1
|
Abdelaziz AM. Alpha-Synuclein drives NURR1 and NLRP3 Inflammasome dysregulation in Parkinson's disease: From pathogenesis to potential therapeutic strategies. Int Immunopharmacol 2025; 156:114692. [PMID: 40267723 DOI: 10.1016/j.intimp.2025.114692] [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: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Parkinson's disease (PD), a progressive neurodegenerative disorder, is characterized by the loss of dopaminergic neurons and pathological aggregation of α-synuclein (α-Syn). Emerging evidence highlights the interplay between genetic susceptibility, neuroinflammation, and transcriptional dysregulation in driving PD pathogenesis. This review brings together the latest information on three important players: α-Syn, the transcription factor Orphan nuclear receptor (NURR1), and the NOD-like receptor 3 (NLRP3) inflammasome. Pathogenic α-syn aggregates cause damage to neurons by disrupting mitochondria and lysosomes and spreading in a way similar to prion proteins. They also turn on the NLRP3 inflammasome, which is a key player in neuroinflammation. NLRP3-driven release of pro-inflammatory cytokines exacerbates neurodegeneration and creates a self-sustaining inflammatory milieu. Meanwhile, reduced NURR1 activity, a pivotal modulator of dopaminergic neuron survival and development, exposes neurons to oxidative stress, neuroinflammation, and α-Syn toxicity, hence exacerbating disease progression. So, targeting this trio exhibits transformative potential against PD pathogenesis.
Collapse
Affiliation(s)
- Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511, Egypt.
| |
Collapse
|
2
|
Matošević A, Opsenica DM, Bartolić M, Maraković N, Stoilković A, Komatović K, Zandona A, Žunec S, Bosak A. Derivatives of Amodiaquine as Potent Human Cholinesterases Inhibitors: Implication for Treatment of Alzheimer's Disease. Molecules 2024; 29:5357. [PMID: 39598746 PMCID: PMC11596630 DOI: 10.3390/molecules29225357] [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: 09/30/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
As some previously reported studies have proven that amodiaquine, in addition to its primary antimalarial activity, also has potential for new applications such as the inhibition of cholinesterases, in our study we focused on the evaluation of the influence of different substituents in the aminoquinoline part of the amodiaquine structure on the inhibition of human acetylcholinesterase and butyrylcholinesterase to investigate the possibility for their use as drugs for the treatment of AD. We synthesized a series of amodiaquine derivatives bearing H-, F-, CF3-, NO2-, CN-, CO2H- or CH3O- groups on the aminoquinoline ring, and determined that all of the tested derivatives were very potent inhibitors of both cholinesterases, with inhibition constants (Ki) in the nM and low μM range and with prominent selectivity (up to 300 times) for the inhibition of acetylcholinesterase. All compounds displayed an ability to chelate biometal ions Fe2+, Zn2+ and Cu2+ and an antioxidant power comparable to that of standard antioxidants. Most of the compounds were estimated to be able to cross the blood-brain barrier by passive transport and were nontoxic toward cells that represent the models of individual organs. Considering all these beneficial features, our study has singled out compound 5, the most potent AChE inhibitor with a CH3O- on C(7) position, followed by 6 and 14, compounds without substituent or hydroxyl groups in the C(17) position, respectively, as the most promising compounds from the series which could be considered as potential multi-target drugs for the treatment of AD.
Collapse
Affiliation(s)
- Ana Matošević
- Institute for Medical Research and Occupational Health, Ksaverska Cesta 2, 10001 Zagreb, Croatia; (A.M.); (M.B.); (N.M.); (A.Z.); (S.Ž.)
| | - Dejan M. Opsenica
- Institute of Chemistry Technology and Metallurgy, University of Belgrade, Njegoševa 12, 11000 Beograd, Serbia;
- Centre of Excellence in Environmental Chemistry and Engineering, Njegoševa 12, 11000 Belgrade, Serbia
| | - Marija Bartolić
- Institute for Medical Research and Occupational Health, Ksaverska Cesta 2, 10001 Zagreb, Croatia; (A.M.); (M.B.); (N.M.); (A.Z.); (S.Ž.)
| | - Nikola Maraković
- Institute for Medical Research and Occupational Health, Ksaverska Cesta 2, 10001 Zagreb, Croatia; (A.M.); (M.B.); (N.M.); (A.Z.); (S.Ž.)
| | - Andriana Stoilković
- Institute of Chemistry Technology and Metallurgy, University of Belgrade, Njegoševa 12, 11000 Beograd, Serbia;
| | - Katarina Komatović
- Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11158 Belgrade, Serbia;
| | - Antonio Zandona
- Institute for Medical Research and Occupational Health, Ksaverska Cesta 2, 10001 Zagreb, Croatia; (A.M.); (M.B.); (N.M.); (A.Z.); (S.Ž.)
| | - Suzana Žunec
- Institute for Medical Research and Occupational Health, Ksaverska Cesta 2, 10001 Zagreb, Croatia; (A.M.); (M.B.); (N.M.); (A.Z.); (S.Ž.)
| | - Anita Bosak
- Institute for Medical Research and Occupational Health, Ksaverska Cesta 2, 10001 Zagreb, Croatia; (A.M.); (M.B.); (N.M.); (A.Z.); (S.Ž.)
| |
Collapse
|
3
|
Kambey PA, Wu J, Liu W, Su M, Buberwa W, Tang C. Targeting serum response factor (SRF) deactivates ΔFosB and mitigates Levodopa-induced dyskinesia in a mouse model of Parkinson's disease. Gene Ther 2024; 31:614-624. [PMID: 39384937 DOI: 10.1038/s41434-024-00492-8] [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: 11/17/2023] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA) is currently the preferred treatment for Parkinson's Disease (PD) and is considered the gold standard. However, prolonged use of L-DOPA in patients can result in involuntary movements known as Levodopa-induced dyskinesia (LID), which includes uncontrollable dystonia affecting the trunk, limbs, and face. The role of ΔFosB protein, a truncated splice variant of the FosB gene, in LID has been acknowledged, but its underlying mechanism has remained elusive. Here, using a mouse model of Parkinson's disease treated with chronic levodopa we demonstrate that serum response factor (SRF) binds to the FosB promoter, thereby activating FosB expression and levodopa induced-dyskinetic movements. Western blot analysis demonstrates a significant increase in SRF expression in the dyskinetic group compared to the control group. Knocking down SRF significantly reduced abnormal involuntary movements (AIMS) and ΔFosB expression compared to the control. Conversely, overexpression of SRF led to an increase in ΔFosB expression and worsened levodopa-induced dyskinesia. To shed light on the regulatory role of the Akt signaling pathway in this phenomenon, we administered the Akt agonist SC79 to PD mouse models via intraperitoneal injection, followed by L-DOPA administration. The expression of SRF, ΔFosB, and phosphorylated Akt (p-Akt) significantly increased in this group compared to the group receiving normal saline to signify that these happen through Akt signaling pathway. Collectively, our findings identify a promising therapeutic target for addressing levodopa-induced dyskinesia.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
- Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Huangpu District, Guangzhou, China.
| | - Jiao Wu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - WenYa Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Mingyu Su
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Wokuheleza Buberwa
- Department of Neurology, The second affiliated hospital of Xi'an Jiaotong University, 710049, Xi'an, China
| | - Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
4
|
Kambey PA, Liu WY, Wu J, Tang C, Buberwa W, Saro A, Nyalali AMK, Gao D. Amphiregulin blockade decreases the levodopa-induced dyskinesia in a 6-hydroxydopamine Parkinson's disease mouse model. CNS Neurosci Ther 2023; 29:2925-2939. [PMID: 37101388 PMCID: PMC10493657 DOI: 10.1111/cns.14229] [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: 01/18/2023] [Revised: 03/09/2023] [Accepted: 04/12/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Levodopa (L-DOPA) is considered the most reliable drug for treating Parkinson's disease (PD) clinical symptoms. Regrettably, long-term L-DOPA therapy results in the emergence of drug-induced abnormal involuntary movements (AIMs) in most PD patients. The mechanisms underlying motor fluctuations and dyskinesia induced by L-DOPA (LID) are still perplexing. METHODS Here, we first performed the analysis on the microarray data set (GSE55096) from the gene expression omnibus (GEO) repository and identified the differentially expressed genes (DEGs) using linear models for microarray analysis (Limma) R packages from the Bioconductor project. 12 genes (Nr4a2, Areg, Tinf2, Ptgs2, Pdlim1, Tes, Irf6, Tgfb1, Serpinb2, Lipg, Creb3l1, Lypd1) were found to be upregulated. Six genes were validated on quantitative polymerase chain reaction and subsequently, Amphiregulin (Areg) was selected (based on log2 fold change) for further experiments to unravel its involvement in LID. Areg LV_shRNA was used to knock down Areg to explore its therapeutic role in the LID model. RESULTS Western blotting and immunofluorescence results show that AREG is significantly expressed in the LID group relative to the control. Dyskinetic movements in LID mice were alleviated by Areg knockdown, and the protein expression of delta FOSB, the commonly attributable protein in LID, was decreased. Moreover, Areg knockdown reduced the protein expression of P-ERK. In order to ascertain whether the inhibition of the ERK pathway (a common pathway known to mediate levodopa-induced dyskinesia) could also impede Areg, the animals were injected with an ERK inhibitor (PD98059). Afterward, the AIMs, AREG, and ERK protein expression were measured relative to the control group. A group treated with ERK inhibitor had a significant decrease of AREG and phosphorylated ERK protein expression relative to the control group. CONCLUSION Taken together, our results indicate unequivocal involvement of Areg in levodopa-induced dyskinesia, thus a target for therapy development.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and AnatomyXuzhou Medical UniversityXuzhouChina
- Organization of African Academic Doctors (OAAD)NairobiKenya
| | - Wen Ya Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and AnatomyXuzhou Medical UniversityXuzhouChina
| | - Jiao Wu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and AnatomyXuzhou Medical UniversityXuzhouChina
| | - Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and AnatomyXuzhou Medical UniversityXuzhouChina
| | - Wokuheleza Buberwa
- Department of PediatricsThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Adonira Saro
- Department of Anatomy and Neurobiology, School of Basic Medical ScienceCentral South UniversityChangshaChina
| | - Alphonce M. K. Nyalali
- Department of Neurosurgery, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Dianshuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and AnatomyXuzhou Medical UniversityXuzhouChina
| |
Collapse
|
5
|
Su J, Yang P, Xing M, Chen B, Xie X, Ding J, Lu M, Liu Y, Guo Y, Hu G. Neuroprotective effects of a lead compound from coral via modulation of the orphan nuclear receptor Nurr1. CNS Neurosci Ther 2022; 29:893-906. [PMID: 36419251 PMCID: PMC9928544 DOI: 10.1111/cns.14025] [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: 05/22/2022] [Revised: 08/03/2022] [Accepted: 08/14/2022] [Indexed: 11/26/2022] Open
Abstract
AIMS To screen coral-derived compounds with neuroprotective activity and clarify the potential mechanism of lead compounds. METHODS The lead compounds with neuroprotective effects were screened by H2 O2 and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPP+ )-induced cell damage models in SH-SY5Y cells. CCK8 and LDH assays were used to detect cell viability. The anti-apoptosis of lead compounds was evaluated by flow cytometry. JC-1 and MitoSox assays were performed to examine the changes in mitochondrial membrane potential and mitochondrial ROS level. Survival of primary cortical and dopaminergic midbrain neurons was measured by MAP2 and TH immunoreactivities. The Caenorhabditis elegans (C. elegans) model was established to determine the effect of lead compounds on dopaminergic neurons and behavior changes. RESULTS Three compounds (No. 63, 68, and 74), derived from marine corals, could markedly alleviate the cell damage and notably reverse the loss of worm dopaminergic neurons. Further investigation indicated that compound 63 could promote the expression of Nurr1 and inhibit neuronal apoptosis signaling pathways. CONCLUSION Lead compounds from marine corals exerted significant neuroprotective effects, which indicated that coral might be a new and potential resource for screening and isolating novel natural compounds with neuroprotective effects. Furthermore, this study also provided a new strategy for the clinical treatment of neurodegenerative diseases such as Parkinson's disease.
Collapse
Affiliation(s)
- Jian‐Wei Su
- Department of PharmacologySchool of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Pei Yang
- Department of PharmacologySchool of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Mei‐Mei Xing
- Department of PharmacologySchool of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Bao Chen
- State Key Laboratory of Drug ResearchShanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Xia‐Hong Xie
- Department of PharmacologySchool of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Jian‐Hua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of PharmacologyNanjing Medical UniversityNanjingChina
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of PharmacologyNanjing Medical UniversityNanjingChina
| | - Yang Liu
- Department of PharmacologySchool of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Yue‐Wei Guo
- State Key Laboratory of Drug ResearchShanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Gang Hu
- Department of PharmacologySchool of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjingJiangsuChina
| |
Collapse
|
6
|
Abdollahi M, Fahnestock M. Nurr1 Is Not an Essential Regulator of BDNF in Mouse Cortical Neurons. Int J Mol Sci 2022; 23:6853. [PMID: 35743300 PMCID: PMC9224520 DOI: 10.3390/ijms23126853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/30/2022] Open
Abstract
Nurr1 and brain-derived neurotrophic factor (BDNF) play major roles in cognition. Nurr1 regulates BDNF in midbrain dopaminergic neurons and cerebellar granule cells. Nurr1 and BDNF are also highly expressed in the cerebral cortex, a brain area important in cognition. Due to Nurr1 and BDNF tissue specificity, the regulatory effect of Nurr1 on BDNF in different brain areas cannot be generalized. The relationship between Nurr1 and BDNF in the cortex has not been investigated previously. Therefore, we examined Nurr1-mediated BDNF regulation in cortical neurons in activity-dependent and activity-independent states. Mouse primary cortical neurons were treated with the Nurr1 agonist, amodiaquine (AQ). Membrane depolarization was induced by KCl or veratridine and reversed by nimodipine. AQ and membrane depolarization significantly increased Nurr1 (p < 0.001) and BDNF (pAQ < 0.001, pKCl < 0.01) as assessed by real-time qRT-PCR. However, Nurr1 knockdown did not affect BDNF gene expression in resting or depolarized neurons. Accordingly, the positive correlation between Nurr1 and BDNF expression in AQ and membrane depolarization experiments does not imply co-regulation because Nurr1 knockdown did not affect BDNF gene expression in resting or depolarized cortical neurons. Therefore, in contrast to midbrain dopaminergic neurons and cerebellar granule cells, Nurr1 does not regulate BDNF in cortical neurons.
Collapse
Affiliation(s)
- Mona Abdollahi
- Medical Sciences Graduate Program, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada;
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
7
|
Hu X, Xiang J, Li Y, Xia Y, Xu S, Gao X, Qiao S. Inhibition of stearoyl-CoA desaturase 1 potentiates anti-tumor activity of amodiaquine in non-small cell lung cancer. Biol Pharm Bull 2022; 45:438-445. [PMID: 35110426 DOI: 10.1248/bpb.b21-00843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer related death with few therapeutic treatment options. Under adverse tumor microenvironment, autophagy is an important mechanism of metabolic adaptations to sustain the survival and proliferation of tumor cells. Therefore, targeting autophagic activity represents a promising opportunity for NSCLC treatment. Here, we found that amodiaquine (AQ) increased autophagosome numbers and LC3BII and p62 at protein levels in A549 lung cancer cells suggesting the blockade of autophagic flux by AQ. To identify the key metabolic vulnerability associated with autophagy inhibition by AQ treatment, we then performed transcriptomics analysis in the presence or absence of AQ in A549 lung cancer cells and found stearoyl-CoA desaturase 1 (SCD) 1 was one of the most highly upregulated with AQ exposure. The induction of SCD1 by AQ exposure at both protein and mRNA level suggests that SCD1 could represent a potential therapeutic target of AQ treatment. Treatment of AQ in combination with SCD1 inhibition by A939572 demonstrated robust synergistic anti-cancer efficacy in cell proliferation assay and a lung cancer mouse xenograft model. Taken together, our study identified SCD1 could be a new therapeutic target upon autophagy inhibition by AQ exposure. Combinational treatment of autophagy inhibition and SCD1 inhibition achieves synergistic anti-tumor effect both in vitro and in vivo. This combinational approach could be a promising strategy for NSCLC treatment.
Collapse
Affiliation(s)
- Xiaolei Hu
- Cancer Institute, Xuzhou Medical University
| | | | - Yibo Li
- Cancer Institute, Xuzhou Medical University
| | - Yan Xia
- Cancer Institute, Xuzhou Medical University
| | - Siyuan Xu
- Cancer Institute, Xuzhou Medical University
| | - Xiaoge Gao
- Cancer Institute, Xuzhou Medical University
| | - Shuxi Qiao
- Cancer Institute, Xuzhou Medical University
| |
Collapse
|
8
|
Zaienne D, Willems S, Schierle S, Heering J, Merk D. Development and Profiling of Inverse Agonist Tools for the Neuroprotective Transcription Factor Nurr1. J Med Chem 2021; 64:15126-15140. [PMID: 34633810 DOI: 10.1021/acs.jmedchem.1c01077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The ligand-sensing transcription factor nuclear receptor related 1 (Nurr1) evolves as an appealing target to treat neurodegenerative diseases. Despite its therapeutic potential observed in various rodent models, potent modulators for Nurr1 are lacking as pharmacological tools. Here, we report the structure-activity relationship and systematic optimization of indole-based inverse Nurr1 agonists. Optimized analogues decreased the receptor's intrinsic transcriptional activity by up to more than 90% and revealed preference for inhibiting Nurr1 monomer activity. In orthogonal cell-free settings, we detected displacement of NCoRs and disruption of the Nurr1 homodimer as molecular modes of action. The inverse Nurr1 agonists reduced the expression of Nurr1-regulated genes in T98G cells, and treatment with an inverse Nurr1 agonist mimicked the effect of Nurr1 silencing on interleukin-6 release from LPS-stimulated human astrocytes. The indole-based inverse Nurr1 agonists valuably extend the toolbox of Nurr1 modulators to further probe the role of Nurr1 in neuroinflammation, cancer, and beyond.
Collapse
Affiliation(s)
- Daniel Zaienne
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany
| | - Sabine Willems
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany
| | - Simone Schierle
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, D-60596 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, D-60438 Frankfurt, Germany.,Department of Pharmacy, Ludwig-Maximilians-Universität München, D-81377 Munich, Germany
| |
Collapse
|
9
|
Mallick S, Marshall PA, Wagner CE, Heck MC, Sabir ZL, Sabir MS, Dussik CM, Grozic A, Kaneko I, Jurutka PW. Evaluating Novel RXR Agonists That Induce ApoE and Tyrosine Hydroxylase in Cultured Human Glioblastoma Cells. ACS Chem Neurosci 2021; 12:857-871. [PMID: 33570383 DOI: 10.1021/acschemneuro.0c00707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
There is considerable interest in identifying effective and safe drugs for neurodegenerative disorders. Cell culture and animal model work have demonstrated that modulating gene expression through RXR-mediated pathways may mitigate or reverse cognitive decline. However, because RXR is a dimeric partner for several transcription factors, activating off-target transcription is a concern with RXR ligands (rexinoids). This off-target gene modulation leads to unwanted side effects that can include low thyroid function and significant hyperlipidemia. There is a need to develop rexinoids that have binding specificity for subsets of RXR heterodimers, to drive desired gene modulation, but that do not induce spurious effects. Herein, we describe experiments in which we analyze a series of novel and previously reported rexinoids for their ability to modulate specific gene pathways implicated in neurodegenerative disorders employing a U87 cell culture model. We demonstrate that, compared to the FDA-approved rexinoid bexarotene (1), several of these compounds are equally or more effective at stimulating gene expression via LXREs or Nurr1/NBREs and are superior at inducing ApoE and/or tyrosine hydroxylase (TH) gene and protein expression, including analogs 8, 9, 13, 14, 20, 23, and 24, suggesting a possible therapeutic role for these compounds in Alzheimer's or Parkinson's disease (PD). A subset of these potent RXR agonists can synergize with a presumed Nurr1 ligand and antimalarial drug (amodiaquine) to further enhance Nurr1/NBREs-directed transcription. This novel discovery has potential clinical implications for treatment of PD since it suggests that the combination of an RXR agonist and a Nurr1 ligand can significantly enhance RXR-Nurr1 heterodimer activity and drive enhanced therapeutic expression of the TH gene to increase endogenous synthesis of dopamine. These data indicate that is it possible and prudent to develop novel rexinoids for testing of gene expression and side effect profiles for use in potential treatment of neurodegenerative disorders, as individual rexinoids can have markedly different gene expression profiles but similar structures.
Collapse
Affiliation(s)
- Sanchita Mallick
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Pamela A. Marshall
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Carl E. Wagner
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Michael C. Heck
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Zhela L. Sabir
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Marya S. Sabir
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Christoper M. Dussik
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Aleksandra Grozic
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Ichiro Kaneko
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Peter W. Jurutka
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| |
Collapse
|