Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
bioRxiv ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38712087

ABSTRACT

Nuclear receptors (NRs) are thought to dynamically alternate between transcriptionally active and repressive conformations, which are stabilized upon ligand binding. Most NR ligand series exhibit limited bias, primarily consisting of transcriptionally active agonists or neutral antagonists, but not repressive inverse agonists-a limitation that restricts understanding of the functional NR conformational ensemble. Here, we report a NR ligand series for peroxisome proliferator-activated receptor gamma (PPARγ) that spans a pharmacological spectrum from repression (inverse agonism) to activation (agonism) where subtle structural modifications switch compound activity. While crystal structures provide snapshots of the fully repressive state, NMR spectroscopy and conformation-activity relationship analysis reveals that compounds within the series shift the PPARγ conformational ensemble between transcriptionally active and repressive conformations that are populated in the apo/ligand-free ensemble. Our findings reveal a molecular framework for minimal chemical modifications that enhance PPARγ inverse agonism and elucidate their influence on the dynamic PPARγ conformational ensemble.

2.
bioRxiv ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38746089

ABSTRACT

We have identified a NMIIA and IIB-specific small molecule inhibitor, MT-125, and have studied its effects in GBM. MT-125 has high brain penetrance and retention and an excellent safety profile; blocks GBM invasion and cytokinesis, consistent with the known roles of NMII; and prolongs survival as a single agent in murine GBM models. MT-125 increases signaling along both the PDGFR- and MAPK-driven pathways through a mechanism that involves the upregulation of reactive oxygen species, and it synergizes with FDA-approved PDGFR and mTOR inhibitors in vitro . Combining MT-125 with sunitinib, a PDGFR inhibitor, or paxalisib, a combined PI3 Kinase/mTOR inhibitor significantly improves survival in orthotopic GBM models over either drug alone, and in the case of sunitinib, markedly prolongs survival in ∼40% of mice. Our results provide a powerful rationale for developing NMII targeting strategies to treat cancer and demonstrate that MT-125 has strong clinical potential for the treatment of GBM. Highlights: MT-125 is a highly specific small molecule inhibitor of non-muscle myosin IIA and IIB, is well-tolerated, and achieves therapeutic concentrations in the brain with systemic dosing.Treating preclinical models of glioblastoma with MT-125 produces durable improvements in survival.MT-125 stimulates PDGFR- and MAPK-driven signaling in glioblastoma and increases dependency on these pathways.Combining MT-125 with an FDA-approved PDGFR inhibitor in a mouse GBM model synergizes to improve median survival over either drug alone, and produces tumor free, prolonged survival in over 40% of mice.

3.
JCI Insight ; 9(2)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38051584

ABSTRACT

Cachexia is a debilitating skeletal muscle wasting condition for which we currently lack effective treatments. In the context of cancer, certain chemotherapeutics cause DNA damage and cellular senescence. Senescent cells exhibit chronic activation of the transcription factor NF-κB, a known mediator of the proinflammatory senescence-associated secretory phenotype (SASP) and skeletal muscle atrophy. Thus, targeting NF-κB represents a logical therapeutic strategy to alleviate unintended consequences of genotoxic drugs. Herein, we show that treatment with the IKK/NF-κB inhibitor SR12343 during a course of chemotherapy reduces markers of cellular senescence and the SASP in liver, skeletal muscle, and circulation and, correspondingly, attenuates features of skeletal muscle pathology. Lastly, we demonstrate that SR12343 mitigates chemotherapy-induced reductions in body weight, lean mass, fat mass, and muscle strength. These findings support senescent cells as a promising druggable target to counteract the SASP and skeletal muscle wasting in the context of chemotherapy.


Subject(s)
Antineoplastic Agents , NF-kappa B , Humans , NF-kappa B/metabolism , Signal Transduction , Cachexia/chemically induced , Cachexia/drug therapy , Senotherapeutics , Muscular Atrophy/chemically induced , Muscular Atrophy/drug therapy , Antineoplastic Agents/adverse effects
4.
J Med Chem ; 66(21): 14815-14823, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37888788

ABSTRACT

Autoimmune diseases affect 50 million Americans, predominantly women, and are thought to be one of the top 10 leading causes of death among women in age groups up to 65 years. A central role for TH17 cells has been highlighted by genome-wide association studies (GWAS) linking genes preferentially expressed in TH17 cells to several human autoimmune diseases. We and others have reported that the nuclear receptors REV-ERBα and ß are cell-intrinsic repressors of TH17 cell development and pathogenicity and might therefore be therapeutic targets for intervention. Herein, we describe detailed SAR studies of a novel REV-ERBα-selective scaffold. Metabolic stability of the ligands was optimized allowing for in vivo interrogation of the receptor in a mouse model of multiple sclerosis (EAE) with a ligand (34). Reduction in frequency and number of T-cells in the CNS as well as key REV-ERB target genes is a measure of target engagement in vivo.


Subject(s)
Genome-Wide Association Study , Multiple Sclerosis , Mice , Animals , Humans , Female , Male , Transcription Factors/genetics , Cell Differentiation , Multiple Sclerosis/drug therapy , Structure-Activity Relationship , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
5.
ACS Chem Biol ; 18(5): 1115-1123, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37146157

ABSTRACT

Inverse agonists of peroxisome proliferator activated receptor γ (PPARγ) have emerged as safer alternatives to full agonists for their reduced side effects while still maintaining impressive insulin-sensitizing properties. To shed light on their molecular mechanism, we characterized the interaction of the PPARγ ligand binding domain with SR10221. X-ray crystallography revealed a novel binding mode of SR10221 in the presence of a transcriptionally repressing corepressor peptide, resulting in much greater destabilization of the activation helix, H12, than without corepressor peptide. Electron paramagnetic resonance provided in-solution complementary protein dynamic data, which revealed that for SR10221-bound PPARγ, H12 adopts a plethora of conformations in the presence of corepressor peptide. Together, this provides the first direct evidence for corepressor-driven ligand conformation for PPARγ and will allow the development of safer and more effective insulin sensitizers suitable for clinical use.


Subject(s)
Insulins , PPAR gamma , Co-Repressor Proteins/metabolism , Drug Inverse Agonism , Ligands , PPAR gamma/metabolism , Protein Conformation
6.
Aging (Albany NY) ; 15(1): 37-52, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36626253

ABSTRACT

Choroidal neovascularization (CNV) causes acute vision loss in neovascular age-related macular degeneration (AMD). Genetic variations of the nuclear receptor RAR-related orphan receptor alpha (RORα) have been linked with neovascular AMD, yet its specific role in pathological CNV development is not entirely clear. In this study, we showed that Rora was highly expressed in the mouse choroid compared with the retina, and genetic loss of RORα in Staggerer mice (Rorasg/sg) led to increased expression levels of Vegfr2 and Tnfa in the choroid and retinal pigment epithelium (RPE) complex. In a mouse model of laser-induced CNV, RORα expression was highly increased in the choroidal/RPE complex post-laser, and loss of RORα in Rorasg/sg eyes significantly worsened CNV with increased lesion size and vascular leakage, associated with increased levels of VEGFR2 and TNFα proteins. Pharmacological inhibition of RORα also worsened CNV. In addition, both genetic deficiency and inhibition of RORα substantially increased vascular growth in isolated mouse choroidal explants ex vivo. RORα inhibition also promoted angiogenic function of human choroidal endothelial cell culture. Together, our results suggest that RORα negatively regulates pathological CNV development in part by modulating angiogenic response of the choroidal endothelium and inflammatory environment in the choroid/RPE complex.


Subject(s)
Choroidal Neovascularization , Wet Macular Degeneration , Mice , Humans , Animals , Angiogenesis Inhibitors , Vascular Endothelial Growth Factor A/metabolism , Visual Acuity , Wet Macular Degeneration/complications , Choroidal Neovascularization/genetics , Choroidal Neovascularization/drug therapy , Lasers , Disease Models, Animal , Mice, Inbred C57BL
7.
SLAS Discov ; 27(4): 242-248, 2022 06.
Article in English | MEDLINE | ID: mdl-35331960

ABSTRACT

NR2F6 is considered an orphan nuclear receptor since its endogenous ligand has yet to be identified. Recently, NR2F6 has emerged as a novel cancer therapeutic target. NR2F6 has been demonstrated to be upregulated or overexpressed in several cancers. Importantly, Nr2f6-/- mice spontaneously reject tumors and develop host-protective immunological memory, a consequence of NR2F6 acting as an immune checkpoint in effector T cells. Collectively, these data suggest that modulation of NR2F6 activity may have important clinical applications in the fight against cancer. The nuclear receptor superfamily of ligand-regulated transcription factors has proven to be an excellent source of targets for therapeutic intervention of a broad range of diseases. Approximately 15% of FDA approved drugs target NRs, demonstrating their clinical efficacy. To identify small molecule regulators of NR2F6 activity, with the overall goal of immuno-oncology, we developed and initiated a high-throughput cell-based assay that specifically measures the transcriptional activity of NR2F6. We completed automated screening of approximately 666,000 compounds and identified 5,008 initial hits. Further screening efforts, including counterscreening assays, confirmed 128 of these hits, most of which had IC50s of equal to or less than 5µM potencies. Here, we report, for the first time, the identification of several small molecule compounds to the orphan nuclear receptor, NR2F6.


Subject(s)
Neoplasms , Orphan Nuclear Receptors , Repressor Proteins , Animals , High-Throughput Screening Assays , Ligands , Mice , Neoplasms/pathology
8.
Redox Biol ; 51: 102261, 2022 05.
Article in English | MEDLINE | ID: mdl-35176707

ABSTRACT

Retinal pigment epithelium (RPE) dysfunction and atrophy occur in dry age-related macular degeneration (AMD), often leading to photoreceptor degeneration and vision loss. Accumulated oxidative stress during aging contributes to RPE dysfunction and degeneration. Here we show that the nuclear receptor REV-ERBα, a redox sensitive transcription factor, protects RPE from age-related degeneration and oxidative stress-induced damage. Genetic deficiency of REV-ERBα leads to accumulated oxidative stress, dysfunction and degeneration of RPE, and AMD-like ocular pathologies in aging mice. Loss of REV-ERBα exacerbates chemical-induced RPE damage, and pharmacological activation of REV-ERBα protects RPE from oxidative damage both in vivo and in vitro. REV-ERBα directly regulates transcription of nuclear factor erythroid 2-related factor 2 (NRF2) and its downstream antioxidant enzymes superoxide dismutase 1 (SOD1) and catalase to counter oxidative damage. Moreover, aged mice with RPE specific knockout of REV-ERBα also exhibit accumulated oxidative stress and fundus and RPE pathologies. Together, our results suggest that REV-ERBα is a novel intrinsic protector of the RPE against age-dependent oxidative stress and a new molecular target for developing potential therapies to treat age-related retinal degeneration.


Subject(s)
Macular Degeneration , Retinal Degeneration , Animals , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1 , Oxidative Stress/physiology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Pigment Epithelium/metabolism
9.
Bioorg Chem ; 119: 105540, 2022 02.
Article in English | MEDLINE | ID: mdl-34902646

ABSTRACT

Liver X Receptors (LXRs) are members of the nuclear receptor family, and they play significant role in lipid and cholesterol metabolism. Moreover, they are key regulators of several inflammatory pathways. Pharmacological modulation of LXRs holds great potential in treatment of metabolic diseases, neurodegenerative diseases, and cancer. We were the first group to identify LXR inverse agonists SR9238 (6) and SR9243 (7) and demonstrate their potential utility in treating liver diseases and cancer. Here, we present the results of structure-activity relationship (SAR) studies, based around SR9238 (6) and SR9243 (7). This study led to identification of 16, 17, 19, and 38, which were more potent inverse agonists than SR9238 (6) and SR9243 (7) and inhibited expression of the fatty acid synthase gene in DU145 cells. We previously demonstrated that inhibition of FASN is correlated to the anticancer activity of SR9243 (7) and this suggests that new inverse agonists have great potential as anticancer agents. We identified compounds with distinct selectivity toward both LXR isoforms, which can be excellent tools to study the pharmacology of both isoforms. We employed molecular dynamic (MD) simulations to better understand the molecular mechanism underlying inverse agonist activity and to guide our future design.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Liver X Receptors/agonists , Sulfonamides/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemistry
10.
Aging Cell ; 20(12): e13486, 2021 12.
Article in English | MEDLINE | ID: mdl-34734460

ABSTRACT

Constitutive NF-κB activation is associated with cellular senescence and stem cell dysfunction and rare variants in NF-κB family members are enriched in centenarians. We recently identified a novel small molecule (SR12343) that inhibits IKK/NF-κB activation by disrupting the association between IKKß and NEMO. Here we investigated the therapeutic effects of SR12343 on senescence and aging in three different mouse models. SR12343 reduced senescence-associated beta-galactosidase (SA-ß-gal) activity in oxidative stress-induced senescent mouse embryonic fibroblasts as well as in etoposide-induced senescent human IMR90 cells. Chronic administration of SR12343 to the Ercc1-/∆ and Zmpste24-/- mouse models of accelerated aging reduced markers of cellular senescence and SASP and improved multiple parameters of aging. SR12343 also reduced markers of senescence and increased muscle fiber size in 2-year-old WT mice. Taken together, these results demonstrate that IKK/NF-κB signaling pathway represents a promising target for reducing markers of cellular senescence, extending healthspan and treating age-related diseases.


Subject(s)
Cellular Senescence/genetics , Gene Expression Regulation/genetics , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Aging , Animals , Disease Models, Animal , Humans , Mice
11.
ACS Chem Biol ; 16(11): 2164-2173, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34558887

ABSTRACT

Myosin IIs, actin-based motors that utilize the chemical energy of adenosine 5'-triphosphate (ATP) to generate force, have potential as therapeutic targets. Their heavy chains differentiate the family into muscle (skeletal [SkMII], cardiac, smooth) and nonmuscle myosin IIs. Despite the therapeutic potential for muscle disorders, SkMII-specific inhibitors have not been reported and characterized. Here, we present the discovery, synthesis, and characterization of "skeletostatins," novel derivatives of the pan-myosin II inhibitor blebbistatin, with selectivity 40- to 170-fold for SkMII over all other myosin II family members. In addition, the skeletostatins bear improved potency, solubility, and photostability, without cytotoxicity. Based on its optimal in vitro profile, MT-134's in vivo tolerability, efficacy, and pharmacokinetics were determined. MT-134 was well-tolerated in mice, impaired motor performance, and had excellent exposure in muscles. Skeletostatins are useful probes for basic research and a strong starting point for drug development.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/chemistry , Myosin Type II/antagonists & inhibitors , Animals , Mice , Molecular Structure , Muscle, Skeletal/metabolism , Myosin Type II/metabolism , Myosin Type II/toxicity
12.
J Mol Biol ; 433(22): 167258, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34547329

ABSTRACT

The retinoic acid receptor-related orphan receptor γ (RORγ) is a ligand-dependent transcription factor of the nuclear receptor super family that underpins metabolic activity, immune function, and cancer progression. Despite being a valuable drug target in health and disease, our understanding of the ligand-dependent activities of RORγ is far from complete. Like most nuclear receptors, RORγ must recruit coregulatory protein to enact the RORγ target gene program. To date, a majority of structural studies have been focused exclusively on the RORγ ligand-binding domain and the ligand-dependent recruitment of small peptide segments of coregulators. Herein, we examine the ligand-dependent assembly of full length RORγ:coregulator complexes on cognate DNA response elements using structural proteomics and small angle x-ray scattering. The results from our studies suggest that RORγ becomes elongated upon DNA recognition, preventing long range interdomain crosstalk. We also determined that the DNA binding domain adopts a sequence-specific conformation, and that coregulatory protein may be able to 'sense' the ligand- and DNA-bound status of RORγ. We propose a model where ligand-dependent coregulator recruitment may be influenced by the sequence of the DNA to which RORγ is bound. Overall, the efforts described herein will illuminate important aspects of full length RORγ and monomeric orphan nuclear receptor target gene regulation through DNA-dependent conformational changes.


Subject(s)
Nuclear Receptor Coactivator 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Response Elements , Animals , Binding Sites , DNA/metabolism , Female , Gene Expression Regulation , Humans , Mass Spectrometry/methods , Mice, Inbred BALB C , Nuclear Receptor Coactivator 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Protein Conformation , Scattering, Small Angle , X-Ray Diffraction
14.
Sci Adv ; 7(5)2021 01.
Article in English | MEDLINE | ID: mdl-33571132

ABSTRACT

Neurotensin receptor 1 (NTSR1) and related G protein-coupled receptors of the ghrelin family are clinically unexploited, and several mechanistic aspects of their activation and inactivation have remained unclear. Enabled by a new crystallization design, we present five new structures: apo-state NTSR1 as well as complexes with nonpeptide inverse agonists SR48692 and SR142948A, partial agonist RTI-3a, and the novel full agonist SRI-9829, providing structural rationales on how ligands modulate NTSR1. The inverse agonists favor a large extracellular opening of helices VI and VII, undescribed so far for NTSR1, causing a constriction of the intracellular portion. In contrast, the full and partial agonists induce a binding site contraction, and their efficacy correlates with the ability to mimic the binding mode of the endogenous agonist neurotensin. Providing evidence of helical and side-chain rearrangements modulating receptor activation, our structural and functional data expand the mechanistic understanding of NTSR1 and potentially other peptidergic receptors.

15.
Nat Commun ; 12(1): 76, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397953

ABSTRACT

Full development of IL-17 producing CD4+ T helper cells (TH17 cells) requires the transcriptional activity of both orphan nuclear receptors RORα and RORγt. However, RORα is considered functionally redundant to RORγt; therefore, the function and therapeutic value of RORα in TH17 cells is unclear. Here, using mouse models of autoimmune and chronic inflammation, we show that expression of RORα is required for TH17 cell pathogenicity. T-cell-specific deletion of RORα reduces the development of experimental autoimmune encephalomyelitis (EAE) and colitis. Reduced inflammation is associated with decreased TH17 cell development, lower expression of tissue-homing chemokine receptors and integrins, and increased frequencies of Foxp3+ T regulatory cells. Importantly, inhibition of RORα with a selective small molecule antagonist mostly phenocopies our genetic data, showing potent suppression of the in vivo development of both chronic/progressive and relapsing/remitting EAE, but with no effect on overall thymic cellularity. Furthermore, use of the RORα antagonist effectively inhibits human TH17 cell differentiation and memory cytokine secretion. Together, these data suggest that RORα functions independent of RORγt in programming TH17 pathogenicity and identifies RORα as a safer and more selective therapeutic target for the treatment of TH17-mediated autoimmunity.


Subject(s)
Inflammation/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Th17 Cells/immunology , Animals , Autoimmunity/drug effects , Body Weight/drug effects , Chronic Disease , Colon/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , HEK293 Cells , Humans , Inflammation/genetics , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Organ Size/drug effects , Severity of Illness Index , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thiophenes/chemistry , Thiophenes/pharmacology
16.
Nat Chem Biol ; 17(3): 307-316, 2021 03.
Article in English | MEDLINE | ID: mdl-33510451

ABSTRACT

Glucocorticoids display remarkable anti-inflammatory activity, but their use is limited by on-target adverse effects including insulin resistance and skeletal muscle atrophy. We used a chemical systems biology approach, ligand class analysis, to examine ligands designed to modulate glucocorticoid receptor activity through distinct structural mechanisms. These ligands displayed diverse activity profiles, providing the variance required to identify target genes and coregulator interactions that were highly predictive of their effects on myocyte glucose disposal and protein balance. Their anti-inflammatory effects were linked to glucose disposal but not muscle atrophy. This approach also predicted selective modulation in vivo, identifying compounds that were muscle-sparing or anabolic for protein balance and mitochondrial potential. Ligand class analysis defined the mechanistic links between the ligand-receptor interface and ligand-driven physiological outcomes, a general approach that can be applied to any ligand-regulated allosteric signaling system.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glucose Transporter Type 4/genetics , Muscular Atrophy/drug therapy , Receptors, Glucocorticoid/chemistry , Signal Transduction/drug effects , A549 Cells , Allosteric Regulation , Animals , Anti-Inflammatory Agents/chemical synthesis , Cell Line, Transformed , Gene Expression Regulation , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , Lipopolysaccharides/administration & dosage , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Rats , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Structure-Activity Relationship
17.
J Med Chem ; 64(2): 980-990, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33434430

ABSTRACT

Despite a myriad of available pharmacotherapies for the treatment of type 2 diabetes (T2D), challenges still exist in achieving glycemic control. Several novel glucose-lowering strategies are currently under clinical investigation, highlighting the need for more robust treatments. Previously, we have shown that suppressing peroxisome proliferator-activated receptor gamma coactivator 1-alpha activity with a small molecule (SR18292, 16) can reduce glucose release from hepatocytes and ameliorate hyperglycemia in diabetic mouse models. Despite structural similarities in 16 to known ß-blockers, detailed structure-activity relationship studies described herein have led to the identification of analogues lacking ß-adrenergic activity that still maintain the ability to suppress glucagon-induced glucose release from hepatocytes and ameliorate hyperglycemia in diabetic mouse models. Hence, these compounds exert their biological effects in a mechanism that does not include adrenergic signaling. These probe molecules may lead to a new therapeutic approach to treat T2D either as a single agent or in combination therapy.


Subject(s)
Glucagon/antagonists & inhibitors , Gluconeogenesis/drug effects , Hypoglycemic Agents/pharmacology , Indoles/pharmacology , Propanols/pharmacology , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Hepatocytes/drug effects , Hepatocytes/metabolism , Hypoglycemic Agents/chemistry , Indoles/chemistry , Lipolysis/drug effects , Liver Glycogen/metabolism , Male , Mice , Mice, Inbred C57BL , PPAR gamma/drug effects , Propanols/chemistry , Receptors, Adrenergic, beta/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Sympathetic Nervous System/drug effects
18.
J Med Chem ; 63(24): 15639-15654, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33289551

ABSTRACT

Nurr1/NR4A2 is an orphan nuclear receptor transcription factor implicated as a drug target for neurological disorders including Alzheimer's and Parkinson's diseases. Previous studies identified small-molecule NR4A nuclear receptor modulators, but it remains unclear if these ligands affect transcription via direct binding to Nurr1. We assessed 12 ligands reported to affect NR4A activity for Nurr1-dependent and Nurr1-independent transcriptional effects and the ability to bind the Nurr1 ligand-binding domain (LBD). Protein NMR structural footprinting data show that amodiaquine, chloroquine, and cytosporone B bind the Nurr1 LBD; ligands that do not bind include C-DIM12, celastrol, camptothecin, IP7e, isoalantolactone, ethyl 2-[2,3,4-trimethoxy-6-(1-octanoyl)phenyl]acetate (TMPA), and three high-throughput screening hit derivatives. Importantly, ligands that modulate Nurr1 transcription also show Nurr1-independent effects on transcription in a cell type-specific manner, indicating that care should be taken when interpreting the functional response of these ligands in transcriptional assays. These findings should help focus medicinal chemistry efforts that desire to optimize Nurr1-binding ligands.


Subject(s)
Ligands , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Amodiaquine/chemistry , Amodiaquine/metabolism , Amodiaquine/pharmacology , Animals , Cell Line , Chloroquine/chemistry , Chloroquine/metabolism , Chloroquine/pharmacology , Humans , Nuclear Magnetic Resonance, Biomolecular , Nuclear Receptor Subfamily 4, Group A, Member 2/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Phenylacetates/chemistry , Phenylacetates/metabolism , Phenylacetates/pharmacology , Protein Binding , Rats , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Transcription, Genetic/drug effects
19.
J Biol Methods ; 7(3): e136, 2020.
Article in English | MEDLINE | ID: mdl-33204739

ABSTRACT

Cytokinesis is the last step of mitotic cell division that separates the cytoplasm of dividing cells. Small molecule inhibitors targeting either the elements of the regulatory pathways controlling cytokinesis, or the terminal effectors have been of interest as potential drug candidates for the treatment of various diseases. Here we present a detailed protocol for a cell-based cytokinesis assay that can be used for the discovery of novel cytokinesis inhibitors. The assay is performed in a 96-well plate format in 48 h. Living cells, nuclei and nuclei of dead cells are identified by a single staining step using three fluorescent dyes, followed by rapid live cell imaging. The primary signal is the nuclei-to-cell ratio (NCR). In the presence of cytokinesis inhibitors, this ratio increases over time, as the ratio of multinucleated cells increases in the population. The ratio of dead nuclei to total nuclei provides a simultaneous measure of cytotoxicity. A screening window coefficient (Z`) of 0.65 indicates that the assay is suitable for screening purposes, as the positive and negative controls are well-separated. EC50 values can be reliably determined in a single 96-well plate by using only six different compound concentrations, enabling the testing of 4 compounds per plate. An excellent test-retest reliability (R 2 = 0.998) was found for EC50 values covering a ~1500-fold range of potencies. Established small molecule inhibitors of cytokinesis operating via direct action on actin dynamics or nonmuscle myosin II are used to demonstrate the robustness, simplicity and flexibility of the assay.

20.
Biochem Biophys Res Commun ; 527(4): 1000-1007, 2020 07 05.
Article in English | MEDLINE | ID: mdl-32439175

ABSTRACT

The nuclear receptors REV-ERBα and REV-ERBß have been demonstrated to play key roles in the regulation of numerous physiological functions, such as metabolism and the circadian rhythm. Recent studies have established the REV-ERBs' roles in immunity, including macrophage and T cell responses. In contrast, their roles in dendritic cells have not been well defined. Dendritic cells are potent antigen presenting cells, connecting microbial sensing and innate immunity to adaptive immune responses. We demonstrate that both REV-ERBα and REV-ERBß expression is upregulated during the course of bone marrow derived dendritic cell (BMDC) differentiation. BMDCs from REV-ERBα and REV-ERBß deficient mice showed enhanced expression of maturation markers like CD86, MHCII, and proinflammatory cytokines. Conversely, treatment of BMDCs with a REV-ERB-specific agonist, SR9009, inhibited the expression of maturation markers and proinflammatory cytokines. Our study suggests the REV-ERBs act as negative regulators of dendritic cell development and activation. These results indicate that pharmacological modulation of REV-ERB activity could be an attractive strategy to modulate DC activation status and for DC-based therapies.


Subject(s)
Bone Marrow Cells/cytology , Dendritic Cells/cytology , Gene Deletion , Gene Expression Regulation, Developmental , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Male , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Pyrrolidines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Repressor Proteins/agonists , Thiophenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...