Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Mol Biol Cell ; 31(8): 793-802, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32023146

ABSTRACT

OSBPL1 encodes the full-length oxysterol-binding protein-related protein ORP1L, which transports LDL-derived cholesterol at membrane contacts between the late endosomes/lysosomes (LEL) and the endoplasmic reticulum (ER). OSBPL1 also encodes the truncated variant ORP1S that contains only the C-terminal lipid binding domain. HeLa cells in which both variants were knocked out (ORP1-null) were used to determine the functional relationship between ORP1L and ORP1S with respect to cellular cholesterol localization and regulation. ORP1-null cells accumulated cholesterol in LEL and had reduced plasma membrane (PM) cholesterol. PM cholesterol was restored by expression of wild-type ORP1S or a phosphatidylinositol phosphate-binding mutant but not by a sterol-binding mutant. Expression of ORP2, another truncated variant, also restored PM cholesterol in ORP1-null cells. Consistent with a LEL-to-PM cholesterol transport activity, a small fraction of ORP1S was detected on the PM. As a consequence of reduced delivery of cholesterol to the PM in ORP1-null cells, cholesterol was diverted to the ER resulting in normalization of de novo cholesterol synthesis. The deficiency in PM cholesterol also reduced ABCA1-dependent cholesterol efflux and LDL receptor activity in ORP1-null cells. We conclude that ORP1S, which lacks discrete membrane-targeting motifs, transports cholesterol from LEL to the PM.


Subject(s)
Cholesterol/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Receptors, Steroid/metabolism , ATP Binding Cassette Transporter 1/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Amino Acid Motifs , Biological Transport , CRISPR-Cas Systems , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Intracellular Membranes/metabolism , Membrane Lipids/metabolism , Protein Domains , Protein Isoforms/metabolism , Receptors, LDL/metabolism , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Sequence Deletion
2.
Biochem Biophys Res Commun ; 521(1): 42-49, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31629475

ABSTRACT

In our previous study, Oxysterol-binding protein-related protein 2 (OSBPL2) was first identified as a new deafness-causative gene contribute to non-syndromic hearing loss. However, the underlying mechanism of OSBPL2-induced hearing loss remains unknown. Here, we used hearing-specific cells and tissues OC-1 cells and zebrafish inner ear tissues as models to identify common transcriptome changes in genes and pathways in the absence of human OSBPL2 orthologues by RNA-seq analysis. In total, 2112 differentially expressed genes (DEGs) were identified between wild-type (WT) and Osbpl2-/- OC-1 cells, and 877 DEGs were identified between WT and osbpl2b-/- zebrafish inner ear tissues. Functional annotation implicated Osbpl2/osbpl2b in lipid metabolism, cell adhesion and the extracellular matrix in both OC-1 cells and zebrafish inner ear tissues. Protein-protein interaction (PPI) analysis indicated that Osbpl2/osbpl2b were also involved in ubiquitination. Further experiments showed that Osbpl2-/- OC-1 cells exhibited an abnormal focal adhesion morphology characterized by inhibited FAK activity and impaired cell adhesion. In conclusion, we identified novel pathways modulated by OSBPL2 orthologues, providing new insight into the mechanism of hearing loss induced by OSBPL2 deficiency.


Subject(s)
Ear, Inner/metabolism , Receptors, Steroid/genetics , Animals , Cell Adhesion , Cells, Cultured , Ear, Inner/pathology , Extracellular Matrix/metabolism , Humans , Lipid Metabolism , Principal Component Analysis , Quality Control , Receptors, Steroid/deficiency , Sequence Analysis, RNA , Transcriptome , Zebrafish/genetics
3.
Proc Natl Acad Sci U S A ; 116(30): 15150-15159, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31285338

ABSTRACT

In response to microbial stimulation, monocytes can differentiate into macrophages or monocyte-derived dendritic cells (MoDCs) but the molecular requirements guiding these possible fates are poorly understood. In addition, the physiological importance of MoDCs in the host cellular and immune responses to microbes remains elusive. Here, we demonstrate that the nuclear orphan receptor NR4A3 is required for the proper differentiation of MoDCs but not for other types of DCs. Indeed, the generation of DC-SIGN+ MoDCs in response to LPS was severely impaired in Nr4a3-/- mice, which resulted in the inability to mount optimal CD8+ T cell responses to gram-negative bacteria. Transcriptomic analyses revealed that NR4A3 is required to skew monocyte differentiation toward MoDCs, at the expense of macrophages, and allows the acquisition of migratory characteristics required for MoDC function. Altogether, our data identify that the NR4A3 transcription factor is required to guide the fate of monocytes toward MoDCs.


Subject(s)
Cell Lineage/immunology , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Lipopolysaccharides/pharmacology , Monocytes/immunology , Nerve Tissue Proteins/genetics , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Differentiation , Cell Lineage/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-4/pharmacology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymphocyte Activation , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Knockout , Monocytes/cytology , Monocytes/drug effects , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/immunology , Primary Cell Culture , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Steroid/deficiency , Receptors, Steroid/immunology , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/immunology , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
4.
Nature ; 567(7749): 530-534, 2019 03.
Article in English | MEDLINE | ID: mdl-30814732

ABSTRACT

T cells expressing chimeric antigen receptors (CAR T cells) targeting human CD19 (hCD19) have shown clinical efficacy against B cell malignancies1,2. CAR T cells have been less effective against solid tumours3-5, in part because they enter a hyporesponsive ('exhausted' or 'dysfunctional') state6-9 triggered by chronic antigen stimulation and characterized by upregulation of inhibitory receptors and loss of effector function. To investigate the function of CAR T cells in solid tumours, we transferred hCD19-reactive CAR T cells into hCD19+ tumour-bearing mice. CD8+CAR+ tumour-infiltrating lymphocytes and CD8+ endogenous tumour-infiltrating lymphocytes expressing the inhibitory receptors PD-1 and TIM3 exhibited similar profiles of gene expression and chromatin accessibility, associated with secondary activation of nuclear receptor transcription factors NR4A1 (also known as NUR77), NR4A2 (NURR1) and NR4A3 (NOR1) by the initiating transcription factor NFAT (nuclear factor of activated T cells)10-12. CD8+ T cells from humans with cancer or chronic viral infections13-15 expressed high levels of NR4A transcription factors and displayed enrichment of NR4A-binding motifs in accessible chromatin regions. CAR T cells lacking all three NR4A transcription factors (Nr4a triple knockout) promoted tumour regression and prolonged the survival of tumour-bearing mice. Nr4a triple knockout CAR tumour-infiltrating lymphocytes displayed phenotypes and gene expression profiles characteristic of CD8+ effector T cells, and chromatin regions uniquely accessible in Nr4a triple knockout CAR tumour-infiltrating lymphocytes compared to wild type were enriched for binding motifs for NF-κB and AP-1, transcription factors involved in activation of T cells. We identify NR4A transcription factors as having an important role in the cell-intrinsic program of T cell hyporesponsiveness and point to NR4A inhibition as a promising strategy for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neoplasms/genetics , Neoplasms/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Chimeric Antigen/immunology , Transcription Factors/metabolism , Adoptive Transfer , Animals , Antigens, CD19/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Female , Gene Expression Profiling , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neoplasms/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 2/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptors, Steroid/deficiency , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/metabolism , Survival Rate , Transcription Factor AP-1/metabolism , Transcription Factors/deficiency
5.
BMC Cardiovasc Disord ; 17(1): 271, 2017 Oct 18.
Article in English | MEDLINE | ID: mdl-29047330

ABSTRACT

BACKGROUND: The NR4A3 orphan nuclear hormone receptor, NOR1, functions as a constitutively active transcription factor to regulate inflammation, proliferation, and cell survival during pathological vascular remodeling. Inflammatory processes represent key mechanisms leading to abdominal aortic aneurysm (AAA) formation. However, a role of NOR1 in AAA formation has not been investigated previously. METHODS: Inflammatory gene expression was analyzed in bone marrow-derived macrophages isolated from NOR1-deficient mice. Low-density lipoprotein receptor-deficient (LDLr-/-) mice were irradiated and reconstituted with hematopoietic stem cells obtained from NOR1-/- or wild-type littermate mice. Animals were infused with angiotensin II and fed a diet enriched in saturated fat to induce AAA formation. Quantification of AAA formation was performed by ultrasound and ex vivo measurements. RESULTS: Among 184 inflammatory genes that were analyzed, 36 genes were differentially regulated in LPS-treated NOR1-deficient macrophages. Albeit this difference in gene regulation, NOR1-deficiency in hematopoietic stem cells did not affect development of AAA formation in bone marrow-derived stem cell transplanted LDLr-deficient mice. CONCLUSION: NOR1 deletion induced differential inflammatory gene transcription in macrophages but did not influence AAA formation in mice.


Subject(s)
Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Deletion , Hematopoietic Stem Cells/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/genetics , Animals , Inflammation/genetics , Inflammation/metabolism , Inflammation/prevention & control , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Biochem Pharmacol ; 142: 194-203, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28756207

ABSTRACT

Obesity has become a significant global health problem, and is a high risk factor for a variety of metabolic diseases. Fibroblast growth factor (FGF) 15 plays an important role in the regulation of metabolism. Xenobiotic-sensing nuclear receptors pregnane X receptor (PXR/NR1I2) and constitutive androstane receptor (CAR/NR1I3) play important roles in xenobiotic detoxification and metabolism, and also are involved in the regulation of energy metabolism. However, the effects that PXR and CAR have on the regulation of FGF15 are unknown. Here, we found that body weight, hepatic triglyceride levels, liver steatosis, and hepatic mRNA expression levels of cholesterol 7α-hydroxylase (CYP7A1) and sterol 12α-hydroxylase (CYP8B1), the key enzymes in the bile acid classical synthesis pathway, were significantly decreased in high fat diet (HFD)-fed PXR knockout (KO) mice compared to HFD-fed wild-type mice. Interestingly, intestinal FGF15 expression levels were significantly elevated in HFD-fed PXR KO mice compared with HFD-fed wild-type mice. Additionally, serum total bile acid levels were significantly decreased in PXR KO mice than those in wild-type mice when fed a control diet or HFD. Total lipids in feces were significantly increased in HFD-fed PXR KO mice compared to HFD-fed wild-type mice. However, these alterations were not found in HFD-fed CAR KO mice. These results indicate that PXR deficiency improves HFD-induced obesity via induction of FGF15 expression, resulting in suppression of bile acid synthesis and reduction of lipid absorption, hepatic lipid accumulation and liver triglyceride levels. Our findings suggest that PXR may negatively regulate FGF15 expression and represent a potential therapeutic target for the treatment for metabolic disorders such as obesity.


Subject(s)
Diet, High-Fat/adverse effects , Fibroblast Growth Factors/genetics , Gene Expression , Obesity/metabolism , Receptors, Steroid/metabolism , Animals , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/blood , Body Weight , Constitutive Androstane Receptor , Feces/chemistry , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/genetics , Organ Size , Pregnane X Receptor , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Triglycerides/blood
7.
Toxicol Lett ; 276: 138-143, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28495613

ABSTRACT

In a National Toxicology Program (NTP) bioassay, inhalation of tetrahydrofuran (THF) induced liver tumors in female B6C3F1 mice but not in male mice or rats of either sex. Since THF is not genotoxic, the NTP concluded this carcinogenic activity was likely mediated via non-genotoxic modes of action (MOA). Based on evidence that THF and phenobarbital share a similar MOA, female Car/Pxr knock-out mice were orally exposed to THF to evaluate the potential role of CAR activation in the MOA for THF-induced liver tumors. Because data from this oral study with Car/Pxr knock-out mice (C57Bl/6) and the inhalation studies with wild type mice (B6C3F1) reported by NTP and others were derived from different strains, oral studies with wild type B6C3F1 and C57Bl/6 mice were conducted to ensure THF responses in both strains were comparable. As seen in inhalation studies with THF, oral exposure of wild type female mice to a maximum tolerated dose of THF increased total P450 content, CAR-related P450 activities, and hepatocyte proliferation; these effects were not observed in Car/Pxr knock-out female mice. This finding supports the hypothesis THF-induced carcinogenicity is likely mediated via CAR activation that has limited, if any, relevance to humans.


Subject(s)
Carcinogens/toxicity , Furans/toxicity , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Administration, Oral , Animals , Carcinogens/administration & dosage , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Female , Furans/administration & dosage , Genotype , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Maximum Tolerated Dose , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnane X Receptor , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Risk Assessment , Species Specificity
8.
Br J Pharmacol ; 174(8): 700-717, 2017 04.
Article in English | MEDLINE | ID: mdl-28128444

ABSTRACT

BACKGROUND AND PURPOSE: Imatinib mesylate (IM) is a first-line treatment for chronic myeloid leukaemia (CML) as a specific inhibitor of BCR-ABL tyrosine kinase. As IM is widely used in CML, in combination with other drugs, the effects of IM on drug-metabolizing enzymes (DMEs) are crucial to the design of rational drug administration. Carboxylesterases (CESs) are enzymes catalysing the hydrolytic biotransformation of several clinically useful drugs. Although IM is known to inhibit cytochromes P450 (CYPs), its effects on DMEs, and CESs in particular, are still largely undefined. EXPERIMENTAL APPROACH: Hepatoma cell lines (HepG2 and Huh7) and primary mouse hepatocytes were used. mRNA and protein expression were evaluated by quantitative RT-PCR and Western blot analysis. Reporter luciferase activity was determined by transient co-transfection experiment. Pregnane X receptor (PXR) expression was regulated by overexpression and RNA interference. The activity of CESs was determined by enzymic and toxicological assays. Mice were treated with a range of doses of IM to analyse expression of CESs in mouse liver. KEY RESULTS: The expression and activity of CESs were markedly repressed by IM, along with the down-regulation of PXR and inhibited expression and activity of CYP3A4 and P-gp. CONCLUSIONS AND IMPLICATIONS: Down-regulation of PXR mediates IM-induced suppression of CESs. IM may inhibit expression of other genes targeted by PXR, thus inducing a wide range of potential drug-drug interactions during treatment of CML. The data deserve further elucidation including clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Carboxylic Ester Hydrolases/antagonists & inhibitors , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Imatinib Mesylate/pharmacology , Receptors, Steroid/biosynthesis , Receptors, Steroid/deficiency , Animals , Antineoplastic Agents/administration & dosage , Carboxylic Ester Hydrolases/metabolism , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Humans , Imatinib Mesylate/administration & dosage , Male , Mice , Mice, Inbred ICR , Pregnane X Receptor , Receptors, Steroid/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
9.
PLoS One ; 10(3): e0119177, 2015.
Article in English | MEDLINE | ID: mdl-25749104

ABSTRACT

Steroid and xenobiotic receptor (SXR) and its murine ortholog, pregnane X receptor (PXR), are nuclear receptors that are expressed at high levels in the liver and the intestine where they function as xenobiotic sensors that induce expression of genes involved in detoxification and drug excretion. Recent evidence showed that SXR and PXR are also expressed in bone tissue where they mediate bone metabolism. Here we report that systemic deletion of PXR results in aging-dependent wearing of articular cartilage of knee joints. Histomorphometrical analysis showed remarkable reduction of width and an enlarged gap between femoral and tibial articular cartilage in PXR knockout mice. We hypothesized that genes induced by SXR in chondrocytes have a protective effect on articular cartilage and identified Fam20a (family with sequence similarity 20a) as an SXR-dependent gene induced by the known SXR ligands, rifampicin and vitamin K2. Lastly, we demonstrated the biological significance of Fam20a expression in chondrocytes by evaluating osteoarthritis-related gene expression of primary articular chondrocytes. Consistent with epidemiological findings, our results indicate that SXR/PXR protects against aging-dependent wearing of articular cartilage and that ligands for SXR/PXR have potential role in preventing osteoarthritis caused by aging.


Subject(s)
Aging/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Osteoarthritis, Knee/metabolism , Receptors, Steroid/deficiency , Aging/genetics , Aging/pathology , Animals , Cartilage, Articular/pathology , Chondrocytes/pathology , Dental Enamel Proteins , Knee Joint/metabolism , Knee Joint/pathology , Mice , Mice, Knockout , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Pregnane X Receptor , Proteins/genetics , Proteins/metabolism
10.
Pharmacol Rep ; 67(2): 299-304, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25712654

ABSTRACT

BACKGROUND: The antiepileptic drug carbamazepine (CBZ) is a typical inducer of cytochrome P450 (CYP) 3A and 2C in the clinic. It is considered a strong constitutive androstane receptor activator, however both CBZ and its main metabolite CBZ 10, 11-epoxide have been reported to be pregnane X receptor (PXR) activators whose maximal efficacy and potency are comparable with the human PXR ligand rifampicin. It is unknown whether or not PXR plays a substantially important role in in vivo induction of CYP by CBZ administration. METHODS: In this study, wild type and Pxr-/- mice were administered with CBZ for 5 days. Serum and liver samples were collected and subjected to hepatotoxicity assessment and CYP induction analysis. RESULTS: CYP2b, 2c and 3a were induced similarly in terms of transcription level, enzyme activity and protein abundance in both wild type and Pxr-/- mice. Inductive profile of CYPs in mice by CBZ administration accorded with those reported in rats, but differed from clinically reported data. CONCLUSIONS: These data suggest that in vivo induction of CYP in mice by multiple administration of CBZ is independent of PXR. Knowledge of the featured CYP induction profile in mice helps us understand species related CYP induction profiles among rodents and humans resulting from administration of CBZ.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Carbamazepine/pharmacology , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/metabolism , Liver/drug effects , Membrane Proteins/biosynthesis , Receptors, Steroid/deficiency , Steroid Hydroxylases/biosynthesis , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Carbamazepine/blood , Carbamazepine/pharmacokinetics , Cytochrome P450 Family 2 , Enzyme Induction/drug effects , Liver/enzymology , Male , Mice , Mice, Knockout , Pregnane X Receptor , Receptors, Steroid/genetics
11.
PLoS One ; 9(10): e109024, 2014.
Article in English | MEDLINE | ID: mdl-25347070

ABSTRACT

INTRODUCTION: Oxysterol binding protein Related Proteins (ORPs) mediate intracellular lipid transport and homeostatic regulation. ORP8 downregulates ABCA1 expression in macrophages and cellular cholesterol efflux to apolipoprotein A-I. In line, ORP8 knockout mice display increased amounts of HDL cholesterol in blood. However, the role of macrophage ORP8 in atherosclerotic lesion development is unknown. METHODS AND RESULTS: LDL receptor knockout (KO) mice were transplanted with bone marrow (BM) from ORP8 KO mice and C57Bl/6 wild type mice. Subsequently, the animals were challenged with a high fat/high cholesterol Western-type diet to induce atherosclerosis. After 9 weeks of Western-Type diet feeding, serum levels of VLDL cholesterol were increased by 50% in ORP8 KO BM recipients compared to the wild-type recipients. However, no differences were observed in HDL cholesterol. Despite the increase in VLDL cholesterol, lesions in mice transplanted with ORP8 KO bone marrow were 20% smaller compared to WT transplanted controls. In addition, ORP8 KO transplanted mice displayed a modest increase in the percentage of macrophages in the lesion as compared to the wild-type transplanted group. ORP8 deficient macrophages displayed decreased production of pro-inflammatory factors IL-6 and TNFα, decreased expression of differentiation markers and showed a reduced capacity to form foam cells in the peritoneal cavity. CONCLUSIONS: Deletion of ORP8 in bone marrow-derived cells, including macrophages, reduces lesion progression after 9 weeks of WTD challenge, despite increased amounts of circulating pro-atherogenic VLDL. Reduced macrophage foam cell formation and lower macrophage inflammatory potential are plausible mechanisms contributing to the observed reduction in atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Cells/metabolism , Receptors, LDL/deficiency , Receptors, Steroid/deficiency , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Atherosclerosis/therapy , Biomarkers , Cholesterol/blood , Cholesterol/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Foam Cells/pathology , Gene Expression , Inflammation Mediators/metabolism , Leukocyte Count , Macrophages/metabolism , Male , Mice , Mice, Knockout , Mustard Gas , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Receptors, LDL/genetics , Receptors, Steroid/genetics , Time Factors , Triglycerides/blood
12.
FEBS Lett ; 588(21): 3947-53, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25240193

ABSTRACT

We show that testicular orphan nuclear receptor 4 (TR4) increases the expression of pyruvate carboxylase (PC) gene in 3T3-L1 adipocytes by direct binding to a TR4 responsive element in the murine PC promoter. While TR4 overexpression increased PC activity, oxaloacetate (OAA) and glycerol levels with enhanced incorporation of (14)C from (14)C-pyruvate into fatty acids in 3T3-L1 adipocytes, PC knockdown by short interfering RNA (siRNA) or inhibition of PC activity by phenylacetic acid (PAA) abolished TR4-enhanced fatty acid synthesis. Moreover, TR4 microRNA reduced PC expression with decreased fatty acid synthesis in 3T3-L1 adipocytes, suggesting that TR4-mediated enhancement of fatty acid synthesis in adipocytes requires increased expression of PC gene.


Subject(s)
Adipocytes/metabolism , Fatty Acids/biosynthesis , Gene Expression Regulation, Enzymologic , Pyruvate Carboxylase/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , 3T3-L1 Cells , Animals , Gene Knockdown Techniques , Lipogenesis , Mice , Promoter Regions, Genetic/genetics , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/genetics
13.
Stem Cells ; 32(9): 2419-29, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24806827

ABSTRACT

The NR4A orphan nuclear receptor NOR1 functions as a constitutively active transcription factor regulating cellular inflammation and proliferation. In this study, we used bone marrow transplantation to determine the selective contribution of NOR1 expression in hematopoietic stem cells to the development of atherosclerosis. Reconstitution of lethally irradiated apoE(-/-) mice with NOR1-deficient hematopoietic stem cells accelerated atherosclerosis formation and macrophage recruitment following feeding a diet enriched in saturated fat. NOR1 deficiency in hematopoietic stem cells induced splenomegaly and monocytosis, specifically the abundance of inflammatory Ly6C(+) monocytes. Bone marrow transplantation studies further confirmed that NOR1 suppresses the proliferation of macrophage and dendritic progenitor (MDP) cells. Expression analysis identified RUNX1, a critical regulator of hematopoietic stem cell expansion, as a target gene suppressed by NOR1 in MDP cells. Finally, in addition to inducing Ly6C(+) monocytosis, NOR1 deletion increased the replicative rate of lesional macrophages and induced local foam cell formation within the atherosclerotic plaque. Collectively, our studies demonstrate that NOR1 deletion in hematopoietic stem cells accelerates atherosclerosis formation by promoting myelopoiesis in the stem cell compartment and by inducing local proatherogenic activities in the macrophage, including lesional macrophage proliferation and foam cell formation.


Subject(s)
DNA-Binding Proteins/deficiency , Hematopoietic Stem Cells/metabolism , Monocytes/metabolism , Nerve Tissue Proteins/deficiency , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Receptors, Steroid/deficiency , Receptors, Thyroid Hormone/deficiency , Animals , Cell Proliferation/physiology , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Plaque, Atherosclerotic/genetics , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Transfection
14.
Genes Cells ; 19(1): 13-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24245814

ABSTRACT

Oligo-astheno-teratozoospermia (OAT), a condition that includes low sperm number, low sperm motility and abnormal sperm morphology, is the commonest cause of male infertility. Because genetic analysis is frequently impeded by the infertility phenotype, the genetic basis of many of OAT conditions has been hard to verify. Here, we show that deficiency of ORP4, a sterol-binding protein in the oxysterol-binding protein (OSBP)-related protein family, causes male infertility due to severe OAT in mice. In ORP4-deficient mice, spermatogonia proliferation and subsequent meiosis occurred normally, but the morphology of elongating and elongated spermatids was severely distorted, with round-shaped head, curled back head or symplast. Spermatozoa derived from ORP4-deficient mice had little or no motility and no fertilizing ability in vitro. In ORP4-deficient testis, postmeiotic spermatids underwent extensive apoptosis, leading to a severely reduced number of spermatozoa. At the ultrastructural level, nascent acrosomes appeared to normally develop in round spermatids, but acrosomes were detached from the nucleus in elongating spermatids. These results suggest that ORP4 is essential for the postmeiotic differentiation of germ cells.


Subject(s)
Asthenozoospermia/genetics , Oligospermia/genetics , Receptors, Steroid/metabolism , Spermatozoa/abnormalities , Animals , Asthenozoospermia/metabolism , Asthenozoospermia/pathology , Female , Male , Mice , Mice, Knockout , Oligospermia/metabolism , Oligospermia/pathology , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Syndrome
15.
PLoS One ; 8(3): e58856, 2013.
Article in English | MEDLINE | ID: mdl-23554939

ABSTRACT

OSBP-related protein 8 (ORP8) encoded by Osbpl8 is an endoplasmic reticulum sterol sensor implicated in cellular lipid metabolism. We generated an Osbpl8(-/-) (KO) C57Bl/6 mouse strain. Wild-type and Osbpl8KO animals at the age of 13-weeks were fed for 5 weeks either chow or high-fat diet, and their plasma lipids/lipoproteins and hepatic lipids were analyzed. The chow-fed Osbpl8KO male mice showed a marked elevation of high-density lipoprotein (HDL) cholesterol (+79%) and phospholipids (+35%), while only minor increase of apolipoprotein A-I (apoA-I) was detected. In chow-fed female KO mice a less prominent increase of HDL cholesterol (+27%) was observed, while on western diet the HDL increment was prominent in both genders. The HDL increase was accompanied by an elevated level of HDL-associated apolipoprotein E in male, but not female KO animals. No differences between genotypes were observed in lecithin:cholesterol acyltransferase (LCAT) or hepatic lipase (HL) activity, or in the fractional catabolic rate of fluorescently labeled mouse HDL injected in chow-diet fed animals. The Osbpl8KO mice of both genders displayed reduced phospholipid transfer protein (PLTP) activity, but only on chow diet. These findings are consistent with a model in which Osbpl8 deficiency results in altered biosynthesis of HDL. Consistent with this hypothesis, ORP8 depleted mouse hepatocytes secreted an increased amount of nascent HDL into the culture medium. In addition to the HDL phenotype, distinct gender-specific alterations in lipid metabolism were detected: Female KO animals on chow diet showed reduced lipoprotein lipase (LPL) activity and increased plasma triglycerides, while the male KO mice displayed elevated plasma cholesterol biosynthetic markers cholestenol, desmosterol, and lathosterol. Moreover, modest gender-specific alterations in the hepatic expression of lipid homeostatic genes were observed. In conclusion, we report the first viable OsbplKO mouse model, demonstrating a HDL elevating effect of Osbpl8 knock-out and additional gender- and/or diet-dependent impacts on lipid metabolism.


Subject(s)
Lipid Metabolism , Lipoproteins, HDL/blood , Receptors, Steroid/deficiency , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein A-I/metabolism , Apolipoproteins/blood , Apolipoproteins E/metabolism , Body Weight , Cholesterol/metabolism , Female , Gene Expression , Gene Order , Gene Targeting , Hepatocytes/metabolism , Kidney/metabolism , Lipid Metabolism/genetics , Lipoprotein Lipase/blood , Liver/metabolism , Male , Mice , Mice, Knockout , Phosphatidylcholine-Sterol O-Acyltransferase/blood , Phospholipid Transfer Proteins/blood , RNA, Messenger , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Sex Factors
16.
Neuron ; 78(2): 312-24, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23622066

ABSTRACT

To what extent dorsal horn interneurons contribute to the modality specific processing of pain and itch messages is not known. Here, we report that loxp/cre-mediated CNS deletion of TR4, a testicular orphan nuclear receptor, results in loss of many excitatory interneurons in the superficial dorsal horn but preservation of primary afferents and spinal projection neurons. The interneuron loss is associated with a near complete absence of supraspinally integrated pain and itch behaviors, elevated mechanical withdrawal thresholds and loss of nerve injury-induced mechanical hypersensitivity, but reflex responsiveness to noxious heat, nerve injury-induced heat hypersensitivity, and tissue injury-induced heat and mechanical hypersensitivity are intact. We conclude that different subsets of dorsal horn excitatory interneurons contribute to tissue and nerve injury-induced heat and mechanical pain and that the full expression of supraspinally mediated pain and itch behaviors cannot be generated solely by nociceptor and pruritoceptor activation of projection neurons; concurrent activation of excitatory interneurons is essential.


Subject(s)
Interneurons/physiology , Pain/genetics , Pain/pathology , Pruritus/pathology , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Spinal Nerve Roots/pathology , Animals , Cell Death/genetics , Cognition Disorders/etiology , Cognition Disorders/genetics , Disease Models, Animal , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/metabolism , Hyperalgesia/genetics , Hyperalgesia/pathology , Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins v-fos/metabolism , Pain Threshold/physiology , Phosphopyruvate Hydratase/metabolism , Pruritus/genetics , Reaction Time/genetics , Receptors, Steroid/deficiency , Receptors, Thyroid Hormone/deficiency , Substance P/metabolism
17.
PLoS One ; 8(4): e61802, 2013.
Article in English | MEDLINE | ID: mdl-23626729

ABSTRACT

Xenobiotic-responsive nuclear receptors pregnane X receptor (PXR), constitutive active/androstane receptor (CAR) and peroxisome proliferator-activated receptor α (PPARα) play pivotal roles in the metabolic functions of the liver such as xenobiotics detoxification and energy metabolism. While CAR or PPARα activation induces hepatocyte proliferation and hepatocarcinogenesis in rodent models, it remains unclear whether PXR activation also shows such effects. In the present study, we have investigated the role of PXR in the xenobiotic-induced hepatocyte proliferation with or without CAR activation by 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) and phenobarbital, or PPARα activation by Wy-14643 in mice. Treatment with TCPOBOP or phenobarbital increased the percentage of Ki-67-positive nuclei as well as mRNA levels of cell proliferation-related genes in livers as expected. On the other hand, treatment with the PXR activator pregnenolone 16α-carbonitrile (PCN) alone showed no such effects. Surprisingly, PCN co-treatment significantly augmented the hepatocyte proliferation induced by CAR activation with TCPOBOP or phenobarbital in wild-type mice but not in PXR-deficient mice. Intriguingly, PXR activation also augmented the hepatocyte proliferation induced by Wy-14643 treatment. Moreover, PCN treatment increased the RNA content of hepatocytes, suggesting the induction of G0/G1 transition, and reduced mRNA levels of Cdkn1b and Rbl2, encoding suppressors of cell cycle initiation. Our present findings indicate that xenobiotic-induced hepatocyte proliferation mediated by CAR or PPARα is enhanced by PXR co-activation despite that PXR activation alone does not cause the cell proliferation in mouse livers. Thus PXR may play a novel and unique role in the hepatocyte/liver hyperplasia upon exposure to xenobiotics.


Subject(s)
Hepatocytes/drug effects , Liver/drug effects , PPAR alpha/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Xenobiotics/pharmacology , Animals , Cell Proliferation/drug effects , Constitutive Androstane Receptor , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Injections, Intraperitoneal , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/agonists , PPAR alpha/metabolism , Phenobarbital/pharmacology , Pregnane X Receptor , Pregnenolone Carbonitrile/pharmacology , Primary Cell Culture , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/agonists , Receptors, Steroid/deficiency , Signal Transduction
18.
Antimicrob Agents Chemother ; 57(3): 1332-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23274663

ABSTRACT

The antifungal efficacy of voriconazole (VRC) differs among host species, with potent efficacy in humans but less in rodents. We investigated the possible involvement of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) in the species-specific efficacy of VRC through pharmacokinetic analyses using genetically modified mice and primary human hepatocytes. VRC (30 mg/kg) was orally administered to wild-type, Pxr-null, Car-null, and Pxr- and Car-null (Pxr/Car-null) mice for 7 days. Hepatic VRC metabolism was significantly increased by VRC administration, and the elimination rates of plasma VRC were much higher on day 7 than on day 1 in wild-type mice. This autoinduction was also observed in Pxr-null and Car-null mice but not in Pxr/Car-null mice, suggesting coordinated roles of PXR and CAR in the autoinduction of VRC metabolism in mice. Hepatic Cyp3a11 mRNA levels were increased by VRC administration, hepatic metabolic activities for VRC were correlated with CYP3A activities, and the induced VRC metabolism was inhibited by ketoconazole (a CYP3A inhibitor). In primary human hepatocytes, VRC barely increased mRNA levels of CYP3A4 and CYP2B6 (human PXR/CAR target genes) at its therapeutic concentrations. In conclusion, these results suggest that VRC is metabolized mainly by CYP3A11 in mouse livers and that PXR- and CAR-mediated CYP3A11 induction, namely, autoinduction of VRC metabolism, is a primary reason for the ineffectiveness of VRC in mice. A limited ability of VRC to activate human PXR/CAR at its clinical concentration might explain the VRC efficacy in humans. Therefore, the ability to activate PXR/CAR might determine the VRC efficacy in different mammalian species.


Subject(s)
Antifungal Agents/pharmacokinetics , Cytochrome P-450 CYP3A/genetics , Hepatocytes/drug effects , Liver/drug effects , Membrane Proteins/genetics , Pyrimidines/pharmacokinetics , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Triazoles/pharmacokinetics , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A/metabolism , Gene Expression Regulation/drug effects , Hepatocytes/cytology , Hepatocytes/metabolism , Host Specificity , Humans , Ketoconazole/pharmacology , Liver/metabolism , Membrane Proteins/agonists , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Pregnane X Receptor , Primary Cell Culture , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Steroid/deficiency , Species Specificity , Voriconazole
19.
Biochimie ; 95(3): 538-47, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22609461

ABSTRACT

Oxysterol-binding protein homologs (ORPs) are implicated in lipid metabolism, vesicle transport and cell signaling. In this study we generated RAW264.7 cells with ORP1L, ORP3, or ORP8 silenced using shRNA lentiviruses. The lipidome of the cells under basal serum-free culture conditions or as treated with oxidized LDL (oxLDL), enzymatically modified LDL (E-LDL), or lipopolysaccharide (LPS) was analyzed by mass spectrometry. Reduction in each ORP resulted in distinct and complex effects on macrophage lipidome. Under basal conditions, ORP1L silencing had strongest effects on phosphatidylinositols (PI, increase), free cholesterol (FC, increase), and cholesteryl esters (CE, increase). ORP3 silencing affected most the glucosyl ceramides (GluCer, decrease) and PE-plasmalogens (PE-pl, decrease), while ORP8 silencing increased FC and CE, and decreased GluCer and PE-pl. Upon LPS treatment, the ORP effects were modified: under these conditions ORP1L silencing caused increase of Cer, ORP3 silencing decrease of PI, and ORP8 silencing decrease of PI and increase of PE, not detectable under basal conditions. The lipid species data were subjected to multivariate statistical analysis of principal components, revealing numerous specific alterations upon ORP silencing. The cells cultured in basal conditions or treated with LPS showed qualitatively different responses. However, in LPS-stimulated cells silencing of any of the three ORPs decreased the relative amount of arachidonic acid-containing PI species, increased the corresponding PE species, and favored 16-carbon sphingomyelin (SM) species at the expense of the 24-carbon ones. As a conclusion, the present study reveals the distinct and sophisticated roles of different ORP proteins as regulators of macrophage lipid composition, with implications for inflammatory signaling.


Subject(s)
Gene Silencing , Lipid Metabolism/genetics , Macrophages/metabolism , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Animals , Atherosclerosis/metabolism , Cell Line , Lentivirus/genetics , Lipid Metabolism/drug effects , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , RNA, Small Interfering/genetics
20.
Drug Metab Dispos ; 41(2): 406-13, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23160821

ABSTRACT

The pregnane X receptor (PXR) has been postulated to play a role in the metabolism of α-tocopherol owing to the up-regulation of hepatic cytochrome P450 (P450) 3A in human cell lines and murine models after α-tocopherol treatment. However, in vivo studies confirming the role of PXR in α-tocopherol metabolism in humans presents significant difficulties and has not been performed. PXR-humanized (hPXR), wild-type, and Pxr-null mouse models were used to determine whether α-tocopherol metabolism is influenced by species-specific differences in PXR function in vivo. No significant difference in the concentration of the major α-tocopherol metabolites was observed among the hPXR, wild-type, and Pxr-null mice through mass spectrometry-based metabolomics. Gene expression analysis revealed significantly increased expression of Cyp3a11 as well as several other P450s only in wild-type mice, suggesting species-specificity for α-tocopherol activation of PXR. Luciferase reporter assay confirmed activation of mouse PXR by α-tocopherol. Analysis of the Cyp2c family of genes revealed increased expression of Cyp2c29, Cyp2c37, and Cyp2c55 in wild-type, hPXR, and Pxr-null mice, which suggests PXR-independent induction of Cyp2c gene expression. This study revealed that α-tocopherol is a partial agonist of PXR and that PXR is necessary for Cyp3a induction by α-tocopherol. The implications of a novel role for α-tocopherol in Cyp2c gene regulation are also discussed.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Liver/drug effects , Receptors, Steroid/drug effects , Receptors, Steroid/metabolism , alpha-Tocopherol/pharmacology , Animals , Biomarkers/urine , Biotransformation , Chromatography, Liquid , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/genetics , Drug Partial Agonism , Gene Expression Regulation, Enzymologic/drug effects , Genes, Reporter , Hep G2 Cells , Humans , Isoenzymes , Liver/enzymology , Male , Mass Spectrometry , Metabolomics/methods , Mice , Mice, Knockout , Mice, Transgenic , Pregnane X Receptor , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Species Specificity , Time Factors , Transfection , alpha-Tocopherol/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...