Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Commun Biol ; 5(1): 51, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35027651

ABSTRACT

Retinoic acid-related orphan receptor alpha (RORA) suppression is associated with autism spectrum disorder (ASD) development, although the mechanism remains unclear. In this study, we aim to investigate the potential effect and mechanisms of RORA suppression on autism-like behavior (ALB) through maternal diabetes-mediated mouse model. Our in vitro study in human neural progenitor cells shows that transient hyperglycemia induces persistent RORA suppression through oxidative stress-mediated epigenetic modifications and subsequent dissociation of octamer-binding transcription factor 3/4 from the RORA promoter, subsequently suppressing the expression of aromatase and superoxide dismutase 2. The in vivo mouse study shows that prenatal RORA deficiency in neuron-specific RORA null mice mimics maternal diabetes-mediated ALB; postnatal RORA expression in the amygdala ameliorates, while postnatal RORA knockdown mimics, maternal diabetes-mediated ALB in offspring. In addition, RORA mRNA levels in peripheral blood mononuclear cells decrease to 34.2% in ASD patients (n = 121) compared to the typically developing group (n = 118), and the related Receiver Operating Characteristic curve shows good sensitivity and specificity with a calculated 84.1% of Area Under the Curve for ASD diagnosis. We conclude that maternal diabetes contributes to ALB in offspring through suppression of RORA and aromatase, RORA expression in PBMC could be a potential marker for ASD screening.


Subject(s)
Aromatase/genetics , Autism Spectrum Disorder/genetics , Diabetes, Gestational/enzymology , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Animals , Aromatase/metabolism , Disease Models, Animal , Female , Mice , Pregnancy
2.
PLoS One ; 16(11): e0248034, 2021.
Article in English | MEDLINE | ID: mdl-34752458

ABSTRACT

Retinoic acid receptor-related orphan nuclear receptor (ROR) γt is a member of the RORC nuclear hormone receptor family of transcription factors. RORγt functions as a critical regulator of thymopoiesis and immune responses. RORγt is expressed in multiple immune cell populations including Th17 cells, where its primary function is regulation of immune responses to bacteria and fungi through IL-17A production. However, excessive IL-17A production has been linked to numerous autoimmune diseases. Moreover, Th17 cells have been shown to elicit both pro- and anti-tumor effects. Thus, modulation of the RORγt/IL-17A axis may represent an attractive therapeutic target for the treatment of autoimmune disorders and some cancers. Herein we report the design, synthesis and characterization of three selective allosteric RORγt inhibitors in preclinical models of inflammation and tumor growth. We demonstrate that these compounds can inhibit Th17 differentiation and maintenance in vitro and Th17-dependent inflammation and associated gene expression in vivo, in a dose-dependent manner. Finally, RORγt inhibitors were assessed for efficacy against tumor formation. While, RORγt inhibitors were shown to inhibit tumor formation in pancreatic ductal adenocarcinoma (PDAC) organoids in vitro and modulate RORγt target genes in vivo, this activity was not sufficient to delay tumor volume in a KP/C human tumor mouse model of pancreatic cancer.


Subject(s)
Gene Expression/drug effects , Inflammation/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Th17 Cells/drug effects , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Inflammation/metabolism , Interleukin-17/metabolism , Mice , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Th17 Cells/metabolism
3.
Cell Death Dis ; 12(10): 886, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34584074

ABSTRACT

Osteoarthritis (OA) is characterized by cartilage destruction, chronic inflammation, and local pain. Evidence showed that retinoic acid receptor-related orphan receptor-α (RORα) is crucial in cartilage development and OA pathogenesis. Here, we investigated the role and molecular mechanism of RORα, an important member of the nuclear receptor family, in regulating the development of OA pathologic features. Investigation into clinical cartilage specimens showed that RORα expression level is positively correlated with the severity of OA and cartilage damage. In an in vivo OA model induced by anterior crucial ligament transaction, intra-articular injection of si-Rora adenovirus reversed the cartilage damage. The expression of cartilage matrix components type II collagen and aggrecan were elevated upon RORα blockade. RNA-seq data suggested that the IL-6/STAT3 pathway is significantly downregulated, manifesting the reduced expression level of both IL-6 and phosphorylated STAT3. RORα exerted its effect on IL-6/STAT3 signaling in two different ways, including interaction with STAT3 and IL-6 promoter. Taken together, our findings indicated the pivotal role of the RORα/IL-6/STAT3 axis in OA progression and confirmed that RORα blockade improved the matrix catabolism in OA chondrocytes. These results may provide a potential treatment target in OA therapy.


Subject(s)
Cartilage, Articular/pathology , Interleukin-6/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction , Aged , Animals , Base Sequence , Benzamides/chemistry , Benzamides/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Down-Regulation/drug effects , Female , Fluorocarbons/chemistry , Fluorocarbons/pharmacology , Humans , Interleukin-6/genetics , Male , Mice, Inbred C57BL , Models, Biological , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Osteoarthritis/genetics , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Severity of Illness Index , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thiophenes/chemistry , Thiophenes/pharmacology
4.
Am J Physiol Cell Physiol ; 321(1): C82-C93, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34038245

ABSTRACT

The resolution of inflammation is closely linked with tissue repair. Recent studies have revealed that macrophages suppress inflammatory reactions by producing lipid mediators, called specialized proresolving mediators (SPMs); however, the biological significance of SPMs in tissue repair remains to be fully elucidated in the heart. In this study, we focused on maresin-1 (MaR1) and examined the reparative effects of MaR1 in cardiomyocytes. The treatment with MaR1 increased cell size in cultured neonatal rat cardiomyocytes. Since the expression of fetal cardiac genes was unchanged by MaR1, physiological hypertrophy was induced by MaR1. SR3335, an inhibitor of retinoic acid-related orphan receptor α (RORα), mitigated MaR1-induced cardiomyocyte hypertrophy, consistent with the recent report that RORα is one of MaR1 receptors. Importantly, in response to MaR1, cardiomyocytes produced IGF-1 via RORα. Moreover, MaR1 activated phosphoinositide 3-kinase (PI3K)/Akt signaling pathway and wortmannin, a PI3K inhibitor, or triciribine, an Akt inhibitor, abrogated MaR1-induced cardiomyocyte hypertrophy. Finally, the blockade of IGF-1 receptor by NVP-AEW541 inhibited MaR-1-induced cardiomyocyte hypertrophy as well as the activation of PI3K/Akt pathway. These data indicate that MaR1 induces cardiomyocyte hypertrophy through RORα/IGF-1/PI3K/Akt pathway. Considering that MaR1 is a potent resolving factor, MaR1 could be a key mediator that orchestrates the resolution of inflammation with myocardial repair.


Subject(s)
Cardiomegaly/genetics , Cardiotonic Agents/pharmacology , Docosahexaenoic Acids/adverse effects , Insulin-Like Growth Factor I/genetics , Myocardial Infarction/genetics , Myocytes, Cardiac/drug effects , Paracrine Communication/genetics , Animals , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Disease Models, Animal , Docosahexaenoic Acids/antagonists & inhibitors , Gene Expression Regulation , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/chemically induced , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Paracrine Communication/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rats , Ribonucleosides/pharmacology , Signal Transduction , Sulfonamides/pharmacology , Thiophenes/pharmacology , Wortmannin/pharmacology
5.
Front Immunol ; 12: 647329, 2021.
Article in English | MEDLINE | ID: mdl-33767712

ABSTRACT

The transcription factor Related Orphan Receptor Alpha (RORα) plays an important role in regulating circadian rhythm, inflammation, metabolism and cellular development. Herein we show that in the absence of functional RORα in mice there is reduced susceptibility to LPS-induced endotoxic shock, with selective decreases in release of pro-inflammatory cytokines. Treatment of mice with a RORα selective synthetic inhibitor also reduced the severity of LPS-induced endotoxemia. The reduction in responses in Rora deficient mice was associated with an alterations in metabolic and pro-inflammatory functions of macrophages, both in vivo peritoneal macrophages and in vitro generated bone marrow derived macrophages. Using LysMCreRorafl/sg mice the reduced susceptibility to LPS was shown to be specific to Rora expression in the macrophages. This study identifies that Rora-mediated regulation of macrophages impacts on the pro-inflammatory responses elicited by LPS.


Subject(s)
Lipopolysaccharides/adverse effects , Macrophage Activation/genetics , Macrophages, Peritoneal/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/deficiency , Shock, Septic/chemically induced , Shock, Septic/immunology , Animals , Cells, Cultured , Chemokines/metabolism , Disease Models, Animal , Female , Inflammation/genetics , Inflammation/metabolism , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Shock, Septic/drug therapy , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfonamides/administration & dosage , Thiophenes/administration & dosage , Treatment Outcome
6.
Nat Commun ; 12(1): 76, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397953

ABSTRACT

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


Subject(s)
Inflammation/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Th17 Cells/immunology , Animals , Autoimmunity/drug effects , Body Weight/drug effects , Chronic Disease , Colon/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , HEK293 Cells , Humans , Inflammation/genetics , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Organ Size/drug effects , Severity of Illness Index , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thiophenes/chemistry , Thiophenes/pharmacology
7.
Prog Lipid Res ; 78: 101034, 2020 04.
Article in English | MEDLINE | ID: mdl-32360520

ABSTRACT

Maresin-1, a pro-resolving lipid mediator, is drawing a great deal of attention in receptor pharmacology, largely because two distinct types of receptor molecules have been reported as the targets of maresin-1. One is retinoic acid-related orphan receptor α (RORα) and the other is leucine-rich repeat domain-containing G protein-coupled receptor 6 (LGR6). RORα is a nuclear receptor and LGR6 is a plasma membrane GPCR. Identification of two different molecular targets raises the following question: What are the pro-resolving functions of each receptor in inflammation resolution, host defense, tissue homeostasis, and wound healing? In this article, I review the new targets from the point of view of pharmacology and maresin-1 resolution along with intracellular signaling molecules.


Subject(s)
Docosahexaenoic Acids/pharmacology , Inflammation Mediators/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Docosahexaenoic Acids/chemistry , Humans , Inflammation Mediators/chemistry , Mice , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
8.
Sci Rep ; 9(1): 11798, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409825

ABSTRACT

Acute liver failure (ALF) is characterized by loss of liver function in response to sustained augmentation of the acute-phase response (APR) in the liver, which can progress even to death. Although the inflammatory interleukin-6 (IL-6)-axis is a crucial factor that drives the hepatic APR by releasing diverse acute-phase proteins (APPs), therapeutic strategies to block the IL-6-STAT3-mediated APR are not well developed. Here, we show that the nuclear receptor retinoic acid-related orphan receptor α (RORα) limits APR-mediated liver injury by inhibiting the hepatic IL-6-STAT3 signaling pathway. Administration of JC1-40, an RORα activator, diminished diethylnitrosamine-induced acute liver injury and repressed transcriptional expression of APPs such as CXCL1 and LCN2 in mice. IL-6-mediated activation of STAT3 was repressed after RORα activation by either adenoviral infusion of RORα or JC1-40 treatment in primary hepatocytes. Activation of RORα decreased transcriptional expression of IL-6 receptor α, an upstream activator of STAT3, both in vitro and in vivo. This may be one mechanism underlying the RORα-mediated inhibition of STAT3. Taken together, our results suggest that RORα is a regulator of the hepatic IL-6-STAT3 signaling pathway and may be a new therapeutic target for treating APR-associated inflammatory ALF.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Interleukin-6/genetics , Liver Failure, Acute/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , STAT3 Transcription Factor/genetics , Acute-Phase Reaction/genetics , Adenoviridae/genetics , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Chemokine CXCL1/genetics , Deoxyuracil Nucleotides/pharmacology , Diethylnitrosamine/toxicity , Disease Models, Animal , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Humans , Hydroxycholesterols/pharmacology , Liver/injuries , Liver/metabolism , Liver/pathology , Liver Failure, Acute/genetics , Liver Failure, Acute/pathology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Primary Cell Culture , Signal Transduction/drug effects
9.
Bioorg Med Chem Lett ; 27(7): 1608-1610, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28242276

ABSTRACT

Targeting the transcriptional activity of nuclear hormone receptors has proven an effective strategy to treat certain human diseases, and they have become a major focus point to develop novel therapies for the treatment of cancer, inflammation, autoimmune diseases, metabolic disorders, and others. One family of nuclear receptors that has attracted most interest in recent years is the retinoic acid receptor-related orphan receptors (RORs), in particular RORγ. RORγ is a critical regulator of the immune system and RORγ antagonists have shown activity in animal models of inflammatory autoimmune diseases. Here we present the synthesis and biological evaluation of dihydroimidazole tethered imidazolinethiones. We have identified several dual RORγ/α and pan-ROR antagonists with significant activity in cellular assays that could serve as starting points for future optimization efforts to generate potent and selective RORγ modulators.


Subject(s)
Imidazolines/pharmacology , Orphan Nuclear Receptors/antagonists & inhibitors , Thiones/pharmacology , Animals , CHO Cells , Cricetulus , Imidazolines/chemical synthesis , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 2/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Thiones/chemical synthesis
10.
Sci Rep ; 7: 39873, 2017 01 04.
Article in English | MEDLINE | ID: mdl-28051153

ABSTRACT

In breast cancer metastasis, the dynamic continuum involving pro- and anti-inflammatory regulators can become compromised. Over 600 genes have been implicated in metastasis to bone, lung or brain but how these genes might contribute to perturbation of immune function is poorly understood. To gain insight, we adopted a gene co-expression network approach that draws on the functional parallels between naturally occurring bone marrow-derived mesenchymal stem cells (BM-MSCs) and cancer stem cells (CSCs). Our network analyses indicate a key role for metastasis suppressor RARRES3, including potential to regulate the immunoproteasome (IP), a specialized proteasome induced under inflammatory conditions. Knockdown of RARRES3 in near-normal mammary epithelial and breast cancer cell lines increases overall transcript and protein levels of the IP subunits, but not of their constitutively expressed counterparts. RARRES3 mRNA expression is controlled by interferon regulatory factor IRF1, an inducer of the IP, and is sensitive to depletion of the retinoid-related receptor RORA that regulates various physiological processes including immunity through modulation of gene expression. Collectively, these findings identify a novel regulatory role for RARRES3 as an endogenous inhibitor of IP expression, and contribute to our evolving understanding of potential pathways underlying breast cancer driven immune modulation.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Receptors, Retinoic Acid/metabolism , Bone Marrow Cells/cytology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Neoplasm Metastasis/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics
11.
J Pineal Res ; 62(3)2017 Apr.
Article in English | MEDLINE | ID: mdl-27862268

ABSTRACT

Diabetic cardiomyopathy is a major complication that significantly contributes to morbidity and mortality in diabetics with few therapies. Moreover, antidiabetic drugs reported inconsistent or even adverse cardiovascular effects, suggesting that it is important to exploit novel therapeutic targets against diabetic cardiomyopathy. Here, we observed that the nuclear melatonin receptor, the retinoic acid-related orphan receptor-α (RORα), was downregulated in diabetic hearts. By utilizing a mouse line with RORα disruption, we demonstrated that RORα deficiency led to significantly augmented diastolic dysfunction and cardiac remodeling induced by diabetes. Microscopic and molecular analyses further indicated that the detrimental effects of RORα deficiency were associated with aggravated myocardial apoptosis, autophagy dysfunction, and oxidative stress by disrupting antioxidant gene expression. By contrast, restoration of cardiac RORα levels in transgenic mice significantly improved cardiac functional and structural parameters at 8 weeks after diabetes induction. Consistent with genetic manipulation, pharmacological activation of RORα by melatonin and SR1078 (a synthetic agonist) showed beneficial effects against diabetic cardiomyopathy, while the RORα inhibitor SR3335 significantly exacerbated cardiac impairments in diabetic mice. Collectively, these findings suggest that cardiac-targeted manipulation of nuclear melatonin receptor RORα may hold promise for delaying diabetic cardiomyopathy development.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Myocardium/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Receptors, Melatonin/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Autophagy/genetics , Benzamides/pharmacology , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Mice , Mice, Mutant Strains , Myocardium/pathology , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Receptors, Melatonin/genetics , Sulfonamides/pharmacology , Thiophenes/pharmacology
12.
J Pathol ; 237(1): 111-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25953430

ABSTRACT

Genetic studies have added to the understanding of complex diseases. Here, we used a combined genetic approach for risk-loci identification in a prototypic, organ-specific, autoimmune disease, namely experimental epidermolysis bullosa acquisita (EBA), in which autoantibodies to type VII collagen (COL7) and neutrophil activation cause mucocutaneous blisters. Anti-COL7 IgG induced moderate blistering in most inbred mouse strains, while some showed severe disease or were completely protected. Using publicly available genotyping data, we identified haplotype blocks that control blistering and confirmed two haplotype blocks in outbred mice. To identify the blistering-associated genes, haplotype blocks encoding genes that are differentially expressed in EBA-affected skin were considered. This procedure identified nine genes, including retinoid-related orphan receptor alpha (RORα), known to be involved in neurological development and function. After anti-COL7 IgG injection, RORα+/- mice showed reduced blistering and homozygous mice were completely resistant to EBA induction. Furthermore, pharmacological RORα inhibition dose-dependently blocked reactive oxygen species (ROS) release from activated neutrophils but did not affect migration or phagocytosis. Thus, forward genomics combined with multiple validation steps identifies RORα to be essential to drive inflammation in experimental EBA.


Subject(s)
Epidermolysis Bullosa Acquisita/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Skin/metabolism , Animals , Autoantibodies/immunology , Collagen Type VII/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Partial Agonism , Epidermolysis Bullosa Acquisita/genetics , Epidermolysis Bullosa Acquisita/immunology , Epidermolysis Bullosa Acquisita/pathology , Genetic Predisposition to Disease , Genomics/methods , Haplotypes , Heterozygote , Homozygote , Immunoglobulin G/immunology , Mice, Knockout , Neutrophil Activation , Neutrophils/immunology , Neutrophils/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/immunology , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction , Skin/drug effects , Skin/immunology , Skin/pathology , Species Specificity , Sulfonamides/pharmacology , Thiophenes/pharmacology , Time Factors
14.
J Nutr Biochem ; 26(2): 112-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25459887

ABSTRACT

Circadian rhythm plays an important role in maintaining homeostasis, and its disruption increases the risk of developing metabolic syndrome. Circadian rhythm is maintained by a central clock in the hypothalamus that is entrained by light, but circadian clocks are also present in peripheral tissues. These peripheral clocks are trained by other cues, such as diet. The aim of this study was to determine whether proanthocyanidins, the most abundant polyphenols in the human diet, modulate the expression of clock and clock-controlled genes in the liver, gut and mesenteric white adipose tissue (mWAT) in healthy and obese rats. Grape seed proanthocyanidin extracts (GSPEs) were administered for 21 days at 5, 25 or 50 mg GSPE/kg body weight in healthy rats and 25 mg GSPE/kg body weight in rats with diet-induced obesity. In healthy animals, GSPE administration led to the overexpression of core clock genes in a positive dose-dependent manner. Moreover, the acetylated BMAL1 protein ratio increased with the same pattern in the liver and mWAT. With regards to clock-controlled genes, Per2 was also overexpressed, whereas Rev-erbα and RORα were repressed in a negative dose-dependent manner. Diet-induced obesity always resulted in the overexpression of some core clock and clock-related genes, although the particular gene affected was tissue specific. GSPE administration counteracted disturbances in the clock genes in the liver and gut but was less effective in normalizing the clock gene disruption in WAT. In conclusion, proanthocyanidins have the capacity to modulate peripheral molecular clocks in both healthy and obese states.


Subject(s)
Chronobiology Disorders/prevention & control , Dietary Supplements , Gene Expression Regulation , Grape Seed Extract/therapeutic use , Obesity/diet therapy , Period Circadian Proteins/metabolism , Peripheral Nervous System Diseases/prevention & control , Proanthocyanidins/therapeutic use , ARNTL Transcription Factors/agonists , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Chronobiology Disorders/etiology , Duodenum/metabolism , Grape Seed Extract/administration & dosage , Hyperlipidemias/etiology , Hyperlipidemias/prevention & control , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/therapeutic use , Intestinal Mucosa/metabolism , Intra-Abdominal Fat/metabolism , Liver/metabolism , Male , Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Obesity/metabolism , Obesity/physiopathology , Organ Specificity , Period Circadian Proteins/agonists , Period Circadian Proteins/antagonists & inhibitors , Period Circadian Proteins/genetics , Peripheral Nervous System Diseases/etiology , Proanthocyanidins/administration & dosage , Random Allocation , Rats, Wistar
15.
Am J Physiol Cell Physiol ; 308(5): C385-96, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25500738

ABSTRACT

Lipid metabolic disturbances are related to many diseases, such as obesity, diabetes, and certain cancers. Notably, lipid metabolic disturbances have been reported to be a risk factor for colorectal cancer. Nuclear receptors act as ligand-dependent transcription regulators and play key roles in the regulation of body lipid metabolism and the development of many cancers. Retinoic acid receptor-related orphan receptor α (RORα) is a nuclear receptor and can regulate several lipid metabolism genes in certain cancers. Herein, we demonstrate that the conditioned medium from adipocytes has a proproliferative and promigratory effect on colorectal cancer cells and enhances angiogenesis in chicken embryonic chorioallantoic membranes. In addition, the conditioned medium leads to a decrease in the expression of RORα and its target genes. Meanwhile, RORα and its target gene expressions are lower in human colorectal tumor tissue compared with control colorectal tissue. Activation of RORα inhibits the effect of conditioned medium on the proliferation and migration of colorectal cancer cells as well as the angiogenesis in chicken embryonic allantoic membranes. In colorectal cancer cells, the putative ligand of RORα, cholesterol sulfate (CS), prevents cell cycle progression at the G1/S boundary and concurrently modulates the expression of cell cycle-regulatory genes in colorectal cancer cell. CS inhibits angiogenesis in chicken embryonic chorioallantoic membranes and concurrently decreases the mRNA expression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α as well as the secretion of VEGF. In addition, lipogenic gene expression is higher in human colorectal tumor tissue compared with control colorectal tissue. CS inhibits the expression of lipogenic genes in colorectal cancer cells. These results suggest that RORα could represent a direct link between local lipid metabolism of colorectal tissue and colorectal cancer. Therefore, the reduction of the expression of RORα could represent a potential warning sign of colorectal cancer.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , Chorioallantoic Membrane/physiology , Colorectal Neoplasms/metabolism , Culture Media, Conditioned/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , 3T3 Cells , Adipocytes/physiology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Colorectal Neoplasms/pathology , Humans , Mice , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors
16.
J Mol Cell Biol ; 6(4): 338-48, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24831657

ABSTRACT

Prostaglandin E2 (PGE2) is known to have a key role in the development of colorectal cancer, but previous experiments showed its contrasting (i.e. tumor-promoting or tumor-suppressive) roles depending on experimental conditions. To elucidate the mechanisms underlying such contrasting roles of PGE2 in tumorigenesis, we investigated all the previous experiments and found a new signal transduction pathway mediated by retinoic acid receptor-related orphan receptor (ROR)α, in which PGE2/PKCα-dependent phosphorylation of RORα attenuates Wnt target gene expression in colon cancer cells. From mathematical simulations combined with biochemical experimentation, we revealed that RORα induces a biphasic response of Wnt target genes to PGE2 stimulation through a regulatory switch formed by an incoherent feedforward loop, which provides a mechanistic explanation on the contrasting roles of PGE2 observed in previous experiments. More interestingly, we found that RORα constitutes another regulatory switch formed by coupled positive and negative feedback loops, which regulates the hysteretic response of Wnt signaling and eventually converts a proliferative cellular state into an anti-proliferative state in a very delicate way. Our results indicate that RORα is the key regulator at the center of these hidden switches that critically regulate cancer cell proliferation and thereby being a promising anti-cancer therapeutic target.


Subject(s)
Cell Proliferation , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Protein Kinase C-alpha/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Blotting, Western , Chromatin Immunoprecipitation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Dinoprostone/metabolism , Feedback, Physiological , Humans , Models, Theoretical , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Phosphorylation , Protein Kinase C-alpha/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Wnt Proteins/genetics , beta Catenin/genetics
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 34(4): 511-8, 2014 Apr.
Article in Chinese | MEDLINE | ID: mdl-24752099

ABSTRACT

OBJECTIVE: Psoriasis is an autoimmune-related chronic inflammatory skin disease strongly associated with the dysfunction of Th17 cells. Retinoic acid-related orphan nuclear receptor γt (RORγt) plays a critical role in the differentiation and maturation of Th17 cells and in cell-derived immunologic derangement. We conducted this study to investigate potential mechanism by which the derivative of digoxin selectively antagonizes RORγt transcriptional activity. METHOD: Using molecular docking in combination with molecular electrostatic potential (MEP), we detected the interaction between the derivative of digoxin (Dhd) and ROR transcription factor (RORα,RORß and RORγt), and the results were further confirmed by bioluminescent assay. RESULT: Molecular docking demonstrated that Dhd could exclusively inhibit the conformation of RORγt; bioluminescent assay further indicated that RORγt was selectively antagonized by Dhd in a dose- and time-dependent manner. CONCLUSION: Dhd can selectively suppress RORγt transcriptional activity.


Subject(s)
Digoxin/pharmacology , Models, Chemical , Molecular Docking Simulation , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Digoxin/analogs & derivatives , Humans , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Transcription, Genetic
18.
Nat Rev Drug Discov ; 13(3): 197-216, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24577401

ABSTRACT

The nuclear receptors REV-ERB (consisting of REV-ERBα and REV-ERBß) and retinoic acid receptor-related orphan receptors (RORs; consisting of RORα, RORß and RORγ) are involved in many physiological processes, including regulation of metabolism, development and immunity as well as the circadian rhythm. The recent characterization of endogenous ligands for these former orphan nuclear receptors has stimulated the development of synthetic ligands and opened up the possibility of targeting these receptors to treat several diseases, including diabetes, atherosclerosis, autoimmunity and cancer. This Review focuses on the latest developments in ROR and REV-ERB pharmacology indicating that these nuclear receptors are druggable targets and that ligands targeting these receptors may be useful in the treatment of several disorders.


Subject(s)
Drug Delivery Systems/methods , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Drug Delivery Systems/trends , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group D, Member 1/chemistry , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
19.
Trends Endocrinol Metab ; 23(12): 619-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22789990

ABSTRACT

The retinoic-acid-receptor-related orphan receptors (RORs) are members of the nuclear receptor (NR) superfamily whose activity has been implicated in several physiological and pathological processes. The RORs, specifically RORα and RORγ, are considered to be master regulators of T(H)17 cells, a recently described subset of CD4(+) T helper cells that have been demonstrated to have a pathological role in autoimmune disease. As with most members of the NR superfamily, RORs are ligand-regulated, suggesting that their activity can be modulated by synthetic ligands. Recent advances in the field have established that selective inhibition of the RORs is a viable therapeutic approach for not only the treatment of autoimmune disorders but also ROR-mediated metabolic disorders.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 1/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/physiology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Circadian Rhythm/physiology , Humans , Ligands , Metabolic Diseases/physiopathology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...