1
|
Ren Y, Mao X, Lin W, Chen Y, Chen R, Sun P. Targeting estrogen-related receptors to mitigate tumor resistance: A comprehensive approach to bridging cellular energy metabolism. Biochim Biophys Acta Rev Cancer 2025; 1880:189256. [PMID: 39743156 DOI: 10.1016/j.bbcan.2024.189256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
The war between humanity and malignant tumors has been ongoing, with continuous advancements in classic chemotherapy and radiotherapy regimens, targeted drugs, endocrine therapy, and immunotherapy. However, tumor cells can develop primary or secondary resistance to these treatment options, making the issue of tumor resistance a major factor affecting patient prognosis and leading to recurrence. Estrogen-related receptors (ERRs) are members of the nuclear receptor superfamily, primarily involved in regulating glucose, lipid, and amino acid metabolism, serving as a central hub for intracellular energy metabolism. ERRs not only mediate insulin resistance but also participate in the mechanisms of drug resistance in various tumors, including breast cancer, osteosarcoma, endometrial cancer, lung cancer, and liver cancer, and even mediate resistance to radiation and immunotherapy. They mainly resist tumor treatment methods through metabolic reprogramming within cells, affecting mitochondrial energy metabolism, regulating metabolites such as cholesterol, glutamine, and lactate, or other signaling pathways, or by influencing the immune microenvironment. ERRs are promising targets for addressing the dilemma of tumor resistance. Currently, electrochemical luminescence biosensors for detecting ERRα in bodily fluids have been developed, making large-scale, low-cost detection of ERRα possible. Additionally, targeted inhibitors of ERRs have shown significant effects in suppressing cancer cell proliferation and reversing tumor resistance. This article reviews the research progress of ERRs in tumor resistance, providing important references for developing more effective anti-tumor treatment strategies.
Collapse
Affiliation(s)
- Yuan Ren
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Xiaodan Mao
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Wenyu Lin
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Yi Chen
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| | - Rongfeng Chen
- National Center for Occupational Safety and Health, Beijing, 102308, China; NHC Key Laboratory for Engineering Control of Dust Hazard, Beijing 102308, China
| | - Pengming Sun
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China; Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; School of Group Medicine and Public Health, Peking Union Medical College, Beijing 100091, China.
| |
Collapse
|
2
|
Fox SN, Savage CH, Amireddy NR, McMeekin LJ, Crossman DK, Detloff PJ, Gray M, Cowell RM. Estrogen-related receptor gamma is a regulator of mitochondrial, autophagy, and immediate-early gene programs in spiny projection neurons: Relevance for transcriptional changes in Huntington disease. Neurobiol Dis 2025; 206:106818. [PMID: 39884587 DOI: 10.1016/j.nbd.2025.106818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025] Open
Abstract
Mitochondrial dysfunction, transcriptional dysregulation, and protein aggregation are hallmarks of multiple neurodegenerative disorders, including Huntington's disease (HD). Strategies are needed to counteract these processes to restore neuronal health and function in HD. Recent evidence indicates that the transcription factor estrogen-related receptor gamma (ERRγ/Esrrg) is required for normal expression of mitochondrial, synaptic, and autophagy genes in neurons. Further, overexpression of Esrrg in dopaminergic neurons reduces synuclein load in the pre-formed fibril model of synucleinopathy. For these reasons, we sought to understand ERRγ's role in transcriptional regulation in spiny projection neurons (SPNs), one of the neuronal populations vulnerable to transcriptional dysregulation, mitochondrial dysfunction, and protein aggregation in HD. Here, we demonstrate that developmental deletion of Esrrg selectively in SPNs causes a transcriptional pattern consistent with a reduction of Drd1 and Drd2-positive neurons in the mouse dorsolateral striatum. To avoid effects of developmental deletion and explore Esrrg's role within adult SPN populations, we deleted or overexpressed Esrrg in adult SPNs. While overexpression was sufficient to increase the expression of mitochondrial and lysosome-related transcripts, Esrrg deletion surprisingly caused increased expression of immediate-early genes and genes with enrichment of binding sites for transcriptional repressors. In contrast, these genes were downregulated by Esrrg overexpression. Concordantly, Esrrg-deficient mice exhibited lack of amphetamine-induced hyperactivity and further upregulation of immediate-early genes. To determine whether the alterations observed with ERRγ modulation have any relevance for understanding transcriptional changes in SPNs in neurodegeneration, we measured Esrrg and its responsive genes in two mouse models of HD. We found an increase in Esrrg expression in HD models, accompanied by a transcriptional profile with similarities to that observed with Esrrg overexpression, suggesting the existence of an ERRγ-dependent, stress-related response. Altogether, these studies suggest that ERRγ is a key activator of mitochondrial and lysosomal transcripts in SPNs with a potential bi-functional role as a mediator of immediate-early gene repression. Ongoing studies are investigating mechanisms underlying ERRγ's roles in transcriptional activation and repression in SPNs to inform strategies to promote neuroprotective actions of ERRγ in SPNs in HD.
Collapse
Affiliation(s)
- Stephanie N Fox
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Southern Research, Birmingham, AL 35205, USA.
| | - Cody H Savage
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Southern Research, Birmingham, AL 35205, USA
| | - Narcy R Amireddy
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | | - David K Crossman
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Peter J Detloff
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michelle Gray
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Rita M Cowell
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Southern Research, Birmingham, AL 35205, USA.
| |
Collapse
|
3
|
Cheng SS, Mody AC, Woo CM. Opportunities for Therapeutic Modulation of O-GlcNAc. Chem Rev 2024; 124:12918-13019. [PMID: 39509538 DOI: 10.1021/acs.chemrev.4c00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
O-Linked β-N-acetylglucosamine (O-GlcNAc) is an essential, dynamic monosaccharide post-translational modification (PTM) found on serine and threonine residues of thousands of nucleocytoplasmic proteins. The installation and removal of O-GlcNAc is controlled by a single pair of enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Since its discovery four decades ago, O-GlcNAc has been found on diverse classes of proteins, playing important functional roles in many cellular processes. Dysregulation of O-GlcNAc homeostasis has been implicated in the pathogenesis of disease, including neurodegeneration, X-linked intellectual disability (XLID), cancer, diabetes, and immunological disorders. These foundational studies of O-GlcNAc in disease biology have motivated efforts to target O-GlcNAc therapeutically, with multiple clinical candidates under evaluation. In this review, we describe the characterization and biochemistry of OGT and OGA, cellular O-GlcNAc regulation, development of OGT and OGA inhibitors, O-GlcNAc in pathophysiology, clinical progress of O-GlcNAc modulators, and emerging opportunities for targeting O-GlcNAc. This comprehensive resource should motivate further study into O-GlcNAc function and inspire strategies for therapeutic modulation of O-GlcNAc.
Collapse
Affiliation(s)
- Steven S Cheng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Alison C Mody
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Affiliate member of the Broad Institute, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
4
|
Sadasivam N, Park WR, Choi B, Seok Jung Y, Choi HS, Kim DK. Exploring the impact of estrogen-related receptor gamma on metabolism and disease. Steroids 2024; 211:109500. [PMID: 39159854 DOI: 10.1016/j.steroids.2024.109500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Estrogen-related receptor gamma (ERRγ) is a member of the ERR orphan nuclear receptor family which possesses three subtypes, α, β, and γ. ERRγ is reportedly predominantly expressed in metabolically active tissues and cells, which promotes positive and negative effects in different tissues. ERRγ overexpression in the liver, pancreas, and thyroid cells is related to liver cancer, oxidative stress, reactive oxygen species (ROS) regulation, and carcinoma. Reduced ERRγ expression in the brain, immune cells, tumor cells, and energy metabolism causes neurological dysfunction, gastric cancer, and obesity. ERRγ is a constitutive receptor; however, its transcriptional activity also depends on co-regulators, agonists, and antagonists, which, when after forming a complex, can play a role in targeting and treating diseases. Moreover, ERRγ has proven crucial in regulating cellular and metabolic activity. However, many functions mediated via ERRγ remain unknown and require further exploration. Hence, considering the importance of ERRγ, this review focuses on the critical findings and interactions between ERRγ and co-regulators, agonists, and antagonists alongside its relationship with downstream and upstream signaling pathways and diseases. This review highlights new findings and provides a path to understanding the current ideas and future studies on ERRγ-mediated cellular activity.
Collapse
Affiliation(s)
- Nanthini Sadasivam
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Woo-Ram Park
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea; Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Byungyoon Choi
- Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Yoon Seok Jung
- Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Republic of Korea; School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Hueng-Sik Choi
- Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Republic of Korea; School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Don-Kyu Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea; Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
5
|
Pathak RK, Kim JM. A computational assay for identifying millet-derived compounds that antagonize the interaction between bisphenols and estrogen-related receptor gamma. Front Pharmacol 2024; 15:1435254. [PMID: 39545070 PMCID: PMC11560427 DOI: 10.3389/fphar.2024.1435254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
The use of Bisphenol A (BPA) and its analogs in industries, as well as the products made from them, is becoming a significant concern for human health. Scientific studies have revealed that BPA functions as an endocrine disruptor. While some analogs of BPA (bisphenols) have been used for a longer time, it was later discovered that they are toxic, similar to BPA. Their widespread use ensures their presence in the environment, and thus, everyone is exposed to them. Scientific research has shown that BPA interacts with estrogen-related receptor gamma (ERRγ), affecting its normal function. ERRγ is involved in biological processes including energy metabolism and mitochondrial function. Therefore, continuous exposure to bisphenols increases the risk of various diseases. In our previous study, we observed that some analogs of BPA had a higher binding affinity to ERRγ compared to BPA itself and analyzed the amino acid residues involved in this interaction. We hypothesized that by antagonizing the interaction between bisphenols and ERRγ, we could neutralize their toxic effects. Taking into account the health benefits of millets and their toxin removal properties, virtual screening of millet-derived compounds was conducted along with prediction of their ADMET profiles. Top five candidates were prioritized for Density Functional Theory (DFT) calculations and further analyses. Long-term molecular dynamics simulation (1 µs) were utilized to evaluate their binding, stability, and antagonizing abilities. Furthermore, reevaluation of their binding energy was conducted using the MM-PBSA method. This study reports millet-derived compounds, namely, Tricin 7-rutinoside, Tricin 7-glucoside, Glucotricin, Kaempferol, and Setarin. These compounds are predicted to be potent competitive inhibitors that can antagonize the interactions between bisphenols and ERRγ. These compounds could potentially assist in the development of future therapeutics. They may also be considered for use as food supplements, although further investigations, including wet-lab experiments and clinical studies, are needed.
Collapse
Affiliation(s)
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong-si, Republic of Korea
| |
Collapse
|
6
|
Losby M, Hayes M, Valfort A, Sopariwala DH, Sanders R, Walker JK, Xu W, Narkar VA, Zhang L, Billon C, Burris TP. The Estrogen Receptor-Related Orphan Receptors Regulate Autophagy through TFEB. Mol Pharmacol 2024; 106:164-172. [PMID: 39168657 PMCID: PMC11413914 DOI: 10.1124/molpharm.124.000889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
Autophagy is an essential self-degradative and recycling mechanism that maintains cellular homeostasis. Estrogen receptor-related orphan receptors (ERRs) are fundamental in regulating cardiac metabolism and function. Previously, we showed that ERR agonists improve cardiac function in models of heart failure and induce autophagy. Here, we characterized a mechanism by which ERRs induce the autophagy pathway in cardiomyocytes. Transcription factor EB (TFEB) is a master regulator of the autophagy-lysosome pathway and has been shown to be crucial regulator of genes that control autophagy. We discovered that TFEB is a direct ERR target gene whose expression is induced by ERR agonists. Activation of ERR results in increased TFEB expression in both neonatal rat ventricular myocytes and C2C12 myoblasts. An ERR-dependent increase in TFEB expression results in increased expression of an array of TFEB target genes, which are critical for the stimulation of autophagy. Pharmacologically targeting ERR is a promising potential method for the treatment of many diseases where stimulation of autophagy may be therapeutic, including heart failure. SIGNIFICANCE STATEMENT: Estrogen receptor-related receptor agonists function as exercise mimetics and also display efficacy in animal models of metabolic disease, obesity, and heart failure.
Collapse
Affiliation(s)
- McKenna Losby
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Matthew Hayes
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Aurore Valfort
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Danesh H Sopariwala
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Ryan Sanders
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - John K Walker
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Weiyi Xu
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Vihang A Narkar
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Lilei Zhang
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Cyrielle Billon
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Thomas P Burris
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| |
Collapse
|
7
|
Chen W, Song YS, Lee HS, Lin CW, Lee J, Kang YE, Kim SK, Kim SY, Park YJ, Park JI. Estrogen-related receptor alpha promotes thyroid tumor cell survival via a tumor subtype-specific regulation of target gene networks. Oncogene 2024; 43:2431-2446. [PMID: 38937602 PMCID: PMC11629884 DOI: 10.1038/s41388-024-03078-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
Mortalin (encoded by HSPA9) is a mitochondrial chaperone often overexpressed in cancer through as-yet-unknown mechanisms. By searching different RNA-sequencing datasets, we found that ESRRA is a transcription factor highly correlated with HSPA9 in thyroid cancer, especially in follicular, but not C cell-originated, tumors. Consistent with this correlation, ESRRA depletion decreased mortalin expression only in follicular thyroid tumor cells. Further, ESRRA expression and activity were relatively high in thyroid tumors with oncocytic characteristics, wherein ESRRA and mortalin exhibited relatively high functional overlap. Mechanistically, ESRRA directly regulated HSPA9 transcription through a novel ESRRA-responsive element located upstream of the HSPA9 promoter. Physiologically, ESRRA depletion suppressed thyroid tumor cell survival via caspase-dependent apoptosis, which ectopic mortalin expression substantially abrogated. ESRRA depletion also effectively suppressed tumor growth and mortalin expression in the xenografts of oncocytic or ESRRA-overexpressing human thyroid tumor cells in mice. Notably, our Bioinformatics analyses of patient data revealed two ESRRA target gene clusters that contrast oncocytic-like and anaplastic features of follicular thyroid tumors. These findings suggest that ESRRA is a tumor-specific regulator of mortalin expression, the ESRRA-mortalin axis has higher significance in tumors with oncocytic characteristics, and ESRRA target gene networks can refine molecular classification of thyroid cancer.
Collapse
Affiliation(s)
- Wenjing Chen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Young Shin Song
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Han Sai Lee
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chien-Wei Lin
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Junguee Lee
- Department of Pathology, Konyang University School of Medicine, Daejeon, Republic of Korea
| | - Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital & College of Medicine, Daejeon, Republic of Korea
| | - Seon-Kyu Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seon-Young Kim
- Korea Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
8
|
Jung YS, Radhakrishnan K, Hammad S, Müller S, Müller J, Noh JR, Kim J, Lee IK, Cho SJ, Kim DK, Kim YH, Lee CH, Dooley S, Choi HS. ERRγ-inducible FGF23 promotes alcoholic liver injury through enhancing CYP2E1 mediated hepatic oxidative stress. Redox Biol 2024; 71:103107. [PMID: 38479224 PMCID: PMC10950689 DOI: 10.1016/j.redox.2024.103107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/24/2024] Open
Abstract
Fibroblast growth factor 23 (FGF23) is a member of endocrine FGF family, along with FGF15/19 and FGF21. Recent reports showed that under pathological conditions, liver produces FGF23, although the role of hepatic FGF23 remains nebulous. Here, we investigated the role of hepatic FGF23 in alcoholic liver disease (ALD) and delineated the underlying molecular mechanism. FGF23 expression was compared in livers from alcoholic hepatitis patients and healthy controls. The role of FGF23 was examined in hepatocyte-specific knock-out (LKO) mice of cannabinoid receptor type 1 (CB1R), estrogen related receptor γ (ERRγ), or FGF23. Animals were fed with an alcohol-containing liquid diet alone or in combination with ERRγ inverse agonist. FGF23 is mainly expressed in hepatocytes in the human liver, and it is upregulated in ALD patients. In mice, chronic alcohol feeding leads to liver damage and induced FGF23 in liver, but not in other organs. FGF23 is transcriptionally regulated by ERRγ in response to alcohol-mediated activation of the CB1R. Alcohol induced upregulation of hepatic FGF23 and plasma FGF23 levels is lost in ERRγ-LKO mice, and an inverse agonist mediated inhibition of ERRγ transactivation significantly improved alcoholic liver damage. Moreover, hepatic CYP2E1 induction in response to alcohol is FGF23 dependent. In line, FGF23-LKO mice display decreased hepatic CYP2E1 expression and improved ALD through reduced hepatocyte apoptosis and oxidative stress. We recognized CBIR-ERRγ-FGF23 axis in facilitating ALD pathology through hepatic CYP2E1 induction. Thus, we propose FGF23 as a potential therapeutic target to treat ALD.
Collapse
Affiliation(s)
- Yoon Seok Jung
- Host-derived Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kamalakannan Radhakrishnan
- Host-derived Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seddik Hammad
- Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany; Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Sebastian Müller
- Center for Alcohol Research (CAR), University of Heidelberg, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| | - Johannes Müller
- Center for Alcohol Research (CAR), University of Heidelberg, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| | - Jung-Ran Noh
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea; Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41940, Republic of Korea
| | - Sung Jin Cho
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Don-Kyu Kim
- Host-derived Antiviral Research Center, Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yong-Hoon Kim
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Chul-Ho Lee
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Steven Dooley
- Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany.
| | - Hueng-Sik Choi
- Host-derived Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
9
|
Jung YS, Radhakrishnan K, Kim HJ, Kim YH, Lee CH, Choi HS. Macrophage stimulating protein is a novel transcriptional target of estrogen related receptor gamma in alcohol-intoxicated mice. Cell Signal 2024; 116:111059. [PMID: 38237793 DOI: 10.1016/j.cellsig.2024.111059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Macrophage stimulating protein (MSP) is a multifunctional serum protein produced in the liver, belonging to the plasminogen-related kringle protein family. It exerts diverse biological functions by activating a transmembrane receptor protein-tyrosine kinase known as RON in humans and SKT in mice. MSP plays a pivotal role in innate immunity and is involved in various activities such as cell survival, migration, and phagocytosis. Elucidating the regulatory mechanisms governing MSP gene expression is of great importance. In this study, we comprehensively elucidate the molecular mechanism underlying hepatic MSP gene expression in response to alcoholism. Exposure to ethanol specifically upregulated the expression of ERRγ and MSP in the liver, while not in other organs. Liver-specific knockout of the cannabinoid receptor type 1 (CB1R), an upstream regulator of ERRγ, inhibited the alcohol-induced upregulation of MSP expression. Overexpression of ERRγ alone was sufficient to enhance MSP expression in hepatic cell lines and in mice. Conversely, knockdown of ERRγ in cell lines or liver-specific knockout of ERRγ in mice reversed ethanol-induced MSP gene expression. Promoter studies revealed the direct binding of ERRγ to the MSP gene promoter at the ERR response element (ERRE), resulting in the positive regulation of MSP gene expression in response to alcohol. This finding was further supported by ERRE-mutated MSP-luciferase reporter assays. Notably, treatment with GSK5182, an ERRγ-specific inverse agonist, significantly suppressed alcohol-induced hepatic MSP expression. Collectively, we exposed a novel mechanistic understanding of how alcohol-induced ERRγ controls the transcriptional regulation of MSP gene expression in the liver.
Collapse
Affiliation(s)
- Yoon Seok Jung
- Host-Directed Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kamalakannan Radhakrishnan
- Host-Directed Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hyo-Jin Kim
- Host-Directed Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yong-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Hueng-Sik Choi
- Host-Directed Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
10
|
Billon C, Schoepke E, Avdagic A, Chatterjee A, Butler AA, Elgendy B, Walker JK, Burris TP. A Synthetic ERR Agonist Alleviates Metabolic Syndrome. J Pharmacol Exp Ther 2024; 388:232-240. [PMID: 37739806 PMCID: PMC10801787 DOI: 10.1124/jpet.123.001733] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/27/2023] [Accepted: 08/07/2022] [Indexed: 09/24/2023] Open
Abstract
Physical exercise induces physiologic adaptations and is effective at reducing the risk of premature death from all causes. Pharmacological exercise mimetics may be effective in the treatment of a range of diseases including obesity and metabolic syndrome. Previously, we described the development of SLU-PP-332, an agonist for the estrogen-related receptor (ERR)α, β, and γ nuclear receptors that activates an acute aerobic exercise program. Here we examine the effects of this exercise mimetic in mouse models of obesity and metabolic syndrome. Diet-induced obese or ob/ob mice were administered SLU-PP-332, and the effects on a range of metabolic parameters were assessed. SLU-PP-332 administration mimics exercise-induced benefits on whole-body metabolism in mice including increased energy expenditure and fatty acid oxidation. These effects were accompanied by decreased fat mass accumulation. Additionally, the ERR agonist effectively reduced obesity and improved insulin sensitivity in models of metabolic syndrome. Pharmacological activation of ERR may be an effective method to treat metabolic syndrome and obesity. SIGNIFICANCE STATEMENT: An estrogen receptor-related orphan receptor agonist, SLU-PP-332, with exercise mimetic activity, holds promise as a therapeutic to treat metabolic diseases by decreasing fat mass in mouse models of obesity.
Collapse
Affiliation(s)
- Cyrielle Billon
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Emmalie Schoepke
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Amer Avdagic
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Arindam Chatterjee
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Andrew A Butler
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Bahaa Elgendy
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - John K Walker
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Thomas P Burris
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| |
Collapse
|
11
|
Zheng Y, Du Y, Zhang H, Lv H, Yan Z, Dong N, Li Q, Wang T. Research Progress in Estrogen-related Receptor Gamma (ERRγ) Agonists and Inverse Agonists. Curr Med Chem 2024; 31:3653-3667. [PMID: 37202889 DOI: 10.2174/0929867330666230518140631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/20/2023]
Abstract
Estrogen-related receptor gamma (ERRγ), one of three members of the ERR family, is an inducible transcription factor. ERRγ has dual functions in different tissues. The decreased expression of ERRγ in the brain, stomach, prostate, and fat cells can cause neuropsychological dysfunction, gastric cancer, prostate cancer, and obesity. However, when ERRγ is present in the liver, pancreas, and thyroid follicular cells, ERRγ overexpression is related to liver cancer, type II diabetes, oxidative liver injury, and anaplastic thyroid carcinoma. Signaling pathway studies have confirmed that ERRγ agonists or inverse agonists can regulate ERRγ expression to treat related diseases. The collision between residue Phe435 and the modulator is a key factor determining the activation or inhibition of ERRγ. Although more than 20 agonists and inverse agonists of ERRγ have been reported, no clinical studies have been found in the literature. This review summarizes the important relationship between ERRγ-related signaling pathways and diseases, research progress, and the structure-activity relationship of modulators. These findings provide guidance for further study on new ERRγ modulators.
Collapse
Grants
- 81872744,81873399,81901399,81872744,81973399,81901399,81872744,81973399,81901399 National Natural Science Foundation of China
- 81973399,81901399,81872744,81973399,81901399,81872744,81973399,81901399,81872744 National Natural Science Foundation of China (NSFC)
Collapse
Affiliation(s)
- Yong Zheng
- School of Chemistry & Chemical Engineering, Qilu University of Technology, Shandong Academy of Sciences, 3501 Da Xue Road, Jinan, 250353, China
| | - Yongli Du
- School of Chemistry & Chemical Engineering, Qilu University of Technology, Shandong Academy of Sciences, 3501 Da Xue Road, Jinan, 250353, China
| | - Haibin Zhang
- School of Chemistry & Chemical Engineering, Qilu University of Technology, Shandong Academy of Sciences, 3501 Da Xue Road, Jinan, 250353, China
| | - Huiting Lv
- School of Chemistry & Chemical Engineering, Qilu University of Technology, Shandong Academy of Sciences, 3501 Da Xue Road, Jinan, 250353, China
| | - Zhijia Yan
- School of Chemistry & Chemical Engineering, Qilu University of Technology, Shandong Academy of Sciences, 3501 Da Xue Road, Jinan, 250353, China
| | - Ning Dong
- School of Chemistry & Chemical Engineering, Qilu University of Technology, Shandong Academy of Sciences, 3501 Da Xue Road, Jinan, 250353, China
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital, Fudan University, 108 Luxiang Road, Shanghai 201907, China
| | - Tianxiao Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, 108 Luxiang Road, Shanghai 201907, China
| |
Collapse
|
12
|
Fan Y, Kim HJ, Jung YS, Na SY, Radhakrishnan K, Choi HS. Chenodeoxycholic acid regulates fibroblast growth factor 23 gene expression via estrogen-related receptor γ in human hepatoma Huh7 cells. Steroids 2023; 197:109257. [DOI: https:/doi.org/10.1016/j.steroids.2023.109257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/18/2023]
|
13
|
Dong N, Du Y, Zheng Y, Zhang H, Lv H, Yan Z. Research progress on tamoxifen and its analogs associated with nuclear receptors. Future Med Chem 2023; 15:1427-1442. [PMID: 37706220 DOI: 10.4155/fmc-2023-0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023] Open
Abstract
Tamoxifen, a triphenylethylene-based selective estrogen-receptor modulator, is a landmark drug for the treatment of breast cancer and is also used for treating liver cancer and osteoporosis. Structural studies of tamoxifen have led to the synthesis of more than 20 novel tamoxifen analogs as receptor modulators, including 16 ERα modulators 2-17, an ERRβ inverse agonist 19 and six ERRγ inverse agonists 20-25. This paper summarizes the research progress and structure-activity relationships of tamoxifen analogs modulating these three nuclear receptors reported in the literature, and introduces the relationship between these three nuclear receptor-mediated diseases and tamoxifen analogs to guide the research of novel tamoxifen analogs.
Collapse
Affiliation(s)
- Ning Dong
- School of Chemistry & Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Yongli Du
- School of Chemistry & Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Yong Zheng
- School of Chemistry & Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Haibin Zhang
- School of Chemistry & Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Huiting Lv
- School of Chemistry & Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Zhijia Yan
- School of Chemistry & Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| |
Collapse
|
14
|
Karri K, Waxman DJ. TCDD dysregulation of lncRNA expression, liver zonation and intercellular communication across the liver lobule. Toxicol Appl Pharmacol 2023; 471:116550. [PMID: 37172768 PMCID: PMC10330769 DOI: 10.1016/j.taap.2023.116550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
The persistent environmental aryl hydrocarbon receptor agonist and hepatotoxin TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) induces hepatic lipid accumulation (steatosis), inflammation (steatohepatitis) and fibrosis. Thousands of liver-expressed, nuclear-localized lncRNAs with regulatory potential have been identified; however, their roles in TCDD-induced hepatoxicity and liver disease are unknown. We analyzed single nucleus (sn)RNA-seq data from control and subchronic (4 wk) TCDD-exposed mouse liver to determine liver cell-type specificity, zonation and differential expression profiles for thousands of lncRNAs. TCDD dysregulated >4000 of these lncRNAs in one or more liver cell types, including 684 lncRNAs specifically dysregulated in liver non-parenchymal cells. Trajectory inference analysis revealed major disruption by TCDD of hepatocyte zonation, affecting >800 genes, including 121 lncRNAs, with strong enrichment for lipid metabolism genes. TCDD also dysregulated expression of >200 transcription factors, including 19 Nuclear Receptors, most notably in hepatocytes and Kupffer cells. TCDD-induced changes in cell-cell communication patterns included marked decreases in EGF signaling from hepatocytes to non-parenchymal cells and increases in extracellular matrix-receptor interactions central to liver fibrosis. Gene regulatory networks constructed from the snRNA-seq data identified TCDD-exposed liver network-essential lncRNA regulators linked to functions such as fatty acid metabolic process, peroxisome and xenobiotic metabolism. Networks were validated by the striking enrichments that predicted regulatory lncRNAs showed for specific biological pathways. These findings highlight the power of snRNA-seq to discover functional roles for many xenobiotic-responsive lncRNAs in both hepatocytes and liver non-parenchymal cells and to elucidate novel aspects of foreign chemical-induced hepatotoxicity and liver disease, including dysregulation of intercellular communication within the liver lobule.
Collapse
Affiliation(s)
- Kritika Karri
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
15
|
Fan Y, Kim HJ, Seok Jung Y, Na SY, Radhakrishnan K, Sik Choi H. Chenodeoxycholic acid regulates fibroblast growth factor 23 gene expression via estrogen-related receptor γ in human hepatoma Huh7 cells. Steroids 2023:109257. [PMID: 37301529 DOI: 10.1016/j.steroids.2023.109257] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
Fibroblast growth factor 23 (FGF23) is a glycoprotein that belongs to the FGF19 subfamily and participates in phosphate and vitamin D homeostasis. Chenodeoxycholic acid (CDCA), one of the primary bile acids, is reported to induce the secretion of FGF19 subfamily members, FGF21 and FGF19, in hepatocytes. However, whether and how CDCA influences FGF23 gene expression are largely unknown. Thus, we performed real-time polymerase chain reaction and Western blot analyses to determine the mRNA and protein expression levels of FGF23 in Huh7 cells. CDCA upregulated estrogen-related receptor γ (ERRγ) alongside FGF23 mRNA and protein levels, while, the knockdown of ERRγ ablated the induction effect of CDCA on FGF23 expression. Promoter studies showed that CDCA-induced FGF23 promoter activity occurred partly through ERRγ binding directly to the ERR response element (ERRE) in the human FGF23 gene promoter. Finally, the inverse agonist of ERRγ, GSK5182 inhibited the induction of FGF23 by CDCA. Overall, our results revealed the mechanism of CDCA-mediated FGF23 gene upregulation in the human hepatoma cell line. Moreover, the ability of GSK5182 to reduce CDCA-induced FGF23 gene expression might represent a therapeutic strategy to control abnormal FGF23 induction in conditions that involve elevated levels of bile acids, such as nonalcoholic fatty liver disease and biliary atresia.
Collapse
Affiliation(s)
- Yiwen Fan
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hyo-Jin Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yoon Seok Jung
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Soon-Young Na
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kamalakannan Radhakrishnan
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hueng Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
16
|
Fan Y, Na SY, Jung YS, Radhakrishnan K, Choi HS. Estrogen-related receptor γ (ERRγ) is a key regulator of lysyl oxidase gene expression in mouse hepatocytes. Steroids 2023; 194:109226. [PMID: 36948345 DOI: 10.1016/j.steroids.2023.109226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 03/24/2023]
Abstract
Lysyl oxidase (LOX), the copper-dependent extracellular enzyme, plays a critical role in the regulation of protein cross-linking in the extracellular matrix (ECM). It is also involved in liver regeneration and liver fibrosis. However, the mechanism of LOX regulation in mouse hepatocytes is still unclear. Here, we identify a molecular mechanism showing that orphan nuclear receptor estrogen-related receptor γ (ERRγ) regulates LOX gene expression in the presence of the pro-inflammatory cytokine, interleukin 6 (IL6). IL6 significantly stimulated the expression of ERRγ and LOX in mouse hepatocytes. Overexpression of ERRγ increased LOX mRNA and protein levels. Moreover, knockdown of ERRγ attenuated IL6-mediated LOX gene expression at mRNA and protein levels. Overexpression of ERRγ or IL6 treatment upregulated LOX gene promoter activity, while knockdown of ERRγ decreased the IL6-induced LOX promoter activity. Furthermore, GSK5182, a specific ERRγ inverse agonist, inhibited the induction effect of IL6 on LOX promoter activity and gene expression in mouse hepatocytes. Overall, our study elucidates the mechanism involved in the LOX gene regulation by nuclear receptor ERRγ in response to IL6 in mouse hepatocytes, suggesting that, in conditions such as chronic inflammation, IL6 may contribute to liver fibrosis via inducing LOX gene expression. Thus, LOX gene regulation by the inverse agonist of ERRγ can be applied to improve liver fibrosis.
Collapse
Affiliation(s)
- Yiwen Fan
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Soon-Young Na
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yoon Seok Jung
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kamalakannan Radhakrishnan
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
17
|
Structure-based discovery of pyrazolamides as novel ERRγ inverse agonists. Eur J Med Chem 2023; 250:115174. [PMID: 36805944 DOI: 10.1016/j.ejmech.2023.115174] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
Estrogen-related receptor-gamma (ERRγ) is an orphan nuclear receptor with high structural similarity to estrogen receptors (ERα and β). The endogenous ligand of the receptor has yet to be identified. Only two classes of molecules-stilbene (diethylstilbestrol, 4-hydroxytamoxifen, and GSK5182) and flavonol (kaempferol) have been known to modulate the transcriptional activity of the receptor to date. Further, these agents lack selectivity to ERRγ suggesting the need for a new inverse agonist. Thus, virtual screening was used to identify pyrazolamide 7 as a novel ERRγ inverse agonist. Structure-based diversification and optimization of the compound further led to the identification of derivative 19 as a potent inverse agonist of ERRγ with selectivity over other nuclear receptors including those of ERR family. Pyrazolamide 19 exhibits strong affinity towards ERRγ and inhibits the expression of hepcidin, fibrinogen and gluconeogenic genes, which suggests that these compounds may have antimicrobial, anti-coagulant and antidiabetic activities.
Collapse
|
18
|
An Inverse Agonist of Estrogen-Related Receptor Gamma, GSK5182, Enhances Na +/I - Symporter Function in Radioiodine-Refractory Papillary Thyroid Cancer Cells. Cells 2023; 12:cells12030470. [PMID: 36766812 PMCID: PMC9914548 DOI: 10.3390/cells12030470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Previously, we reported that an inverse agonist of estrogen-related receptor gamma (ERRγ), GSK5182, enhances sodium iodide (Na+/I-) symporter (NIS) function through mitogen-activated protein (MAP) kinase signaling in anaplastic thyroid cancer cells. This finding helped us to further investigate the effects of GSK5182 on NIS function in papillary thyroid cancer (PTC) refractory to radioactive iodine (RAI) therapy. Herein, we report the effects of ERRγ on the regulation of NIS function in RAI-resistant PTC cells using GSK5182. RAI-refractory BCPAP cells were treated with GK5182 for 24 h at various concentrations, and radioiodine avidity was determined with or without potassium perchlorate (KClO4) as an NIS inhibitor. We explored the effects of GSK5182 on ERRγ, the mitogen-activated protein (MAP) kinase pathway, and iodide metabolism-related genes. We examined whether the MAP pathway affected GSK5182-mediated NIS function using U0126, a selective MEK inhibitor. A clonogenic assay was performed to evaluate the cytotoxic effects of I-131. GSK5182 induced an increase in radioiodine avidity in a dose-dependent manner, and the enhanced uptake was completely inhibited by KClO4 in BCPAP cells. We found that ERRγ was downregulated and phosphorylated extracellular signal-regulated kinase (ERK)1/2 was upregulated in BCPAP cells, with an increase in total and membranous NIS and iodide metabolism-related genes. MEK inhibitors reversed the increase in radioiodine avidity induced by GSK5182. Clonogenic examination revealed the lowest survival in cells treated with a combination of GSK5182 and I-131 compared to those treated with either GSK518 or I-131 alone. We demonstrate that an inverse agonist of ERRγ, GSK5182, enhances the function of NIS protein via the modulation of ERRγ and MAP kinase signaling, thereby leading to increased responsiveness to radioiodine in RAI-refractory papillary thyroid cancer cells.
Collapse
|
19
|
Karri K, Waxman DJ. TCDD dysregulation of lncRNA expression, liver zonation and intercellular communication across the liver lobule. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.523119. [PMID: 36711947 PMCID: PMC9881922 DOI: 10.1101/2023.01.07.523119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The persistent environmental aryl hydrocarbon receptor agonist and hepatotoxin TCDD (2,3,7,8-tetrachlorodibenzo- p -dioxin) induces hepatic lipid accumulation (steatosis), inflammation (steatohepatitis) and fibrosis. Thousands of liver-expressed, nuclear-localized lncRNAs with regulatory potential have been identified; however, their roles in TCDD-induced hepatoxicity and liver disease are unknown. We analyzed single nucleus (sn)RNA-seq data from control and chronic TCDD-exposed mouse liver to determine liver cell-type specificity, zonation and differential expression profiles for thousands of IncRNAs. TCDD dysregulated >4,000 of these lncRNAs in one or more liver cell types, including 684 lncRNAs specifically dysregulated in liver non-parenchymal cells. Trajectory inference analysis revealed major disruption by TCDD of hepatocyte zonation, affecting >800 genes, including 121 IncRNAs, with strong enrichment for lipid metabolism genes. TCDD also dysregulated expression of >200 transcription factors, including 19 Nuclear Receptors, most notably in hepatocytes and Kupffer cells. TCDD-induced changes in cellâ€"cell communication patterns included marked decreases in EGF signaling from hepatocytes to non-parenchymal cells and increases in extracellular matrix-receptor interactions central to liver fibrosis. Gene regulatory networks constructed from the snRNA-seq data identified TCDD-exposed liver network-essential lncRNA regulators linked to functions such as fatty acid metabolic process, peroxisome and xenobiotic metabolic. Networks were validated by the striking enrichments that predicted regulatory IncRNAs showed for specific biological pathways. These findings highlight the power of snRNA-seq to discover functional roles for many xenobiotic-responsive lncRNAs in both hepatocytes and liver non-parenchymal cells and to elucidate novel aspects of foreign chemical-induced hepatotoxicity and liver disease, including dysregulation of intercellular communication within the liver lobule.
Collapse
|
20
|
Na SY, Kim KS, Jung YS, Kim DK, Kim J, Cho SJ, Lee IK, Chung J, Kim JS, Choi HS. An Inverse Agonist GSK5182 Increases Protein Stability of the Orphan Nuclear Receptor ERRγ via Inhibition of Ubiquitination. Int J Mol Sci 2022; 24:ijms24010096. [PMID: 36613556 PMCID: PMC9820335 DOI: 10.3390/ijms24010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The orphan nuclear receptor, estrogen-related receptor γ (ERRγ) is a constitutively active transcription factor involved in mitochondrial metabolism and energy homeostasis. GSK5182, a specific inverse agonist of ERRγ that inhibits transcriptional activity, induces a conformational change in ERRγ, resulting in a loss of coactivator binding. However, the molecular mechanism underlying the stabilization of the ERRγ protein by its inverse agonist remains largely unknown. In this study, we found that GSK5182 inhibited ubiquitination of ERRγ, thereby stabilizing the ERRγ protein, using cell-based assays and confocal image analysis. Y326 of ERRγ was essential for stabilization by GSK5182, as ligand-induced stabilization of ERRγ was not observed with the ERRγ-Y326A mutant. GSK5182 suppressed ubiquitination of ERRγ by the E3 ligase Parkin and subsequent degradation. The inhibitory activity of GSK5182 was strong even when the ERRγ protein level was elevated, as ERRγ bound to GSK5182 recruited a corepressor, small heterodimer partner-interacting leucine zipper (SMILE), through the activation function 2 (AF-2) domain, without alteration of the nuclear localization or DNA-binding ability of ERRγ. In addition, the AF-2 domain of ERRγ was critical for the regulation of protein stability. Mutants in the AF-2 domain were present at higher levels than the wild type in the absence of GSK5182. Furthermore, the ERRγ-L449A/L451A mutant was no longer susceptible to GSK5182. Thus, the AF-2 domain of ERRγ is responsible for the regulation of transcriptional activity and protein stability by GSK5182. These findings suggest that GSK5182 regulates ERRγ by a unique molecular mechanism, increasing the inactive form of ERRγ via inhibition of ubiquitination.
Collapse
Affiliation(s)
- Soon-Young Na
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ki-Sun Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yoon Seok Jung
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Don-Kyu Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jina Kim
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Sung Jin Cho
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41940, Republic of Korea
| | - Jongkyeong Chung
- SRC Center for Systems Geroscience, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Sun Kim
- Department of Chemistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
- Correspondence: ; Tel.: +82-62-530-0503
| |
Collapse
|
21
|
Melnik BC, Schmitz G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int J Mol Sci 2022; 23:ijms231911503. [PMID: 36232796 PMCID: PMC9569743 DOI: 10.3390/ijms231911503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic β cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with β cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic β cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal β cell proliferation and glycolysis, but attenuate β cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching β cells from proliferation to TGF-β/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse β cell differentiation, promoting β cell de-differentiation. Whereas MEX miR signaling supports postnatal β cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate β cells back to the postnatal phenotype (diabetes induction).
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-52-4198-8060
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
22
|
Choi B, Park WR, Kim YJ, Mun S, Park SJ, Jeong JH, Choi HS, Kim DK. Nuclear receptor estrogen-related receptor modulates antimicrobial peptide expression for host innate immunity in Tribolium castaneum. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 148:103816. [PMID: 35926689 DOI: 10.1016/j.ibmb.2022.103816] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 07/15/2022] [Accepted: 07/26/2022] [Indexed: 06/15/2023]
Abstract
Antimicrobial peptides (AMPs) are core components of innate immunity to protect insects against microbial infections. Nuclear receptors (NRs) are ligand-dependent transcription factors that can regulate the expression of genes critical for insect development including molting and metamorphosis. However, the role of NRs in host innate immune response to microbial infection remains poorly understood in Tribolium castaneum (T. castaneum). Here, we show that estrogen-related receptor (ERR), an insect ortholog of the mammalian ERR family of NRs, is a novel transcriptional regulator of AMP genes for innate immune response of T. castaneum. Tribolium ERR (TcERR) expression was induced by immune deficiency (IMD)-Relish signaling in response to infection by Escherichia coli (E. coli), a Gram-negative bacterium, as demonstrated in TcIMD-deficient beetles. Interestingly, genome-wide transcriptome analysis of TcERR-deficient old larvae using RNA-sequencing analysis showed that TcERR expression was positively correlated with gene transcription levels of AMPs including attacins, defensins, and coleoptericin. Moreover, chromatin immunoprecipitation analysis revealed that TcERR could directly bind to ERR-response elements on promoters of genes encoding defensin3 and coleoptericin, critical for innate immune response of T. castaneum. Finally, TcERR-deficient old larvae infected with E. coli displayed enhanced bacterial load and significantly less host survival. These findings suggest that TcERR can coordinate transcriptional regulation of AMPs and host innate immune response to bacterial infection.
Collapse
Affiliation(s)
- Byungyoon Choi
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Woo-Ram Park
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Yu-Ji Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Seulgi Mun
- Department of Applied Biology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Su-Jin Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea.
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Gwangju, 61468, Republic of Korea.
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Don-Kyu Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
23
|
Oxidative Stress, Genomic Integrity, and Liver Diseases. Molecules 2022; 27:molecules27103159. [PMID: 35630636 PMCID: PMC9147071 DOI: 10.3390/molecules27103159] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Excess reactive oxygen species production and free radical formation can lead to oxidative stress that can damage cells, tissues, and organs. Cellular oxidative stress is defined as the imbalance between ROS production and antioxidants. This imbalance can lead to malfunction or structure modification of major cellular molecules such as lipids, proteins, and DNAs. During oxidative stress conditions, DNA and protein structure modifications can lead to various diseases. Various antioxidant-specific gene expression and signal transduction pathways are activated during oxidative stress to maintain homeostasis and to protect organs from oxidative injury and damage. The liver is more vulnerable to oxidative conditions than other organs. Antioxidants, antioxidant-specific enzymes, and the regulation of the antioxidant responsive element (ARE) genes can act against chronic oxidative stress in the liver. ARE-mediated genes can act as the target site for averting/preventing liver diseases caused by oxidative stress. Identification of these ARE genes as markers will enable the early detection of liver diseases caused by oxidative conditions and help develop new therapeutic interventions. This literature review is focused on antioxidant-specific gene expression upon oxidative stress, the factors responsible for hepatic oxidative stress, liver response to redox signaling, oxidative stress and redox signaling in various liver diseases, and future aspects.
Collapse
|
24
|
Are BPA Substitutes as Obesogenic as BPA? Int J Mol Sci 2022; 23:ijms23084238. [PMID: 35457054 PMCID: PMC9031831 DOI: 10.3390/ijms23084238] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolic diseases, such as obesity, Type II diabetes and hepatic steatosis, are a significant public health concern affecting more than half a billion people worldwide. The prevalence of these diseases is constantly increasing in developed countries, affecting all age groups. The pathogenesis of metabolic diseases is complex and multifactorial. Inducer factors can either be genetic or linked to a sedentary lifestyle and/or consumption of high-fat and sugar diets. In 2002, a new concept of “environmental obesogens” emerged, suggesting that environmental chemicals could play an active role in the etiology of obesity. Bisphenol A (BPA), a xenoestrogen widely used in the plastic food packaging industry has been shown to affect many physiological functions and has been linked to reproductive, endocrine and metabolic disorders and cancer. Therefore, the widespread use of BPA during the last 30 years could have contributed to the increased incidence of metabolic diseases. BPA was banned in baby bottles in Canada in 2008 and in all food-oriented packaging in France from 1 January 2015. Since the BPA ban, substitutes with a similar structure and properties have been used by industrials even though their toxic potential is unknown. Bisphenol S has mainly replaced BPA in consumer products as reflected by the almost ubiquitous human exposure to this contaminant. This review focuses on the metabolic effects and targets of BPA and recent data, which suggest comparable effects of the structural analogs used as substitutes.
Collapse
|
25
|
Park WR, Choi B, Kim YJ, Kim YH, Park MJ, Kim DI, Choi HS, Kim DK. Melatonin Regulates Iron Homeostasis by Inducing Hepcidin Expression in Hepatocytes. Int J Mol Sci 2022; 23:ijms23073593. [PMID: 35408955 PMCID: PMC8998539 DOI: 10.3390/ijms23073593] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/10/2022] Open
Abstract
The pineal hormone, melatonin, plays important roles in circadian rhythms and energy metabolism. The hepatic peptide hormone, hepcidin, regulates iron homeostasis by triggering the degradation of ferroportin (FPN), the protein that transfers cellular iron to the blood. However, the role of melatonin in the transcriptional regulation of hepcidin is largely unknown. Here, we showed that melatonin upregulates hepcidin gene expression by enhancing the melatonin receptor 1 (MT1)-mediated c-Jun N-terminal kinase (JNK) activation in hepatocytes. Interestingly, hepcidin gene expression was increased during the dark cycle in the liver of mice, whereas serum iron levels decreased following hepcidin expression. In addition, melatonin significantly induced hepcidin gene expression and secretion, as well as the subsequent FPN degradation in hepatocytes, which resulted in cellular iron accumulation. Melatonin-induced hepcidin expression was significantly decreased by the melatonin receptor antagonist, luzindole, and by the knockdown of MT1. Moreover, melatonin activated JNK signaling and upregulated hepcidin expression, both of which were significantly decreased by SP600125, a specific JNK inhibitor. Chromatin immunoprecipitation analysis showed that luzindole significantly blocked melatonin-induced c-Jun binding to the hepcidin promoter. Finally, melatonin induced hepcidin expression and secretion by activating the JNK-c-Jun pathway in mice, which were reversed by the luzindole treatment. These findings reveal a previously unrecognized role of melatonin in the circadian regulation of hepcidin expression and iron homeostasis.
Collapse
Affiliation(s)
- Woo-Ram Park
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Korea; (W.-R.P.); (B.C.); (Y.-J.K.)
| | - Byungyoon Choi
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Korea; (W.-R.P.); (B.C.); (Y.-J.K.)
| | - Yu-Ji Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Korea; (W.-R.P.); (B.C.); (Y.-J.K.)
| | - Yong-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea;
| | - Min-Jung Park
- Department of Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea; (M.-J.P.); (D.-I.K.)
| | - Dong-Il Kim
- Department of Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea; (M.-J.P.); (D.-I.K.)
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Korea;
| | - Don-Kyu Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Korea; (W.-R.P.); (B.C.); (Y.-J.K.)
- Correspondence: ; Tel.: +82-62-530-2166; Fax: +82-62-530-2160
| |
Collapse
|
26
|
Welch RD, Billon C, Losby M, Bedia-Diaz G, Fang Y, Avdagic A, Elgendy B, Burris TP, Griffett K. Emerging Role of Nuclear Receptors for the Treatment of NAFLD and NASH. Metabolites 2022; 12:238. [PMID: 35323681 PMCID: PMC8953348 DOI: 10.3390/metabo12030238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
Non-alcoholic fatty liver (NAFLD) over the past years has become a metabolic pandemic linked to a collection of metabolic diseases. The nuclear receptors ERRs, REV-ERBs, RORs, FXR, PPARs, and LXR are master regulators of metabolism and liver physiology. The characterization of these nuclear receptors and their biology has promoted the development of synthetic ligands. The possibility of targeting these receptors to treat NAFLD is promising, as several compounds including Cilofexor, thiazolidinediones, and Saroglitazar are currently undergoing clinical trials. This review focuses on the latest development of the pharmacology of these metabolic nuclear receptors and how they may be utilized to treat NAFLD and subsequent comorbidities.
Collapse
Affiliation(s)
- Ryan D. Welch
- Biology and Chemistry Department, Blackburn College, Carlinville, IL 62626, USA;
| | - Cyrielle Billon
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University in St. Louis, St. Louis, MO 63110, USA; (C.B.); (G.B.-D.); (Y.F.); (A.A.); (B.E.)
| | - McKenna Losby
- Biochemistry, Biophysics and Structural Biology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA;
| | - Gonzalo Bedia-Diaz
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University in St. Louis, St. Louis, MO 63110, USA; (C.B.); (G.B.-D.); (Y.F.); (A.A.); (B.E.)
| | - Yuanying Fang
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University in St. Louis, St. Louis, MO 63110, USA; (C.B.); (G.B.-D.); (Y.F.); (A.A.); (B.E.)
| | - Amer Avdagic
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University in St. Louis, St. Louis, MO 63110, USA; (C.B.); (G.B.-D.); (Y.F.); (A.A.); (B.E.)
| | - Bahaa Elgendy
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University in St. Louis, St. Louis, MO 63110, USA; (C.B.); (G.B.-D.); (Y.F.); (A.A.); (B.E.)
- Department of Anesthesiology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Thomas P. Burris
- UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA;
| | - Kristine Griffett
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
27
|
Kim IY, Choi B, Park WR, Kim YJ, Kim BE, Mun S, Choi HS, Kim DK. Nuclear receptor HR96 up-regulates cytochrome P450 for insecticide detoxification in Tribolium castaneum. PEST MANAGEMENT SCIENCE 2022; 78:230-239. [PMID: 34472702 DOI: 10.1002/ps.6626] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/02/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Red flour beetle, Tribolium castaneum (T. castaneum), is a major agricultural pest that causes significant damage to stored grains and products. Although hormone receptor 96 (HR96) is known to be the single ortholog corresponding to mammalian constitutive androstane receptor and pregnane X receptor, the structural features of Tribolium HR96 (TcHR96) and its role in insecticide-mediated transcription control of cytochrome P450 enzyme genes in T. castaneum have not been elucidated yet. RESULTS We cloned full-length complementary DNA encoding TcHR96 and revealed the role of TcHR96 in transcriptional control of cytochrome P450 enzyme genes. Interestingly, genome-wide transcriptome analysis of HR96-deficient beetles using RNA sequencing showed a positive correlation between TcHR96 and gene transcription of metabolizing enzymes involved in phase I detoxification processes. Moreover, TcHR96 overexpression significantly increased the promoter activity of genes encoding phase I P450 enzymes such as CYP4Q4, CYP4G7, CYP4BR3, and CYP345A1. Chromatin immunoprecipitation analysis showed that TcHR96 could directly bind to the promoter of gene encoding CYP345A1, an enzyme for metabolizing insecticides in T. castaneum. Furthermore, imidacloprid, a neonicotinoid insecticide, significantly increased gene expression of phase I P450 enzymes in old larvae of T. castaneum, which were reversed by TcHR96 knockdown. Finally, TcHR96 knockdown significantly decreased the resistance of old larvae to imidacloprid concomitant with reduction of imidacloprid-mediated phase I P450 enzyme gene expression. CONCLUSION TcHR96 plays a major role in transcriptional control of P450 enzyme for imidacloprid detoxification. Controlling TcHR96 might facilitate the regulation of insecticide tolerance in T. castaneum, thus providing a promising new strategy to manage pest beetle populations. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- In-Young Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, South Korea
| | - Byungyoon Choi
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, South Korea
| | - Woo-Ram Park
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, South Korea
| | - Yu-Ji Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, South Korea
| | - Bo-Eun Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, South Korea
| | - Seulgi Mun
- Department of Applied Biology, Chonnam National University, Gwangju, South Korea
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea
| | - Don-Kyu Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
28
|
Molecular dynamics of estrogen-related receptors and their regulatory proteins: roles in transcriptional control for endocrine and metabolic signaling. Anat Sci Int 2021; 97:15-29. [PMID: 34609710 DOI: 10.1007/s12565-021-00634-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022]
Abstract
Estrogen-related receptor (ERR) is a member of the nuclear receptor (NR) superfamily and has three subtypes α, β, and γ. Despite their strong homology with estrogen receptor (ER) α, ERRs cannot accommodate endogenous hormones. However, they are able to regulate gene expression without ligand binding. ERRα and ERRγ orchestrate the expression of genes involved in bioenergetic pathways, while ERRβ controls placental development and stem cell maintenance. Evidence from recent studies, including clinical research, has also demonstrated close associations of ERRs with the pathophysiology of hormone-related cancers and metabolic disorders including type 2 diabetes mellitus. This review summarizes the basic knowledge and recent advances in ERRs and their associated proteins, focusing on the subcellular dynamics involved in transcriptional regulation. Fluorescent protein labeling enabled monitoring of ERRs in living cells and revealed previously unrecognized characteristics. Using this technique, we demonstrated a role of ERRβ in controlling estrogen signaling by regulating the subnuclear dynamics of ligand-activated ERα. Visualization of ERRs and related proteins and subsequent analyses also revealed a function of ERRγ in promoting liver lactate metabolism in association with LRPGC1, a recently identified lactic acid-responsive protein. These findings suggest that ERRs activate unique transregulation mechanisms in response to extracellular stimuli such as hormones and metabolic signals, implying an adaptive system behind the cellular homeostatic regulation by orphan NRs. Control of subcellular ERR dynamics will contribute toward the development of therapeutic approaches to treat various diseases including hormone-related cancers and metabolic disorders associated with abnormal ERR signaling pathways.
Collapse
|
29
|
Growth hormone promotes hepatic gluconeogenesis by enhancing BTG2-YY1 signaling pathway. Sci Rep 2021; 11:18999. [PMID: 34556771 PMCID: PMC8460702 DOI: 10.1038/s41598-021-98537-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
Growth hormone (GH) is one of the critical factors in maintaining glucose metabolism. B-cell translocation gene 2 (BTG2) and yin yang 1 (YY1) are key regulators of diverse metabolic processes. In this study, we investigated the link between GH and BTG2–YY1 signaling pathway in glucose metabolism. GH treatment elevated the expression of hepatic Btg2 and Yy1 in primary mouse hepatocytes and mouse livers. Glucose production in primary mouse hepatocytes and serum blood glucose levels were increased during GH exposure. Overexpression of hepatic Btg2 and Yy1 induced key gluconeogenic enzymes phosphoenolpyruvate carboxykinase 1 (PCK1) and glucose-6 phosphatase (G6PC) as well as glucose production in primary mouse hepatocytes, whereas this phenomenon was markedly diminished by knockdown of Btg2 and Yy1. Here, we identified the YY1-binding site on the Pck1 and G6pc gene promoters using reporter assays and point mutation analysis. The regulation of hepatic gluconeogenic genes induced by GH treatment was clearly linked with YY1 recruitment on gluconeogenic gene promoters. Overall, this study demonstrates that BTG2 and YY1 are novel regulators of GH-dependent regulation of hepatic gluconeogenic genes and glucose production. BTG2 and YY1 may be crucial therapeutic targets to intervene in metabolic dysfunction in response to the GH-dependent signaling pathway.
Collapse
|
30
|
Jung YS, Kim YH, Radhakrishnan K, Kim J, Lee IK, Cho SJ, Kim DK, Dooley S, Lee CH, Choi HS. Orphan nuclear receptor ERRγ regulates hepatic TGF-β2 expression and fibrogenic response in CCl4-induced acute liver injury. Arch Toxicol 2021; 95:3071-3084. [DOI: https:/doi.org/10.1007/s00204-021-03112-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 06/22/2021] [Indexed: 09/18/2023]
|
31
|
Orphan nuclear receptor ERRγ regulates hepatic TGF-β2 expression and fibrogenic response in CCl 4-induced acute liver injury. Arch Toxicol 2021; 95:3071-3084. [PMID: 34191077 DOI: 10.1007/s00204-021-03112-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
Acute liver injury results from the complex interactions of various pathological processes. The TGF-β superfamily plays a crucial role in orchestrating fibrogenic response. In contrast to TGF-β1, a role of TGF-β2 in hepatic fibrogenic response has not been fully investigated. In this study, we showed that TGF-β2 gene expression and secretion are induced in the liver of CCl4 (1 ml/kg)-treated WT mice. Studies with hepatocyte specific ERRγ knockout mice or treatment with an ERRγ-specific inverse agonist, GSK5182 (40 mg/kg), indicated that CCl4-induced hepatic TGF-β2 production is ERRγ dependent. Moreover, IL6 was found as upstream signal to induce hepatic ERRγ and TGF-β2 gene expression in CCl4-mediated acute toxicity model. Over-expression of ERRγ was sufficient to induce hepatic TGF-β2 expression, whereas ERRγ depletion markedly reduces IL6-induced TGF-β2 gene expression and secretion in vitro and in vivo. Promoter assays showed that ERRγ directly binds to an ERR response element in the TGF-β2 promoter to induce TGF-β2 transcription. Finally, GSK5182 diminished CCl4-induced fibrogenic response through inhibition of ERRγ-mediated TGF-β2 production. Taken together, these results firstly demonstrate that ERRγ can regulate the TGF-β2-mediated fibrogenic response in a mouse model of CC14-induced acute liver injury.
Collapse
|
32
|
ERRγ enhances cardiac maturation with T-tubule formation in human iPSC-derived cardiomyocytes. Nat Commun 2021; 12:3596. [PMID: 34155205 PMCID: PMC8217550 DOI: 10.1038/s41467-021-23816-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/17/2021] [Indexed: 01/17/2023] Open
Abstract
One of the earliest maturation steps in cardiomyocytes (CMs) is the sarcomere protein isoform switch between TNNI1 and TNNI3 (fetal and neonatal/adult troponin I). Here, we generate human induced pluripotent stem cells (hiPSCs) carrying a TNNI1EmGFP and TNNI3mCherry double reporter to monitor and isolate mature sub-populations during cardiac differentiation. Extensive drug screening identifies two compounds, an estrogen-related receptor gamma (ERRγ) agonist and an S-phase kinase-associated protein 2 inhibitor, that enhances cardiac maturation and a significant change to TNNI3 expression. Expression, morphological, functional, and molecular analyses indicate that hiPSC-CMs treated with the ERRγ agonist show a larger cell size, longer sarcomere length, the presence of transverse tubules, and enhanced metabolic function and contractile and electrical properties. Here, we show that ERRγ-treated hiPSC-CMs have a mature cellular property consistent with neonatal CMs and are useful for disease modeling and regenerative medicine. Cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSCs) suffer from limited maturation. Here the authors identify ERRγ agonist as a factor that enhances cardiac morphological, metabolic, contractile and electrical maturation of hiPSC-derived CMs with T-tubule formation.
Collapse
|
33
|
Orphan Nuclear Receptor ERRγ Is a Transcriptional Regulator of CB1 Receptor-Mediated TFR2 Gene Expression in Hepatocytes. Int J Mol Sci 2021; 22:ijms22116021. [PMID: 34199599 PMCID: PMC8199698 DOI: 10.3390/ijms22116021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 11/23/2022] Open
Abstract
Orphan nuclear receptor estrogen-related receptor γ (ERRγ) is an important transcription factor modulating gene transcription involved in endocrine control of liver metabolism. Transferrin receptor 2 (TFR2), a carrier protein for transferrin, is involved in hepatic iron overload in alcoholic liver disease (ALD). However, TFR2 gene transcriptional regulation in hepatocytes remains largely unknown. In this study, we described a detailed molecular mechanism of hepatic TFR2 gene expression involving ERRγ in response to an endocannabinoid 2-arachidonoylglycerol (2-AG). Treatment with 2-AG and arachidonyl-2′-chloroethylamide, a selective cannabinoid receptor type 1 (CB1) receptor agonist, increased ERRγ and TFR2 expression in hepatocytes. Overexpression of ERRγ was sufficient to induce TFR2 expression in both human and mouse hepatocytes. In addition, ERRγ knockdown significantly decreased 2-AG or alcohol-mediated TFR2 gene expression in cultured hepatocytes and mouse livers. Finally, deletion and mutation analysis of the TFR2 gene promoter demonstrated that ERRγ directly modulated TFR2 gene transcription via binding to an ERR-response element. This was further confirmed by chromatin immunoprecipitation assay. Taken together, these results reveal a previously unrecognized role of ERRγ in the transcriptional regulation of TFR2 gene expression in response to alcohol.
Collapse
|
34
|
Kim HJ, Yoon HJ, Lee DK, Jin X, Che X, Choi JY. The estrogen-related receptor γ modulator, GSK5182, inhibits osteoclast differentiation and accelerates osteoclast apoptosis. BMB Rep 2021. [PMID: 33612148 PMCID: PMC8167243 DOI: 10.5483/bmbrep.2021.54.5.243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Estrogen-related receptor γ (ERRγ), a member of the orphan nuclear receptor family, is a key mediator in cellular metabolic processes and energy homeostasis. Therefore, ERRγ has become an attractive target for treating diverse metabolic disorders. We recently reported that ERRγ acts as a negative regulator of osteoclastogenesis induced by receptor activator of nuclear factor-κB ligand (RANKL). In the present study, we explored the effects of an ERRγ-specific modulator, GSK5182, on ERRγ-regulated osteoclast differentiation and survival. Interestingly, GSK5182 increased ERRγ protein levels much as does GSK4716, which is an ERRγ agonist. GSK5182 inhibited osteoclast generation from bone-marrow-derived macrophages without affecting cytotoxicity. GSK5182 also attenuated RANKL-mediated expression of c-Fos and nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), pivotal transcription factors for osteoclastogenesis. Arrested osteoclast differentiation was associated with reduced RANK expression, but not with the M-CSF receptor, c-Fms. GSK5182 strongly blocked the phosphorylation of IκBα, c-Jun N-terminal kinase, and extracellular signal-regulated kinase in response to RANKL. GSK5182 also suppressed NF-κB promoter activity in a dose-dependent manner. In addition to osteoclastogenesis, GSK5182 accelerated osteoclast apoptosis by caspase-3 activation. Together, these results suggest that GSK5182, a synthetic ERRγ modulator, may have potential in treating disorders related to bone resorption.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hye-Jin Yoon
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Dong-Kyo Lee
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Xian Jin
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Xiangguo Che
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
35
|
Kim SE, Ji SB, Kim E, Jeong M, Kim J, Lee GM, Seo HJ, Bae S, Jeong Y, Lee S, Kim S, Lee T, Cho SJ, Liu KH. Nontargeted Metabolomics by High-Resolution Mass Spectrometry to Study the In Vitro Metabolism of a Dual Inverse Agonist of Estrogen-Related Receptors β and γ, DN203368. Pharmaceutics 2021; 13:pharmaceutics13060776. [PMID: 34072800 PMCID: PMC8230175 DOI: 10.3390/pharmaceutics13060776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
DN203368 ((E)-3-[1-(4-[4-isopropylpiperazine-1-yl]phenyl) 3-methyl-2-phenylbut-1-en-1-yl] phenol) is a 4-hydroxy tamoxifen analog that is a dual inverse agonist of estrogen-related receptor β/γ (ERRβ/γ). ERRγ is an orphan nuclear receptor that plays an important role in development and homeostasis and holds potential as a novel therapeutic target in metabolic diseases such as diabetes mellitus, obesity, and cancer. ERRβ is also one of the orphan nuclear receptors critical for many biological processes, such as development. We investigated the in vitro metabolism of DN203368 by conventional and metabolomic approaches using high-resolution mass spectrometry. The compound (100 μM) was incubated with rat and human liver microsomes in the presence of NADPH. In the metabolomic approach, the m/z value and retention time information obtained from the sample and heat-inactivated control group were statistically evaluated using principal component analysis and orthogonal partial least-squares discriminant analysis. Significant features responsible for group separation were then identified using tandem mass spectra. Seven metabolites of DN203368 were identified in rat liver microsomes and the metabolic pathways include hydroxylation (M1-3), N-oxidation (M4), N-deisopropylation (M5), N,N-dealkylation (M6), and oxidation and dehydrogenation (M7). Only five metabolites (M2, M3, and M5-M7) were detected in human liver microsomes. In the conventional approach using extracted ion monitoring for values of mass increase or decrease by known metabolic reactions, only five metabolites (M1-M5) were found in rat liver microsomes, whereas three metabolites (M2, M3, and M5) were found in human liver microsomes. This study revealed that nontargeted metabolomics combined with high-resolution mass spectrometry and multivariate analysis could be a more efficient tool for drug metabolite identification than the conventional approach. These results might also be useful for understanding the pharmacokinetics and metabolism of DN203368 in animals and humans.
Collapse
Affiliation(s)
- Sin-Eun Kim
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Seung-Bae Ji
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Euihyeon Kim
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Minseon Jeong
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (M.J.); (J.K.)
| | - Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (M.J.); (J.K.)
| | - Gyung-Min Lee
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Hyung-Ju Seo
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Subin Bae
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Yeojin Jeong
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Sangkyu Lee
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea;
| | - Sunghwan Kim
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea;
- Department of Chemistry, Kyungpook National University, Daegu 41566, Korea
| | - Taeho Lee
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Sung Jin Cho
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, Korea
- Correspondence: (S.J.C.); (K.-H.L.); Tel.: +82-2-958-5137 (S.-J.C.); +82-53-950-8567 (K.-H.L.); Fax: +82-2-958-5137 (S.J.C.); +82-53-950-8557 (K.-H.L.)
| | - Kwang-Hyeon Liu
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea;
- Correspondence: (S.J.C.); (K.-H.L.); Tel.: +82-2-958-5137 (S.-J.C.); +82-53-950-8567 (K.-H.L.); Fax: +82-2-958-5137 (S.J.C.); +82-53-950-8557 (K.-H.L.)
| |
Collapse
|
36
|
Kim KS, Kim DK, Na SY, Jung YS, Cho SJ, Kim J, Lee IK, Kim YH, Lee CH, Jeong WI, Jo EK, Choi HS. Frontline Science: Estrogen-related receptor γ increases poly(I:C)-mediated type I IFN expression in mouse macrophages. J Leukoc Biol 2021; 109:865-875. [PMID: 33615540 DOI: 10.1002/jlb.2hi1219-762r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 10/19/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
Although type I IFNs (IFN-I) are important for the innate and adaptive immune responses to suppress viral replication, prolonged IFN-I signaling in macrophages suppresses the immune response. Nuclear receptor estrogen-related receptor γ (ERRγ) regulates the transcription of genes involved in endocrine and metabolic functions. However, the role of ERRγ in macrophage immune responses to viruses remains largely unknown. ERRγ expression was significantly induced in mouse bone marrow-derived macrophages (BMDMs) treated with polyinosinic-polycytidylic acid (poly(I:C)). Our results indicated that the induction of ERRγ expression by poly(I:C) is mediated through activation of the cytoplasmic dsRNA receptors, retinoic acid-inducible gene I and melanoma differentiation-associated protein 5. In BMDMs, overexpression of ERRγ significantly increased gene expression and secretion of the IFN-I genes, IFN-α and IFN-β, whereas abolition of ERRγ significantly attenuated poly(I:C)-mediated IFN-I secretion. Chromatin immunoprecipitation assays and mutation analyses of the IFN-I promoters revealed that ERRγ regulates the transcription of IFN-α and IFN-β by binding to a conserved ERR response element in each promoter region. Finally, GSK5182 significantly suppressed poly(I:C)-mediated induction of IFN-I gene expression and secretion in BMDMs. Taken together, these findings reveal a previously unrecognized role for ERRγ in the transcriptional control of innate and adaptive immune response to dsRNA virus replication.
Collapse
Affiliation(s)
- Ki-Sun Kim
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Don-Kyu Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Soon-Young Na
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Yoon Seok Jung
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Sung Jin Cho
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - In-Kyu Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Young-Hoon Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Chul-Ho Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Won-Il Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
37
|
Radhakrishnan K, Kim YH, Jung YS, Kim DK, Na SY, Lim D, Kim DH, Kim J, Kim HS, Choy HE, Cho SJ, Lee IK, Ayvaz Ş, Nittka S, Fliser D, Schunk SJ, Speer T, Dooley S, Lee CH, Choi HS. Orphan nuclear receptor ERR-γ regulates hepatic FGF23 production in acute kidney injury. Proc Natl Acad Sci U S A 2021; 118. [DOI: https:/doi.org/10.1073/pnas.2022841118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2023] Open
Abstract
Significance
Bone is the main source of fibroblast growth factor 23 (FGF23), which is important for phosphate and vitamin D homeostasis. In acute kidney injury (AKI), high blood levels of FGF23 are positively correlated with disease progression and increased risk of mortality. Reducing adverse plasma FGF23 levels in AKI patients is favorable. We showed here that hepatocytes are the major source of circulating FGF23, and orphan nuclear receptor ERR-γ is a novel transcriptional regulator of hepatic FGF23 production in AKI. Liver-specific depletion of ERR-γ or ERR-γ inverse agonist, GSK5182, significantly reduced plasma levels of FGF23 in AKI. This study reveals liver is the source of FGF23 and a therapeutic strategy to control pathologically adverse plasma FGF23 levels in AKI.
Collapse
Affiliation(s)
- Kamalakannan Radhakrishnan
- School of Biological Sciences and Technology, Chonnam National University, 61186 Gwangju, Republic of Korea
| | - Yong-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 34141 Daejeon, Republic of Korea
- Department of Functional Genomics, Korea Research Institute of Biosciences and Biotechnology School of Bioscience, University of Science and Technology, 34141 Daejeon, Republic of Korea
| | - Yoon Seok Jung
- School of Biological Sciences and Technology, Chonnam National University, 61186 Gwangju, Republic of Korea
| | - Don-Kyu Kim
- Department of Molecular Biotechnology, Chonnam National University, 61186 Gwangju, Republic of Korea
| | - Soon-Young Na
- School of Biological Sciences and Technology, Chonnam National University, 61186 Gwangju, Republic of Korea
| | - Daejin Lim
- Department of Microbiology, Chonnam National University Medical School, 61468 Gwangju, Republic of Korea
| | - Dong Hun Kim
- Department of Biomedical Science, Graduate School, Kyungpook National University, 41404 Daegu, Republic of Korea
| | - Jina Kim
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, 41061 Daegu, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, 61468 Gwangju, Republic of Korea
| | - Hyon E. Choy
- Department of Microbiology, Chonnam National University Medical School, 61468 Gwangju, Republic of Korea
| | - Sung Jin Cho
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, 41061 Daegu, Republic of Korea
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, 41404 Daegu, Republic of Korea
| | - In-Kyu Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, 41404 Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, 41944 Daegu, Republic of Korea
| | - Şamil Ayvaz
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Stefanie Nittka
- Institute for Clinical Chemistry, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University, D-66421 Homburg/Saar, Germany
| | - Stefan J. Schunk
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University, D-66421 Homburg/Saar, Germany
| | - Thimoteus Speer
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University, D-66421 Homburg/Saar, Germany
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 34141 Daejeon, Republic of Korea
- Department of Functional Genomics, Korea Research Institute of Biosciences and Biotechnology School of Bioscience, University of Science and Technology, 34141 Daejeon, Republic of Korea
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, 61186 Gwangju, Republic of Korea
| |
Collapse
|
38
|
Orphan nuclear receptor ERR-γ regulates hepatic FGF23 production in acute kidney injury. Proc Natl Acad Sci U S A 2021; 118:2022841118. [PMID: 33853949 DOI: 10.1073/pnas.2022841118] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23), a hormone generally derived from bone, is important in phosphate and vitamin D homeostasis. In acute kidney injury (AKI) patients, high-circulating FGF23 levels are associated with disease progression and mortality. However, the organ and cell type of FGF23 production in AKI and the molecular mechanism of its excessive production are still unidentified. For insight, we investigated folic acid (FA)-induced AKI in mice. Interestingly, simultaneous with FGF23, orphan nuclear receptor ERR-γ expression is increased in the liver of FA-treated mice, and ectopic overexpression of ERR-γ was sufficient to induce hepatic FGF23 production. In patients and in mice, AKI is accompanied by up-regulated systemic IL-6, which was previously identified as an upstream regulator of ERR-γ expression in the liver. Administration of IL-6 neutralizing antibody to FA-treated mice or of recombinant IL-6 to healthy mice confirms IL-6 as an upstream regulator of hepatic ERR-γ-mediated FGF23 production. A significant (P < 0.001) interconnection between high IL-6 and FGF23 levels as a predictor of AKI in patients that underwent cardiac surgery was also found, suggesting the clinical relevance of the finding. Finally, liver-specific depletion of ERR-γ or treatment with an inverse ERR-γ agonist decreased hepatic FGF23 expression and plasma FGF23 levels in mice with FA-induced AKI. Thus, inverse agonist of ERR-γ may represent a therapeutic strategy to reduce adverse plasma FGF23 levels in AKI.
Collapse
|
39
|
Park WR, Lim DJ, Sang H, Kim E, Moon JH, Choi HS, Kim IS, Kim DK. Aphid estrogen-related receptor controls glycolytic gene expression and fecundity. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 130:103529. [PMID: 33485935 DOI: 10.1016/j.ibmb.2021.103529] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/13/2021] [Accepted: 01/13/2021] [Indexed: 06/12/2023]
Abstract
Aphids, the major insect pests of agricultural crops, reproduce sexually and asexually depending upon environmental factors such as the photoperiod and temperature. Nuclear receptors, a unique family of ligand-dependent transcription factors, control insect development and growth including morphogenesis, molting, and metamorphosis. However, the structural features and biological functions of the aphid estrogen-related receptor (ERR) are largely unknown. Here, we cloned full-length cDNA encoding the ERR in the green peach aphid, Myzus persicae, (Sulzer) (Hemiptera: Aphididae) (MpERR) and demonstrated that the MpERR modulated glycolytic gene expression and aphid fecundity. The phylogenetic analysis revealed that the MpERR originated in a unique evolutionary lineage distinct from those of hemipteran insects. Moreover, the AF-2 domain of the MpERR conferred nuclear localization and transcriptional activity. The overexpression of the MpERR significantly upregulated the gene expression of rate-limiting enzymes involved in glycolysis such as phosphofructokinase and pyruvate kinase by directly binding to ERR-response elements in their promoters. Moreover, ERR-deficient viviparous female aphids showed decreased glycolytic gene expression and produced fewer offspring. These results suggest that the aphid ERR plays a pivotal role in glycolytic transcriptional control and fecundity.
Collapse
Affiliation(s)
- Woo-Ram Park
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Da Jung Lim
- Department of Agricultural Chemistry, Institute of Environmentally Friendly Agriculture, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Hyunkyu Sang
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Eunae Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| | - Jae-Hak Moon
- Department of Food Science and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - In Seon Kim
- Department of Agricultural Chemistry, Institute of Environmentally Friendly Agriculture, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Don-Kyu Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
40
|
Sijbesma E, Somsen BA, Miley GP, Leijten-van de Gevel IA, Brunsveld L, Arkin MR, Ottmann C. Fluorescence Anisotropy-Based Tethering for Discovery of Protein-Protein Interaction Stabilizers. ACS Chem Biol 2020; 15:3143-3148. [PMID: 33196173 PMCID: PMC7754187 DOI: 10.1021/acschembio.0c00646] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein-protein interaction (PPI) networks are fundamental for cellular processes. Small-molecule PPI enhancers have been shown to be powerful tools to fundamentally study PPIs and as starting points for potential new therapeutics. Yet, systematic approaches for their discovery are not widely available, and the design prerequisites of "molecular glues" are poorly understood. Covalent fragment-based screening can identify chemical starting points for these enhancers at specific sites in PPI interfaces. We recently reported a mass spectrometry-based disulfide-trapping (tethering) approach for a cysteine residue in the hub protein 14-3-3, an important regulator of phosphorylated client proteins. Here, we invert the strategy and report the development of a functional read-out for systematic identification of PPI enhancers based on fluorescence anisotropy (FA-tethering) with the reactive handle now on a client-derived peptide. Using the DNA-binding domain of the nuclear receptor Estrogen Related Receptor gamma (ERRγ), we target a native cysteine positioned at the 14-3-3 PPI interface and identify several fragments that form a disulfide bond to ERRγ and stabilize the complex up to 5-fold. Crystallography indicates that fragments bind in a pocket comprised of 14-3-3 and the ERRγ phosphopeptide. FA-tethering presents a streamlined methodology to discover molecular glues for protein complexes.
Collapse
Affiliation(s)
- Eline Sijbesma
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Bente A. Somsen
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Galen P. Miley
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Iris A. Leijten-van de Gevel
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Michelle R. Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center (SMDC), University of California, San Francisco, United States
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Chemistry, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
41
|
Min Y, Kim D, Suminda GGD, Zhao X, Kim M, Zhao Y, Son YO. GSK5182, 4-Hydroxytamoxifen Analog, a New Potential Therapeutic Drug for Osteoarthritis. Pharmaceuticals (Basel) 2020; 13:ph13120429. [PMID: 33261216 PMCID: PMC7761342 DOI: 10.3390/ph13120429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 11/16/2022] Open
Abstract
Estrogen-related receptors (ERRs) are the first identified orphan nuclear receptors. The ERR family consists of ERRα, ERRβ, and ERRγ, regulating diverse isoform-specific functions. We have reported the importance of ERRγ in osteoarthritis (OA) pathogenesis. However, therapeutic approaches with ERRγ against OA associated with inflammatory mechanisms remain limited. Herein, we examined the therapeutic potential of a small-molecule ERRγ inverse agonist, GSK5182 (4-hydroxytamoxifen analog), in OA, to assess the relationship between ERRγ expression and pro-inflammatory cytokines in mouse articular chondrocyte cultures. ERRγ expression increased following chondrocyte exposure to various pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. Pro-inflammatory cytokines dose-dependently increased ERRγ protein levels. In mouse articular chondrocytes, adenovirus-mediated ERRγ overexpression upregulated matrix metalloproteinase (MMP)-3 and MMP-13, which participate in cartilage destruction during OA. Adenovirus-mediated ERRγ overexpression in mouse knee joints or ERRγ transgenic mice resulted in OA. In mouse joint tissues, genetic ablation of Esrrg obscured experimental OA. These results indicate that ERRγ is involved in OA pathogenesis. In mouse articular chondrocytes, GSK5182 inhibited pro-inflammatory cytokine-induced catabolic factors. Consistent with the in vitro results, GSK5182 significantly reduced cartilage degeneration in ERRγ-overexpressing mice administered intra-articular Ad-Esrrg. Overall, the ERRγ inverse agonist GSK5182 represents a promising therapeutic small molecule for OA.
Collapse
Affiliation(s)
- Yunhui Min
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
| | - Dahye Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City 63243, Korea; (D.K.); (M.K.)
| | - Godagama Gamaarachchige Dinesh Suminda
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
| | - Xiangyu Zhao
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
| | - Mangeun Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City 63243, Korea; (D.K.); (M.K.)
| | - Yaping Zhao
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Young-Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City 63243, Korea; (D.K.); (M.K.)
- Bio-Health Materials Core-Facility Center, Jeju National University, Jeju City 63243, Korea
- Practical Translational Research Center, Jeju National University, Jeju City 63243, Korea
- Correspondence: ; Tel.: +82-(64)-754-3331
| |
Collapse
|
42
|
Orphan Nuclear Receptor ERRγ Is a Novel Transcriptional Regulator of IL-6 Mediated Hepatic BMP6 Gene Expression in Mice. Int J Mol Sci 2020; 21:ijms21197148. [PMID: 32998264 PMCID: PMC7582774 DOI: 10.3390/ijms21197148] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/23/2022] Open
Abstract
Bone morphogenetic protein 6 (BMP6) is a multifunctional growth factor involved in organ development and homeostasis. BMP6 controls expression of the liver hormone, hepcidin, and thereby plays a crucial role in regulating iron homeostasis. BMP6 gene transcriptional regulation in liver is largely unknown, but would be of great help to externally modulate iron load in pathologic conditions. Here, we describe a detailed molecular mechanism of hepatic BMP6 gene expression by an orphan nuclear receptor, estrogen-related receptor γ (ERRγ), in response to the pro-inflammatory cytokine interleukin 6 (IL-6). Recombinant IL-6 treatment increases hepatic ERRγ and BMP6 expression. Overexpression of ERRγ is sufficient to increase BMP6 gene expression in hepatocytes, suggesting that IL-6 is upstream of ERRγ. In line, knock-down of ERRγ in cell lines or a hepatocyte specific knock-out of ERRγ in mice significantly decreases IL-6 mediated BMP6 expression. Promoter studies show that ERRγ directly binds to the ERR response element (ERRE) in the mouse BMP6 gene promoter and positively regulates BMP6 gene transcription in IL-6 treatment conditions, which is further confirmed by ERRE mutated mBMP6-luciferase reporter assays. Finally, an inverse agonist of ERRγ, GSK5182, markedly inhibits IL-6 induced hepatic BMP6 expression in vitro and in vivo. Taken together, these results reveal a novel molecular mechanism on ERRγ mediated transcriptional regulation of hepatic BMP6 gene expression in response to IL-6.
Collapse
|
43
|
Jeon JH, Thoudam T, Choi EJ, Kim MJ, Harris RA, Lee IK. Loss of metabolic flexibility as a result of overexpression of pyruvate dehydrogenase kinases in muscle, liver and the immune system: Therapeutic targets in metabolic diseases. J Diabetes Investig 2020; 12:21-31. [PMID: 32628351 PMCID: PMC7779278 DOI: 10.1111/jdi.13345] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Good health depends on the maintenance of metabolic flexibility, which in turn is dependent on the maintenance of regulatory flexibility of a large number of regulatory enzymes, but especially the pyruvate dehydrogenase complex (PDC), because of its central role in carbohydrate metabolism. Flexibility in regulation of PDC is dependent on rapid changes in the phosphorylation state of PDC determined by the relative activities of the pyruvate dehydrogenase kinases (PDKs) and the pyruvate dehydrogenase phosphatases. Inactivation of the PDC by overexpression of PDK4 contributes to hyperglycemia, and therefore the serious health problems associated with diabetes. Loss of regulatory flexibility of PDC occurs in other disease states and pathological conditions that have received less attention than diabetes. These include cancers, non‐alcoholic fatty liver disease, cancer‐induced cachexia, diabetes‐induced nephropathy, sepsis and amyotrophic lateral sclerosis. Overexpression of PDK4, and in some situations, the other PDKs, as well as under expression of the pyruvate dehydrogenase phosphatases, leads to inactivation of the PDC, mitochondrial dysfunction and deleterious effects with health consequences. The possible basis for this phenomenon, along with evidence that overexpression of PDK4 results in phosphorylation of “off‐target” proteins and promotes excessive transport of Ca2+ into mitochondria through mitochondria‐associated endoplasmic reticulum membranes are discussed. Recent efforts to find small molecule PDK inhibitors with therapeutic potential are also reviewed.
Collapse
Affiliation(s)
- Jae-Han Jeon
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea.,Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Korea
| | - Eun Jung Choi
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Korea
| | - Min-Ji Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea.,Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Korea.,Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Korea.,Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Korea
| |
Collapse
|
44
|
Tanida T, Matsuda KI, Tanaka M. Novel metabolic system for lactic acid via LRPGC1/ERRγ signaling pathway. FASEB J 2020; 34:13239-13256. [PMID: 32851675 DOI: 10.1096/fj.202000492r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022]
Abstract
Lactic acid (LA) is a byproduct of glycolysis resulting from intense exercise or a metabolic defect in aerobic processes. LA metabolism is essential to prevent lactic acidosis, but the mechanism through which LA regulates its own metabolism is largely unknown. Here, we identified a LA-responsive protein, named LRPGC1, which has a distinct role from PGC1α, a key metabolic regulator, and report that LRPGC1 particularly mediates LA response to activate liver LA metabolism. Following LA stimulation, LRPGC1, but not PGC1α, translocates from the cytoplasm to the nucleus through deactivation of nuclear export signals, interacts with the nuclear receptor ERRγ, and upregulates TFAM, which ensures mitochondrial biogenesis. Knockout of PGC1 gene in HepG2 hepatocarcinoma cells decreased the LA consumption and TFAM expression, which were rescued by LRPGC1 expression, but not by PGC1α. These LRPGC1-induced effects were mediated by ERRγ, concomitantly with mitochondrial activation. The response element for LRPGC1/ERRγ signaling pathway was identified in TFAM promoter. Notably, the survival rate of a mouse model of lactic acidosis was reduced by the liver-targeted silencing of Lrpgc1, while it was significantly ameliorated by the pharmacological activation of ERRγ. These findings demonstrate LA-responsive transactivation via LRPGC1 that highlight an intrinsic molecular mechanism for LA homeostasis.
Collapse
Affiliation(s)
- Takashi Tanida
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Ichi Matsuda
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masaki Tanaka
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
45
|
Han HS, Kwon Y, Koo SH. Role of CRTC2 in Metabolic Homeostasis: Key Regulator of Whole-Body Energy Metabolism? Diabetes Metab J 2020; 44:498-508. [PMID: 32174060 PMCID: PMC7453979 DOI: 10.4093/dmj.2019.0200] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/18/2019] [Indexed: 12/18/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) signaling is critical for regulating metabolic homeostasis in mammals. In particular, transcriptional regulation by cAMP response element-binding protein (CREB) and its coactivator, CREB-regulated transcription coactivator (CRTC), is essential for controlling the expression of critical enzymes in the metabolic process, leading to more chronic changes in metabolic flux. Among the CRTC isoforms, CRTC2 is predominantly expressed in peripheral tissues and has been shown to be associated with various metabolic pathways in tissue-specific manners. While initial reports showed the physiological role of CRTC2 in regulating gluconeogenesis in the liver, recent studies have further delineated the role of this transcriptional coactivator in the regulation of glucose and lipid metabolism in various tissues, including the liver, pancreatic islets, endocrine tissues of the small intestines, and adipose tissues. In this review, we discuss recent studies that have utilized knockout mouse models to delineate the role of CRTC2 in the regulation of metabolic homeostasis.
Collapse
Affiliation(s)
- Hye Sook Han
- Division of Life Sciences, College of Life Sciences & Biotechnology, Korea University, Seoul, Korea
| | - Yongmin Kwon
- Division of Life Sciences, College of Life Sciences & Biotechnology, Korea University, Seoul, Korea
| | - Seung Hoi Koo
- Division of Life Sciences, College of Life Sciences & Biotechnology, Korea University, Seoul, Korea.
| |
Collapse
|
46
|
Meng J, Lv Z, Sun C, Qiao X, Chen C. An extract of Lycium barbarum mimics exercise to improve muscle endurance through increasing type IIa oxidative muscle fibers by activating ERRγ. FASEB J 2020; 34:11460-11473. [PMID: 33411401 DOI: 10.1096/fj.202000136r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022]
Abstract
Lycium barbarum berry (gouqi, Goji, goji berry, or wolfberry), a traditional medicine and functional food, has a wide range of biological effects, including immuno-modulation, anti-aging, antitumor, neuro-protection, and hepato-protection. However, thus far, little is known about the traditional effects of L. barbarum on strengthening muscles. Therefore, this study focused on the effects of an extract of L. barbarum on skeletal muscles. First, the extract of L. barbarum significantly increased the mass of the tibial anterior muscle and gastrocnemius muscle and improved the average running distance of mice. Then, in vivo and in vitro experiments showed that the extract enhanced muscle endurance by increasing the proportion of type IIa oxidative muscle fibers and aerobic respiration. In an in-depth study of the molecular mechanism of these effects, we found that the extract upregulated the proportion of type IIa oxidative muscle fibers by activating ERRγ and that the PKA-CREB signaling pathway was involved in its activation. This study is the first to show that L. barbarum extract modulates skeletal muscle remodeling and has mimetic effects on skeletal muscles in a manner similar to exercise. It provides a scientific explanation based on modern biological technologies and concepts for the traditional function of L. barbarum in improving muscle fitness. This study lays a theoretical foundation for the application of L. barbarum in skeletal muscles as an exercise mimetic.
Collapse
Affiliation(s)
- Jiao Meng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Lv
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chuanxin Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
47
|
Kim J, Hwang H, Yoon H, Lee JE, Oh JM, An H, Ji HD, Lee S, Cha E, Ma MJ, Kim DS, Lee SJ, Kadayat TM, Song J, Lee SW, Jeon JH, Park KG, Lee IK, Jeon YH, Chin J, Cho SJ. An orally available inverse agonist of estrogen-related receptor gamma showed expanded efficacy for the radioiodine therapy of poorly differentiated thyroid cancer. Eur J Med Chem 2020; 205:112501. [PMID: 32758860 DOI: 10.1016/j.ejmech.2020.112501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/29/2020] [Accepted: 05/22/2020] [Indexed: 12/24/2022]
Abstract
Estrogen-related receptor gamma (ERRγ) is the NR3B subgroup of associated transcription factors. In this report, a new generation of a potent and selective ERRγ inverse agonist (25) with good biocompatibility was proposed. We also explored the potential of the newly developed compound 25 in the PDTC model to expand the original indications from ATC. In addition, an X-ray crystallographic study of the ligand and ERRγ co-complex showed that 25 completely binds to the target protein (PDB 6KNR). Its medicinal chemistry, including a distinctive structural study to in vivo results, denotes that 25 may be directed towards the development of a pivotal treatment for ERRγ-related cancers.
Collapse
Affiliation(s)
- Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea; College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Hayoung Hwang
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea
| | - Heeseok Yoon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea
| | - Jae-Eon Lee
- Department of Biomaterials Science, College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Pusan, South Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Hongchan An
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea
| | - Hyun Dong Ji
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Seungmi Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, 41404, South Korea
| | - Eunju Cha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea
| | - Min Jung Ma
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea
| | - Dong-Su Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea
| | - Su-Jeong Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea
| | - Tara Man Kadayat
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea
| | - Jaeyoung Song
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea
| | - Sang Woo Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Jae-Han Jeon
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, 41404, South Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Keun-Gyu Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - In-Kyu Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, 41404, South Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea.
| | - Jungwook Chin
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea.
| | - Sung Jin Cho
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, 41404, South Korea.
| |
Collapse
|
48
|
Jung YS, Kim YH, Radhakrishnan K, kim J, Kim DK, Lee JH, Oh H, Lee IK, Kim W, Cho SJ, Choi CS, Dooley S, Egan JM, Lee CH, Choi HS. An inverse agonist of estrogen-related receptor γ regulates 2-arachidonoylglycerol synthesis by modulating diacylglycerol lipase expression in alcohol-intoxicated mice. Arch Toxicol 2020; 94:427-438. [DOI: https:/doi.org/10.1007/s00204-019-02648-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/12/2019] [Indexed: 09/18/2023]
|
49
|
An inverse agonist of estrogen-related receptor γ regulates 2-arachidonoylglycerol synthesis by modulating diacylglycerol lipase expression in alcohol-intoxicated mice. Arch Toxicol 2020; 94:427-438. [PMID: 31912162 PMCID: PMC10131092 DOI: 10.1007/s00204-019-02648-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/12/2019] [Indexed: 01/01/2023]
Abstract
Chronic alcohol feeding increases the levels of 2-arachidonoylglycerol (2-AG) in the liver, which activates hepatic cannabinoid receptor type 1 (CB1R), leading to oxidative liver injury. 2-AG biosynthesis is catalyzed by diacylglycerol lipase (DAGL). However, the mechanisms regulating hepatic DAGL gene expression and 2-AG production are largely unknown. In this study, we show that CB1R-induced estrogen-related receptor γ (ERRγ) controls hepatic DAGL gene expression and 2-AG levels. Arachidonyl-2'-chloroethylamide (ACEA), a synthetic CB1R agonist, significantly upregulated ERRγ, DAGLα, and DAGLβ, and increased 2-AG levels in the liver (10 mg/kg) and hepatocytes (10 μM) of wild-type (WT) mice. ERRγ overexpression upregulated DAGLα and DAGLβ expressions and increased 2-AG levels, whereas ERRγ knockdown abolished ACEA-induced DAGLα, DAGLβ, and 2-AG in vitro and in vivo. Promoter assays showed that ERRγ positively regulated DAGLα and DAGLβ transcription by binding to the ERR response element in the DAGLα and DAGLβ promoters. Chronic alcohol feeding (27.5% of total calories) induced hepatic steatosis and upregulated ERRγ, leading to increased DAGLα, DAGLβ, or 2-AG in WT mice, whereas these alcohol-induced effects did not occur in hepatocyte-specific CB1R knockout mice or in those treated with the ERRγ inverse agonist GSK5182 (40 mg/kg in mice and 10 μM in vitro). Taken together, these results indicate that suppression of alcohol-induced DAGLα and DAGLβ gene expressions and 2-AG levels by an ERRγ-specific inverse agonist may be a novel and attractive therapeutic approach for the treatment of alcoholic liver disease.
Collapse
|
50
|
Thouennon E, Delfosse V, Bailly R, Blanc P, Boulahtouf A, Grimaldi M, Barducci A, Bourguet W, Balaguer P. Insights into the activation mechanism of human estrogen-related receptor γ by environmental endocrine disruptors. Cell Mol Life Sci 2019; 76:4769-4781. [PMID: 31127318 PMCID: PMC11105698 DOI: 10.1007/s00018-019-03129-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/08/2019] [Accepted: 05/02/2019] [Indexed: 12/19/2022]
Abstract
The estrogen-related receptor γ (ERRγ, NR3B3) is a constitutively active nuclear receptor which has been proposed to act as a mediator of the low-dose effects of a number of environmental endocrine-disrupting chemicals (EDCs) such as the xenoestrogen bisphenol-A (BPA). To better characterize the ability of exogenous compounds to bind and activate ERRγ, we used a combination of cell-based, biochemical, structural and computational approaches. A purposely created stable cell line allowed for the determination of the EC50s for over 30 environmental ERRγ ligands, including previously unknown ones. Interestingly, affinity constants (Kds) of the most potent compounds measured by isothermal titration calorimetry were in the 50-500 nM range, in agreement with their receptor activation potencies. Crystallographic analysis of the interaction between the ERRγ ligand-binding domain (LBD) and compounds of the bisphenol, alkylphenol and naphthol families revealed a partially shared binding mode and minimal alterations of the receptor conformation upon ligand binding. Further biophysical characterizations coupled to molecular dynamics simulations suggested a mechanism through which ERRγ ligands would exhibit their agonistic properties by preserving the transcriptionally active form of the receptor while rigidifying some loop regions with associated functions. This unique mechanism contrasts with the classical one involving a ligand-induced repositioning and stabilization of the C-terminal activation helix H12.
Collapse
Affiliation(s)
- Erwan Thouennon
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm, Univ Montpellier, ICM, Montpellier, France
| | - Vanessa Delfosse
- Centre de Biochimie Structurale (CBS), Inserm, CNRS, Univ Montpellier, Montpellier, France
| | - Rémy Bailly
- Centre de Biochimie Structurale (CBS), Inserm, CNRS, Univ Montpellier, Montpellier, France
| | - Pauline Blanc
- Centre de Biochimie Structurale (CBS), Inserm, CNRS, Univ Montpellier, Montpellier, France
| | - Abdelhay Boulahtouf
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm, Univ Montpellier, ICM, Montpellier, France
| | - Marina Grimaldi
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm, Univ Montpellier, ICM, Montpellier, France
| | - Alessandro Barducci
- Centre de Biochimie Structurale (CBS), Inserm, CNRS, Univ Montpellier, Montpellier, France
| | - William Bourguet
- Centre de Biochimie Structurale (CBS), Inserm, CNRS, Univ Montpellier, Montpellier, France.
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm, Univ Montpellier, ICM, Montpellier, France.
| |
Collapse
|