Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Med Chem ; 204: 112608, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32717483

ABSTRACT

Nur77 is a potential target for the treatment of cancer such as HCC. Herein, we detailed the discovery of a novel series of 5-((8-methoxy-2-methylquinolin-4-yl)amino)-1H-indole-2-carbohydrazide derivatives as potential Nur77 modulators. The studies of antiproliferative activity and Nur77-binding affinity of target compounds resulted in the discovery of a lead candidate (10g), which was a good Nur77 binder (KD = 3.58 ± 0.16 µM) with a broad-spectrum antiproliferative activity against all tested hepatoma cells (IC50 < 2.0 µM) and was low toxic to normal LO2 cells. 10g could up-regulate Nur77 expression and mediate sub-cellular localization of Nur77 to induce apoptosis in hepatocellular carcinoma cell lines, which relied on 10g inducing Nur77-dependent autophagy and endoplasmic reticulum stress as the upstream of apoptosis. Moreover, the in vivo assays verified that 10g significantly inhibited xenograft tumor growth. These results indicate that 10g has the potential to be developed as a novel Nur77-targeting anti-hepatoma drug.


Subject(s)
Drug Design , Hydrazines/chemistry , Hydrazines/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Humans , Hydrazines/chemical synthesis , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Mice , Molecular Docking Simulation , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
2.
Am J Respir Crit Care Med ; 201(8): 934-945, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31834999

ABSTRACT

Rationale: The role of FSTL-1 (follistatin-like 1) in lung homeostasis is unknown.Objectives: We aimed to define the impact of FSTL-1 attenuation on lung structure and function and to identify FSTL-1-regulated transcriptional pathways in the lung. Further, we aimed to analyze the association of FSTL-1 SNPs with lung disease.Methods: FSTL-1 hypomorphic (FSTL-1 Hypo) mice underwent lung morphometry, pulmonary function testing, and micro-computed tomography. Fstl1 expression was determined in wild-type lung cell populations from three independent research groups. RNA sequencing of wild-type and FSTL-1 Hypo mice identified FSTL-1-regulated gene expression, followed by validation and mechanistic in vitro examination. FSTL1 SNP analysis was performed in the COPDGene (Genetic Epidemiology of Chronic Obstructive Pulmonary Disease) cohort.Measurements and Main Results: FSTL-1 Hypo mice developed spontaneous emphysema, independent of smoke exposure. Fstl1 is highly expressed in the lung by mesenchymal and endothelial cells but not immune cells. RNA sequencing of whole lung identified 33 FSTL-1-regulated genes, including Nr4a1, an orphan nuclear hormone receptor that negatively regulates NF-κB (nuclear factor-κB) signaling. In vitro, recombinant FSTL-1 treatment of macrophages attenuated NF-κB p65 phosphorylation in an Nr4a1-dependent manner. Within the COPDGene cohort, several SNPs in the FSTL1 region corresponded to chronic obstructive pulmonary disease and lung function.Conclusions: This work identifies a novel role for FSTL-1 protecting against emphysema development independent of smoke exposure. This FSTL-1-deficient emphysema implicates regulation of immune tolerance in lung macrophages through Nr4a1. Further study of the mechanisms involving FSTL-1 in lung homeostasis, immune regulation, and NF-κB signaling may provide additional insight into the pathophysiology of emphysema and inflammatory lung diseases.


Subject(s)
Follistatin-Related Proteins/genetics , Lung/diagnostic imaging , Pulmonary Emphysema/genetics , Smoke/adverse effects , Animals , Endothelial Cells/metabolism , Follistatin-Related Proteins/pharmacology , Gene Expression Regulation , Gene Knockdown Techniques , Humans , In Vitro Techniques , Lung/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mutation , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phosphorylation/drug effects , Polymorphism, Single Nucleotide , Positron Emission Tomography Computed Tomography , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Emphysema/diagnostic imaging , Pulmonary Emphysema/metabolism , Single Photon Emission Computed Tomography Computed Tomography , Nicotiana , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism , X-Ray Microtomography
3.
Nat Commun ; 8(1): 1903, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29199269

ABSTRACT

Dendrogenin A (DDA) is a newly discovered cholesterol metabolite with tumor suppressor properties. Here, we explored its efficacy and mechanism of cell death in melanoma and acute myeloid leukemia (AML). We found that DDA induced lethal autophagy in vitro and in vivo, including primary AML patient samples, independently of melanoma Braf status or AML molecular and cytogenetic classifications. DDA is a partial agonist on liver-X-receptor (LXR) increasing Nur77, Nor1, and LC3 expression leading to autolysosome formation. Moreover, DDA inhibited the cholesterol biosynthesizing enzyme 3ß-hydroxysterol-Δ8,7-isomerase (D8D7I) leading to sterol accumulation and cooperating in autophagy induction. This mechanism of death was not observed with other LXR ligands or D8D7I inhibitors establishing DDA selectivity. The potent anti-tumor activity of DDA, its original mechanism of action and its low toxicity support its clinical evaluation. More generally, this study reveals that DDA can direct control a nuclear receptor to trigger lethal autophagy in cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cholestanols/pharmacology , Imidazoles/pharmacology , Leukemia, Myeloid, Acute , Liver X Receptors/drug effects , Melanoma , Animals , Cell Death/drug effects , Cell Line, Tumor , Drug Partial Agonism , Gene Expression/drug effects , HEK293 Cells , HL-60 Cells , Humans , In Vitro Techniques , Liver X Receptors/metabolism , Melanoma, Experimental , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/genetics , Mice , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
4.
Ann Otol Rhinol Laryngol ; 126(8): 581-588, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28635301

ABSTRACT

OBJECTIVES: Interactions between mesenchymal stem cells (MSCs) and native vocal fold fibroblasts (VFFs) have not been described in spite of promising preliminary data regarding the effects of MSCs on vocal fold repair in vivo. The current study employed a conditioned media (CM) model to investigate the paracrine effects of bone marrow-derived mesenchymal stem cells (BMSCs) on VFFs. METHODS: Human VFFs were treated with transforming growth factor-ß1 (TGF-ß1; 10 ng/mL), CM from human BMSCs following 48 hours of TGF-ß1 stimulation, or CM+TGF-ß1. Proliferation, immunocytochemistry for alpha smooth muscle actin (αSMA), migration, and collagen gel contraction were quantified as well as transcription of components of the TGF-ß signaling pathway. RESULTS: Transforming growth factor-ß1 accelerated proliferation and induced αSMA in VFFs; these effects were suppressed with CM ( P = .009, P < .001, respectively). The CM+TGF-ß1 condition increased cell migration ( P = .02) and decreased gel contraction; CM+TGF-ß1 also inhibited TGF-ß signaling via significant upregulation of NR4A1 as well as downregulation of S MAD3 and TGF-ß1 relative to TGF-ß1 stimulation in the absence of CM ( P = .002, P < .001, and P = .005, respectively). CONCLUSIONS: Conditioned media affected many profibrotic cell activities in TGF-ß1-stimulated VFFs, likely related to altered TGF-ß signaling. These data provide preliminary insight regarding the antifibrotic effects of MSCs and further support their progression to clinical utility.


Subject(s)
Actins/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Fibroblasts/drug effects , Mesenchymal Stem Cells/physiology , Transforming Growth Factor beta1/pharmacology , Vocal Cords/cytology , Actins/metabolism , Cell Line , Collagen/drug effects , Collagen/metabolism , Culture Media, Conditioned , Down-Regulation , Fibroblasts/physiology , Gels , Gene Expression/drug effects , Humans , Immunohistochemistry , In Vitro Techniques , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Paracrine Communication , Signal Transduction , Smad3 Protein/drug effects , Smad3 Protein/metabolism , Up-Regulation
5.
Inflammation ; 40(3): 806-817, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28213866

ABSTRACT

The aim of this study is to investigate the effect of 5-aminosalicylic acid (5-ASA) on monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) in rats and its molecular mechanism. Sixty male Sprague-Dawley rats (250-300 g) were evenly randomized into six groups: control group; PAH group induced by MCT intraperitoneal injection (50 mg/kg) on day 1; and four PAH groups treated for 30 days from day 2 with 5-ASA at 50 (5-ASA-50 group), 100 (5-ASA-100 group), 150 (5-ASA-150 group), and 200 mg/kg/day (5-ASA-200 group), respectively. Body mass, weight increment, survival rates, pulmonary artery pressure (PAP), right ventricular hypertrophy index (RVHI), and the signal pathway regulated by 5-ASA were assessed. (1) Compared with the control group, the PAH group had lower body mass and weight increment, and relative to the latter, 5-ASA-treated groups had larger body mass and weight increment except for groups 5-ASA-150 and 5-ASA-200 and greater overall survival rates; (2) SPAP, DPAP, MPAP, and RVHI in 5-ASA-treated groups, except for MPAP and RVHI in 5-ASA-200 group, were lower than those in the PAH group; (3) compared with the PAH group, Nur77 expression in the pulmonary arteries of 5-ASA-treated groups was increased; and (4) expression of inflammatory mediators (NF-κB p65) was lower, while that of IκBα was higher in the pulmonary arteries of 5-ASA-treated groups and control group than that in the PAH group (all P < 0.05). 5-ASA attenuates PAH in MCT-injected rats, reducing pulmonary arterial pressures and right ventricular hypertrophy and improving survival rates, via the Nur77-NF-κB/IκBα pathway involved in modulating the pulmonary vascular remodeling.


Subject(s)
Hypertension, Pulmonary/drug therapy , Mesalamine/pharmacology , Monocrotaline/toxicity , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Hypertension, Pulmonary/chemically induced , Hypertrophy, Right Ventricular/drug therapy , Male , NF-kappa B/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Survival Rate
6.
Neuropsychopharmacology ; 42(6): 1243-1253, 2017 May.
Article in English | MEDLINE | ID: mdl-27834392

ABSTRACT

Nr4a nuclear receptors contribute to long-term memory formation and are required for long-term memory enhancement by a class of broad-acting drugs known as histone deacetylase (HDAC) inhibitors. Understanding the molecular mechanisms that regulate these genes and identifying ways to increase their activity may provide novel therapeutic approaches for ameliorating cognitive dysfunction. In the present study, we find that Nr4a gene expression after learning requires the cAMP-response element binding (CREB) interaction domain of the histone acetyltransferase CREB-binding protein (CBP). These gene expression deficits emerge at a time after learning marked by promoter histone acetylation in wild-type mice. Further, mutation of the CREB-CBP interaction domain reduces Nr4a promoter acetylation after learning. As memory enhancement by HDAC inhibitors requires CREB-CBP interaction and Nr4a gene function, these data support the notion that the balance of histone acetylation at the Nr4a promoters is critical for memory formation. NR4A ligands have recently been described, but the effect of these drugs on synaptic plasticity or memory has not been investigated. We find that the 'C-DIM' NR4A ligands, para-phenyl substituted di-indolylmethane compounds, enhance long-term contextual fear memory and increase the duration of long-term potentiation (LTP), a form of hippocampal synaptic plasticity. LTP enhancement by these drugs is eliminated in mice expressing a dominant negative form of NR4A and attenuated in mice with mutation of the CREB-CBP interaction domain. These data define the molecular connection between histone acetylation and Nr4a gene expression after learning. In addition, they suggest that NR4A-activating C-DIM compounds may serve as a potent and selective means to enhance memory and synaptic plasticity.


Subject(s)
CREB-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/drug effects , Indoles/pharmacology , Long-Term Potentiation/drug effects , Memory, Long-Term/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Acetylation/drug effects , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
Nat Chem Biol ; 11(5): 339-46, 2015 May.
Article in English | MEDLINE | ID: mdl-25822914

ABSTRACT

Sepsis, a hyperinflammatory response that can result in multiple organ dysfunctions, is a leading cause of mortality from infection. Here, we show that orphan nuclear receptor Nur77 (also known as TR3) can enhance resistance to lipopolysaccharide (LPS)-induced sepsis in mice by inhibiting NF-κB activity and suppressing aberrant cytokine production. Nur77 directly associates with p65 to block its binding to the κB element. However, this function of Nur77 is countered by the LPS-activated p38α phosphorylation of Nur77. Dampening the interaction between Nur77 and p38α would favor Nur77 suppression of the hyperinflammatory response. A compound, n-pentyl 2-[3,5-dihydroxy-2-(1-nonanoyl) phenyl]acetate, screened from a Nur77-biased library, blocked the Nur77-p38α interaction by targeting the ligand-binding domain of Nur77 and restored the suppression of the hyperinflammatory response through Nur77 inhibition of NF-κB. This study associates the nuclear receptor with immune homeostasis and implicates a new therapeutic strategy to treat hyperinflammatory responses by targeting a p38α substrate to modulate p38α-regulated functions.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/prevention & control , Lipopolysaccharides/toxicity , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Phenylacetates/pharmacology , p38 Mitogen-Activated Protein Kinases/drug effects , Animals , Diabetes Mellitus, Type 2/complications , Drug Evaluation, Preclinical , Homeostasis/drug effects , Inflammation/chemically induced , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Conformation , Sepsis/drug therapy , Sepsis/genetics , Transcription Factor RelA/antagonists & inhibitors
8.
Diabetes ; 63(6): 1870-80, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24647738

ABSTRACT

B-cell translocation gene 2 (BTG2) is a member of an emerging gene family that is involved in cellular functions. In this study, we demonstrate that BTG2 regulates glucose homeostasis via upregulation of Nur77 in diabetic mice. Hepatic BTG2 gene expression was elevated by fasting and forskolin. Overexpression of Btg2 increased the expression of hepatic gluconeogenic genes and blood glucose output and subsequently impaired glucose and insulin tolerance. Upregulation of the transcriptional activity of Nur77, gluconeogenic genes, and glucose production by forskolin was observed by Btg2 transduction, but not in Btg2 knockdown. BTG2-stimulated glucose production and glucose-6-phosphatase promoter activity were attenuated by dominant-negative Nur77. Coimmunoprecipitation and chromatin immunoprecipitation assays showed that BTG2 induced Nur77 occupancy on the glucose-6-phosphatase promoter via a physical interaction. Btg2 gene expression was increased in streptozotocin-treated and db/db mice. Finally, impairment of glucose homeostasis, such as the increase of blood glucose, glucose intolerance, and insulin intolerance, was elevated in diabetic mice, whereas this phenomenon was abolished in knockdown of Btg2. Together, these data suggest that BTG2 participates in the regulation of hepatic glucose homeostasis, which means that BTG2 might serve as a potential therapeutic target for combating metabolic dysfunction.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Immediate-Early Proteins/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Tumor Suppressor Proteins/pharmacology , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/drug therapy , Gene Expression Regulation , Gluconeogenesis , Homeostasis , Immediate-Early Proteins/genetics , Insulin-Secreting Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Promoter Regions, Genetic , Transcriptional Activation/drug effects , Tumor Suppressor Proteins/genetics , Up-Regulation
9.
J Nat Prod ; 76(9): 1771-80, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-24033101

ABSTRACT

Phytochemical investigation of the latex of Antiaris toxicaria resulted in the isolation of 15 new [antiarosides J-X (1-15)] and 17 known cardiac glycosides. The effects of the cardiac glycosides on apoptosis and the expression of orphan nuclear receptor Nur77 were examined in human NIH-H460 lung cancer cells. Several of the cardiac glycosides induced apoptosis in lung cancer cells, which was accompanied by induction of Nur77 protein expression. Treatment of cancer cells with the cardiac glycosides resulted in translocation of the Nur77 protein from the nucleus to the cytoplasm and subsequent targeting to mitochondria. The results show that the cardiac glycosides exert their apoptotic effect through the Nur77-dependent apoptotic pathway.


Subject(s)
Antiaris/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Cardiac Glycosides/isolation & purification , Cardiac Glycosides/pharmacology , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/pharmacology , Latex/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Cardiac Glycosides/chemistry , Drug Screening Assays, Antitumor , Drugs, Chinese Herbal/chemistry , Humans , Mitochondria/metabolism , Nuclear Magnetic Resonance, Biomolecular , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...