Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nature ; 456(7223): 819-23, 2008 Dec 11.
Article in English | MEDLINE | ID: mdl-18849970

ABSTRACT

DNA double-strand breaks are generated by genotoxic agents and by cellular endonucleases as intermediates of several important physiological processes. The cellular response to genotoxic DNA breaks includes the activation of transcriptional programs known primarily to regulate cell-cycle checkpoints and cell survival. DNA double-strand breaks are generated in all developing lymphocytes during the assembly of antigen receptor genes, a process that is essential for normal lymphocyte development. Here we show that in murine lymphocytes these physiological DNA breaks activate a broad transcriptional program. This program transcends the canonical DNA double-strand break response and includes many genes that regulate diverse cellular processes important for lymphocyte development. Moreover, the expression of several of these genes is regulated similarly in response to genotoxic DNA damage. Thus, physiological DNA double-strand breaks provide cues that can regulate cell-type-specific processes not directly involved in maintaining the integrity of the genome, and genotoxic DNA breaks could disrupt normal cellular functions by corrupting these processes.


Subject(s)
B-Lymphocytes/metabolism , DNA Breaks, Double-Stranded , Gene Expression Regulation, Developmental/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/drug effects , Cell Cycle Proteins/drug effects , Cell Line , DNA-Binding Proteins/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Mice, SCID , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/drug effects , Tumor Suppressor Proteins/drug effects
2.
PLoS Genet ; 6(1): e1000812, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20090835

ABSTRACT

The neurodegenerative disease Friedreich's ataxia (FRDA) is the most common autosomal-recessively inherited ataxia and is caused by a GAA triplet repeat expansion in the first intron of the frataxin gene. In this disease, transcription of frataxin, a mitochondrial protein involved in iron homeostasis, is impaired, resulting in a significant reduction in mRNA and protein levels. Global gene expression analysis was performed in peripheral blood samples from FRDA patients as compared to controls, which suggested altered expression patterns pertaining to genotoxic stress. We then confirmed the presence of genotoxic DNA damage by using a gene-specific quantitative PCR assay and discovered an increase in both mitochondrial and nuclear DNA damage in the blood of these patients (p<0.0001, respectively). Additionally, frataxin mRNA levels correlated with age of onset of disease and displayed unique sets of gene alterations involved in immune response, oxidative phosphorylation, and protein synthesis. Many of the key pathways observed by transcription profiling were downregulated, and we believe these data suggest that patients with prolonged frataxin deficiency undergo a systemic survival response to chronic genotoxic stress and consequent DNA damage detectable in blood. In conclusion, our results yield insight into the nature and progression of FRDA, as well as possible therapeutic approaches. Furthermore, the identification of potential biomarkers, including the DNA damage found in peripheral blood, may have predictive value in future clinical trials.


Subject(s)
DNA Damage , Friedreich Ataxia/genetics , Friedreich Ataxia/pathology , Gene Expression , RNA/blood , Adolescent , Adult , Cells, Cultured , Child , Cohort Studies , Female , Humans , Iron-Binding Proteins/genetics , Male , Middle Aged , RNA/genetics , Young Adult , Frataxin
3.
Brain Behav Immun ; 25(6): 1063-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21435392

ABSTRACT

In the periphery, exercise induces interleukin (IL)-6 to downregulate tumor necrosis factor (TNF), elevate interleukin-1 receptor antagonist (IL-1RA), decreasing inflammation. Exercise also offers neuroprotection and facilitates brain repair. IL-6 production in the hippocampus following exercise suggests the potential of a similar protective role as in the periphery to down-regulate TNFα and inflammation. Using a chemical-induced model of hippocampal dentate granule cell death (trimethyltin, TMT 2.4 mg/kg, ip) dependent upon TNF receptor signaling, we demonstrate neuroprotection in mice with 2 weeks access to running wheel. Exercise attenuated neuronal death and diminished elevations in TNFα, TNF receptor 1, myeloid differentiation primary response gene (MyD) 88, transforming growth factor ß, chemokine (C-C motif) ligand 2 (CCL2), and CCL3. Elevated mRNA levels for IL-1α, IL-1RA, occurred with injury and protection. mRNA and protein levels of IL-6 and neuronal expression of IL-6 receptor α, were elevated with injury and protection. Microarray pathway analysis supported an up-regulation of TNFα cell death signaling pathways with TMT and inhibition by exercise. IL-6 pathway recruitment occurred in both conditions. IL-6 downstream signal events differed in the level of STAT3 activation. Exercise did not increase mRNA levels of brain derived neurotrophic factor, nerve growth factor, or glial derived neurotrophic factor. In IL-6 deficient mice, exercise did not attenuate TMT-induced tremor and a diminished level of neuroprotection was observed. These data suggest a contributory role for IL-6 induced by exercise for neuroprotection in the CNS similar to that seen in the periphery.


Subject(s)
Hippocampus/drug effects , Interleukin-6/physiology , Neurons/drug effects , Neurotoxins/toxicity , Physical Conditioning, Animal/physiology , Receptors, Tumor Necrosis Factor/drug effects , Trimethyltin Compounds/toxicity , Animals , Chemokines/biosynthesis , Chemokines/genetics , Epilepsy, Tonic-Clonic/chemically induced , Hippocampus/metabolism , Hippocampus/pathology , Interleukin-6/biosynthesis , Interleukin-6/deficiency , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Myeloid Differentiation Factor 88/biosynthesis , Myeloid Differentiation Factor 88/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radiation Chimera , Random Allocation , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/physiology , Specific Pathogen-Free Organisms , Tremor/chemically induced , Tumor Necrosis Factor-alpha/physiology , Up-Regulation
4.
Mol Cell Biol ; 27(20): 7143-60, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17682054

ABSTRACT

Glucocorticoids are widely used in the treatment of inflammatory and other diseases. However, high-dose or chronic administration often triggers troublesome side effects such as metabolic syndrome and osteoporosis. We recently described that one glucocorticoid receptor gene produces eight translational glucocorticoid receptor isoforms that have distinct gene-regulatory abilities. We show here that specific, but not all, glucocorticoid receptor isoforms induced apoptosis in human osteosarcoma U-2 OS bone cells. Whole human genome microarray analysis revealed that the majority of the glucocorticoid target genes were selectively regulated by specific glucocorticoid receptor isoforms. Real-time PCR experiments confirmed that proapoptotic enzymes necessary for cell death, granzyme A and caspase-6, were induced by specific glucocorticoid receptor isoforms. Chromatin immunoprecipitation assays further suggested that glucocorticoid receptor isoform-dependent induction of proapoptotic genes was likely due to selective coregulator recruitment and chromatin modification. Interestingly, the capabilities to transrepress proinflammatory genes were similar among glucocorticoid receptor isoforms. Together, these findings provide new evidence that translational glucocorticoid receptor isoforms can elicit distinct glucocorticoid responses and may be useful for the development of safe glucocorticoids with reduced side effects.


Subject(s)
Apoptosis/physiology , Bone and Bones/cytology , Protein Isoforms/metabolism , Receptors, Glucocorticoid/metabolism , Animals , COS Cells , Caspases/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Cluster Analysis , Dexamethasone/metabolism , Gene Expression Profiling , Glucocorticoids/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Osteosarcoma , Protein Biosynthesis , Protein Isoforms/genetics , Receptors, Glucocorticoid/genetics
5.
Mol Cell Biol ; 27(6): 2266-82, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17242213

ABSTRACT

Human glucocorticoid receptor (hGR) is expressed as two alternately spliced C-terminal isoforms, alpha and beta. In contrast to the canonical hGRalpha, hGRbeta is a nucleus-localized orphan receptor thought not to bind ligand and not to affect gene transcription other than by acting as a dominant negative to hGRalpha. Here we used confocal microscopy to examine the cellular localization of transiently expressed fluorescent protein-tagged hGRbeta in COS-1 and U-2 OS cells. Surprisingly, yellow fluorescent protein (YFP)-hGRbeta was predominantly located in the cytoplasm and translocated to the nucleus following application of the glucocorticoid antagonist RU-486. This effect of RU-486 was confirmed with transiently expressed wild-type hGRbeta. Confocal microscopy of coexpressed YFP-hGRbeta and cyan fluorescent protein-hGRalpha in COS-1 cells indicated that the receptors move into the nucleus independently. Using a ligand binding assay, we confirmed that hGRbeta bound RU-486 but not the hGRalpha ligand dexamethasone. Examination of the cellular localization of YFP-hGRbeta in response to a series of 57 related compounds indicated that RU-486 is thus far the only identified ligand that interacts with hGRbeta. The selective interaction of RU-486 with hGRbeta was also supported by molecular modeling and computational docking studies. Interestingly, microarray analysis indicates that hGRbeta, expressed in the absence of hGRalpha, can regulate gene expression and furthermore that occupation of hGRbeta with the antagonist RU-486 diminishes that capacity despite the lack of helix 12 in the ligand binding domain.


Subject(s)
Mifepristone/chemistry , Mifepristone/pharmacology , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Animals , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Computational Biology , Gene Expression Profiling , Gene Expression Regulation , Humans , Ligands , Mifepristone/metabolism , Models, Molecular , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , Receptors, Glucocorticoid/genetics
6.
Mol Reprod Dev ; 76(8): 733-50, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19484750

ABSTRACT

Both ovarian and pituitary hormones are required for the pubertal development of the mouse mammary gland. Estradiol directs ductal elongation and branching, while progesterone leads to tertiary branching and alveolar development. The purpose of this investigation was to identify estrogen-responsive genes associated with pubertal ductal growth in the mouse mammary gland in the absence of other ovarian hormones and at different stages of development. We hypothesized that the estrogen-induced genes and their associated functions at early stages of ductal elongation would be distinct from those induced after significant ductal elongation had occurred. Therefore, ovariectomized prepubertal mice were exposed to 17beta-estradiol from two to 28 days, and mammary gland global gene expression analyzed by microarray analysis at various times during this period. We found that: (a) gene expression changes in our estrogen-only model mimic those changes that occur in normal pubertal development in intact mice, (b) both distinct and overlapping gene profiles were observed at varying extents of ductal elongation, and (c) cell proliferation, the immune response, and metabolism/catabolism were the most common functional categories associated with mammary ductal growth. Particularly striking was the novel observation that genes active during carbohydrate metabolism were rapidly and robustly decreased in response to estradiol. Lastly, we identified mammary estradiol-responsive genes that are also co-expressed with estrogen receptor alpha in human breast cancer. In conclusion, our genomic data support the physiological observation that estradiol is one of the primary hormonal signals driving ductal elongation during pubertal mammary development.


Subject(s)
Estradiol/pharmacology , Gene Expression Regulation, Developmental/physiology , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/physiology , Animals , Breast Neoplasms/genetics , Carbohydrate Metabolism/genetics , Cell Growth Processes/genetics , Cluster Analysis , Estrogen Receptor alpha/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/drug effects , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Ovariectomy , Sexual Maturation/genetics
7.
Toxicol Pathol ; 37(4): 502-11, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19395590

ABSTRACT

Carbonyl sulfide (COS) is an odorless gas that produces highly reproducible lesions in the central nervous system. In the present study, the time course for the development of the neurotoxicological lesions was defined and the gene expression changes occurring in the posterior colliculus upon exposure to COS were characterized. Fischer 344 rats were exposed to 0 or 500 ppm COS for one, two, three, four, five, eight, or ten days, six hours per day. On days 1 and 2, no morphological changes were detected; on day 3, 10/10 (100%) rats had necrosis in the posterior colliculi; and on day 4 and later, necrosis was observed in numerous areas of the brain. Important gene expression changes occurring in the posterior colliculi after one or two days of COS exposure that were predictive of the subsequent morphological findings included up-regulation of genes associated with DNA damage and G1/S checkpoint regulation (KLF4, BTG2, GADD45g), apoptosis (TGM2, GADD45g, RIPK3), and vascular mediators (ADAMTS, CTGF, CYR61, VEGFC). Proinflammatory mediators (CCL2, CEBPD) were up-regulated prior to increases in expression of the astrocytic marker GFAP and macrophage marker CSF2rb1. These gene expression findings were predictive of later CNS lesions caused by COS exposure and serve as a model for future investigations into the mechanisms of disease in the central nervous system.


Subject(s)
Brain Diseases/chemically induced , Brain/metabolism , DNA Damage/drug effects , Gene Expression/drug effects , Nerve Degeneration/metabolism , Sulfur Oxides/toxicity , Administration, Inhalation , Animals , Apoptosis/drug effects , Brain/pathology , Brain Diseases/genetics , Brain Diseases/metabolism , Brain Diseases/pathology , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Inflammation/metabolism , Kruppel-Like Factor 4 , Male , Necrosis , Nerve Degeneration/pathology , Oligonucleotide Array Sequence Analysis , Protein Glutamine gamma Glutamyltransferase 2 , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Sulfur Oxides/administration & dosage
8.
Cancer Res ; 67(16): 7929-36, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699800

ABSTRACT

Farnesol (FOH) and other isoprenoid alcohols induce apoptosis in various carcinoma cells and inhibit tumorigenesis in several in vivo models. However, the mechanisms by which they mediate their effects are not yet fully understood. In this study, we show that FOH is an effective inducer of apoptosis in several lung carcinoma cells, including H460. This induction is associated with activation of several caspases and cleavage of poly(ADP-ribose) polymerase (PARP). To obtain insight into the mechanism involved in FOH-induced apoptosis, we compared the gene expression profiles of FOH-treated and control H460 cells by microarray analysis. This analysis revealed that many genes implicated in endoplasmic reticulum (ER) stress signaling, including ATF3, DDIT3, HERPUD1, HSPA5, XBP1, PDIA4, and PHLDA1, were highly up-regulated within 4 h of FOH treatment, suggesting that FOH-induced apoptosis involves an ER stress response. This was supported by observations showing that treatment with FOH induces splicing of XBP1 mRNA and phosphorylation of eIF2alpha. FOH induces activation of several mitogen-activated protein kinase (MAPK) pathways, including p38, MAPK/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK, and c-jun NH2-terminal kinase (JNK). Inhibition of MEK1/2 by U0126 inhibited the induction of ER stress response genes. In addition, knockdown of the MEK1/2 and JNK1/2 expression by short interfering RNA (siRNA) effectively inhibited the cleavage of caspase-3 and PARP and apoptosis induced by FOH. However, only MEK1/2 siRNAs inhibited the induction of ER stress-related genes, XBP1 mRNA splicing, and eIF2alpha phosphorylation. Our results show that FOH-induced apoptosis is coupled to ER stress and that activation of MEK1/2 is an early upstream event in the FOH-induced ER stress signaling cascade.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum/drug effects , Farnesol/pharmacology , Lung Neoplasms/drug therapy , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , RNA, Small Interfering/genetics
9.
Mol Carcinog ; 47(11): 845-85, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18381591

ABSTRACT

Steroid hormone receptors, like glucocorticoid (GR) and estrogen receptors (ER), are master regulators of genes that control many biological processes implicated in health and disease. Gene expression is dependent on receptor levels which are tightly regulated by the ubiquitin-proteasome system. Previous studies have shown that proteasome inhibition increases GR, but decreases ER-mediated gene expression. At the gene expression level this divergent role of the proteasome in receptor-dependent transcriptional regulation is not well understood. We have used a genomic approach to examine the impact of proteasome activity on GR- and ER-mediated gene expression in MCF-7 breast cancer cells treated with dexamethasone (DEX) or 17beta-estradiol (E2), the proteasome inhibitor MG132 (MG) or MG132 and either hormone (MD or ME2) for 24 h. Transcript profiling reveals that inhibiting proteasome activity modulates gene expression by GR and ER in a similar manner in that several GR and ER target genes are upregulated and downregulated after proteasome inhibition. In addition, proteasome inhibition modulates receptor-dependent genes involved in the etiology of a number of human pathological states, including multiple myeloma, leukemia, breast/prostate cancer, HIV/AIDS, and neurodegenerative disorders. Importantly, our analysis reveals that a number of transcripts encoding histone and DNA modifying enzymes, prominently histone/DNA methyltransferases and demethylases, are altered after proteasome inhibition. As proteasome inhibitors are currently in clinical trials as therapy for multiple myeloma, HIV/AIDS and leukemia, the possibility that some of the target molecules are hormone regulated and chromatin modifying enzymes is intriguing in this era of epigenetic therapy.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Chromatin/metabolism , Genome, Human/genetics , Proteasome Inhibitors , Receptors, Estrogen/metabolism , Transcription, Genetic/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin/drug effects , Chromatin/genetics , DNA/genetics , DNA/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glucocorticoids/metabolism , Humans , Proteasome Endopeptidase Complex/metabolism , RNA Polymerase II/metabolism , Receptors, Estrogen/antagonists & inhibitors , Transcription, Genetic/drug effects
10.
Toxicol Pathol ; 36(5): 743-52, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18648096

ABSTRACT

National Toxicology Program (NTP) inhalation studies demonstrated that cumene significantly increased the incidence of alveolar/bronchiolar adenomas and carcinomas in B6C3F1 mice. Cumene or isopropylbenzene is a component of crude oil used primarily in the production of phenol and acetone. The authors performed global gene expression analysis to distinguish patterns of gene regulation between cumene-induced tumors and normal lung tissue and to look for patterns based on the presence or absence of K-ras and p53 mutations in the tumors. Principal component analysis segregated the carcinomas into groups with and without K-ras mutations, but failed to separate the tumors based on p53 mutation status. Expression of genes associated with the Erk MAP kinase signaling pathway was significantly altered in carcinomas with K-ras mutations compared to tumors without K-ras mutations or normal lung. Gene expression analysis also suggested that cumene-induced carcinomas with K-ras mutations have greater malignant potential than those without mutations. In addition, significance analysis of function and expression (SAFE) demonstrated expression changes of genes regulated by histone modification in carcinomas with K-ras mutations. The gene expression analysis suggested the formation of alveolar/bronchiolar carcinomas in cumene-exposed mice typically involves mutation of K-ras, which results in increased Erk MAP kinase signaling and modification of histones.


Subject(s)
Benzene Derivatives/toxicity , Genes, ras/genetics , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/genetics , Adenocarcinoma, Bronchiolo-Alveolar/chemically induced , Adenocarcinoma, Bronchiolo-Alveolar/genetics , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Animals , Female , Gene Expression Regulation, Neoplastic , Immunohistochemistry , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred Strains , Signal Transduction/physiology
11.
PLoS One ; 13(2): e0191105, 2018.
Article in English | MEDLINE | ID: mdl-29462216

ABSTRACT

Changes in gene expression can help reveal the mechanisms of disease processes and the mode of action for toxicities and adverse effects on cellular responses induced by exposures to chemicals, drugs and environment agents. The U.S. Tox21 Federal collaboration, which currently quantifies the biological effects of nearly 10,000 chemicals via quantitative high-throughput screening(qHTS) in in vitro model systems, is now making an effort to incorporate gene expression profiling into the existing battery of assays. Whole transcriptome analyses performed on large numbers of samples using microarrays or RNA-Seq is currently cost-prohibitive. Accordingly, the Tox21 Program is pursuing a high-throughput transcriptomics (HTT) method that focuses on the targeted detection of gene expression for a carefully selected subset of the transcriptome that potentially can reduce the cost by a factor of 10-fold, allowing for the analysis of larger numbers of samples. To identify the optimal transcriptome subset, genes were sought that are (1) representative of the highly diverse biological space, (2) capable of serving as a proxy for expression changes in unmeasured genes, and (3) sufficient to provide coverage of well described biological pathways. A hybrid method for gene selection is presented herein that combines data-driven and knowledge-driven concepts into one cohesive method. Our approach is modular, applicable to any species, and facilitates a robust, quantitative evaluation of performance. In particular, we were able to perform gene selection such that the resulting set of "sentinel genes" adequately represents all known canonical pathways from Molecular Signature Database (MSigDB v4.0) and can be used to infer expression changes for the remainder of the transcriptome. The resulting computational model allowed us to choose a purely data-driven subset of 1500 sentinel genes, referred to as the S1500 set, which was then augmented using a knowledge-driven selection of additional genes to create the final S1500+ gene set. Our results indicate that the sentinel genes selected can be used to accurately predict pathway perturbations and biological relationships for samples under study.


Subject(s)
Gene Expression Profiling/methods , High-Throughput Screening Assays/methods , Computational Biology , Databases, Genetic , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Oligonucleotide Array Sequence Analysis , Transcriptome
12.
Physiol Genomics ; 31(2): 281-94, 2007 Oct 22.
Article in English | MEDLINE | ID: mdl-17666523

ABSTRACT

Retinoid-related orphan receptors alpha (ROR alpha) and gamma (ROR gamma) are both expressed in liver; however, their physiological functions in this tissue have not yet been clearly defined. The ROR alpha1 and ROR gamma 1 isoforms, but not ROR alpha 4, show an oscillatory pattern of expression during circadian rhythm. To obtain insight into the physiological functions of ROR receptors in liver, we analyzed the gene expression profiles of livers from WT, ROR alpha-deficient staggerer (sg) mice (ROR alpha(sg/sg)), ROR gamma(-/-), and ROR alpha(sg/sg)ROR gamma(-/-) double knockout (DKO) mice by microarray analysis. DKO mice were generated to study functional redundancy between ROR alpha and ROR gamma. These analyses demonstrated that ROR alpha and ROR gamma affect the expression of a number of genes. ROR alpha and ROR gamma are particularly important in the regulation of genes encoding several phase I and phase II metabolic enzymes, including several 3beta-hydroxysteroid dehydrogenases, cytochrome P450 enzymes, and sulfotransferases. In addition, our results indicate that ROR alpha and ROR gamma each affect the expression of a specific set of genes but also exhibit functional redundancy. Our study shows that ROR alpha and ROR gamma receptors influence the regulation of several metabolic pathways, including those involved in the metabolism of steroids, bile acids, and xenobiotics, suggesting that RORs are important in the control of metabolic homeostasis.


Subject(s)
Gene Expression Profiling , Metabolic Networks and Pathways/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Retinoic Acid/physiology , Receptors, Thyroid Hormone/physiology , Trans-Activators/physiology , Animals , Bile Acids and Salts/metabolism , Cells, Cultured/metabolism , Circadian Rhythm/genetics , Cytochrome P-450 Enzyme System/metabolism , Female , Hepatocytes/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Nuclear Receptor Subfamily 1, Group F, Member 1 , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Retinoic Acid/deficiency , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/genetics , Recombinant Fusion Proteins/metabolism , Steroids/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics , Transfection , Xenobiotics/metabolism
13.
Biochem J ; 397(1): 89-99, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16533169

ABSTRACT

RORgamma2 (retinoid-related orphan receptor gamma2) plays a critical role in the regulation of thymopoiesis. Microarray analysis was performed in order to uncover differences in gene expression between thymocytes of wild-type and RORgamma-/- mice. This analysis identified a novel gene encoding a 22 kDa protein, referred to as NABP1 (nucleic-acid-binding protein 1). This subsequently led to the identification of an additional protein, closely related to NABP1, designated NABP2. Both proteins contain an OB (oligonucleotide/oligosaccharide binding) motif at their N-terminus. This motif is highly conserved between the two proteins. NABP1 is highly expressed in the thymus of wild-type mice and is greatly suppressed in RORgamma-/- mice. During thymopoiesis, NABP1 mRNA expression is restricted to CD4+CD8+ thymocytes, an expression pattern similar to that observed for RORgamma2. These observations appear to suggest that NABP1 expression is regulated either directly or indirectly by RORgamma2. Confocal microscopic analysis showed that the NABP1 protein localizes to the nucleus. Analysis of nuclear proteins by size-exclusion chromatography indicated that NABP1 is part of a high molecular-mass protein complex. Since the OB-fold is frequently involved in the recognition of nucleic acids, the interaction of NABP1 with various nucleic acids was examined. Our results demonstrate that NABP1 binds single-stranded nucleic acids, but not double-stranded DNA, suggesting that it functions as a single-stranded nucleic acid binding protein.


Subject(s)
Carrier Proteins/genetics , Thymus Gland/physiology , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/isolation & purification , Cell Nucleus/chemistry , Chromatography, Gel , DNA/metabolism , Escherichia coli , Gene Expression Profiling , Mice , Microscopy, Confocal , Molecular Sequence Data , Nuclear Receptor Subfamily 1, Group F, Member 3 , Nucleic Acids/metabolism , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Receptors, Retinoic Acid/physiology , Receptors, Thyroid Hormone/physiology , Thymus Gland/cytology
14.
Physiol Genomics ; 21(3): 351-61, 2005 May 11.
Article in English | MEDLINE | ID: mdl-15888570

ABSTRACT

Gene expression was measured during t10c12-CLA-induced body fat reduction in a polygenic obese line of mice. Adult mice (n = 185) were allotted to a 2 x 2 factorial experiment consisting of either nonobese (ICR-control) or obese (M16-selected) mice fed a 7% fat, purified diet containing either 1% linoleic acid (LA) or 1% t10c12-CLA. Body weight (BW) by day 14 was 12% lower in CLA- compared with LA-fed mice (P < 0.0001). By day 14, t10c12-CLA reduced weights of epididymal, mesenteric, and brown adipose tissues, as a percentage of BW, in both lines by 30, 27, and 58%, respectively, and increased liver weight/BW by 34% (P < 0.0001). Total RNA was isolated and pooled (4 pools per tissue per day) from epididymal adipose (days 5 and 14) of the obese mice to analyze gene expression profiles using Agilent mouse oligo microarray slides representing > 20,000 genes. Numbers of genes differentially expressed by greater than or equal to twofold in epididymal adipose (days 5 and 14) were 29 and 125, respectively. It was concluded that, in adipose tissue, CLA increased expression of uncoupling proteins (1 and 2), carnitine palmitoyltransferase system, tumor necrosis factor-alpha (P < 0.05), and caspase-3 but decreased expression of peroxisome proliferator-activated receptor-gamma, glucose transporter-4, perilipin, caveolin-1, adiponectin, resistin, and Bcl-2 (P < 0.01). In conclusion, this experiment has revealed candidate genes that will be useful in elucidating mechanisms of adipose delipidation.


Subject(s)
Gene Expression Regulation/drug effects , Lipids/physiology , Obesity/genetics , alpha-Linolenic Acid/analogs & derivatives , alpha-Linolenic Acid/therapeutic use , Animals , Base Sequence , Body Weight/drug effects , DNA Primers , Genomics , Male , Mice , Mice, Inbred ICR , Mice, Obese , Obesity/drug therapy , Oligonucleotide Array Sequence Analysis , Organ Size/drug effects , Reverse Transcriptase Polymerase Chain Reaction
15.
Sci Signal ; 3(143): ra74, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20940427

ABSTRACT

Males and females show differences in the prevalence of many major diseases that have important inflammatory components to their etiology. These gender-specific diseases, which include autoimmune diseases, hepatocellular carcinoma, diabetes, and osteoporosis, are largely considered to reflect the actions of sex hormones on the susceptibility to inflammatory stimuli. However, inflammation reflects a balance between pro- and anti-inflammatory signals, and investigation of gender-specific responses to the latter has been neglected. Glucocorticoids are the primary physiological anti-inflammatory hormones in mammals, and synthetic derivatives of these hormones are prescribed as anti-inflammatory agents, irrespective of patient gender. We explored the possibility that sexually dimorphic actions of glucocorticoid regulation of gene expression may contribute to the dimorphic basis of inflammatory disease by evaluating the rat liver, a classic glucocorticoid-responsive organ. Surprisingly, glucocorticoid administration expanded the set of hepatic sexually dimorphic genes. Eight distinct patterns of glucocorticoid-regulated gene expression were identified, which included sex-specific genes. Our experiments also defined specific genes with altered expression in response to glucocorticoid treatment in both sexes, but in opposite directions. Pathway analysis identified sex-specific glucocorticoid-regulated gene expression in several canonical pathways involved in susceptibility to and progression of diseases with gender differences in prevalence. Moreover, a comparison of the number of genes involved in inflammatory disorders between sexes revealed 84 additional glucocorticoid-responsive genes in the male, suggesting that the anti-inflammatory actions of glucocorticoids are more effective in males. These gender-specific actions of glucocorticoids in liver were substantiated in vivo with a sepsis model of systemic inflammation.


Subject(s)
Gene Expression Regulation/physiology , Glucocorticoids/physiology , Inflammation/physiopathology , Sex Characteristics , Signal Transduction/physiology , Adrenalectomy , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Apoptosis/drug effects , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/physiology , Gonadal Steroid Hormones/physiology , Inflammation/epidemiology , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/metabolism , Male , Oligonucleotide Array Sequence Analysis , Ovariectomy , Prevalence , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Systemic Inflammatory Response Syndrome/chemically induced , Systemic Inflammatory Response Syndrome/drug therapy , Systemic Inflammatory Response Syndrome/physiopathology
16.
J Nutr Biochem ; 21(9): 848-55, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19800780

ABSTRACT

The trans-10, cis-12 isomer of conjugated linoleic acid (CLA) causes a rapid reduction of body and adipose mass in mice. In addition to changes in adipose tissue, numerous studies have reported alterations in hepatic lipid metabolism. Livers of CLA-fed mice gain mass, partly due to lipid accumulation; however, the precise molecular mechanisms are unknown. To elucidate these mechanisms, we examined fatty acid composition and gene expression profiles of livers from a polygenic obese line of mice fed 1% trans-10, cis-12-CLA for 14 days. Analysis of gene expression data led to the identification of 1393 genes differentially expressed in the liver of CLA-fed male mice at a nominal P value of .01, and 775 were considered significant using a false discovery rate (FDR) threshold of .05. While surprisingly few genes in lipid metabolism were impacted, pathway analysis found that protein kinase A (PKA) and cyclic adenosine monophosphate (cAMP) pathways signaling pathways were affected by CLA treatment and 98 of the 775 genes were found to be regulated by hepatocyte nuclear factor 4alpha, a transcription factor important in controlling liver metabolic status.


Subject(s)
Fatty Liver/metabolism , Linoleic Acids, Conjugated/pharmacology , Liver/metabolism , Obesity/metabolism , Animals , Fatty Liver/genetics , Gene Expression Profiling , Hepatocyte Nuclear Factor 4/genetics , Male , Mice , Obesity/genetics
17.
Cell Metab ; 11(6): 493-502, 2010 Jun 09.
Article in English | MEDLINE | ID: mdl-20519121

ABSTRACT

Crosstalk exists in mammalian cells between cholesterol trafficking and innate immune signaling. Apolipoprotein A-I (apoA-I), a serum apolipoprotein that induces antiatherogenic efflux of macrophage cholesterol, is widely described as anti-inflammatory because it neutralizes bacterial lipopolysaccharide. Conversely, lipopolysaccharide-induced inflammation is proatherogenic. However, whether innate immunity plays an endogenous, physiological role in host cholesterol homeostasis in the absence of infection is undetermined. We report that apoA-I signals in the macrophage through Toll-like receptor (TLR)2, TLR4, and CD14, utilizing myeloid differentiation primary response protein 88 (MyD88)-dependent and -independent pathways, to activate nuclear factor-kappaB and induce cytokines. MyD88 plays a critical role in reverse cholesterol transport in vitro and in vivo, in part through promoting ATP-binding cassette A1 transporter upregulation. Taken together, this work identifies apoA-I as an endogenous stimulus of innate immunity that couples cholesterol trafficking to inflammation through MyD88 and identifies innate immunity as a physiologic signal in cholesterol homeostasis.


Subject(s)
Cholesterol/metabolism , Inflammation/metabolism , Myeloid Differentiation Factor 88/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein A-I/pharmacology , Biological Transport , Cell Differentiation , Cytokines/metabolism , Immunity, Innate , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/toxicity , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
18.
Mol Endocrinol ; 23(7): 955-65, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19324971

ABSTRACT

Granulosa cells of preovulatory follicles differentiate in response to FSH, and this differentiation is augmented by estradiol. We have previously shown that FSH-mediated granulosa cell differentiation requires functional estrogen receptor-beta (ERbeta) by demonstrating that the granulosa cells of ERbeta(-/-) FSH-treated mice are unable to maximally induce expression of the LH receptor (an indicator of granulosa cell differentiation) compared with ERbeta(+/+) controls. As a result, FSH-primed ERbeta(-/-) granulosa cells exhibit a reduced response to a subsequent ovulatory dose of LH. In this study, we further characterized the attenuated response of ERbeta(-/-) granulosa cells to stimulation by LH and FSH using isolated mouse granulosa cells and primary granulosa cell cultures. We observed a 50% reduction in cAMP levels in cultured ERbeta(-/-) granulosa cells exposed to LH compared with ERbeta(+/+) controls. We also observed an attenuated genomic response in granulosa cells isolated from FSH-primed ERbeta(-/-) mice compared with ERbeta(+/+) controls. Our data indicate that this attenuated response may result from inadequate levels of cAMP, because cAMP levels in cultured ERbeta(-/-) granulosa cells exposed to forskolin were approximately 50% lower than in ERbeta(+/+) granulosa cells. Phosphorylation of cAMP regulatory element binding protein, an indicator of protein kinase A activity, was also reduced in FSH-treated ERbeta(-/-) granulosa cells compared with ERbeta(+/+) controls. These are the first data to indicate that ERbeta plays a role in the induction of the cAMP pathway in mouse granulosa cells and that disruption of proper ERbeta signaling associated with this pathway may cause negative effects on ovulation and fertility.


Subject(s)
Cyclic AMP/metabolism , Estrogen Receptor beta/physiology , Granulosa Cells/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Fertility/genetics , Granulosa Cells/drug effects , Luteinizing Hormone/pharmacology , Mice , Mice, Knockout , Ovulation/genetics , Ovulation/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism
19.
Mol Cell Biol ; 28(7): 2358-67, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18227149

ABSTRACT

To obtain insight into the physiological functions of the Krüppel-like zinc finger protein Gli-similar 2 (Glis2), mice deficient in Glis2 expression were generated. Glis2 mutant (Glis2(mut)) mice exhibit significantly shorter life spans than do littermate wild-type (WT) mice due to the development of progressive chronic kidney disease with features resembling nephronophthisis. Glis2(mut) mice develop severe renal atrophy involving increased cell death and basement membrane thickening in the proximal convoluted tubules. This development is accompanied by infiltration of lymphocytic inflammatory cells and interstitial/glomerular fibrosis. The severity of the fibrosis, inflammatory infiltrates, and glomerular and tubular changes progresses with age. Blood urea nitrogen and creatinine increase, and Glis2(mut) mice develop proteinuria and ultimately die prematurely of renal failure. A comparison of the gene expression profiles of kidneys from 25-day-old/60-day-old WT and Glis2(mut) mice by microarray analysis showed increased expressions of many genes involved in immune responses/inflammation and fibrosis/tissue remodeling in kidneys of Glis2(mut) mice, including several cytokines and adhesion and extracellular matrix proteins. Our data demonstrate that a deficiency in Glis2 expression leads to tubular atrophy and progressive fibrosis, similar to nephronophthisis, that ultimately results in renal failure. Our study indicates that Glis2 plays a critical role in the maintenance of normal kidney architecture and functions.


Subject(s)
Kidney Failure, Chronic/genetics , Kidney/physiology , Kruppel-Like Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Age Factors , Animals , Atrophy , Cell Line , Disease Progression , Exons/genetics , Fibrosis , Gene Expression Profiling , Genes, Reporter , Humans , Kidney/pathology , Kidney Failure, Chronic/pathology , Kidney Failure, Chronic/urine , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Proteinuria/genetics , Proteinuria/pathology , Transcription, Genetic , Zinc Fingers/genetics
20.
Genes Dev ; 22(14): 1921-33, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18628398

ABSTRACT

The Negative Elongation Factor (NELF) is a transcription regulatory complex that induces stalling of RNA polymerase II (Pol II) during early transcription elongation and represses expression of several genes studied to date, including Drosophila Hsp70, mammalian proto-oncogene junB, and HIV RNA. To determine the full spectrum of NELF target genes in Drosophila, we performed a microarray analysis of S2 cells depleted of NELF and discovered that NELF RNAi affects many rapidly inducible genes involved in cellular responses to stimuli. Surprisingly, only one-third of NELF target genes were, like Hsp70, up-regulated by NELF-depletion, whereas the majority of target genes showed decreased expression levels upon NELF RNAi. Our data reveal that the presence of stalled Pol II at this latter group of genes enhances gene expression by maintaining a permissive chromatin architecture around the promoter-proximal region, and that loss of Pol II stalling at these promoters is accompanied by a significant increase in nucleosome occupancy and a decrease in histone H3 Lys 4 trimethylation. These findings identify a novel, positive role for stalled Pol II in regulating gene expression and suggest that there is a dynamic interplay between stalled Pol II and chromatin structure.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , DNA Footprinting , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Profiling , Genes, Dominant , Luciferases/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA Polymerase II/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL