1
|
Yang Y, Zhou J, Lv Q, Ni Q, Hu B, Wang Y, Qu S, Li G, Yang W, Zhong R, Chen X, Zhu Y. Canagliflozin alleviates progestin resistance by suppressing RARβ/CRABP2 signaling in THRB knockout endometrial cancer cells. Front Pharmacol 2025; 16:1573032. [PMID: 40371351 PMCID: PMC12075957 DOI: 10.3389/fphar.2025.1573032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/11/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction Progestin resistance has emerged as a significant barrier to the conservative management of endometrial cancer (EC). The mechanisms underlying progestin resistance in endocrine therapy remain incompletely understood. Previous studies have suggested that silencing thyroid hormone receptor B (THRB) is associated with progestin resistance in EC cells. Methods THRB-knockout RL95-2 (THRB(-/-)/RL95-2) cells were constructed to investigate progestin resistance mechanisms. Cell proliferation and apoptosis were assessed in RL95-2 and THRB(-/-)/RL95-2 cells treated with canagliflozin (CANA), medroxyprogesterone acetate (MPA), and their combination using CCK-8, EdU, and flow cytometry assays. In vivo, nude mouse xenograft models were used to evaluate the efficacy of CANA and MPA. Transcriptomic and proteomic analyses were performed to identify pathways associated with progestin resistance. Molecular dynamics simulations, along with western blotting and immunohistochemistry were utilized to validate the targets of CANA. Electrophoretic mobility shift assays and dual luciferase reporter assays were employed to investigate the interactions between TRβ, RARβ, and CRABP2. Results THRB(-/-)/RL95-2 cells were successfully constructed. CANA demonstrated a strong binding affinity for TRβ. Both MPA and CANA suppressed proliferation in RL95-2 cells, but MPA was ineffective in THRB(-/-)/RL95-2 cells, indicating that THRB deficiency induced progestin resistance. CANA significantly inhibited proliferation and promoted apoptosis in THRB(-/-)/RL95-2 cells. In vivo, CANA, either alone or in combination with MPA, significantly reduced tumor growth in xenograft models derived from both wild-type and THRB-knockout RL95-2 cells. Transcriptomic and proteomic analyses revealed that progestin resistance in EC was linked to the retinoic acid signaling pathways. Western blotting confirmed that the expressions of RARβ, RXRA and CRABP2 were significantly elevated in THRB(-/-)/RL95-2 cells. Treatment with CANA, alone or in combination with MPA, effectively reduced the expression of these proteins. Immunohistochemical analysis demonstrated that RARβ expression was significantly increased in uterine tissues from patients with progestin-insensitive EC or endometrial atypical hyperplasia. Electrophoretic mobility shift assays and dual luciferase reporter assays demonstrated that TRβ negatively modulated RARβ expression by binding to its promoter, while RARβ positively regulated CRABP2 expression. Conclusion THRB knockout activated retinoic acid pathway, leading to progestin resistance. CANA targeted RARβ and RXRA, downregulated CRABP2, restored BAX levels, and counteracted progestin resistance. The combination of CANA and MPA presented a novel strategy for alleviating progestin resistance and enhancing clinical efficacy.
Collapse
Affiliation(s)
- Ye Yang
- School of Pharmacy, Fudan University, Shanghai, China
- Laboratory of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jieyun Zhou
- Laboratory of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Qiaoying Lv
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qicheng Ni
- School of Pharmacy, Fudan University, Shanghai, China
- Laboratory of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Baichun Hu
- Key Laboratory of Structure Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
| | - Yulong Wang
- Key Laboratory of Structure Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
| | - Shuning Qu
- Key Laboratory of Structure Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
| | - Guoting Li
- Laboratory of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Wenjie Yang
- Laboratory of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Ruihua Zhong
- Laboratory of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Xiaojun Chen
- Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yan Zhu
- Laboratory of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| |
Collapse
|
2
|
Zhang Y, Gailloud L, Shin A, Fewkes J, Pinckney R, Whiting A, Chazot P. A Comparative Study of a Potent CNS-Permeable RARβ-Modulator, Ellorarxine, in Neurons, Glia and Microglia Cells In Vitro. Int J Mol Sci 2025; 26:3551. [PMID: 40332055 PMCID: PMC12027090 DOI: 10.3390/ijms26083551] [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: 01/21/2025] [Revised: 02/28/2025] [Accepted: 03/20/2025] [Indexed: 05/08/2025] Open
Abstract
Vitamin A (retinol) and its derivatives (retinoids) assume critical roles in neural development, cellular differentiation, axon elongation, programmed cell apoptosis and various fundamental cellular processes. Retinoids function by binding to specific nuclear receptors, such as retinoic acid receptors (RARs) and retinoid X receptors (RXRs), activating specific signalling pathways in the cells. The disruption of the retinoic acid signalling pathway can result in neuroinflammation, oxidative and ER stress and mitochondrial dysfunction and has been implicated in a wide range of neurodegenerative diseases. The present study explored the potential therapeutic application of our innovative CNS-permeable synthetic retinoid, Ellorarxine, for the treatment of neurodegenerative disorders in vitro. An MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) tetrazolium assay, lactate dehydrogenase (LDH) assay, enzyme-linked immunosorbent assay (ELISA), immunocytochemistry and immunofluorescence staining were performed. Ellorarxine increased Cyp26 and, selectively, RARβ protein expression in neurons, glia and microglia. Ellorarxine significantly reduced cell death (neurons, glia), increased mitochondrial viability (neurons), modulated cytokine release (microglia), and positively regulated cellular autophagy (neurons, glia, microglia). These results suggest that Ellorarxine is a promising drug candidate that should be further investigated in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yunxi Zhang
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (Y.Z.); (L.G.); (A.S.); (J.F.); (R.P.)
| | - Lilie Gailloud
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (Y.Z.); (L.G.); (A.S.); (J.F.); (R.P.)
- Department of Pharmacology, University College London, London WC1E 6BT, UK
| | - Alexander Shin
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (Y.Z.); (L.G.); (A.S.); (J.F.); (R.P.)
| | - Jessica Fewkes
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (Y.Z.); (L.G.); (A.S.); (J.F.); (R.P.)
| | - Rosella Pinckney
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (Y.Z.); (L.G.); (A.S.); (J.F.); (R.P.)
| | - Andrew Whiting
- Department of Chemistry, Science Laboratories, Durham University, South Road, Durham DH1 3LE, UK;
| | - Paul Chazot
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (Y.Z.); (L.G.); (A.S.); (J.F.); (R.P.)
| |
Collapse
|
3
|
Chitranshi N, Dheer Y, Kumar S, Graham SL, Gupta V. Molecular docking, dynamics, and pharmacology studies on bexarotene as an agonist of ligand-activated transcription factors, retinoid X receptors. J Cell Biochem 2019; 120:11745-11760. [PMID: 30746761 DOI: 10.1002/jcb.28455] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 01/24/2023]
Abstract
Retinoid X receptors (RXRs) belong to the nuclear receptor superfamily, and upon ligand activation, these receptors control gene transcription via either homodimerization with themselves or heterodimerization with the partner-nuclear receptor. The protective effects of RXRs and RXR agonists have been reported in several neurodegenerative diseases, including in the retina. This study was aimed to prioritize compounds from natural and synthetic origin retinoids as potential RXR agonists by molecular docking and molecular dynamic simulation strategies. The docking studies indicated bexarotene as a lead compound that can activate various RXR receptor isoforms (α, β, and γ) and has a strong binding affinity to the receptor protein than retinoic acid, which is known as a natural endogenous RXR agonist. Dynamic simulation studies confirmed that the hydrogen bonding and hydrophobic interactions were highly stable in all the three isoforms of the RXR-bexarotene complex. To further validate the significance of the RXR receptor in neurons, in vitro pharmacological treatment of neuronal SH-SY5Y cells with bexarotene was performed. In vitro data from SH-SY5Y cells confirmed that bexarotene activated RXR-simulated neurite outgrowth significantly. We conclude that bexarotene could be potentially used as an exogenous activator of RXRs and emerge as a good drug target for several neurodegenerative disorders.
Collapse
Affiliation(s)
- Nitin Chitranshi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales
| | - Yogita Dheer
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales
| | - Sanjay Kumar
- Bioinformatics Centre, Biotech Park, Jankipuram, Lucknow, Uttar Pradesh, India
| | - Stuart L Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales
- Save Sight Institute, Sydney University, Sydney, New South Wales, Australia
| | - Vivek Gupta
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales
| |
Collapse
|
4
|
Kovalevich J, Yen W, Ozdemir A, Langford D. Cocaine induces nuclear export and degradation of neuronal retinoid X receptor-γ via a TNF-α/JNK- mediated mechanism. J Neuroimmune Pharmacol 2015; 10:55-73. [PMID: 25586717 PMCID: PMC4336643 DOI: 10.1007/s11481-014-9573-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 11/26/2014] [Indexed: 12/14/2022]
Abstract
Cocaine abuse represents an immense societal health and economic burden for which no effective treatment currently exists. Among the numerous intracellular signaling cascades impacted by exposure to cocaine, increased and aberrant production of pro-inflammatory cytokines in the CNS has been observed. Additionally, we have previously reported a decrease in retinoid-X-receptor-gamma (RXR-γ) in brains of mice chronically exposed to cocaine. Through obligate heterodimerization with a number of nuclear receptors, RXRs serve as master regulatory transcription factors, which can potentiate or suppress expression of a wide spectrum of genes. Little is known about the regulation of RXR levels, but previous studies indicate cellular stressors such as cytokines negatively regulate levels of RXRs in vitro. To evaluate the mechanism underlying the cocaine-induced decreases in RXR-γ levels observed in vivo, we exposed neurons to cocaine in vitro and examined pathways which may contribute to disruption in RXR signaling, including activation of stress pathways by cytokine induction. In these studies, we provide the first evidence that cocaine exposure disrupts neuronal RXR-γ signaling in vitro by promoting its nuclear export and degradation. Furthermore, we demonstrate this effect may be mediated, at least in part, by cocaine-induced production of TNF-α and its downstream effector c-Jun-NH-terminal kinase (JNK). Findings from this study are therefore applicable to both cocaine abuse and to pathological conditions characterized by neuroinflammatory factors, such as neurodegenerative disease.
Collapse
Affiliation(s)
- Jane Kovalevich
- Department of Neuroscience, Temple University School of Medicine, Medical Education Research Building, 3500 North Broad Street, Philadelphia, PA, 19140, USA
| | | | | | | |
Collapse
|
5
|
Devanna P, Middelbeek J, Vernes SC. FOXP2 drives neuronal differentiation by interacting with retinoic acid signaling pathways. Front Cell Neurosci 2014; 8:305. [PMID: 25309332 PMCID: PMC4176457 DOI: 10.3389/fncel.2014.00305] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/10/2014] [Indexed: 11/14/2022] Open
Abstract
FOXP2 was the first gene shown to cause a Mendelian form of speech and language disorder. Although developmentally expressed in many organs, loss of a single copy of FOXP2 leads to a phenotype that is largely restricted to orofacial impairment during articulation and linguistic processing deficits. Why perturbed FOXP2 function affects specific aspects of the developing brain remains elusive. We investigated the role of FOXP2 in neuronal differentiation and found that FOXP2 drives molecular changes consistent with neuronal differentiation in a human model system. We identified a network of FOXP2 regulated genes related to retinoic acid signaling and neuronal differentiation. FOXP2 also produced phenotypic changes associated with neuronal differentiation including increased neurite outgrowth and reduced migration. Crucially, cells expressing FOXP2 displayed increased sensitivity to retinoic acid exposure. This suggests a mechanism by which FOXP2 may be able to increase the cellular differentiation response to environmental retinoic acid cues for specific subsets of neurons in the brain. These data demonstrate that FOXP2 promotes neuronal differentiation by interacting with the retinoic acid signaling pathway and regulates key processes required for normal circuit formation such as neuronal migration and neurite outgrowth. In this way, FOXP2, which is found only in specific subpopulations of neurons in the brain, may drive precise neuronal differentiation patterns and/or control localization and connectivity of these FOXP2 positive cells.
Collapse
Affiliation(s)
- Paolo Devanna
- Language and Genetics Department, Max Planck Institute for Psycholinguistics Nijmegen, Netherlands
| | - Jeroen Middelbeek
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Netherlands
| | - Sonja C Vernes
- Language and Genetics Department, Max Planck Institute for Psycholinguistics Nijmegen, Netherlands ; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Netherlands
| |
Collapse
|
6
|
Dummer R, Beyer M, Hymes K, Epping MT, Bernards R, Steinhoff M, Sterry W, Kerl H, Heath K, Ahern JD, Hardwick JS, Garcia-Vargas J, Baumann K, Rizvi S, Frankel SR, Whittaker SJ, Assaf C. Vorinostat combined with bexarotene for treatment of cutaneous T-cell lymphoma:in vitroand phase I clinical evidence supporting augmentation of retinoic acid receptor/retinoid X receptor activation by histone deacetylase inhibition. Leuk Lymphoma 2012; 53:1501-8. [DOI: 10.3109/10428194.2012.656625] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
7
|
Ishizawa M, Kagechika H, Makishima M. NR4A nuclear receptors mediate carnitine palmitoyltransferase 1A gene expression by the rexinoid HX600. Biochem Biophys Res Commun 2012; 418:780-5. [PMID: 22310716 DOI: 10.1016/j.bbrc.2012.01.102] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 01/21/2012] [Indexed: 12/24/2022]
Abstract
Retinoid X receptors (RXRs) are members of the nuclear receptor superfamily and can be activated by 9-cis retinoic acid (9CRA). RXRs form homodimers and heterodimers with other nuclear receptors such as the retinoic acid receptor and NR4 subfamily nuclear receptors, Nur77 and NURR1. Potential physiological roles of the Nur77-RXR and NURR1-RXR heterodimers have not been elucidated. In this study, we identified a gene regulated by these heterodimers utilizing HX600, a selective RXR agonist for Nur77-RXR and NURR1-RXR. While 9CRA induced many genes, including RAR-target genes, HX600 effectively induced only carnitine palmitoyltransferase 1A (CPT1A) in human teratocarcinoma NT2/D1 cells, which express RXRα, Nur77 and NURR1. HX600 also increased CPT1A expression in human embryonic kidney (HEK) 293 cells and hepatocyte-derived HepG2 cells. Although HX600 induced CPT1A less effectively than 9CRA, overexpression of Nur77 or NURR1 increased the HX600 response to levels similar to 9CRA in NT2/D1 and HEK293 cells. A dominant-negative form of Nur77 or NURR1 repressed the induction of CPT1A by HX600. A protein synthesis inhibitor did not alter HX600-dependent CPT1A induction. Thus, the rexinoid HX600 directly induces expression of CPT1A through a Nur77 or NURR1-mediated mechanism. CPT1A, a gene involved in fatty acid β-oxidation, could be a target of RXR-NR4 receptor heterodimers.
Collapse
Affiliation(s)
- Michiyasu Ishizawa
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | | | | |
Collapse
|
8
|
Hewson QDC, Lovat PE, Corazzari M, Catterall JB, Redfern CPF. The NF-kappaB pathway mediates fenretinide-induced apoptosis in SH-SY5Y neuroblastoma cells. Apoptosis 2005; 10:493-8. [PMID: 15909111 DOI: 10.1007/s10495-005-1878-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Fenretinide induces apoptosis in SH-SY5Y neuroblastoma cells via a signaling pathway involving the production of reactive oxygen species (ROS), 12-lipoxygenase activity and the induction of the GADD153 transcription factor. NF-kappa B is a key element of many cell signaling pathways and adopts a pro- or anti-apoptotic role in different cell types. Studies have suggested that NF-kappa B may play a pro-apoptotic role in SH-SY5Y cells, and in other cell types NF-kappa B activation may be linked to lipoxygenase activity. The aim of this study was to test the hypothesis that NF-kappa B activity mediates fenretinide-induced apoptosis in SH-SY5Y neuroblastoma cells. Using a dominant-negative construct for Ikappa Balpha stably transfected into SH-SY5Y cells, we show that apoptosis, but not the induction of ROS, in response to fenretinide was blocked by abrogation of NF-kappa B activity. In parental SH-SY5Y cells, fenretinide induced NF-kappa B activity and Ikappa Balpha phosphorylation. These results suggest that NF-kappa B activity links fenretinide-induced ROS to the induction of apoptosis in SH-SH5Y cells, and may be a target for the future development of drugs for neuroblastoma therapy.
Collapse
Affiliation(s)
- Q D Campbell Hewson
- Northern Institute for Cancer Research and School of Clinical Medical Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne, NE2 4HH, UK
| | | | | | | | | |
Collapse
|