Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 50(1): 43-48, 2022 01.
Article in English | MEDLINE | ID: mdl-34697082

ABSTRACT

Carboxylesterase 1 (CES1) is the predominant carboxylesterase in the human liver, involved in metabolism of both xenobiotics and endogenous substrates. Genetic or epigenetic factors that alter CES1 activity or expression are associated with changes in drug response, lipid, and glucose homeostasis. However, the transcriptional regulation of CES1 in the human liver remains uncertain. By applying both the random forest and Sobol's Sensitivity Indices (SSI) to analyze existing liver RNA expression microarray data (GSE9588), we identified nuclear receptor subfamily 1 group H member 3 (NR1H3) liver X receptor (LXR)α as a key factor regulating constitutive CES1 expression. This model prediction was validated using small interfering RNA (siRNA) knockdown and CRISPR-mediated transcriptional activation of NR1H3 in Huh7 and HepG2 cells. We found that NR1H3's activation of CES1 is splice isoform-specific, namely that increased expression of the NR1H3-211 isoform increased CES1 expression whereas NR1H3-201 did not. Also, in human liver samples, expression of NR1H3-211 and CES1 are correlated, whereas NR1H3-201 and CES1 are not. This trend also occurs during differentiation of induced pluripotent stem cells (iPSCs) to hepatocytes, where only expression of the NR1H3-211 isoform parallels expression of CES1 Moreover, we found that treatment with the NR1H3 agonist T0901317 in HepG2 cells had no effect on CES1 expression. Overall, our results demonstrate a key role of NR1H3 in maintaining the constitutive expression of CES1 in the human liver. Furthermore, our results support that the effect of NR1H3 is splice isoform-specific and appears to be ligand independent. SIGNIFICANCE STATEMENT: Despite the central role of carboxylesterase 1 (CES1) in metabolism of numerous medications, little is known about its transcriptional regulation. This study identifies nuclear receptor subfamily 1 group H member 3 as a key regulator of constitutive CES1 expression and therefore is a potential target for future studies to understand interperson variabilities in CES1 activity and drug metabolism.


Subject(s)
Carboxylic Ester Hydrolases/biosynthesis , Carboxylic Ester Hydrolases/genetics , Gene Expression Regulation, Enzymologic/physiology , Liver X Receptors/genetics , Liver X Receptors/physiology , Liver/enzymology , Aged , Cell Line , Female , Gene Expression Regulation , Gene Knockdown Techniques , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells , Isoenzymes/genetics , Isoenzymes/metabolism , Liver X Receptors/agonists , Male , Middle Aged , RNA, Small Interfering , Transcriptional Activation/genetics
2.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33373442

ABSTRACT

T cells increase cholesterol biosynthesis upon activation to generate substrates for cellular growth and proliferation. The ubiquitously expressed liver X receptor ß (LXRß) encoded by the Nr1h2 gene is a critical regulator of cholesterol homeostasis in mammalian cells; however, its cell-intrinsic role in T cell biology remains poorly understood. We report that ablation of LXRß in T cells leads to spontaneous T cell activation and T lymphocytopenia. Unexpectedly, analysis of mixed bone marrow chimeric mice revealed a cell-autonomous survival defect that reduced the fitness of LXRß-deficient effector T cells, suggesting that the heightened immune activation in mice harboring LXRß-deficient T cells was due to impaired regulatory T (T reg) cell functionality. Indeed, we found that single-copy deletion of Nr1h2 in T reg cells disrupted activated T reg cell metabolism and fitness and resulted in early-onset fatal autoimmune disease. Our study demonstrated an indispensable requirement for T reg cell-intrinsic LXRß function in immune homeostasis and provides a basis for immunological therapies through targeting of this receptor.


Subject(s)
Autoimmune Diseases/immunology , Homeostasis/immunology , Liver X Receptors/physiology , Lymphocyte Activation/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytopenia, Idiopathic CD4-Positive/immunology , Animals , Autoimmune Diseases/genetics , Cells, Cultured , Cholesterol/metabolism , Female , Forkhead Transcription Factors/genetics , Homeostasis/genetics , Liver X Receptors/genetics , Male , Mice , Mice, Inbred C57BL , Radiation Chimera/immunology , Signal Transduction/genetics , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytopenia, Idiopathic CD4-Positive/genetics
3.
Hepatology ; 73(4): 1307-1326, 2021 04.
Article in English | MEDLINE | ID: mdl-32557804

ABSTRACT

BACKGROUND AND AIMS: Fat accumulation results from increased fat absorption and/or defective fat metabolism. Currently, the lipid-sensing nuclear receptor that controls fat utilization in hepatocytes is elusive. Liver X receptor alpha (LXRα) promotes accumulation of lipids through the induction of several lipogenic genes. However, its effect on lipid degradation is open for study. Here, we investigated the inhibitory role of LXRα in autophagy/lipophagy in hepatocytes and the underlying basis. APPROACH AND RESULTS: In LXRα knockout mice fed a high-fat diet, or cell models, LXRα activation suppressed the function of mitochondria by inhibiting autophagy/lipophagy and induced hepatic steatosis. Gene sets associated with "autophagy" were enriched in hepatic transcriptome data. Autophagy flux was markedly augmented in the LXRα knockout mouse liver and primary hepatocytes. Mechanistically, LXRα suppressed autophagy-related 4B cysteine peptidase (ATG4B) and Rab-8B, responsible for autophagosome and -lysosome formation, by inducing let-7a and microRNA (miR)-34a. Chromatin immunoprecipitation assay enabled us to find LXRα as a transcription factor of let-7a and miR-34a. Moreover, 3' untranslated region luciferase assay substantiated the direct inhibitory effects of let-7a and miR-34a on ATG4B and Rab-8B. Consistently, either LXRα activation or the let-7a/miR-34a transfection lowered mitochondrial oxygen consumption rate and mitochondrial transmembrane potential and increased fat levels. In obese animals or nonalcoholic fatty liver disease (NAFLD) patients, let-7a and miR-34a levels were elevated with simultaneous decreases in ATG4B and Rab-8B levels. CONCLUSIONS: LXRα inhibits autophagy in hepatocytes through down-regulating ATG4B and Rab-8B by transcriptionally activating microRNA let-7a-2 and microRNA 34a genes and suppresses mitochondrial biogenesis and fuel consumption. This highlights a function of LXRα that culminates in the progression of liver steatosis and steatohepatitis, and the identified targets may be applied for a therapeutic strategy in the treatment of NAFLD.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy/physiology , Cysteine Endopeptidases/metabolism , Hepatocytes/metabolism , Liver X Receptors/metabolism , Mitochondria/physiology , rab GTP-Binding Proteins/metabolism , Activation, Metabolic , Animals , Autophagy/genetics , Autophagy-Related Proteins/genetics , Cysteine Endopeptidases/genetics , Disease Models, Animal , Disease Progression , Down-Regulation , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/physiopathology , Hep G2 Cells/metabolism , Hep G2 Cells/physiology , Hepatocytes/physiology , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Liver/metabolism , Liver/physiology , Liver/physiopathology , Liver X Receptors/genetics , Liver X Receptors/physiology , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , MicroRNAs/physiology , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology , Organelle Biogenesis , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Transcriptome , rab GTP-Binding Proteins/genetics
4.
J Exp Med ; 217(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32716519

ABSTRACT

The thymus is a primary lymphoid organ necessary for optimal T cell development. Here, we show that liver X receptors (LXRs)-a class of nuclear receptors and transcription factors with diverse functions in metabolism and immunity-critically contribute to thymic integrity and function. LXRαß-deficient mice develop a fatty, rapidly involuting thymus and acquire a shrunken and prematurely immunoinhibitory peripheral T cell repertoire. LXRαß's functions are cell specific, and the resulting phenotypes are mutually independent. Although thymic macrophages require LXRαß for cholesterol efflux, thymic epithelial cells (TECs) use LXRαß for self-renewal and thymocytes for negative selection. Consequently, TEC-derived LXRαß protects against homeostatic premature involution and orchestrates thymic regeneration following stress, while thymocyte-derived LXRαß limits cell disposal during negative selection and confers heightened sensitivity to experimental autoimmune encephalomyelitis. These results identify three distinct but complementary mechanisms by which LXRαß governs T lymphocyte education and illuminate LXRαß's indispensable roles in adaptive immunity.


Subject(s)
Liver X Receptors/physiology , Liver/metabolism , T-Lymphocytes/physiology , Thymus Gland/physiology , Adaptive Immunity , Animals , Apoptosis , Female , Flow Cytometry , Homeostasis , Humans , Lipid Metabolism , Liver X Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , T-Lymphocytes/metabolism , Thymus Gland/metabolism
5.
Cell Signal ; 74: 109720, 2020 10.
Article in English | MEDLINE | ID: mdl-32711110

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignancies with a high rate of mortality. Highly upregulated in liver cancer (HULC), the specifically overexpressed long non-coding RNA in human HCC, plays important roles in promoting the growth and metastasis of HCC cells. So downregulating HULC will be benefit to HCC treatment. The nuclear receptor LXR (liver X receptor), consist of α and ß isoforms, exerts significant anti-HCC effects, but the corresponding mechanisms are not well known, especially, it's unclear whether LXR is involved in the regulation of HULC. In this study, we found that LXR inhibited HCC cell growth by downregulating HULC, and LXRα (but not LXRß) caused HULC downregulation. Luciferase reporter assays showed that LXR suppressed transcriptional activity of HULC gene promoter, and chromatin immunoprecipitation assays revealed that LXRα (but not LXRß) bound to HULC promoter region. Furthermore, LXR increased miR-134-5p while decreased FOXM1 by reducing HULC. Additionally, HULC upregulated FOXM1 via sequestrating miR-134-5p, and miR-134-5p downregulated FOXM1 by targeting 3'-UTR of its mRNA. The in vivo experiments showed that LXR repressed the growth of HCC xenografts, and decreased HULC and FOXM1 while increased miR-134-5p in the xenografts. In summary, these findings for the first time demonstrate that LXR inhibits HCC cell growth by modulating HULC/miR-134-5p/FOXM1 axis, suggesting that the pathway LXR/HULC/miR-134-5p/FOXM1 may serve as a novel target for HCC treatment.


Subject(s)
Forkhead Box Protein M1/metabolism , Liver X Receptors/physiology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Animals , Carcinoma, Hepatocellular , Cell Proliferation , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms , Mice , Mice, Nude
6.
PLoS One ; 15(2): e0223813, 2020.
Article in English | MEDLINE | ID: mdl-32109243

ABSTRACT

Cholesterol metabolism is greatly affected in fish fed plant-based diet. The regulation of cholesterol metabolism is mediated by both transcriptional factors such as sterol regulatory element-binding proteins (SREBPs) and liver X receptors (LXRs), and posttranscriptional factors including miRNAs. In mammals, SREBP-2 and LXRα are involved in the transcriptional regulation of cholesterol synthesis and elimination, respectively. In mammals, miR-33a is reported to directly target genes involved in cholesterol catabolism. The present study aims to investigate the regulation of cholesterol metabolism by SREBP-2 and LXRα and miR-33a in rainbow trout using in vivo and in vitro approaches. In vivo, juvenile rainbow trout of ~72 g initial body weight were fed a total plant-based diet (V) or a marine diet (M) containing fishmeal and fish oil. In vitro, primary cell culture hepatocytes were stimulated by graded concentrations of 25-hydroxycholesterol (25-HC). The hepatic expression of cholesterol synthetic genes, srebp-2 and miR-33a as well as miR-33a level in plasma were increased in fish fed the plant-based diet, reversely, their expression in hepatocytes were inhibited with the increasing 25-HC in vitro. However, lxrα was not affected neither in vivo nor in vitro. Our results suggest that SREBP-2 and miR-33a synergistically enhance the expression of cholesterol synthetic genes but do not support the involvement of LXRα in the regulation of cholesterol elimination. As plasma level of miR-33a appears as potential indicator of cholesterol synthetic capacities, this study also highlights circulating miRNAs as promising noninvasive biomarker in aquaculture.


Subject(s)
Cholesterol/metabolism , Liver X Receptors/physiology , MicroRNAs/physiology , Oncorhynchus mykiss/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , Aquaculture/methods , Cells, Cultured , Hepatocytes/metabolism , Lipid Metabolism
7.
Sci Rep ; 9(1): 15458, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664073

ABSTRACT

Cholesterol is a critical component of membranes and a precursor for hormones and other signaling molecules. Previously, we showed that unlike astrocytes, glioblastoma cells do not downregulate cholesterol synthesis when plated at high density. In this report, we show that high cell density induces ABCA1 expression in glioblastoma cells, enabling them to get rid of excess cholesterol generated by an activated cholesterol biosynthesis pathway. Because oxysterols are agonists for Liver X Receptors (LXRs), we investigated whether increased cholesterol activates LXRs to maintain cholesterol homeostasis in highly-dense glioblastoma cells. We observed that dense cells had increased oxysterols, which activated LXRß to upregulate ABCA1. Cells with CRISPR-mediated knockdown of LXRß, but not ABCA1, had decreased cell cycle progression and cell survival, and decreased feedback repression of the mevalonate pathway in densely-plated glioma cells. LXRß gene expression poorly correlates with ABCA1 in glioblastoma patients, and expression of each gene correlates with poor patient prognosis in different prognostic subtypes. Finally, gene expression and lipidomics analyses cells revealed that LXRß regulates the expression of immune response gene sets and lipids known to be involved in immune modulation. Thus, therapeutic targeting of LXRß in glioblastoma might be effective through diverse mechanisms.


Subject(s)
ATP Binding Cassette Transporter 1/physiology , Brain Neoplasms/pathology , Cell Proliferation/physiology , Glioblastoma/pathology , Lipid Metabolism , Liver X Receptors/physiology , ATP Binding Cassette Transporter 1/genetics , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cholesterol/metabolism , Glioblastoma/immunology , Glioblastoma/metabolism , Homeostasis , Humans , Liver X Receptors/metabolism , Mevalonic Acid/metabolism , Signal Transduction , Transcription, Genetic
8.
FASEB J ; 33(9): 10077-10088, 2019 09.
Article in English | MEDLINE | ID: mdl-31237775

ABSTRACT

Bone morphogenetic protein (BMP)-9 has been reported to regulate energy balance in vivo. However, the mechanisms underlying BMP9-mediated regulation of energy balance remain incompletely understood. Here, we investigated the role of BMP9 in energy metabolism. In the current study, we found that hepatic BMP9 expression was down-regulated in insulin resistance (IR) mice and in patients who are diabetic. In mice fed a high-fat diet (HFD), the overexpression of hepatic BMP9 improved glucose tolerance and IR. The expression of gluconeogenic genes was down-regulated, whereas the level of insulin signaling molecule phosphorylation was increased in the livers of Adenovirus-BMP9-treated mice and glucosamine-treated hepatocytes. Furthermore, BMP9 overexpression ameliorated triglyceride accumulation and inhibited the expression of lipogenic genes in both human hepatocellular carcinoma HepG2 cells treated with a fatty acid mixture as well as the livers of HFD-fed mice. In hepatocytes isolated from sterol regulatory element-binding protein (SREBP)-1c knockout mice, the effects of BMP9 were ablated. Mechanistically, BMP9 inhibited SREBP-1c expression through the inhibition of liver X receptor response element 1 activity in the SREBP-1c promoter. Taken together, our results show that BMP9 is an important regulator of hepatic glucose and lipid metabolism.-Yang, M., Liang, Z., Yang, M., Jia, Y., Yang, G., He, Y., Li, X., Gu, H. F., Zheng, H., Zhu, Z., Li, L. Role of bone morphogenetic protein-9 in the regulation of glucose and lipid metabolism.


Subject(s)
Glucose/metabolism , Growth Differentiation Factor 2/physiology , Lipid Metabolism/physiology , Liver/metabolism , Animals , Bone Morphogenetic Protein Receptors/physiology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cells, Cultured , Diet, High-Fat/adverse effects , Fatty Acids/pharmacology , Gene Expression Regulation , Growth Differentiation Factor 2/biosynthesis , Growth Differentiation Factor 2/genetics , Hepatocytes/metabolism , Humans , Insulin Resistance , Lipid Metabolism/genetics , Lipogenesis/genetics , Liver/drug effects , Liver Neoplasms/pathology , Liver X Receptors/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , Receptors, Leptin/deficiency , Recombinant Proteins/metabolism , Response Elements/genetics , Sterol Regulatory Element Binding Protein 1/deficiency , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
9.
Cell Rep ; 25(2): 271-277.e4, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30304667

ABSTRACT

Obesity is associated with many complications, including type 2 diabetes and painful neuropathy. There is no cure or prevention for obesity-induced pain, and the neurobiology underlying the onset of the disease is still obscure. In this study, we observe that western diet (WD)-fed mice developed early allodynia with an increase of ER stress markers in the sensory neurons of the dorsal root ganglia (DRG). Using cell-specific approaches, we demonstrate that neuronal liver X receptor (LXR) activation delays ER stress and allodynia in WD-fed mice. Our findings suggest that lipid-binding nuclear receptors expressed in the sensory neurons of the DRG play a role in the onset of obesity-induced hypersensitivity. The LXR and lipid-sensor pathways represent a research avenue to identify targets to prevent debilitating complications affecting the peripheral nerve system in obesity.


Subject(s)
Endoplasmic Reticulum Stress , Ganglia, Spinal/drug effects , Hyperalgesia/etiology , Liver X Receptors/physiology , Obesity/complications , Sensory Receptor Cells/drug effects , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Benzoates/pharmacology , Benzylamines/pharmacology , Diet, Western/adverse effects , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Liver X Receptors/agonists , Male , Mice , Mice, Knockout , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism
10.
Int J Mol Sci ; 19(8)2018 Jul 25.
Article in English | MEDLINE | ID: mdl-30044452

ABSTRACT

A close relationship exists between cholesterol and female reproductive physiology. Indeed, cholesterol is crucial for steroid synthesis by ovary and placenta, and primordial for cell structure during folliculogenesis. Furthermore, oxysterols, cholesterol-derived ligands, play a potential role in oocyte maturation. Anomalies of cholesterol metabolism are frequently linked to infertility. However, little is known about the molecular mechanisms. In parallel, increasing evidence describing the biological roles of liver X receptors (LXRs) in the regulation of steroid synthesis and inflammation, two processes necessary for follicle maturation and ovulation. Both of the isoforms of LXRs and their bona fide ligands are present in the ovary. LXR-deficient mice develop late sterility due to abnormal oocyte maturation and increased oocyte atresia. These mice also have an ovarian hyper stimulation syndrome in response to gonadotropin stimulation. Hence, further studies are necessary to explore their specific roles in oocyte, granulosa, and theca cells. LXRs also modulate estrogen signaling and this could explain the putative protective role of the LXRs in breast cancer growth. Altogether, clinical studies would be important for determining the physiological relevance of LXRs in reproductive disorders in women.


Subject(s)
Cholesterol/metabolism , Infertility, Female/metabolism , Liver X Receptors/physiology , Metabolic Syndrome/metabolism , Obesity/metabolism , Animals , Breast Neoplasms/metabolism , Estrogens/metabolism , Female , Humans , Infertility, Female/complications , Infertility, Female/genetics , Liver X Receptors/genetics , Metabolic Syndrome/complications , Metabolic Syndrome/genetics , Mice , Obesity/complications , Obesity/genetics , Ovarian Hyperstimulation Syndrome/genetics , Ovarian Hyperstimulation Syndrome/metabolism , Ovary/physiology , Placenta/physiology , Pregnancy
11.
Mol Cell Biol ; 38(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29507185

ABSTRACT

The liver X receptors (LXRs) are ligand-activated nuclear receptors with established roles in the maintenance of lipid homeostasis in multiple tissues. LXRs exert additional biological functions as negative regulators of inflammation, particularly in macrophages. However, the transcriptional responses controlled by LXRs in other myeloid cells, such as dendritic cells (DCs), are still poorly understood. Here we used gain- and loss-of-function models to characterize the impact of LXR deficiency on DC activation programs. Our results identified an LXR-dependent pathway that is important for DC chemotaxis. LXR-deficient mature DCs are defective in stimulus-induced migration in vitro and in vivo Mechanistically, we show that LXRs facilitate DC chemotactic signaling by regulating the expression of CD38, an ectoenzyme important for leukocyte trafficking. Pharmacological or genetic inactivation of CD38 activity abolished the LXR-dependent induction of DC chemotaxis. Using the low-density lipoprotein receptor-deficient (LDLR-/-) LDLR-/- mouse model of atherosclerosis, we also demonstrated that hematopoietic CD38 expression is important for the accumulation of lipid-laden myeloid cells in lesions, suggesting that CD38 is a key factor in leukocyte migration during atherogenesis. Collectively, our results demonstrate that LXRs are required for the efficient emigration of DCs in response to chemotactic signals during inflammation.


Subject(s)
Chemotaxis/physiology , Dendritic Cells/physiology , Liver X Receptors/physiology , ADP-ribosyl Cyclase 1/metabolism , Animals , Cells, Cultured , Dendritic Cells/cytology , Inflammation , Lipid Metabolism , Liver X Receptors/genetics , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear , Signal Transduction
12.
Sci Rep ; 8(1): 2524, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29410501

ABSTRACT

Reactive oxygen species (ROS) modify proteins and lipids leading to deleterious outcomes. Thus, maintaining their homeostatic levels is vital. This study highlights the endogenous role of LXRs (LXRα and ß) in the regulation of oxidative stress in peripheral nerves. We report that the genetic ablation of both LXR isoforms in mice (LXRdKO) provokes significant locomotor defects correlated with enhanced anion superoxide production, lipid oxidization and protein carbonylation in the sciatic nerves despite the activation of Nrf2-dependant antioxidant response. Interestingly, the reactive oxygen species scavenger N-acetylcysteine counteracts behavioral, electrophysical, ultrastructural and biochemical alterations in LXRdKO mice. Furthermore, Schwann cells in culture pretreated with LXR agonist, TO901317, exhibit improved defenses against oxidative stress generated by tert-butyl hydroperoxide, implying that LXRs play an important role in maintaining the redox homeostasis in the peripheral nervous system. Thus, LXR activation could be a promising strategy to protect from alteration of peripheral myelin resulting from a disturbance of redox homeostasis in Schwann cell.


Subject(s)
Homeostasis , Liver X Receptors/physiology , Myelin Sheath/metabolism , Oxidative Stress , Schwann Cells , Sciatic Nerve , Animals , Cell Line , Hydrocarbons, Fluorinated/chemistry , Lipid Metabolism , Liver X Receptors/antagonists & inhibitors , Liver X Receptors/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Protein Carbonylation , Reactive Oxygen Species/metabolism , Schwann Cells/cytology , Schwann Cells/metabolism , Sciatic Nerve/cytology , Sciatic Nerve/metabolism , Sulfonamides/chemistry , tert-Butylhydroperoxide/chemistry
13.
Anesth Analg ; 127(3): 775-783, 2018 09.
Article in English | MEDLINE | ID: mdl-29324503

ABSTRACT

BACKGROUND: Neuropathic pain is an intractable and complex disease. Recent studies have shown a close relationship between endoplasmic reticulum (ER) stress and neuropathic pain. Here, we investigated the effect of α-asarone, an ER stress inhibitor, on chronic constriction injury (CCI)-induced neuropathic pain. METHODS: Two parts were included in this study. In part 1, rats were assigned to 7 groups: the sham group, the sham + α-asarone 20 mg/kg group, the CCI group, the CCI + vehicle group, the CCI + α-asarone 5 mg/kg group, the CCI + α-asarone 10 mg/kg group, and the CCI + α-asarone 20 mg/kg group. After surgery, the rats were treated with α-asarone or normal saline daily. Pain thresholds were measured, and samples of the L3-6 spinal cord were taken for western blotting and immunofluorescence on day 7. In part 2, rats were intrathecally implanted with PE-10 tubes and divided into 4 groups: the CCI + α-asarone 20 mg/kg group, the CCI + α-asarone 20 mg/kg + vehicle group, the CCI + α-asarone 20 mg/kg + SR9243 group, and the CCI group. Five rats in each group were separated for behavioral tests 1 hour after intrathecal injection. The rest of them were killed for western blotting on day 7. RESULTS: In this study, CCI surgery significantly induced mechanical allodynia and thermal hyperalgesia. CCI surgery significantly induced activation of ER stress (PERK-eIF2α, IRE1α, CHOP, and XBP-1s) in rats. However, treatment with 20 mg/kg of α-asarone significantly alleviated CCI-induced activation of ER stress. Behavioral results showed that daily treatment with 20 mg/kg of α-asarone significantly alleviated CCI-induced nociceptive behaviors, on day 7 (mechanical allodynia, P = .016, 95% confidence interval, 0.645-5.811; thermal hyperalgesia, P = .012, 95% confidence interval, 0.860-6.507). Furthermore, α-asarone induced upregulated expression of liver X receptor ß (LXRß) and downstream proteins in the spinal cord. The LXR antagonist SR9243 completely inhibited the anti-ER stress and antinociceptive effects of α-asarone in rats. CONCLUSIONS: α-Asarone relieved CCI-induced neuropathic pain in an LXR-dependent manner. α-Asarone may be a potential agent for treatment of neuropathic pain.


Subject(s)
Anisoles/administration & dosage , Endoplasmic Reticulum Stress/physiology , Liver X Receptors/physiology , Neuralgia/drug therapy , Sciatic Neuropathy/drug therapy , Allylbenzene Derivatives , Animals , Constriction , Endoplasmic Reticulum Stress/drug effects , Fibrinolytic Agents/administration & dosage , Injections, Spinal , Liver X Receptors/agonists , Liver X Receptors/antagonists & inhibitors , Male , Neuralgia/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/pathology , Sulfonamides/administration & dosage
14.
Am J Chin Med ; 46(1): 87-106, 2018.
Article in English | MEDLINE | ID: mdl-29298513

ABSTRACT

oxLDL is involved in the pathogenesis of atherosclerotic lesions through cholesterol accumulation in macrophage foam cells. Andrographolide, the bioactive component of Andrographis paniculata, possesses several biological activities such as anti-inflammatory, anti-oxidant, and anticancer functions. Scavenger receptors (SRs), including class A SR (SR-A) and CD36, are responsible for the internalization of oxLDL. In contrast, receptors for reverse cholesterol transport, including ABCA1 and ABCG1, mediate the efflux of cholesterol from macrophage foam cells. Transcription factor liver X receptor [Formula: see text] (LXR[Formula: see text] plays a key role in lipid metabolism and inflammation as well as in the regulation of ABCA1 and ABCG1 expression. Because of the contribution of inflammation to macrophage foam cell formation and the potent anti-inflammatory activity of andrographolide, we hypothesized that andrographolide might inhibit oxLDL-induced macrophage foam cell formation. The results showed that andrographolide reduced oxLDL-induced lipid accumulation in macrophage foam cells. Andrographolide decreased the mRNA and protein expression of CD36 by inducing the degradation of CD36 mRNA; however, andrographolide had no effect on SR-A expression. In contrast, andrographolide increased the mRNA and protein expression of ABCA1 and ABCG1, which were dependent on LXR[Formula: see text]. Andrographolide enhanced LXR[Formula: see text] nuclear translocation and DNA binding activity. Treatment with the LXR[Formula: see text] antagonist GGPP and transfection with LXR[Formula: see text] siRNA reversed the ability of andrographolide to stimulate ABCA1 and ABCG1 protein expression. In conclusion, inhibition of CD36-mediated oxLDL uptake and induction of ABCA1- and ABCG1-dependent cholesterol efflux are two working mechanisms by which andrographolide inhibits macrophage foam cell formation, which suggests that andrographolide could be a potential candidate to prevent atherosclerosis.


Subject(s)
Andrographis/chemistry , Cholesterol/metabolism , Diterpenes/pharmacology , Foam Cells/metabolism , Lipoproteins, LDL/adverse effects , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Animals , Anti-Inflammatory Agents , Antineoplastic Agents, Phytogenic , Antioxidants , Atherosclerosis/etiology , Biological Transport/genetics , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Line , Gene Expression/drug effects , Liver X Receptors/physiology , Mice , RNA, Messenger/metabolism , Receptors, Scavenger/physiology
15.
Transl Res ; 193: 13-30, 2018 03.
Article in English | MEDLINE | ID: mdl-29172034

ABSTRACT

Dysfunctional macrophages underlie the development of several diseases including atherosclerosis where accumulation of cholesteryl esters and persistent inflammation are 2 of the critical macrophage processes that regulate the progression as well as stability of atherosclerotic plaques. Ligand-dependent activation of liver-x-receptor (LXR) not only enhances mobilization of stored cholesteryl ester but also exerts anti-inflammatory effects mediated via trans-repression of proinflammatory transcription factor nuclear factor kappa B. However, increased hepatic lipogenesis by systemic administration of LXR ligands (LXR-L) has precluded their therapeutic use. The objective of the present study was to devise a strategy to selectively deliver LXR-L to atherosclerotic plaque-associated macrophages while limiting hepatic uptake. Mannose-functionalized dendrimeric nanoparticles (mDNP) were synthesized to facilitate active uptake via the mannose receptor expressed exclusively by macrophages using polyamidoamine dendrimer. Terminal amine groups were used to conjugate mannose and LXR-L T091317 via polyethylene glycol spacers. mDNP-LXR-L was effectively taken up by macrophages (and not by hepatocytes), increased expression of LXR target genes (ABCA1/ABCG1), and enhanced cholesterol efflux. When administered intravenously to LDLR-/- mice with established plaques, significant accumulation of fluorescently labeled mDNP-LXR-L was seen in atherosclerotic plaque-associated macrophages. Four weekly injections of mDNP-LXR-L led to significant reduction in atherosclerotic plaque progression, plaque necrosis, and plaque inflammation as assessed by expression of nuclear factor kappa B target gene matrix metalloproteinase 9; no increase in hepatic lipogenic genes or plasma lipids was observed. These studies validate the development of a macrophage-specific delivery platform for the delivery of anti-atherosclerotic agents directly to the plaque-associated macrophages to attenuate plaque burden.


Subject(s)
Atherosclerosis/drug therapy , Dendrimers/administration & dosage , Macrophages/metabolism , Mannose/metabolism , Nanoparticles/administration & dosage , Animals , Cells, Cultured , Female , Liver X Receptors/physiology , Male , Mice , Receptors, LDL/physiology
16.
Pharmacol Ther ; 181: 1-12, 2018 01.
Article in English | MEDLINE | ID: mdl-28720427

ABSTRACT

The Liver X Receptors (LXRs) are oxysterol-activated transcription factors that upregulate a suite of genes that together promote coordinated mobilization of excess cholesterol from cells and from the body. The LXRs, like other nuclear receptors, are anti-inflammatory, inhibiting signal-dependent induction of pro-inflammatory genes by nuclear factor-κB, activating protein-1, and other transcription factors. Synthetic LXR agonists have been shown to ameliorate atherosclerosis and a wide range of inflammatory disorders in preclinical animal models. Although this has suggested potential for application to human disease, systemic LXR activation is complicated by hepatic steatosis and hypertriglyceridemia, consequences of lipogenic gene induction in the liver by LXRα. The past several years have seen the development of multiple advanced LXR therapeutics aiming to avoid hepatic lipogenesis, including LXRß-selective agonists, tissue-selective agonists, and transrepression-selective agonists. Although several synthetic LXR agonists have made it to phase I clinical trials, none have progressed due to unforeseen adverse reactions or undisclosed reasons. Nonetheless, several sophisticated pharmacologic strategies, including structure-guided drug design, cell-specific drug targeting, as well as non-systemic drug routes have been initiated and remain to be comprehensively explored. In addition, recent studies have identified potential utility for targeting the LXRs during therapy with other agents, such as glucocorticoids and rexinoids. Despite the pitfalls encountered to date in translation of LXR agonists to human disease, it appears likely that this accelerating field will ultimately yield effective and safe applications for LXR targeting in humans.


Subject(s)
Atherosclerosis/drug therapy , Inflammation/drug therapy , Liver X Receptors/agonists , Liver X Receptors/physiology , Animals , Benzoates/pharmacology , Benzoates/therapeutic use , Benzylamines/pharmacology , Benzylamines/therapeutic use , Gene Expression Regulation/drug effects , Humans , Hydrocarbons, Fluorinated/pharmacology , Hydrocarbons, Fluorinated/therapeutic use , Models, Biological , Molecular Targeted Therapy/methods , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
17.
Reproduction ; 154(6): 827-842, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28971895

ABSTRACT

Lipid metabolism disorders (dyslipidemia) are causes of male infertility, but little is known about their impact on male gametes when considering post-testicular maturation events, given that studies concentrate most often on endocrine dysfunctions and testicular consequences. In this study, three-month-old wild-type (wt) and Liver-X-Receptors knock out (Lxrα;ß-/- ) males were fed four weeks with a control or a lipid-enriched diet containing 1.25% cholesterol (high cholesterol diet (HCD)). The HCD triggered a dyslipidemia leading to sperm post-testicular alterations and infertility. Sperm lipids were analyzed by LC-MS and those from Lxrα;ß-/- males fed the HCD showed higher chol/PL and PC/PE ratios compared to wt-HCD (P < 0.05) and lower oxysterol contents compared to wt (P < 0.05) or Lxrα;ß-/- (P < 0.05). These modifications impaired membrane-associated events triggering the tyrosine phosphorylation normally occurring during the capacitation process, as shown by phosphotyrosine Western blots. Using flow cytometry, we showed that a smaller subpopulation of spermatozoa from Lxrα;ß-/- -HCD males could raise their membrane fluidity during capacitation (P < 0.05 vs wt or wt-HCD) as well as their intracellular calcium concentration (P < 0.05 vs Lxrα;ß-/- and P < 0.001 vs wt). The accumulation of the major sperm calcium efflux pump (PMCA4) was decreased in Lxrα;ß-/- males fed the HCD (P < 0.05 vs Lxrα;ß-/- and P < 0.001 vs wt). This study is the first showing an impact of dyslipidemia on post-testicular sperm maturation with consequences on the capacitation signaling cascade. It may lead to the identification of fertility prognostic markers in this pathophysiological situation, which could help clinicians to better understand male infertilities which are thus far classified as idiopathic.


Subject(s)
Dyslipidemias/complications , Infertility, Male/etiology , Liver X Receptors/physiology , Sperm Capacitation , Sperm Maturation , Spermatozoa/pathology , Animals , Fertility , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Mice , Mice, Knockout , Signal Transduction
18.
Biochem Pharmacol ; 144: 108-119, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28807695

ABSTRACT

The anthracycline-mediated cardiotoxicity is still not completely understood. To examine the impact of cholesterol metabolism and transport in this context, cholesterol and oxysterol levels as well as the expression of the cholesterol transporters ABCA1 and ABCG1 were analyzed in doxorubicin-treated HL-1 murine cardiomyocytes as well as in mouse model for acute doxorubicin-induced cardiotoxicity. Doxorubicin-treated HL-1 cells exhibited enhanced cholesterol (153±20% of control), oxysterol (24S-hydroxycholesterol: 206±29% of control) and cholesterol precursor levels (lathosterol: 122±12% of control; desmosterol: 188±10% of control) indicating enhanced cholesterol synthesis. Moreover, abca1 and abcg1 were upregulated on mRNA, protein and functional level caused by a doxorubicin-mediated activation of the nuclear receptor LXR. In addition, the oxysterols not only induced the abca1 and abcg1 in HL-1 cells but also enhanced the expression of endothelin-1 and transforming growth factor-ß, which have already been identified as important factors in doxorubicin-induced cardiotoxicity. These in vitro findings were verified in a murine model for acute doxorubicin-induced cardiotoxicity, demonstrating elevated cardiac (2.1±0.2vs. 3.6±1.0ng/mg) and systemic cholesterol levels (105.0±8.4vs. 130.0±4.3mg/dl), respectively, as well as enhanced oxysterol levels such as cardiac 24S-hydroxycholesterol (2.1±0.2vs. 3.6±1.0ng/mg). In line with these findings cardiac mRNA expression of abca1 (303% of control) and abcg1 (161% of control) was induced. Taken together, our data demonstrate enhanced cholesterol and oxysterol levels by doxorubicin, resulting in a LXR-dependent upregulation of abca1 and abcg1. In this context, the cytotoxic effects of oxysterols and their impact on cardiac gene expression should be considered as an important factor in doxorubicin-induced cardiotoxicity.


Subject(s)
ATP Binding Cassette Transporter 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 1/biosynthesis , Doxorubicin/pharmacology , Liver X Receptors/physiology , Myocytes, Cardiac/metabolism , Oxysterols/metabolism , Animals , Cells, Cultured , Cholesterol/metabolism , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
20.
Mol Nutr Food Res ; 61(9)2017 09.
Article in English | MEDLINE | ID: mdl-28296229

ABSTRACT

SCOPE: Previous studies have proposed that phytosterols activate liver X receptors (LXR) in the intestine, thereby reducing intestinal cholesterol absorption and promoting fecal cholesterol excretion. METHODS AND RESULTS: In the present study, we examined the effects of dietary phytosterol supplementation on intestinal cholesterol absorption and fecal neutral sterol excretion in LXRαß-deficient mice, and wild-type mice treated with synthetic high-affinity LXRαß agonists. LXRαß deficiency led to an induction of intestinal cholesterol absorption and liver cholesterol accumulation. Phytosterol feeding resulted in an approximately 40% reduction of intestinal cholesterol absorption both in wild-type and LXRαß-deficient mice, reduced dietary cholesterol accumulation in liver and promoted the excretion of fecal cholesterol-derived compounds. Furthermore, phytosterols produced additive inhibitory effects on cholesterol absorption in mice treated with LXRαß agonists. CONCLUSIONS: Our data confirm the effect of LXR in regulating intestinal cholesterol absorption and demonstrate that the cholesterol-lowering effects of phytosterols occur in an LXR-independent manner.


Subject(s)
Cholesterol/metabolism , Intestinal Absorption/drug effects , Liver X Receptors/physiology , Phytosterols/pharmacology , Animals , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...