Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Life Sci Alliance ; 4(7)2021 07.
Article in English | MEDLINE | ID: mdl-34099540

ABSTRACT

Recent studies suggested that dysregulated YY1 plays a pivotal role in many liver diseases. To obtain a detailed view of genes and pathways regulated by YY1 in the liver, we carried out RNA sequencing in HepG2 cells after YY1 knockdown. A rigid set of 2,081 differentially expressed genes was identified by comparing the YY1-knockdown samples (n = 8) with the control samples (n = 14). YY1 knockdown significantly decreased the expression of several key transcription factors and their coactivators in lipid metabolism. This is illustrated by YY1 regulating PPARA expression through binding to its promoter and enhancer regions. Our study further suggest that down-regulation of the key transcription factors together with YY1 knockdown significantly decreased the cooperation between YY1 and these transcription factors at various regulatory regions, which are important in regulating the expression of genes in hepatic lipid metabolism. This was supported by the finding that the expression of SCD and ELOVL6, encoding key enzymes in lipogenesis, were regulated by the cooperation between YY1 and PPARA/RXRA complex over their promoters.


Subject(s)
Lipid Metabolism/genetics , Liver/metabolism , YY1 Transcription Factor/metabolism , Base Sequence , Fatty Acid Elongases , Hep G2 Cells , Humans , Lipid Metabolism/physiology , PPAR alpha/genetics , Promoter Regions, Genetic/genetics , Retinoid X Receptor alpha , Stearoyl-CoA Desaturase , Transcription Factors/genetics , YY1 Transcription Factor/genetics , YY1 Transcription Factor/physiology
2.
Nutr Res ; 85: 84-98, 2021 01.
Article in English | MEDLINE | ID: mdl-33453499

ABSTRACT

L-carnitine is an indispensable metabolite facilitating the transport of fatty acids into the mitochondrial matrix and has been previously postulated to exert a nutrigenomic effect. However, the underlying molecular mechanisms remain mostly unclear. We hypothesized that L-carnitine interacts with nuclear receptors involved in metabolic regulation, thereby modulating downstream targets of cellular metabolism. Therefore, we investigated the effect of L-carnitine supplementation on protein activity, mRNA expression, and binding affinities of nuclear receptors as well as mRNA expression of downstream targets in skeletal muscle cells, hepatocytes, and differentiated adipocytes. L-carnitine supplementation to hepatocytes increased the protein activity of multiple nuclear receptors (RAR, RXR, VDR, PPAR, HNF4, ER, LXR). Diverging effects on the mRNA expression of PPAR-α, PPAR-δ, PPAR-γ, RAR-ß, LXR-α, and RXR-α were observed in adipocytes, hepatocytes, and skeletal muscle cells. mRNA levels of PPAR-α, a key regulator of lipolysis and ß-oxidation, were significantly upregulated, emphasizing a role of L-carnitine as a promoter of lipid catabolism. L-carnitine administration to hepatocytes modulated the transcription of key nuclear receptor target genes, including ALDH1A1, a promoter of adipogenesis, and OGT, a contributor to insulin resistance. Electrophoretic mobility shift assays proved L-carnitine to increase binding affinities of nuclear receptors to their promoter target sequences, suggesting a molecular mechanism for the observed transcriptional modulation. Overall, these findings indicate that L-carnitine modulates the activity and expression of nuclear receptors, thereby promoting lipolytic gene expression and decreasing transcription of target genes linked to adipogenesis and insulin resistance.


Subject(s)
Adipocytes/metabolism , Carnitine/metabolism , Carnitine/pharmacology , Cell Nucleus/metabolism , Hepatocytes/metabolism , Muscle Fibers, Skeletal/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , 3T3-L1 Cells , Animals , Binding Sites , Carnitine O-Acetyltransferase/genetics , Carnitine O-Acetyltransferase/metabolism , Cells, Cultured , Culture Media , Humans , Liver X Receptors/genetics , Mice , Nutrigenomics , PPAR alpha/genetics , PPAR alpha/metabolism , Promoter Regions, Genetic , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Signal Transduction , Transcription, Genetic
3.
Bioorg Chem ; 107: 104578, 2021 02.
Article in English | MEDLINE | ID: mdl-33383323

ABSTRACT

Hyperelodione D (1), an undescribed polyprenylated phloroglucinol derivative possessing 6/6/5/5 fused tetracyclic core, together with hyperelodiones E-F (2-3), two unreported analogues bearing 6/5/5 fused tricyclic structure, were isolated from Hypericum elodeoides Choisy. Their planar structures were elucidated by spectroscopic analysis (HRESIMS, 1D and 2D NMR) and their absolute configurations were determined by comparison of experimental and calculated ECD data. The cytotoxicity and retinoid X receptor-α (RXRα) related activities of the isolates were evaluated and the plausible biogenetic pathways of 1-3 were proposed.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Hypericum/chemistry , Phloroglucinol/pharmacology , Retinoid X Receptor alpha/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Line , Cell Survival/drug effects , Density Functional Theory , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Phloroglucinol/chemistry , Phloroglucinol/isolation & purification , Retinoid X Receptor alpha/metabolism , Structure-Activity Relationship
4.
J Med Chem ; 63(11): 5841-5855, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32391701

ABSTRACT

We reported recently that berberine (Ber), a traditional oriental medicine to treat gastroenteritis, binds and activates retinoid X receptor α (RXRα) for suppressing the growth of colon cancer cells. Here, we extended our studies based on the binding mode of Ber with RXRα by design, synthesis, and biological evaluation of a focused library of 15 novel Ber analogues. Among them, 3,9-dimethoxy-5,6-dihydroisoquinolino[3,2-a]isoquinolin-7-ium chloride (B-12) was identified as the optimal RXRα activator. More efficiently than Ber, B-12 bound and altered the conformation of RXRα/LBD, thereby suppressing the Wnt/ß-catenin pathway and colon cancer cell growth via RXRα mediation. In addition, B-12 not only preserved Ber's tumor selectivity but also greatly improved its bioavailability. Remarkably, in mice, B-12 did not show obvious side effects including hypertriglyceridemia as other RXRα agonists or induce hepatorenal toxicity. Together, our study describes an approach for the rational design of Ber-derived RXRα activators as novel effective antineoplastic agents for colon cancer.


Subject(s)
Antineoplastic Agents/chemistry , Berberine/analogs & derivatives , Retinoid X Receptor alpha/agonists , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Berberine/metabolism , Binding Sites , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Evaluation, Preclinical , Half-Life , Humans , Male , Mice , Mice, Nude , Molecular Docking Simulation , Protein Structure, Tertiary , Rats , Retinoid X Receptor alpha/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Transplantation, Heterologous
5.
J Ethnopharmacol ; 257: 112859, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32294506

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Danqi Pill (DQP), commonly known as a routinely prescribed traditional Chinese medicine (TCM), is composed of Salviae Miltiorrhizae Radix et Rhizoma and Notoginseng Radix et Rhizoma and effective in treating heart failure (HF) clinically due to their multicompound and multitarget properties. However, the exact active compounds and corresponding targets of DQP are still unknown. AIM OF THE STUDY: This study aimed to investigate active compounds and drug targets of DQP in heart failure based on the PPARs-RXRα pathway. MATERIALS AND METHODS: Network pharmacology was used to predict the compound-target interactions of DQP. Left anterior descending (LAD)-induced HF mouse model and oxygen-glucose deprivation/recovery (OGD/R)-induced H9C2 model were constructed to screen the active compounds of DQP. RESULTS: According to BATMAN-TCM (a bioinformatics analysis tool for molecular mechanism of traditional Chinese medicine we previously developed), 24 compounds in DQP were significantly enriched in the peroxisome proliferator activated receptors-retinoid X receptor α (PPARs-RXRα) pathway. Among them, Ginsenoside Rb3 (G-Rb3) had the best pharmacodynamics against OGD/R-induced loss of cell viability, and it was selected to verify the compound-target interaction. In HF mice, G-Rb3 protected cardiac functions and activated the PPARs-RXRα pathway. In vitro, G-Rb3 protected against OGD/R-induced reactive oxygen species (ROS) production, promoted the expressions of RXRα and sirtuin 3 (SIRT3), thereafter improved the intracellular adenosine triphosphate (ATP) level. Immunofluorescent staining demonstrated that G-Rb3 could activate RXRα, and facilitate RXRα shifting to the nucleus. HX531, the specific inhibitor of RXRα, could abolish the protective effects of G-Rb3 on RXRα translocation. Consistently, the effect was also confirmed on RXRα siRNA cardiomyocytes model. Moreover, surface plasmon resonance (SPR) assays identified that G-Rb3 bound directly to RXRα with the affinity of KD = 10 × 10-5 M. CONCLUSION: By integrating network pharmacology and experimental validation, we identified that as the major active compound of DQP, G-Rb3 could ameliorate ROS-induced energetic metabolism dysfunction, maintain mitochondrial function and facilitate energy metabolism via directly targeting on RXRα. This study provides a promising strategy to dissect the effective patterns for TCM and finally promote the modernization of TCM.


Subject(s)
Cardiovascular Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Ginsenosides/pharmacology , Heart Failure/drug therapy , Myocytes, Cardiac/drug effects , Peroxisome Proliferator-Activated Receptors/metabolism , Retinoid X Receptor alpha/metabolism , Animals , Cell Line , Disease Models, Animal , Gene Regulatory Networks , Heart Failure/metabolism , Heart Failure/pathology , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Peroxisome Proliferator-Activated Receptors/genetics , Protein Interaction Maps , Rats , Retinoid X Receptor alpha/genetics , Signal Transduction , Systems Biology
6.
Cancer Lett ; 473: 107-117, 2020 03 31.
Article in English | MEDLINE | ID: mdl-31874245

ABSTRACT

Radiation therapy is a common treatment for prostate cancer, however recurrence remains a problem. MicroRNA expression is altered in prostate cancer and may promote therapy resistance. Through bioinformatic analyses of TCGA and CPC-GENE patient cohorts, we identified higher miR-191 expression in tumor versus normal tissue, and increased expression in higher Gleason scores. In vitro and in vivo experiments demonstrated that miR-191 overexpression promotes radiation survival, and contributes to a more aggressive phenotype. Retinoid X receptor alpha, RXRA, was discovered to be a novel target of miR-191, and knockdown recapitulated radioresistance. Furthermore, treatment of prostate cancer cells with the RXRA agonist 9-cis-retinoic acid restored radiosensitivity. Supporting this relationship, patients with high miR-191 and low RXRA abundance experienced quicker biochemical recurrence. Reduced RXRA translated to a higher risk of distant failure after radiotherapy. Notably, this miR-191/RXRA interaction was conserved in a novel primary cell line derived from radiorecurrent prostate cancer. Together, our findings demonstrate that miR-191 promotes prostate cancer survival after radiotherapy, and highlights retinoids as a potential option to improve radiotherapy response.


Subject(s)
Biomarkers, Tumor/metabolism , MicroRNAs/metabolism , Neoplasm Recurrence, Local/genetics , Prostatic Neoplasms/therapy , Radiation Tolerance/genetics , Retinoid X Receptor alpha/genetics , Alitretinoin/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Chemoradiotherapy, Adjuvant/methods , Disease-Free Survival , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Kallikreins/blood , Kaplan-Meier Estimate , Male , Mice , MicroRNAs/agonists , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Primary Cell Culture , Prognosis , Prostate/pathology , Prostate/surgery , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Radiation Tolerance/drug effects , Retinoid X Receptor alpha/agonists , Survival Rate , Time Factors , Xenograft Model Antitumor Assays
7.
Phytochemistry ; 170: 112216, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31841782

ABSTRACT

Hyperelodiones A-C, three undescribed monoterpenoid polyprenylated acylphloroglucinols possessing 6/6/6 fused tricyclic core, were isolated from Hypericum elodeoides Choisy. Their gross structures were elucidated by HRESIMS and NMR data. The absolute configurations of hyperelodiones A-C were assigned by their calculated and compared electronic circular dichroism (ECD) spectra combined with their common biosynthetic origin. A fluorescence quenching assay suggested that hyperelodiones A-C could bind to RXRα-LBD, whereas hyperelodione C showed the strongest interaction with a KD of 12.81 µΜ. In addition, hyperelodiones A-C dose-dependently inhibited RXRα transactivation and the growth of HeLa and MCF-7 cells. Among them, hyperelodione C showed the most potent inhibitory activities and dose-dependent PARP cleavage. Molecular docking results suggested that hyperelodione C showed a different interaction mode compared with hyperelodione A and hyperelodione B. Thus, hyperelodione C can be considered as a promising lead compound for cancer therapy, which can bind to RXRα-LBD and induce HeLa and MCF-7 cell apoptosis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Hypericum/chemistry , Monoterpenes/pharmacology , Phytochemicals/pharmacology , Retinoid X Receptor alpha/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HeLa Cells , Humans , MCF-7 Cells , Molecular Conformation , Molecular Docking Simulation , Monoterpenes/chemistry , Monoterpenes/isolation & purification , Phytochemicals/chemistry , Phytochemicals/isolation & purification , Retinoid X Receptor alpha/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Methods Mol Biol ; 2019: 95-108, 2019.
Article in English | MEDLINE | ID: mdl-31359391

ABSTRACT

This chapter outlines the materials, methods, and procedures for the in vitro biological evaluation of retinoid-X-receptor (RXR) agonists including 6-(ethyl(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)amino)nicotinic acid (NEt-TMN), as well as several NEt-TMN analog compounds recently reported by our group. These methods have general applicability beyond this NEt-TMN case study, and can be employed to characterize and biologically evaluate other putative RXR agonists (rexinoids), and benchmarked against perhaps the most common rexinoid known as bexarotene (Bex), a drug awarded FDA approval for the treatment of cutaneous T-cell lymphoma in 1999 but that is also prescribed for non-small cell lung cancer and continues to be explored in multiple human cancer types. The side-effect profile of Bex treatment includes hypothyroidism and hypertriglyceridemia arising from the inhibition or activation of additional nuclear receptors that partner with RXR. Because rexinoids often exhibit selectivity for RXR activation, versus activating the retinoic-acid-receptor (RAR), rexinoid treatment avoids the cutaneous toxicity commonly associated as a side effect with retinoids. There are many examples of other potent rexinoids, where biological evaluation has contributed useful insight into qSAR studies on these compounds, often also benchmarked to Bex, as potential treatments for cancer. Because of differential pleiotropy in other pathways, even closely related rexinoids display unique side-effect and activity profiles. Notable examples of potent rexinoids in addition to Bex and NEt-TMN include CD3254, LGD100268, and 9-cis-UAB30. Indeed, the methods described herein to evaluate NEt-TMN and analogous rexinoids are generally applicable to a wider variety of potent, moderate, and even weak RXR ligands.In terms of in vitro biological evaluation, methods for a rapid and preliminary assessment of rexinoid activity are described by employing a biologically relevant, RXR-responsive element (RXRE)-mediated transcription assay in mammalian cells. In addition, a second, more sensitive assay is also detailed that utilizes activation of RXR-RXR homodimers in the context of a mammalian two-hybrid (M2H) luciferase assay. Methods for applying the M2H assay at different rexinoid concentrations are further described for the determination of EC50 values for rexinoids from dose-response curves.


Subject(s)
Retinoid X Receptor alpha/agonists , Tetrahydronaphthalenes/pharmacology , Coumaric Acids/pharmacology , Drug Evaluation, Preclinical , Gene Expression Regulation , HEK293 Cells , Humans , Retinoids/pharmacology , Signal Transduction
9.
J Nat Prod ; 82(5): 1072-1080, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31038949

ABSTRACT

To find small-molecule regulators of RXRα, a phytochemical study of Hypericum elodeoides was conducted. Fifteen compounds, including the new 1 and 6, were isolated from the whole plant of H. elodeoides. The absolute configuration of 1 was assigned by comparison of experimental and calculated ECD data. Compounds 1 and 6 exhibited concentration-dependent inhibitory effects on RXRα transcription and selectively inhibited the proliferation of HeLa cells. Western blot analysis suggested that 1 and 6 induced apoptosis of HeLa cells with time- and dose-dependent PARP cleavage. A caspase activation assay indicated that these two compounds triggered caspase-8 activation to induce apoptosis by the extrinsic pathway. Molecular docking results suggested that 1 and 6 interacted with the Arg319 moiety of RXRα-LBD. Ligands binding to RXRα have shown promise in the discovery of anticancer drugs. A fluorescence quenching assay indicated the binding of 1 and 6 to the RXRα with the binding constant ( KD) fitted as 68.3 and 14.0 µM, respectively. A preliminary SAR study of the isolates was conducted to enhance the knowledge of the RXRα ligands. Thus, 1 and 6 might act as the small-molecule regulators of RXRα, which target RXRα and mediate HeLa cell apoptosis through the extrinsic pathways.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Caspase 8/metabolism , Hypericum/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Retinoid X Receptor alpha/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Molecular Docking Simulation , Structure-Activity Relationship
10.
Bioorg Chem ; 85: 413-419, 2019 04.
Article in English | MEDLINE | ID: mdl-30665035

ABSTRACT

Retinoid X receptor alpha (RXRα), a central member of the nuclear receptor superfamily and a key regulator of many signal transduction pathways, has been an attractive drug target. We previously discovered that an N-terminally truncated form of RXRα can be induced by specific ligands to form homotetramers, which, as a result of conformational selection, forms the basis for inhibiting the nongenomic activation of RXRα. Here, we report the identification and characterization of atorvastatin as a new RXRα tetramer stabilizer by using structure-based virtual screening and demonstrate that virtual library screening can be used to aid in identifying RXRα ligands that can induce its tetramerization. In this study, docking was applied to screen the FDA-approved small molecule drugs in the DrugBank 4.0 collection. Two compounds were selected and purchased for testing. We showed that the selected atorvastatin could bind to RXRα to promote RXRα-LBD tetramerization. We also showed that atorvastatin possessed RXRα-dependent apoptotic effects. In addition, we used a chemical approach to aid in the studies of the binding mode of atorvastatin.


Subject(s)
Atorvastatin/pharmacology , Protein Multimerization/drug effects , Retinoid X Receptor alpha/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Atorvastatin/chemistry , Atorvastatin/metabolism , Binding Sites , Drug Evaluation, Preclinical , Humans , Ligands , MCF-7 Cells , Protein Binding/drug effects , Protein Domains , Protein Stability/drug effects , Sulindac/analogs & derivatives , Sulindac/metabolism
11.
J Bone Miner Res ; 34(2): 231-240, 2019 02.
Article in English | MEDLINE | ID: mdl-30321476

ABSTRACT

We have previously demonstrated inverse associations between maternal 25(OH)-vitamin D status and perinatal DNA methylation at the retinoid-X-receptor-alpha (RXRA) locus and between RXRA methylation and offspring bone mass. In this study, we used an existing randomized trial to test the hypothesis that maternal gestational vitamin D supplementation would lead to reduced perinatal RXRA locus DNA methylation. The Maternal Vitamin D Osteoporosis Study (MAVIDOS) was a multicenter, double-blind, randomized, placebo-controlled trial of 1000 IU/day cholecalciferol or matched placebo from 14 weeks' gestation until delivery. Umbilical cord (fetal) tissue was collected at birth and frozen at -80°C (n = 453). Pyrosequencing was used to undertake DNA methylation analysis at 10 CpG sites within the RXRA locus (identified previously). T tests were used to assess differences between treatment groups in methylation at the three most representative CpG sites. Overall, methylation levels were significantly lower in the umbilical cord from offspring of cholecalciferol-supplemented mothers, reaching statistical significance at four CpG sites, represented by CpG5: mean difference in % methylation between the supplemented and placebo groups was -1.98% (95% CI, -3.65 to -0.32, p = 0.02). ENCODE (Encyclopedia of DNA Elements) evidence supports the functionality of this locus with strong DNase hypersensitivity and enhancer chromatin within biologically relevant cell types including osteoblasts. Enrichment of the enhancer-related H3K4me1 histone mark is also seen in this region, as are binding sites for a range of transcription factors with roles in cell proliferation, response to stress, and growth factors. Our findings are consistent with previous observational results and provide new evidence that maternal gestational supplementation with cholecalciferol leads to altered perinatal epigenetic marking, informing mechanistic understanding of early life mechanisms related to maternal vitamin D status, epigenetic marks, and bone development. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.


Subject(s)
CpG Islands , DNA Methylation/drug effects , Dietary Supplements , Genetic Loci , Retinoid X Receptor alpha , Vitamin D/analogs & derivatives , Adult , Double-Blind Method , Female , Humans , Infant, Newborn , Male , Pregnancy , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Vitamin D/administration & dosage
12.
Phytochemistry ; 158: 67-76, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30476898

ABSTRACT

Veramyosides A-J, eleven undescribed stigmastane-type steroids, including one aglycone and ten glycosides, along with three known homologues were isolated from the twigs of Vernonia amygdalina Delile (compositae). All compounds featured a stigmastane-type steroid skeleton with a unique conjugated Δ7,9(11) diene segment and highly oxygenated side chains with a γ-lactone or an α, ß-unsaturated five-membered lactone ring. The structures of veramyosides A-J and their absolute configurations were unambiguously elucidated by HR-ESI-MS, extensive NMR spectroscopy, in situ dimolybdenum CD methods, modified Mosher's method, quantum chemical calculation of their ECD curves, and CD comparison methods on basis of their biogenetic pathway. In addition, all isolates were investigated for their effects on RXRα transcription, and their effects on the NF-κB signaling pathway were also evaluated.


Subject(s)
Steroids/chemistry , Steroids/pharmacology , Vernonia/chemistry , Circular Dichroism , Drug Evaluation, Preclinical/methods , Glycosides/chemistry , Glycosides/isolation & purification , Glycosides/pharmacology , HEK293 Cells , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Oxygen/chemistry , Retinoid X Receptor alpha/antagonists & inhibitors , Retinoid X Receptor alpha/genetics , Steroids/isolation & purification , Structure-Activity Relationship
13.
Free Radic Biol Med ; 131: 376-381, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30578920

ABSTRACT

Chronic kidney disease (CKD) is a worldwide public health problem with an estimated prevalence of 8.2%. This study reports glutathione deficiency, excess oxidative stress, and altered vitamin D metabolism in the kidney of mice fed a high-fat diet (HFD). The levels of GCLC and GCLM gene expression were significantly downregulated and the protein carbonylation level, a hallmark of oxidative damage, was significantly increased in the kidney of HFD-fed mice. While the levels of VD-regulatory genes 1-alpha-hydroxylase (CYP27B1), VDR, and RXRα were significantly downregulated in the kidney of mice fed a HFD, those of 24-hydroxylase (CYP24A1) were significantly elevated. In vitro, GSH deficiency per se causes excess oxidative damage (protein carbonylation), and significantly decreases the levels of VD-regulatory genes (CYP27B1, VDR, and RXRα), but increases levels of CYP24A1 in human renal proximal tubule epithelial cells (RPTEC), similar to findings in the kidney of HFD-fed diabetic mice. L-cysteine supplementation restores GSH and prevents oxidative damage in RPTEC. These studies suggest a potential role of GSH precursor in reducing excess oxidative stress and renal injury that commonly accompanies obesity/diabetes.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Diabetes Mellitus, Experimental/enzymology , Glutathione/deficiency , Receptors, Calcitriol/genetics , Renal Insufficiency, Chronic/enzymology , Vitamin D3 24-Hydroxylase/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Animals , Cysteine/pharmacology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat/adverse effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Primary Cell Culture , Protein Carbonylation , Receptors, Calcitriol/metabolism , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Signal Transduction , Vitamin D3 24-Hydroxylase/metabolism
14.
J Cell Biochem ; 119(10): 8623-8635, 2018 11.
Article in English | MEDLINE | ID: mdl-30058740

ABSTRACT

Non-small-cell lung cancer (NSCLC) appears to be a significant threat to public health worldwide. MicroRNAs have been identified as significant regulators for the development of NSCLC. Previous reports have suggested that hsa-mir-485-5p is dysregulated in various cancers. RXRα, as a kind of nuclear receptor, is an effective target of cancer treatment. Cancer stem cells (CSCs) are recognized as the main cause for tumor metastasis, recurrence, and chemotherapy resistance. However, the mechanism by which hsa-mir-485-5p and RXRα modulate CSCs in NSCLC remains unknown. Here, we found that hsa-mir-485-5p was decreased in serum samples from patients with NSCLC and NSCLC cells. Meanwhile, epigallocatechin-3-gallate (EGCG), an effective anticancer compound extracted from green tea, can enhance hsa-mir-485-5p expression. Hsa-mir-485-5p mimics markedly inhibited NSCLC cell growth and induced cell apoptosis. However, inhibition of hsa-mir-485-5p significantly enriched CSC-like traits. Moreover, bioinformatics analysis predicted the binding correlation between hsa-mir-485-5p and RXRα, which was confirmed by a dual-luciferase reporter assay. We observed that RXRα was increased in NSCLC and EGCG could inhibit RXRα levels dose dependently. In addition, RXRα upregulation or activation expanded the CSC-like properties of NSCLC cells, whereas RXRα inhibition or inactivation could exert a reverse phenomenon. Consistently, in vivo experiments also validated that EGCG could repress the CSC-like characteristics by modulating the hsa-mir-485-5p/RXRα axis. Our findings may reveal a novel molecular mechanism for the treatment of NSCLC.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/drug therapy , Catechin/analogs & derivatives , Lung Neoplasms/blood , Lung Neoplasms/drug therapy , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Retinoid X Receptor alpha/metabolism , A549 Cells , Analysis of Variance , Animals , Catechin/therapeutic use , Down-Regulation , Gene Knockdown Techniques , Gene Silencing , HEK293 Cells , Humans , Mice , Mice, Nude , MicroRNAs/chemistry , MicroRNAs/genetics , Molecular Mimicry/genetics , Retinoid X Receptor alpha/genetics , Signal Transduction/drug effects , Transfection , Up-Regulation , Xenograft Model Antitumor Assays
15.
J Anim Physiol Anim Nutr (Berl) ; 102(1): e279-e287, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28503816

ABSTRACT

This study was designed to investigate effects of xanthophylls on serum lipid profile (triglyceride, TG; cholesterol, CHO; high-density lipoprotein cholesterol, HDLC; and low-density lipoprotein cholesterol, LDLC) and nuclear factor (peroxisome proliferator-activated receptor gamma, PPARγ; PPAR gamma coactivator 1 alpha, PGC1α; retinoid X receptor gamma, RXRγ; and retinoic acid receptor alpha, RARα) gene expression of breeding hens and chicks. In experiment 1, 432 hens were divided into three groups and fed diets supplemented with 0 (as control group), 20 or 40 mg/kg xanthophylls. Blood was sampled at 7, 14, 21, 28 and 35 days of trial. Liver, duodenum, jejunum and ileum were sampled at 35 days of trial. Results showed that serum HDLC level of hens was increased after dietary 40 mg/kg xanthophyll addition for 21, 28 and 35 days, while serum TG, CHO and LDLC were not affected. Xanthophyll addition also increased PPARγ expression in jejunum, RXRγ expression in duodenum and jejunum, and RARα expression in liver and duodenum. Experiment 2 was a 2 × 2 factorial design. Male chicks hatched from 0 or 40 mg/kg xanthophyll diet of hens were fed diet containing either 0 or 40 mg/kg xanthophylls. Liver, duodenum, jejunum and ileum were sampled at 0, 7, 14 and 21 days after hatching. Blood samples were also collected at 21 days. Results showed that in ovo xanthophylls elevated PPARγ in duodenum and jejunum, and RXRγ and RARα in liver of chicks mainly within 1 week after hatching, while dietary xanthophylls increased serum HDLC level and PPARγ and RXRγ in liver from 2 weeks onwards. In conclusion, our research suggested xanthophylls can regulate serum lipid profile and nuclear factor expression in hens and chicks.


Subject(s)
Chickens/metabolism , Cholesterol, HDL/blood , PPAR gamma/metabolism , Retinoid X Receptor gamma/metabolism , Xanthophylls/pharmacology , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Chickens/blood , Diet/veterinary , Dietary Supplements , Female , Gene Expression Regulation/drug effects , Male , PPAR gamma/genetics , Retinoid X Receptor alpha , Retinoid X Receptor gamma/genetics
16.
J Mol Graph Model ; 77: 121-129, 2017 10.
Article in English | MEDLINE | ID: mdl-28850894

ABSTRACT

The treatment of many diseases may require drugs that are capable to attack multiple targets simultaneously. Obviously, the virtual screening of multi-target drug candidates is much more time consuming compared to the single-target case. This, in particular, concerns the last step of virtual screening where the binding free energy is computed by conventional molecular dynamics simulation. To overcome this difficulty we propose a simple protocol which is relied on the fast steered molecular dynamics simulation and on available experimental data on binding affinity of reference ligand to a given target. Namely, first we compute non-equilibrium works generated during pulling ligands from the binding site using the steered molecular dynamics method. Then as top leads we choose only those compounds that have the non-equilibrium work larger than that of a reference compound for which the binding free energy has been already known from experiment. Despite many efforts no cures for AD (Alzheimer's disease) have been found. One of possible reasons for this failure is that drug candidates were developed for a single target, while there are exist many possible pathways to AD. Applying our new protocol to five targets including amyloid beta fibril, peroxisome proliferator-activated receptor γ, retinoic X receptor α, ß- and γ-secretases, we have found two potential drugs (CID 16040294 and CID 9998128) for AD from the large PubChem database. We have also shown that these two ligands can interfere with the activity of popular Acetylcholinesterase target through strong binding towards it.


Subject(s)
Alzheimer Disease/drug therapy , Drug Design , Drug Evaluation, Preclinical , User-Computer Interface , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Binding Sites , Drug Delivery Systems , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Retinoid X Receptor alpha/antagonists & inhibitors , Retinoid X Receptor alpha/chemistry
17.
Oncogene ; 36(50): 6906-6918, 2017 12 14.
Article in English | MEDLINE | ID: mdl-28846104

ABSTRACT

Berberine, an isoquinoline alkaloid, is a traditional oriental medicine used to treat diarrhea and gastroenteritis. Recently, we reported that it could inhibit the growth of intestinal polyp in animals and in patients with the familial adenomatous polyposis by downregulating ß-catenin signaling. However, the intracellular target mediating the effects of berberine remains elusive. Here, we provide evidence that berberine inhibits ß-catenin function via directly binding to a unique region comprising residues Gln275, Arg316 and Arg371 in nuclear receptor retinoid X receptor alpha (RXRα), where berberine concomitantly binding to and synergistically activating RXRα with 9-cis-retinoic acid (9-cis-RA), a natural ligand binding to the classical ligand-binding pocket of RXRα. Berberine binding promotes RXRα interaction with nuclear ß-catenin, leading to c-Cbl mediated degradation of ß-catenin, and consequently inhibits the proliferation of colon cancer cells. Furthermore, berberine suppresses the growth of human colon carcinoma xenograft in nude mice in an RXRα-dependent manner. Together, our study not only identifies RXRα as a direct protein target for berberine but also dissects their binding mode and validates that berberine indeed suppresses ß-catenin signaling and cell growth in colon cancer via binding RXRα, which provide new strategies for the design of new RXRα-based antitumor agents and drug combinations.


Subject(s)
Berberine/pharmacology , Colonic Neoplasms/pathology , Retinoid X Receptor alpha/metabolism , Signal Transduction/physiology , beta Catenin/physiology , Animals , Berberine/metabolism , Berberine/therapeutic use , Colonic Neoplasms/drug therapy , Humans , Mice , Mice, Inbred BALB C , Xenograft Model Antitumor Assays
18.
Nat Commun ; 8(1): 103, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740126

ABSTRACT

Muscle-invasive bladder cancer (MIBC) is an aggressive disease with limited therapeutic options. Although immunotherapies are approved for MIBC, the majority of patients fail to respond, suggesting existence of complementary immune evasion mechanisms. Here, we report that the PPARγ/RXRα pathway constitutes a tumor-intrinsic mechanism underlying immune evasion in MIBC. Recurrent mutations in RXRα at serine 427 (S427F/Y), through conformational activation of the PPARγ/RXRα heterodimer, and focal amplification/overexpression of PPARγ converge to modulate PPARγ/RXRα-dependent transcription programs. Immune cell-infiltration is controlled by activated PPARγ/RXRα that inhibits expression/secretion of inflammatory cytokines. Clinical data sets and an in vivo tumor model indicate that PPARγHigh/RXRαS427F/Y impairs CD8+ T-cell infiltration and confers partial resistance to immunotherapies. Knockdown of PPARγ or RXRα and pharmacological inhibition of PPARγ significantly increase cytokine expression suggesting therapeutic approaches to reviving immunosurveillance and sensitivity to immunotherapies. Our study reveals a class of tumor cell-intrinsic "immuno-oncogenes" that modulate the immune microenvironment of cancer.Muscle-invasive bladder cancer (MIBC) is a potentially lethal disease. Here the authors characterize diverse genetic alterations in MIBC that convergently lead to constitutive activation of PPARgamma/RXRalpha and result in immunosurveillance escape by inhibiting CD8+ T-cell recruitment.


Subject(s)
Immune Evasion/immunology , Monitoring, Immunologic , PPAR gamma/immunology , Retinoid X Receptor alpha/immunology , Urinary Bladder Neoplasms/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Gene Expression Profiling/methods , HCT116 Cells , Humans , Immunoblotting , Immunotherapy/methods , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Mice , Microscopy, Fluorescence , Mutation/immunology , Neoplasm Invasiveness , PPAR gamma/chemistry , PPAR gamma/genetics , Protein Multimerization/immunology , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy
19.
Clin Epigenetics ; 9: 16, 2017.
Article in English | MEDLINE | ID: mdl-28191262

ABSTRACT

BACKGROUND: Maternal nutrition during pregnancy and infant nutrition in the early postnatal period (lactation) are critically involved in the development and health of the newborn infant. The Maternal Nutrition and Offspring's Epigenome (MANOE) study was set up to assess the effect of maternal methyl-group donor intake (choline, betaine, folate, methionine) on infant DNA methylation. Maternal intake of dietary methyl-group donors was assessed using a food-frequency questionnaire (FFQ). Before and during pregnancy, we evaluated maternal methyl-group donor intake through diet and supplementation (folic acid) in relation to gene-specific (IGF2 DMR, DNMT1, LEP, RXRA) buccal epithelial cell DNA methylation in 6 months old infants (n = 114) via pyrosequencing. In the early postnatal period, we determined the effect of maternal choline intake during lactation (in mothers who breast-fed for at least 3 months) on gene-specific buccal DNA methylation (n = 65). RESULTS: Maternal dietary and supplemental intake of methyl-group donors (folate, betaine, folic acid), only in the periconception period, was associated with buccal cell DNA methylation in genes related to growth (IGF2 DMR), metabolism (RXRA), and appetite control (LEP). A negative association was found between maternal folate and folic acid intake before pregnancy and infant LEP (slope = -1.233, 95% CI -2.342; -0.125, p = 0.0298) and IGF2 DMR methylation (slope = -0.706, 95% CI -1.242; -0.107, p = 0.0101), respectively. Positive associations were observed for maternal betaine (slope = 0.875, 95% CI 0.118; 1.633, p = 0.0241) and folate (slope = 0.685, 95% CI 0.245; 1.125, p = 0.0027) intake before pregnancy and RXRA methylation. Buccal DNMT1 methylation in the infant was negatively associated with maternal methyl-group donor intake in the first and second trimester of pregnancy and negatively in the third trimester. We found no clear association between maternal choline intake during lactation and buccal infant DNA methylation. CONCLUSIONS: This study suggests that maternal dietary and supplemental intake of methyl-group donors, especially in the periconception period, can influence infant's buccal DNA methylation in genes related to metabolism, growth, appetite regulation, and maintenance of DNA methylation reactions.


Subject(s)
Betaine/administration & dosage , Choline/administration & dosage , DNA Methylation/drug effects , Folic Acid/administration & dosage , Maternal Nutritional Physiological Phenomena , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Dietary Supplements , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Infant , Insulin-Like Growth Factor II/genetics , Leptin/genetics , Nutrition Surveys , Pregnancy , Retinoid X Receptor alpha/genetics , Sequence Analysis, DNA/methods
20.
Bioorg Med Chem Lett ; 27(4): 1055-1061, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28089347

ABSTRACT

Retinoid X receptor alpha (RXRα), an important ligand-dependent transcription factor, plays a critical role in the development of various cancers and metabolic and neurodegenerative diseases. Therefore, RXRα represents one of the most important targets in modern drug discovery. In this study, Drugbank 2.0 with 1280 old drugs were virtually screened by Glide according to the crystal structure of ligand-binding domain (LBP) of RXRα. 15 compounds selected were tested for their binding and transcriptional activity toward RXRα by Biacore and reporter gene assay, respectively. The identified new scafford ligand of RXRα, Pitavastatin (1), was chemically optimized. Our results demonstrated that statin compounds Pitavastatin (1) and Fluvastatin (4) could bind to the LBP of RXRα (KD=13.30µM and 11.04µM, respectively) and serve as transcriptional antagonists of RXRα. On the contrary, compound (12) (domperidone) and (13) (rosiglitazone maleate) could bind to the LBP of RXRα (KD=8.80µM and 15.01µM, respectively) but serve as transcriptional agonists of RXRα.


Subject(s)
Databases, Factual , Retinoid X Receptor alpha/antagonists & inhibitors , Crystallography, X-Ray , Drug Evaluation, Preclinical , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Indoles/chemistry , Indoles/pharmacology , Ligands , Quinolines/chemistry , Quinolines/pharmacology , Retinoid X Receptor alpha/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL