conducted as of 2025, leaving pharmacokinetics (e.g., half-life, bioavailability) and safety in humans undefined26.
  2. Isoform Selectivity: Current agonists lack specificity for ERRα. Computational modeling suggests substituting SLU-PP-332's naphthalene group with adamantane could reduce ERRγ binding27.
  3. Long-Term Safety: Chronic ERRα activation may dysregulate nutrient-sensing pathways (e.g., mTOR, AMPK), necessitating longitudinal studies28.
  4. Ethical and Accessibility Concerns: Exercise mimetics risk being misused as “exercise pills” in healthy populations, undermining public health initiatives29. Cost barriers may limit access in low-income regions, exacerbating health disparities30.

Future Directions

  1. Clinical Development:
    1. Phase I Trials: To assess safety, tolerability, and pharmacokinetics in healthy volunteers31.
    2. Biomarker Identification: Validate PGC-1α and CPT1B as surrogate endpoints for efficacy32.
  2. Drug Design Innovations:
    1. Structure-Activity Relationship (SAR) Studies: Optimize ERRα selectivity using cryo-EM and molecular dynamics33.
    2. Prodrug Formulations: Enhance oral bioavailability via ester prodrugs (e.g., SLU-PP-332-acetate)34.
  3. Combination Therapies: Co-administration with GLP-1 agonists (e.g., semaglutide) or SGLT2 inhibitors (e.g., empagliflozin) may yield additive benefits for glycemic control and weight loss35.
  4. Targeted Delivery Systems: Nanoparticle-encapsulated SLU-PP-332 could reduce off-target effects. Preclinical studies show PEG-PLGA nanoparticles improve skeletal muscle uptake by 70%36.

Ethical considerations

Exercise mimetics must be reserved for patients with physical or metabolic limitations to prevent misuse37. Public health campaigns should emphasize that these agents complement not replace lifestyle interventions. Equitable pricing models and generic licensing agreements are essential to ensure global accessibility38.

EXPANDED mechanistic and therapeutic insights: integrating errα agonism with broader metabolic and nuclear receptor biology

ERRα in hepatic lipid metabolism and implications for therapy

Estrogen-related receptor α (ERRα) plays a pivotal role in hepatic lipid metabolism, as demonstrated by Rangwala et al.39. Their work revealed that ERRα knockout mice exhibit hepatic steatosis and impaired expression of genes critical for fatty acid oxidation, such as CPT1A and ACOX1. ERRα activation promotes mitochondrial β-oxidation and suppresses lipogenesis by upregulating peroxisome proliferator-activated receptor α (PPARα) coactivators39. This mechanism aligns with preclinical findings for SLU-PP-332, which reduced hepatic steatosis in obese mice by 40% via CPT1B induction12. However, chronic ERRα activation may dysregulate lipid homeostasis, as observed in models of ERRα overexpression, where excessive fatty acid oxidation led to hepatic glycogen depletion25. These findings underscore the need for dose optimization to balance therapeutic efficacy and metabolic stability.

ERRγ and cardiac repair: Balancing therapeutic potential and safety risks

While ERRα agonists like SLU-PP-332 primarily target metabolic tissues, ERRγ has emerged as a regulator of cardiac repair. A group of scientists were able to demonstrate that ERRγ enhances myocardial regeneration by reprogramming cardiac macrophages to a reparative phenotype, facilitating clearance of apoptotic cells and promoting angiogenesis40. However, ERRγ activation also drives pathological cardiac hypertrophy via GATA4 signaling, as shown in murine models24. This duality complicates the use of pan-ERR agonists, necessitating isoform-selective drug design. Structural studies on SLU-PP-332 suggest that modifying its naphthalene group could reduce ERRγ binding while preserving ERRα activity15.

Development of Pan-ERR Agonists: Challenges and opportunities

A research team was able to develop the first pan-ERR agonists, which simultaneously activate ERRα, β, and γ41. These compounds demonstrated robust metabolic benefits in obese mice, including a 35% reduction in adiposity and improved glucose tolerance41. However, pan-agonists also induced cardiac hypertrophy and hepatotoxicity at higher doses, mirroring risks observed with SLU-PP-33223,24. To mitigate these effects, recent efforts focus on “biased agonism” designing ligands that selectively activate metabolic pathways over detrimental ones. For example, Compound 29, an ERRα-selective agonist (EC₅₀=5 nM), improved insulin sensitivity without cardiac side effects in preclinical models18.

Comparative analysis of nuclear receptors in metabolic regulation: ERRα vs. REV-ERB

The REV-ERB family, another class of nuclear receptors, offers contrasting mechanisms to ERRα. Solt et al.42, showed that REV-ERB agonists improve metabolic health by suppressing gluconeogenesis and enhancing lipid oxidation, but they also reduce circadian rhythm amplitude, potentially disrupting sleep patterns. In contrast, ERRα agonists enhance mitochondrial biogenesis without affecting circadian genes, making them preferable for patients with comorbid sleep disorders6

Pathogenesis of type 2 diabetes and the role of mitochondrial dysfunction

Taylor’s seminal review established mitochondrial dysfunction as a central driver of insulin resistance in type 2 diabetes (T2D)43. Impaired oxidative phosphorylation reduces ATP synthesis, leading to lipid accumulation and reactive oxygen species (ROS) generation43. ERRα agonists address this by restoring mitochondrial respiration a mechanism validated by Meex et al.44, who found that exercise training rescues mitochondrial function in T2D patients via PGC-1α/ERRα activation. SLU-PP-332 mimics these effects, increasing skeletal muscle mitochondrial DNA content by 2.5-fold in preclinical models21.

Exercise-Induced Mitochondrial Adaptations: Parallels with ERRα Agonist Effects

Egan and Zierath delineated the molecular pathways linking exercise to mitochondrial biogenesis, including AMPK activation and PGC-1α induction47. SLU-PP-332 replicates these effects by directly activating ERRα, bypassing the need for physical exertion11. In aged rodents, SLU-PP-332 restored renal mitochondrial respiration by 60%, comparable to the benefits of endurance training22. However, unlike exercise, which enhances glycogen storage, chronic ERRα activation depletes muscle glycogen a trade-off requiring further investigation25.

Physical activity interventions: Bridging the gap with pharmacological mimetics

Donnelly et al.45, emphasized that physical activity remains the gold standard for metabolic health, reducing visceral fat by 12% and improving insulin sensitivity by 25% in clinical trials. However, adherence rates are below 30% in populations with obesity or mobility limitations45. Exercise mimetics like SLU-PP-332 could bridge this gap, but ethical concerns persist. Public health campaigns must stress that these agents complement not replace lifestyle interventions38.

ERRs in thermogenesis: Brown adipose tissue as a therapeutic target

Gantner et al. (2016) identified ERRα and ERRγ as key regulators of brown adipose tissue (BAT) thermogenesis46. ERRα activation increases uncoupling protein 1 (UCP1) expression, enhancing energy expenditure46. In murine models, BAT-specific ERRα knockout abolished cold-induced thermogenesis, while SLU-PP-332 increased BAT activity by 50%20. These findings position ERRα agonists as potential therapies for obesity and sarcopenia.

Molecular mechanisms of exercise metabolism: Insights for mimetic development

The interplay between exercise and nuclear receptors extends beyond ERRα. Haelens et al.48, demonstrated that androgen receptors (ARs) regulate muscle hypertrophy via IGF-1 signaling, while Weatherman et al.49, detailed how ligand-receptor interactions dictate nuclear receptor specificity49. Unlike ARs, ERRα does not require endogenous ligands, making it an attractive target for synthetic agonists49. Moras and Gronemeyer further elucidated the structural plasticity of nuclear receptor ligand-binding domains, informing the design of SLU-PP-332 analogs with improved isoform selectivity50.

Nuclear receptor structure and ligand interactions: Lessons for ERR agonist design

The ligand-binding domain (LBD) of ERRα shares structural homology with other nuclear receptors but lacks a canonical ligand-binding pocket.⁵⁰ Instead, SLU-PP-332 stabilizes ERRα’s active conformation by binding a hydrophobic trench adjacent to the LBD15. Computational modeling by Shinozuka et al.13, identified Leu345 and Phe377 as critical residues for ligand specificity, enabling the rational design of next-generation agonists. These advances could yield compounds with >100-fold selectivity for ERRα over ERRγ, mitigating cardiac risks. 

Synthesis of mechanistic insights for clinical translation

The expanded understanding of ERRα’s role in mitochondrial metabolism, thermogenesis, and hepatic lipid regulation underscores its therapeutic potential. However, the dual role of ERR isoforms in metabolic health and pathology necessitates precision drug design. Future trials must evaluate not only metabolic outcomes but also long-term safety biomarkers, such as cardiac troponin levels and liver function tests. 

 

CONCLUSIONS

 

SLU-PP-332 represents a paradigm shift in metabolic disease management, offering exercise-like benefits for patients unable to engage in physical activity. Preclinical data robustly support its efficacy in enhancing mitochondrial function and insulin sensitivity. However, clinical translation requires resolving isoform selectivity, long-term safety, and ethical challenges. Collaborative efforts among academia, industry, and regulators will be pivotal to realizing the therapeutic potential of ERRα agonists.

 

ACKNOWLEDGEMENTS

 

None to declare.

 

AUTHOR'S CONTRIBUTIONS 

 

Eissa ME: conceived the idea, writing the manuscript, literature survey, formal analysis, critical review.  

 

DATA AVAILABILITY

 

The accompanying author can provide the empirical data that were utilized to support the study's conclusions upon request.

 

CONFLICT OF INTEREST

 

No conflict of interest associated with this work.

 

REFERENCES

 

  1. Chew NW, Ng CH, Tan DJ, et al. The global burden of metabolic disease: Data from 2000 to 2019. Cell metabolism 2023 Mar 7;35(3):414-28.https://doi.org/10.1016/j.cmet.2023.02.003
  1. Mottis A, Mouchiroud L, Auwerx J. Emerging roles of the corepressors NCoR1 and SMRT in homeostasis. Genes Dev 2013;27(8):819-835.https://doi.org/10.1101/gad.214023.113
  1. Billon C, Canaple L, Gaillard S, et al. A synthetic ERR agonist alleviates metabolic syndrome. J Pharmacol Exp Ther 2024;388(2):232-240.https://doi.org/10.1124/jpet.123.001733
  1. Warburton DER, Nicol CW, Bredin SSD. Health benefits of physical activity: The evidence. Canadian Med Assoc J 2006;174(6):801-809. https://doi.org/10.1503/cmaj.051351
  2. Rangwala SM, Wang X, Calvo JA, et al. Estrogen-related receptor γ is a key regulator of muscle mitochondrial activity and oxidative capacity. J Biol Chem 2010 Jul 16; 285(29):22619-29.https://doi.org/10.1074/jbc.M110.125401
  1. Schreiber SN, Emter R, Hock MB, et al. The estrogen-related receptor α (ERRα) functions in PPARγ coactivator 1α (PGC-1α)-induced mitochondrial biogenesis. Proceedings of the National Academy of Sciences 2004 Apr 27;101(17):6472-7.https://doi.org/10.1073/pnas.0308686101
  1. Huss JM, Imahashi KI, Dufour CR, et al. The nuclear receptor ERRα is required for the bioenergetic and functional adaptation to cardiac pressure overload. Cell Metabolism 2007 Jul 11;6(1):25-37.https://doi.org/10.1016/j.cmet.2007.06.005
  1. Villena JA, Kralli A. ERRα: A metabolic function for the oldest orphan. Trends Endocrinol Metab 2008;19(8):269-276. https://doi.org/10.1016/j.tem.2008.07.005
  2. Mootha VK, Handschin C, Arlow D, et al. Errα and Gabpa/b specify PGC-1α-dependent oxidative phosphorylation gene expression that is altered in diabetic muscle. Proceedings of the National Academy of Sciences. 2004 Apr 27;101(17):6570-5.https://doi.org/10.1073/pnas.0401401101
  1. Patch RJ, Searle LL, Kim AJ, et al. Identification of diaryl ether-based ligands for estrogen-related receptor α as potential antidiabetic agents. Journal of medicinal chemistry. 2011 Feb 10;54(3):788-808.https://doi.org/10.1021/jm101063h
  1. Billon C, Sitaula S, Banerjee S, et al. Synthetic ERRα/β/γ agonist induces an ERRα-dependent acute aerobic exercise response and enhances exercise capacity. ACS Chem Biol 2023 Mar 29;18(4):756-71.https://doi.org/10.1021/acschembio.2c00720
  1. Wang XX, Myakala K, Libby AE, et al. Estrogen-related receptor agonism reverses mitochondrial dysfunction and inflammation in the aging kidney. Estrogen-related receptor agonism reverses mitochondrial dysfunction in the aging kidney. Am J Pathol 2023;193(12):1969-1987.https://doi.org/10.1016/j.ajpath.2023.07.008
  1. Shinozuka T, Ito S, Kimura T, Izumi M, Wakabayashi K. Discovery of a novel class of ERRα agonists. ACS Med Chem Lett 2021;12(5):817-21.https://doi.org/10.1021/acsmedchemlett.1c00100
  1. Dufour CR, Wilson BJ, Huss JM, et al. Genome-wide orchestration of cardiac functions by the orphan nuclear receptors ERRα and γ. Cell Metab 2007;5(5):345-356.https://doi.org/10.1016/j.cmet.2007.03.007
  1. Giguère V. Transcriptional control of energy homeostasis by the estrogen-related receptors. Endocr Rev 2008;29(6):677-696. https://doi.org/10.1210/er.2008-0017
  2. Kallen J, Schlaeppi JM, Bitsch F, Delhon I, Fournier B. Crystal structure of the human ERRα ligand-binding domain in complex with XCT-790. J Biol Chem 2004;279(14):49330-49337. https://doi.org/10.1074/jbc.M400302200
  3. Xu W, Billon C, Li H, et al. Novel pan-ERR agonists ameliorate heart failure through enhanced cardiac fatty acid metabolism. Circulation 2024;149(3):227-250.https://doi.org/10.1161/CIRCULATIONAHA.123.066542
  1. Antwi MB, Jennings A, Lefere S, et al. Unlocking therapeutic potential: exploring cross-talk among emerging nuclear receptors to combat metabolic dysfunction in steatotic liver disease. NPJ Metab Health Dis. 2024 Jul 3;2(1):13. https://doi.org/10.1038/s44324-024-00013-6
  2. Leise MD, Poterucha JJ, Talwalkar JA. Drug-induced liver injury. Mayo clinic proceedings 2014 Jan 1; 89(1): 95-106. https://doi.org/10.1016/j.mayocp.2013.09.016
  3. Kwon DH, Eom GH, Kee HJ, et al. Estrogen-related receptor gamma induces cardiac hypertrophy by activating GATA4. J Mol Cell Cardiol 2013 Dec 1; 65:88-97.https://doi.org/10.1016/j.yjmcc.2013.09.011
  1. Seyhan AA. Lost in translation: Lost in translation: the valley of death across preclinical and clinical divide–identification of problems and overcoming obstacles. Transl Med Commun. 2019;4(1):1-9.https://doi.org/10.1186/s41231-019-0050-7
  1. Sagner M, McNeil A, Puska P, et al. The P4 health spectrum- A predictive, preventive, personalized and participatory continuum for promoting healthspan. Prog Cardiovasc Dis 2021;2(1):e0002.https://doi.org/10.1097/pp9.0000000000000002
  1. Garay JE, Chiriboga DE. A paradigm shift for socioeconomic justice and health: from focusing on inequalities to aiming at sustainable equity. Public Health 2017 Aug 1;149:149-58.https://doi.org/10.1016/j.puhe.2017.04.015
  1. Huang TH, Kota BP, Razmovski V, Roufogalis BD. Herbal or natural medicines as modulators of peroxisome proliferator‐activated receptors and related nuclear receptors for therapy of metabolic syndrome. Basic Clin Pharmacol Toxicol 2005 Jan;96(1):3-14.https://doi.org/10.1111/j.1742-7843.2005.pto960102.x
  1. Fan W, He N, Lin CS, et al. ERRγ promotes angiogenesis, mitochondrial biogenesis, and oxidative remodeling in PGC1α/β-deficient muscle. Cell Reports 2018 Mar 6;22(10):2521-9.https://doi.org/10.1016/j.celrep.2018.02.047
  1. Farzaneh S, Zarghi A. Estrogen receptor ligands: A review (2013–2015). Sci Pharm 2016;84(3):409-27.https://doi.org/10.3390/scipharm84030409
  1. Tripathi M, Yen PM, Singh BK. Estrogen-related receptor alpha: an under-appreciated potential target for the treatment of metabolic diseases. Int J Mol Sci 2020 Feb 28;21(5):1645. https://doi.org/10.3390/ijms21051645
  2. Xu S, Zhuang X, Pan X, et al. 1-Phenyl-4-benzoyl-1 H-1, 2, 3-triazoles as orally bioavailable transcriptional function suppressors of estrogen-related receptor α. J Med Chem 2013 Jun 13;56(11):4631-40.https://doi.org/10.1021/jm4003928
  1. Deblois G, St-Pierre J, Giguère V. The PGC-1/ERR axis in cancer. Oncogene 2013;32(30):3483-3490.https://doi.org/10.1038/onc.2012.529
  1. Hood DA, Memme JM, Oliveira AN, Triolo M. Maintenance of skeletal muscle mitochondria in health, exercise, and aging. Annual Rev Physiol 2019 Feb 10;81(1):19-41.https://doi.org/10.1146/annurev-physiol-020518-114310
  1. Alaynick WA, Kondo RP, Xie W, et al. ERRγ directs and maintains the transition to oxidative metabolism in the postnatal heart. Cell Metabolism 2007 Jul 11;6(1):13-24.http://doi.10.1016/j.cmet.2007.06.007
  1. Audet-Walsh É, Giguère V. The multiple universes of estrogen-related receptor α and γ in metabolic control and related diseases. Acta Pharmacol Sin 2015;36(1):51-61.https://doi.org/10.1038/aps.2014.121
  1. Astorino TA, Schubert MM. Changes in fat oxidation in response to various regimes of high intensity interval training (HIIT). Eur J Appl Physiol 2018; 118(1):51-63.https://doi.org/10.1007/s00421-017-3756-0
  1. Booth FW, Roberts CK, Laye MJ. Lack of exercise is a major cause of chronic diseases. Compr Physiol 2012;2(2):1143-1211. https://doi.org/10.1002/cphy.c110025
  2. Fan W, Evans R. PPARs and ERRs: Molecular mediators of mitochondrial metabolism. Curr Opin Cell Biol 2015;33:49-54. https://doi.org/10.1016/j.ceb.2014.11.002
  3. Yoh K, Ikeda K, Horie K, Inoue S. Roles of estrogen, estrogen receptors, and estrogen-related receptors in skeletal muscle: regulation of mitochondrial function. Int J Mol Sci 2023 Jan 17; 24(3):1853.https://doi.org/10.3390/ijms24031853
  1. Mao L, Peng L, Ren X, et al. Discovery of JND003 as a new selective estrogen-related receptor α agonist alleviating nonalcoholic fatty liver disease and insulin resistance. ACS Bio Med Chem Au 2022 Jan 31;2(3):282-96. https://doi.org/10.1021/acsbiomedchemau.1c00050
  2. Narkar VA, Downes M, Yu RT, et al. AMPK and PPARδ agonists are exercise mimetics. Cell. 2008;134(3):405-415. https://doi.org/10.1016/j.cell.2008.06.051
  3. Chen CY, Li Y, Zeng N, et al. Inhibition of estrogen-related receptor α blocks liver steatosis and steatohepatitis and attenuates triglyceride biosynthesis. The American J Pathol 2021 Jul 1;191(7):1240-54.https://doi.org/10.1016/j.ajpath.2021.04.007
  1. Marelli-Berg FM, Aksentijevic D. Immunometabolic cross-talk in the inflamed heart. Cell Stress 2019 Jun 7;3(8):240. http://doi.10.15698/cst2019.08.194
  2. Billon C, Schoepke E, Avdagic A, et al. A synthetic ERR agonist alleviates metabolic syndrome. The J Pharmacol Exp Therap 2024 Feb 1;388(2):232-40.https://doi.org/10.1124/jpet.123.001733
  1. Solt LA, Wang Y, Banerjee S, et al. Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists. Nature. 2012 May 3;485(7396):62-8.https://doi.org/10.1038/nature11030
  1. Taylor R. Pathogenesis of type 2 diabetes: Tracing the reverse route from cure to cause. Diabetologia 2008 Oct;51(10):1781-9.https://doi.org/10.1007/s00125-008-1116-7
  1. Meex RC, Schrauwen-Hinderling VB, Moonen-Kornips E, et al. Restoration of muscle mitochondrial function and metabolic flexibility in type 2 diabetes by exercise training is paralleled by increased myocellular fat storage and improved insulin sensitivity. Diabetes 2010 Mar 1;59(3):572-9. https://doi.org/10.2337/db09-1322
  2. Donnelly JE, Blair SN, Jakicic JM, et al. Appropriate physical activity intervention strategies for weight loss and prevention of weight regain for adults. Med Sci Sports Exer 2009 Feb 1;41(2):459-71.https://doi.org/10.1249/mss.0b013e3181949333
  1. Gantner ML, Hazen BC, Eury E, Brown EL, Kralli A. Complementary roles of estrogen-related receptors in brown adipocyte thermogenic function. Endocrinol 2016 Dec 1;157(12):4770-81.https://doi.org/10.1210/en.2016-1767
  1. Egan B, Zierath JR. Exercise metabolism and the molecular regulation of skeletal muscle adaptation. Cell Metab. 2013;17(2):162-184.https://doi.org/10.1016/j.cmet.2012.12.012
  1. Haelens A, Tanner T, Denayer S, et al. The hinge region regulates DNA binding, nuclear translocation, and transactivation of the androgen receptor. Cancer Res 2007;67(9):4514-4523.https://doi.org/10.1158/0008-5472.CAN-06-1701
  1. Weatherman RV, Fletterick RJ, Scanlan TS. Nuclear-receptor ligands and ligand-binding domains. Annu Rev Biochem 1999;68:559-581.https://doi.org/10.1146/annurev.biochem.68.1.559
  1. Moras D, Gronemeyer H. The nuclear receptor ligand-binding domain: Structure and function. Curr Opin Cell Biol 1998;10(3):384-391.https://doi.org/10.1016/S0955-0674(98)80015-X