201
|
Insel PA, Wilderman A, Zambon AC, Snead AN, Murray F, Aroonsakool N, McDonald DS, Zhou S, McCann T, Zhang L, Sriram K, Chinn AM, Michkov AV, Lynch RM, Overland AC, Corriden R. G Protein-Coupled Receptor (GPCR) Expression in Native Cells: "Novel" endoGPCRs as Physiologic Regulators and Therapeutic Targets. Mol Pharmacol 2015; 88:181-7. [PMID: 25737495 DOI: 10.1124/mol.115.098129] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/02/2015] [Indexed: 12/24/2022] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of signaling receptors in the human genome, are also the largest class of targets of approved drugs. Are the optimal GPCRs (in terms of efficacy and safety) currently targeted therapeutically? Especially given the large number (∼ 120) of orphan GPCRs (which lack known physiologic agonists), it is likely that previously unrecognized GPCRs, especially orphan receptors, regulate cell function and can be therapeutic targets. Knowledge is limited regarding the diversity and identity of GPCRs that are activated by endogenous ligands and that native cells express. Here, we review approaches to define GPCR expression in tissues and cells and results from studies using these approaches. We identify problems with the available data and suggest future ways to identify and validate the physiologic and therapeutic roles of previously unrecognized GPCRs. We propose that a particularly useful approach to identify functionally important GPCRs with therapeutic potential will be to focus on receptors that show selective increases in expression in diseased cells from patients and experimental animals.
Collapse
Affiliation(s)
- Paul A Insel
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Andrea Wilderman
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Alexander C Zambon
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Aaron N Snead
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Fiona Murray
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Nakon Aroonsakool
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Daniel S McDonald
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Shu Zhou
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Thalia McCann
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Lingzhi Zhang
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Krishna Sriram
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Amy M Chinn
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Alexander V Michkov
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Rebecca M Lynch
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Aaron C Overland
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Ross Corriden
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| |
Collapse
|
202
|
Neeland MR, Elhay MJ, Powell DR, Rossello FJ, Meeusen ENT, de Veer MJ. Transcriptional profile in afferent lymph cells following vaccination with liposomes incorporating CpG. Immunology 2015; 144:518-529. [PMID: 25308816 DOI: 10.1111/imm.12401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/14/2014] [Accepted: 10/02/2014] [Indexed: 12/17/2022] Open
Abstract
Vaccine formulations incorporating innate immune stimulants are highly immunogenic; however, the biological signals that originate in the peripheral tissues at the site of injection and are transmitted to the local lymph node to induce immunity remain unclear. By directly cannulating the ovine afferent lymphatic vessels, we have previously shown that it takes 72 hr for mature antigen-loaded dendritic cells and monocytes to appear within afferent lymph following injection of a liposomal formulation containing the Toll-like receptor ligand CpG. In this present study, we characterize the global transcriptional signatures at this time-point in ovine afferent lymph cells as they migrate from the injection site into the lymphatics following vaccination with a liposome antigen formulation incorporating CpG. We show that at 72 hr post vaccination, liposomes alone induce no changes in gene expression and inflammatory profiles within afferent lymph; however, the incorporation of CpG drives interferon, antiviral and cytotoxic gene programmes. This study also measures the expression of key genes within individual cell types in afferent lymph. Antiviral gene signatures are most prominent in lymphocytes, which may play a significant and unexpected role in sustaining the immune response to vaccination at the site of injection. These findings provide a comprehensive analysis of the in vivo immunological pathways that connect the injection site with the local draining lymph node following vaccination.
Collapse
Affiliation(s)
- Melanie R Neeland
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Clayton, Vic., Australia
| | - Martin J Elhay
- Zoetis Research and Manufacturing Australia P/L, Parkville, Vic., Australia
| | - David R Powell
- Victorian Bioinformatics Consortium, Monash University, Clayton, Vic., Australia.,Victorian Life Sciences Computation Initiative, Life Sciences Computation Centre, Carlton, Vic., Australia
| | - Fernando J Rossello
- Victorian Bioinformatics Consortium, Monash University, Clayton, Vic., Australia.,Victorian Life Sciences Computation Initiative, Life Sciences Computation Centre, Carlton, Vic., Australia
| | - Els N T Meeusen
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Clayton, Vic., Australia.,Department of Microbiology, Monash University, Clayton, Vic., Australia
| | - Michael J de Veer
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Clayton, Vic., Australia
| |
Collapse
|
203
|
Adijanto J, Du J, Moffat C, Seifert EL, Hurle JB, Philp NJ. The retinal pigment epithelium utilizes fatty acids for ketogenesis. J Biol Chem 2015; 289:20570-82. [PMID: 24898254 DOI: 10.1074/jbc.m114.565457] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Every day, shortly after light onset, photoreceptor cells shed approximately a tenth of their outer segment. The adjacent retinal pigment epithelial (RPE) cells phagocytize and digest shed photoreceptor outer segment, which provides a rich source of fatty acids that could be utilized as an energy substrate. From a microarray analysis, we found that RPE cells express particularly high levels of the mitochondrial HMG-CoA synthase 2 (Hmgcs2) compared with all other tissues (except the liver and colon), leading to the hypothesis that RPE cells, like hepatocytes, can produce β-hydroxybutyrate (β-HB) from fatty acids. Using primary human fetal RPE (hfRPE) cells cultured on Transwell filters with separate apical and basal chambers, we demonstrate that hfRPE cells can metabolize palmitate, a saturated fatty acid that constitutes .15% of all lipids in the photoreceptor outer segment, to produce β-HB. Importantly, we found that hfRPE cells preferentially release β-HB into the apical chamber and that this process is mediated primarily by monocarboxylate transporter isoform 1 (MCT1). Using a GC-MS analysis of (13)C-labeled metabolites, we showed that retinal cells can take up and metabolize (13)C-labeled β-HB into various TCA cycle intermediates and amino acids. Collectively, our data support a novel mechanism of RPE-retina metabolic coupling in which RPE cells metabolize fatty acids to produce β-HB, which is transported to the retina for use as a metabolic substrate.
Collapse
|
204
|
Bickert A, Ginkel C, Kol M, vom Dorp K, Jastrow H, Degen J, Jacobs RL, Vance DE, Winterhager E, Jiang XC, Dörmann P, Somerharju P, Holthuis JCM, Willecke K. Functional characterization of enzymes catalyzing ceramide phosphoethanolamine biosynthesis in mice. J Lipid Res 2015; 56:821-35. [PMID: 25667419 DOI: 10.1194/jlr.m055269] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Besides bulk amounts of SM, mammalian cells produce small quantities of the SM analog ceramide phosphoethanolamine (CPE). Little is known about the biological role of CPE or enzymes responsible for CPE production. Heterologous expression studies revealed that SM synthase (SMS)2 is a bifunctional enzyme producing both SM and CPE, whereas SMS-related protein (SMSr) serves as monofunctional CPE synthase. Acute disruption of SMSr catalytic activity in cultured cells causes a rise in endoplasmic reticulum (ER) ceramides, fragmentation of ER exit sites, and induction of mitochondrial apoptosis. To address the relevance of CPE biosynthesis in vivo, we analyzed the tissue-specific distribution of CPE in mice and generated mouse lines lacking SMSr and SMS2 catalytic activity. We found that CPE levels were >300-fold lower than SM in all tissues examined. Unexpectedly, combined inactivation of SMSr and SMS2 significantly reduced, but did not eliminate, tissue-specific CPE pools and had no obvious impact on mouse development or fertility. While SMSr is widely expressed and serves as the principal CPE synthase in the brain, blocking its catalytic activity did not affect ceramide levels or secretory pathway integrity in the brain or any other tissue. Our data provide a first inventory of CPE species and CPE-biosynthetic enzymes in mammals.
Collapse
Affiliation(s)
- Andreas Bickert
- Molecular Genetics, Life, and Medical Sciences Institute University of Bonn, 53115 Bonn, Germany
| | - Christina Ginkel
- Molecular Genetics, Life, and Medical Sciences Institute University of Bonn, 53115 Bonn, Germany
| | - Matthijs Kol
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Katharina vom Dorp
- Institute of Molecular Physiology and Biotechnology of Plants, University of Bonn, 53115 Bonn, Germany
| | - Holger Jastrow
- Imaging Center Essen, Electron Microscopy Unit, University Hospital University of Duisburg-Essen, 45147 Essen, Germany
| | - Joachim Degen
- Molecular Genetics, Life, and Medical Sciences Institute University of Bonn, 53115 Bonn, Germany
| | - René L Jacobs
- Departments of Agricultural, Food, and Nutritional Science, Molecular and Cell Biology of Lipids, University of Alberta, T6G 2S2 Edmonton, Canada
| | - Dennis E Vance
- Biochemistry, University of Alberta, T6G 2S2 Edmonton, Canada
| | - Elke Winterhager
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Xian-Cheng Jiang
- Department of Anatomy and Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203
| | - Peter Dörmann
- Institute of Molecular Physiology and Biotechnology of Plants, University of Bonn, 53115 Bonn, Germany
| | - Pentti Somerharju
- Medical Biochemistry, Institute of Biomedicine, University of Helsinki, 00014 Helsinki, Finland
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Klaus Willecke
- Molecular Genetics, Life, and Medical Sciences Institute University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
205
|
Inferring biological tasks using Pareto analysis of high-dimensional data. Nat Methods 2015; 12:233-5, 3 p following 235. [PMID: 25622107 DOI: 10.1038/nmeth.3254] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 11/17/2014] [Indexed: 01/16/2023]
Abstract
We present the Pareto task inference method (ParTI; http://www.weizmann.ac.il/mcb/UriAlon/download/ParTI) for inferring biological tasks from high-dimensional biological data. Data are described as a polytope, and features maximally enriched closest to the vertices (or archetypes) allow identification of the tasks the vertices represent. We demonstrate that human breast tumors and mouse tissues are well described by tetrahedrons in gene expression space, with specific tumor types and biological functions enriched at each of the vertices, suggesting four key tasks.
Collapse
|
206
|
PKA reduces the rat and human KCa3.1 current, CaM binding, and Ca2+ signaling, which requires Ser332/334 in the CaM-binding C terminus. J Neurosci 2015; 34:13371-83. [PMID: 25274816 DOI: 10.1523/jneurosci.1008-14.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The Ca(2+)-dependent K(+) channel, KCa3.1 (KCNN4/IK/SK4), is widely expressed and contributes to cell functions that include volume regulation, migration, membrane potential, and excitability. KCa3.1 is now considered a therapeutic target for several diseases, including CNS disorders involving microglial activation; thus, we need to understand how KCa3.1 function is regulated. KCa3.1 gating and trafficking require calmodulin binding to the two ends of the CaM-binding domain (CaMBD), which also contains three conserved sites for Ser/Thr kinases. Although cAMP protein kinase (PKA) signaling is important in many cells that use KCa3.1, reports of channel regulation by PKA are inconsistent. We first compared regulation by PKA of native rat KCa3.1 channels in microglia (and the microglia cell line, MLS-9) with human KCa3.1 expressed in HEK293 cells. In all three cells, PKA activation with Sp-8-Br-cAMPS decreased the current, and this was prevented by the PKA inhibitor, PKI14-22. Inhibiting PKA with Rp-8-Br-cAMPS increased the current in microglia. Mutating the single PKA site (S334A) in human KCa3.1 abolished the PKA-dependent regulation. CaM-affinity chromatography showed that CaM binding to KCa3.1 was decreased by PKA-dependent phosphorylation of S334, and this regulation was absent in the S334A mutant. Single-channel analysis showed that PKA decreased the open probability in wild-type but not S334A mutant channels. The same decrease in current for native and wild-type expressed KCa3.1 channels (but not S334A) occurred when PKA was activated through the adenosine A2a receptor. Finally, by decreasing the KCa3.1 current, PKA activation reduced Ca(2+)-release-activated Ca(2+) entry following activation of metabotropic purinergic receptors in microglia.
Collapse
|
207
|
West DB, Pasumarthi RK, Baridon B, Djan E, Trainor A, Griffey SM, Engelhard EK, Rapp J, Li B, de Jong PJ, Lloyd KCK. A lacZ reporter gene expression atlas for 313 adult KOMP mutant mouse lines. Genome Res 2015; 25:598-607. [PMID: 25591789 PMCID: PMC4381530 DOI: 10.1101/gr.184184.114] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/05/2015] [Indexed: 02/04/2023]
Abstract
Expression of the bacterial beta-galactosidase reporter gene (lacZ) in the vector used for the Knockout Mouse Project (KOMP) is driven by the endogenous promoter of the target gene. In tissues from KOMP mice, histochemical staining for LacZ enzyme activity can be used to determine gene expression patterns. With this technique, we have produced a comprehensive resource of gene expression using both whole mount (WM) and frozen section (FS) LacZ staining in 313 unique KOMP mutant mouse lines. Of these, ∼80% of mutants showed specific staining in one or more tissues, while ∼20% showed no specific staining, ∼13% had staining in only one tissue, and ∼25% had staining in >6 tissues. The highest frequency of specific staining occurred in the brain (∼50%), male gonads (42%), and kidney (39%). The WM method was useful for rapidly identifying whole organ and some substructure staining, while the FS method often revealed substructure and cellular staining specificity. Both staining methods had >90% repeatability in biological replicates. Nonspecific LacZ staining occurs in some tissues due to the presence of bacteria or endogenous enzyme activity. However, this can be effectively distinguished from reporter gene activity by the combination of the WM and FS methods. After careful annotation, LacZ staining patterns in a high percentage of mutants revealed a unique structure-function not previously reported for many of these genes. The validation of methods for LacZ staining, annotation, and expression analysis reported here provides unique insights into the function of genes for which little is currently known.
Collapse
Affiliation(s)
- David B West
- Children's Hospital of Oakland Research Institute (CHORI), Oakland, California 94609, USA;
| | - Ravi K Pasumarthi
- Mouse Biology Program, University of California, Davis, California 95618, USA
| | - Brian Baridon
- Mouse Biology Program, University of California, Davis, California 95618, USA
| | - Esi Djan
- Mouse Biology Program, University of California, Davis, California 95618, USA
| | - Amanda Trainor
- Mouse Biology Program, University of California, Davis, California 95618, USA
| | - Stephen M Griffey
- Mouse Biology Program, University of California, Davis, California 95618, USA
| | - Eric K Engelhard
- Mouse Biology Program, University of California, Davis, California 95618, USA
| | - Jared Rapp
- Mouse Biology Program, University of California, Davis, California 95618, USA
| | - Bowen Li
- Mouse Biology Program, University of California, Davis, California 95618, USA
| | - Pieter J de Jong
- Children's Hospital of Oakland Research Institute (CHORI), Oakland, California 94609, USA
| | - K C Kent Lloyd
- Mouse Biology Program, University of California, Davis, California 95618, USA
| |
Collapse
|
208
|
Abstract
The sialic acids N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc) differ by a single oxygen atom and are widely found at the terminal position of glycans on vertebrate cell surfaces. In animals capable of synthesizing Neu5Gc, most tissues and cell types express both sialic acids, in proportions that vary between species. However, it has long been noted that Neu5Gc is consistently expressed at trace to absent levels in the brains of all vertebrates studied to date. Although several reports have claimed to find low levels of Neu5Gc-containing glycans in neural tissue, no study definitively excludes the possibility of contamination with glycans from non-neural cell types. This distribution of a molecule - prominently but variably expressed in extraneural tissues but very low or absent in the brain - is, to our knowledge, unique. The evolutionarily conserved brain-specific suppression of Neu5Gc may indicate that its presence is toxic to this organ; however, no studies to date have directly addressed this very interesting question. Here we provide a historical background to this issue and discuss potential mechanisms causing the suppression of Neu5Gc expression in brain tissue, as well as mechanisms by which Neu5Gc may exert the presumed toxicity. Finally, we discuss future approaches towards understanding the mechanisms and implications of this unusual finding.
Collapse
Affiliation(s)
- Leela R L Davies
- Glycobiology Research and Training Center, Center for Academic Research and Training in Anthropogeny, Biomedical Sciences Graduate Program, Departments of Medicine and Cellular and Molecular Medicine, University of California at San Diego, 9500 Gilman Dr., MC 0687, La Jolla, CA, 92093-0687, USA
| | | |
Collapse
|
209
|
Predicting the phenotypic values of physiological traits using SNP genotype and gene expression data in mice. PLoS One 2014; 9:e115532. [PMID: 25541966 PMCID: PMC4277360 DOI: 10.1371/journal.pone.0115532] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 11/25/2014] [Indexed: 01/22/2023] Open
Abstract
Predicting phenotypes using genome-wide genetic variation and gene expression data is useful in several fields, such as human biology and medicine, as well as in crop and livestock breeding. However, for phenotype prediction using gene expression data for mammals, studies remain scarce, as the available data on gene expression profiling are currently limited. By integrating a few sources of relevant data that are available in mice, this study investigated the accuracy of phenotype prediction for several physiological traits. Gene expression data from two tissues as well as single nucleotide polymorphisms (SNPs) were used. For the studied traits, the variance of the effects of the expression levels was more likely to differ among the genes than were the effects of SNPs. For the glucose concentration, the total cholesterol amount, and the total tidal volume, the accuracy by cross validation tended to be higher when the gene expression data rather than the SNP genotype data were used, and a statistically significant increase in the accuracy was obtained when the gene expression data from the liver were used alone or jointly with the SNP genotype data. For these traits, there were no additional gains in accuracy from using the gene expression data of both the liver and lung compared to that of individual use. The accuracy of prediction using genes that were selected differently was examined; the use of genes with a higher tissue specificity tended to result in an accuracy that was similar to or greater than that associated with the use of all of the available genes for traits such as the glucose concentration and total cholesterol amount. Although relatively few animals were evaluated, the current results suggest that gene expression levels could be used as explanatory variables. However, further studies are essential to confirm our findings using additional animal samples.
Collapse
|
210
|
PET/CT imaging reveals unrivaled placental avidity for glucose compared to other tissues. Placenta 2014; 36:115-20. [PMID: 25555498 DOI: 10.1016/j.placenta.2014.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/05/2014] [Accepted: 12/12/2014] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The goal of this study was to define the kinetics of glucose transport from maternal blood to placenta to fetus using real time imaging. METHODS Positron emission tomography (PET) imaging of the glucose-tracer [(18)F]fluorodeoxyglucose (FDG) was used to temporally and spatially define, in vivo, the kinetics of glucose transport from maternal blood into placentae and fetuses, in the late gestational gravid rat. Computed tomography (CT), with intravenous contrast, co-registered to the PET images allowed anatomic differentiation of placentae from fetal and maternal tissues. RESULTS FDG was rapidly taken up by placentae and subsequently appeared in fetuses with minimal temporal lag. FDG standardized uptake values in placentae and fetuses approached that of maternal brain. In both anesthetized and awake dams, one quarter of the administered FDG ultimately was accrued in the collective fetuses and placentae. Accordingly, kinetic modeling demonstrated that the placentae had very high avidity for FDG, 2-fold greater than that of the fetus and maternal brain, when accounting for the fact that fetal FDG necessarily must first be taken up by placentae. Consistent with this, placental expression of glucose transporter 1 exceeded that of all other tissues. DISCUSSION Fetal and placental tissues place a substantial glucose metabolic burden on the mother, owing to very high avidity of placentae for glucose coupled with the large relative mass of fetal and placental tissues. CONCLUSIONS The placenta has a tremendous capacity to uptake and transport glucose. PET/CT imaging is an ideal means to study metabolite transport kinetics in the fetoplacental unit.
Collapse
|
211
|
Sansom SN, Shikama-Dorn N, Zhanybekova S, Nusspaumer G, Macaulay IC, Deadman ME, Heger A, Ponting CP, Holländer GA. Population and single-cell genomics reveal the Aire dependency, relief from Polycomb silencing, and distribution of self-antigen expression in thymic epithelia. Genome Res 2014; 24:1918-31. [PMID: 25224068 PMCID: PMC4248310 DOI: 10.1101/gr.171645.113] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 09/12/2014] [Indexed: 12/18/2022]
Abstract
Promiscuous gene expression (PGE) by thymic epithelial cells (TEC) is essential for generating a diverse T cell antigen receptor repertoire tolerant to self-antigens, and thus for avoiding autoimmunity. Nevertheless, the extent and nature of this unusual expression program within TEC populations and single cells are unknown. Using deep transcriptome sequencing of carefully identified mouse TEC subpopulations, we discovered a program of PGE that is common between medullary (m) and cortical TEC, further elaborated in mTEC, and completed in mature mTEC expressing the autoimmune regulator gene (Aire). TEC populations are capable of expressing up to 19,293 protein-coding genes, the highest number of genes known to be expressed in any cell type. Remarkably, in mouse mTEC, Aire expression alone positively regulates 3980 tissue-restricted genes. Notably, the tissue specificities of these genes include known targets of autoimmunity in human AIRE deficiency. Led by the observation that genes induced by Aire expression are generally characterized by a repressive chromatin state in somatic tissues, we found these genes to be strongly associated with H3K27me3 marks in mTEC. Our findings are consistent with AIRE targeting and inducing the promiscuous expression of genes previously epigenetically silenced by Polycomb group proteins. Comparison of the transcriptomes of 174 single mTEC indicates that genes induced by Aire expression are transcribed stochastically at low cell frequency. Furthermore, when present, Aire expression-dependent transcript levels were 16-fold higher, on average, in individual TEC than in the mTEC population.
Collapse
Affiliation(s)
- Stephen N Sansom
- MRC Computational Genomics Analysis and Training Programme, MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, United Kingdom;
| | - Noriko Shikama-Dorn
- Paediatric Immunology, Department of Biomedicine, University of Basel, and The Basel University Children's Hospital, Basel, 4058, Switzerland
| | - Saule Zhanybekova
- Paediatric Immunology, Department of Biomedicine, University of Basel, and The Basel University Children's Hospital, Basel, 4058, Switzerland
| | - Gretel Nusspaumer
- Paediatric Immunology, Department of Biomedicine, University of Basel, and The Basel University Children's Hospital, Basel, 4058, Switzerland
| | - Iain C Macaulay
- Wellcome Trust Sanger Institute-EBI Single Cell Genomics Centre, Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Mary E Deadman
- Developmental Immunology, Department of Paediatrics, and the Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Andreas Heger
- MRC Computational Genomics Analysis and Training Programme, MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, United Kingdom
| | - Chris P Ponting
- MRC Computational Genomics Analysis and Training Programme, MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, United Kingdom; Wellcome Trust Sanger Institute-EBI Single Cell Genomics Centre, Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Georg A Holländer
- Paediatric Immunology, Department of Biomedicine, University of Basel, and The Basel University Children's Hospital, Basel, 4058, Switzerland; Developmental Immunology, Department of Paediatrics, and the Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| |
Collapse
|
212
|
Beck S, Lee BK, Rhee C, Song J, Woo AJ, Kim J. CpG island-mediated global gene regulatory modes in mouse embryonic stem cells. Nat Commun 2014; 5:5490. [PMID: 25405324 PMCID: PMC4236720 DOI: 10.1038/ncomms6490] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 10/03/2014] [Indexed: 12/13/2022] Open
Abstract
Both transcriptional and epigenetic regulations are fundamental for the control of eukaryotic gene expression. Here we perform a compendium analysis of >200 large sequencing data sets to elucidate the regulatory logic of global gene expression programs in mouse embryonic stem (ES) cells. We define four major classes of DNA-binding proteins (Core, PRC, MYC and CTCF) based on their target co-occupancy, and discover reciprocal regulation between the MYC and PRC classes for the activity of nearly all genes under the control of the CpG island (CGI)-containing promoters. This CGI-dependent regulatory mode explains the functional segregation between CGI-containing and CGI-less genes during early development. By defining active enhancers based on the co-occupancy of the Core class, we further demonstrate their additive roles in CGI-containing gene expression and cell type-specific roles in CGI-less gene expression. Altogether, our analyses provide novel insights into previously unknown CGI-dependent global gene regulatory modes.
Collapse
Affiliation(s)
- Samuel Beck
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Bum-Kyu Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Catherine Rhee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Jawon Song
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, Texas 78758, USA
| | - Andrew J Woo
- School of Medicine and Pharmacology, Royal Perth Hospital Unit, The University of Western Australia, Perth, WA 6000, Australia
| | - Jonghwan Kim
- 1] Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA [2] Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, USA [3] Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas 78712, USA
| |
Collapse
|
213
|
Garcia RA, Yan M, Search D, Zhang R, Carson NL, Ryan CS, Smith-Monroy C, Zheng J, Chen J, Kong Y, Tang H, Hellings SE, Wardwell-Swanson J, Dinchuk JE, Psaltis GC, Gordon DA, Glunz PW, Gargalovic PS. P2Y6 receptor potentiates pro-inflammatory responses in macrophages and exhibits differential roles in atherosclerotic lesion development. PLoS One 2014; 9:e111385. [PMID: 25360548 PMCID: PMC4216081 DOI: 10.1371/journal.pone.0111385] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/23/2014] [Indexed: 12/15/2022] Open
Abstract
Background P2Y6, a purinergic receptor for UDP, is enriched in atherosclerotic lesions and is implicated in pro-inflammatory responses of key vascular cell types and macrophages. Evidence for its involvement in atherogenesis, however, has been lacking. Here we use cell-based studies and three murine models of atherogenesis to evaluate the impact of P2Y6 deficiency on atherosclerosis. Methodology/Principal Findings Cell-based studies in 1321N1 astrocytoma cells, which lack functional P2Y6 receptors, showed that exogenous expression of P2Y6 induces a robust, receptor- and agonist-dependent secretion of inflammatory mediators IL-8, IL-6, MCP-1 and GRO1. P2Y6-mediated inflammatory responses were also observed, albeit to a lesser extent, in macrophages endogenously expressing P2Y6 and in acute peritonitis models of inflammation. To evaluate the role of P2Y6 in atherosclerotic lesion development, we used P2Y6-deficient mice in three mouse models of atherosclerosis. A 43% reduction in aortic arch plaque was observed in high fat-fed LDLR knockout mice lacking P2Y6 receptors in bone marrow-derived cells. In contrast, no effect on lesion development was observed in fat-fed whole body P2Y6xLDLR double knockout mice. Interestingly, in a model of enhanced vascular inflammation using angiotensin II, P2Y6 deficiency enhanced formation of aneurysms and exhibited a trend towards increased atherosclerosis in the aorta of LDLR knockout mice. Conclusions P2Y6 receptor augments pro-inflammatory responses in macrophages and exhibits a pro-atherogenic role in hematopoietic cells. However, the overall impact of whole body P2Y6 deficiency on atherosclerosis appears to be modest and could reflect additional roles of P2Y6 in vascular disease pathophysiologies, such as aneurysm formation.
Collapse
Affiliation(s)
- Ricardo A. Garcia
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Mujing Yan
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Debra Search
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Rongan Zhang
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Nancy L. Carson
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Carol S. Ryan
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Constance Smith-Monroy
- Applied Genomics, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Joanna Zheng
- Pharmaceutical Compound Optimization: Discovery Toxicology, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Jian Chen
- Applied Genomics, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Yan Kong
- Lead Evaluation, Bristol-Myers Squibb Company, Lawrenceville, New Jersey, United States of America
| | - Huaping Tang
- Lead Evaluation, Bristol-Myers Squibb Company, Lawrenceville, New Jersey, United States of America
| | - Samuel E. Hellings
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Judith Wardwell-Swanson
- Applied Genomics, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Joseph E. Dinchuk
- Applied Genomics, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - George C. Psaltis
- Veterinary Sciences, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - David A. Gordon
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Peter W. Glunz
- Discovery Chemistry, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Peter S. Gargalovic
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
214
|
Español AJ, Maddaleno MO, Lombardi MG, Cella M, Martínez Pulido P, Sales ME. Treatment with LPS plus INF-γ induces the expression and function of muscarinic acetylcholine receptors, modulating NIH3T3 cell proliferation: participation of NOS and COX. Br J Pharmacol 2014; 171:5154-67. [PMID: 24990429 DOI: 10.1111/bph.12834] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 06/17/2014] [Accepted: 06/24/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE LPS and IFN-γ are potent stimuli of inflammation, a process in which fibroblasts are frequently involved. We analysed the effect of treatment with LPS plus IFN-γ on the expression and function of muscarinic acetylcholine receptors in NIH3T3 fibroblasts with regards to proliferation of these cells. We also investigated the participation of NOS and COX, and the role of NF-κB in this process. EXPERIMENTAL APPROACH NIH3T3 cells were treated with LPS (10 ng·mL(-1)) plus IFN-γ (0.5 ng·mL(-1)) for 72 h (iNIH3T3 cells). Cell proliferation was evaluated with MTT and protein expression by Western blot analysis. NOS and COX activities were measured by the Griess method and radioimmunoassay respectively. KEY RESULTS The cholinoceptor agonist carbachol was more effective at stimulating proliferation in iNIH3T3 than in NIH3T3 cells, probably due to the de novo induction of M3 and M5 muscarinic receptors independently of NF-κB activation. iNIH3T3 cells produced higher amounts of NO and PGE2 than NIH3T3 cells, concomitantly with an up-regulation of NOS1 and COX-2, and with the de novo induction of NOS2/3 in inflamed cells. We also found a positive feedback between NOS and COX that could potentiate inflammation. CONCLUSIONS AND IMPLICATIONS Inflammation induced the expression of muscarinic receptors and, therefore,stimulated carbachol-induced proliferation of fibroblasts. Inflammation also up-regulated the expression of NOS and COX-2, thus potentiating the effect of carbachol on NO and PGE2 production. A positive crosstalk between NOS and COX triggered by carbachol in inflamed cells points to muscarinic receptors as potential therapeutic targets in inflammation.
Collapse
Affiliation(s)
- A J Español
- Facultad de Medicina, Universidad de Buenos Aires, Centro de Estudios Farmacológicos y Botánicos (CEFYBO)-CONICET, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
215
|
Tseng HY, Thorausch N, Ziegler T, Meves A, Fässler R, Böttcher RT. Sorting Nexin 31 Binds Multiple β Integrin Cytoplasmic Domains and Regulates β1 Integrin Surface Levels and Stability. J Mol Biol 2014; 426:3180-3194. [DOI: 10.1016/j.jmb.2014.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 06/17/2014] [Accepted: 07/03/2014] [Indexed: 11/24/2022]
|
216
|
Botman D, Tigchelaar W, Van Noorden CJF. Determination of phosphate-activated glutaminase activity and its kinetics in mouse tissues using metabolic mapping (quantitative enzyme histochemistry). J Histochem Cytochem 2014; 62:813-26. [PMID: 25163927 PMCID: PMC4230542 DOI: 10.1369/0022155414551177] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Phosphate-activated glutaminase (PAG) converts glutamine to glutamate as part of the glutaminolysis pathway in mitochondria. Two genes, GLS1 and GLS2, which encode for kidney-type PAG and liver-type PAG, respectively, differ in their tissue-specific activities and kinetics. Tissue-specific PAG activity and its kinetics were determined by metabolic mapping using a tetrazolium salt and glutamate dehydrogenase as an auxiliary enzyme in the presence of various glutamine concentrations. In kidney and brain, PAG showed Michaelis-Menten kinetics with a Km of 0.6 mM glutamine and a Vmax of 1.1 µmol/mL/min when using 5 mM glutamine. PAG activity was high in the kidney cortex and inner medulla but low in the outer medulla, papillary tip, glomeruli and the lis of Henle. In brain tissue sections, PAG was active in the grey matter and inactive in myelin-rich regions. Liver PAG showed allosteric regulation with a Km of 11.6 mM glutamine and a Vmax of 0.5 µmol/mL/min when using 20 mM glutamine. The specificity of the method was shown after incubation of brain tissue sections with the PAG inhibitor 6-diazo-5-oxo-L-norleucine. PAG activity was decreased to 22% in the presence of 2 mM 6-diazo-5-oxo-L-norleucine. At low glutamine concentrations, PAG activity was higher in periportal regions, indicating a lower Km for periportal PAG. When compared with liver, kidney and brain, other tissues showed 3-fold to 6-fold less PAG activity. In conclusion, PAG is mainly active in mouse kidney, brain and liver, and shows different kinetics depending on which type of PAG is expressed.
Collapse
Affiliation(s)
- Dennis Botman
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (DB, WT, CJFVN)
| | - Wikky Tigchelaar
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (DB, WT, CJFVN)
| | - Cornelis J F Van Noorden
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (DB, WT, CJFVN)
| |
Collapse
|
217
|
Raeven RHM, Brummelman J, Pennings JLA, Nijst OEM, Kuipers B, Blok LER, Helm K, van Riet E, Jiskoot W, van Els CACM, Han WGH, Kersten GFA, Metz B. Molecular signatures of the evolving immune response in mice following a Bordetella pertussis infection. PLoS One 2014; 9:e104548. [PMID: 25137043 PMCID: PMC4138111 DOI: 10.1371/journal.pone.0104548] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/14/2014] [Indexed: 12/21/2022] Open
Abstract
Worldwide resurgence of pertussis necessitates the need for improvement of pertussis vaccines and vaccination strategies. Since natural infections induce a longer-lasting immunity than vaccinations, detailed knowledge of the immune responses following natural infection can provide important clues for such improvement. The purpose was to elucidate the kinetics of the protective immune response evolving after experimental Bordetella pertussis (B. pertussis) infection in mice. Data were collected from (i) individual analyses, i.e. microarray, flow cytometry, multiplex immunoassays, and bacterial clearance; (ii) twelve time points during the infection; and (iii) different tissues involved in the immune responses, i.e. lungs, spleen and blood. Combined data revealed detailed insight in molecular and cellular sequence of events connecting different phases (innate, bridging and adaptive) of the immune response following the infection. We detected a prolonged acute phase response, broad pathogen recognition, and early gene signatures of subsequent T-cell recruitment in the lungs. Activation of particular transcription factors and specific cell markers provided insight into the time course of the transition from innate towards adaptive immune responses, which resulted in a broad spectrum of systemic antibody subclasses and splenic Th1/Th17 memory cells against B. pertussis. In addition, signatures preceding the local generation of Th1 and Th17 cells as well as IgA in the lungs, considered key elements in protection against B. pertussis, were established. In conclusion, molecular and cellular immunological processes in response to live B. pertussis infection were unraveled, which may provide guidance in selecting new vaccine candidates that should evoke local and prolonged protective immune responses.
Collapse
Affiliation(s)
- René H. M. Raeven
- Intravacc, Bilthoven, The Netherlands
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Jolanda Brummelman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Jeroen L. A. Pennings
- Centre for Health Protection (GZB), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - Betsy Kuipers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - Kina Helm
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - Wim Jiskoot
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Cecile A. C. M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Wanda G. H. Han
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Gideon F. A. Kersten
- Intravacc, Bilthoven, The Netherlands
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | | |
Collapse
|
218
|
Circadian regulation of myocardial sarcomeric Titin-cap (Tcap, telethonin): identification of cardiac clock-controlled genes using open access bioinformatics data. PLoS One 2014; 9:e104907. [PMID: 25121604 PMCID: PMC4133362 DOI: 10.1371/journal.pone.0104907] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/17/2014] [Indexed: 12/26/2022] Open
Abstract
Circadian rhythms are important for healthy cardiovascular physiology and are regulated at the molecular level by a circadian clock mechanism. We and others previously demonstrated that 9–13% of the cardiac transcriptome is rhythmic over 24 h daily cycles; the heart is genetically a different organ day versus night. However, which rhythmic mRNAs are regulated by the circadian mechanism is not known. Here, we used open access bioinformatics databases to identify 94 transcripts with expression profiles characteristic of CLOCK and BMAL1 targeted genes, using the CircaDB website and JTK_Cycle. Moreover, 22 were highly expressed in the heart as determined by the BioGPS website. Furthermore, 5 heart-enriched genes had human/mouse conserved CLOCK:BMAL1 promoter binding sites (E-boxes), as determined by UCSC table browser, circadian mammalian promoter/enhancer database PEDB, and the European Bioinformatics Institute alignment tool (EMBOSS). Lastly, we validated findings by demonstrating that Titin cap (Tcap, telethonin) was targeted by transcriptional activators CLOCK and BMAL1 by showing 1) Tcap mRNA and TCAP protein had a diurnal rhythm in murine heart; 2) cardiac Tcap mRNA was rhythmic in animals kept in constant darkness; 3) Tcap and control Per2 mRNA expression and cyclic amplitude were blunted in ClockΔ19/Δ19 hearts; 4) BMAL1 bound to the Tcap promoter by ChIP assay; 5) BMAL1 bound to Tcap promoter E-boxes by biotinylated oligonucleotide assay; and 6) CLOCK and BMAL1 induced tcap expression by luciferase reporter assay. Thus this study identifies circadian regulated genes in silico, with validation of Tcap, a critical regulator of cardiac Z-disc sarcomeric structure and function.
Collapse
|
219
|
Botman D, Tigchelaar W, Van Noorden CJF. Determination of glutamate dehydrogenase activity and its kinetics in mouse tissues using metabolic mapping (quantitative enzyme histochemistry). J Histochem Cytochem 2014; 62:802-12. [PMID: 25124006 PMCID: PMC4230541 DOI: 10.1369/0022155414549071] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Glutamate dehydrogenase (GDH) catalyses the reversible conversion of glutamate into α-ketoglutarate with the concomitant reduction of NAD(P)(+) to NAD(P)H or vice versa. GDH activity is subject to complex allosteric regulation including substrate inhibition. To determine GDH kinetics in situ, we assessed the effects of various glutamate concentrations in combination with either the coenzyme NAD(+) or NADP(+) on GDH activity in mouse liver cryostat sections using metabolic mapping. NAD(+)-dependent GDH V(max) was 2.5-fold higher than NADP(+)-dependent V(max), whereas the K(m) was similar, 1.92 mM versus 1.66 mM, when NAD(+) or NADP(+) was used, respectively. With either coenzyme, V(max) was determined at 10 mM glutamate and substrate inhibition was observed at higher glutamate concentrations with a K(i) of 12.2 and 3.95 for NAD(+) and NADP(+) used as coenzyme, respectively. NAD(+)- and NADP(+)-dependent GDH activities were examined in various mouse tissues. GDH activity was highest in liver and much lower in other tissues. In all tissues, the highest activity was found when NAD(+) was used as a coenzyme. In conclusion, GDH activity in mice is highest in the liver with NAD(+) as a coenzyme and highest GDH activity was determined at a glutamate concentration of 10 mM.
Collapse
Affiliation(s)
- Dennis Botman
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (DB, WT, CJFVN)
| | - Wikky Tigchelaar
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (DB, WT, CJFVN)
| | - Cornelis J F Van Noorden
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (DB, WT, CJFVN)
| |
Collapse
|
220
|
Bennett TM, Mackay DS, Siegfried CJ, Shiels A. Mutation of the melastatin-related cation channel, TRPM3, underlies inherited cataract and glaucoma. PLoS One 2014; 9:e104000. [PMID: 25090642 PMCID: PMC4121231 DOI: 10.1371/journal.pone.0104000] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 07/04/2014] [Indexed: 11/19/2022] Open
Abstract
Inherited forms of cataract are a clinically important and genetically heterogeneous cause of visual impairment that usually present at an early age with or without systemic and/or other ocular abnormalities. Here we have identified a new locus for inherited cataract and high-tension glaucoma with variable anterior segment defects, and characterized an underlying mutation in the gene coding for transient receptor potential cation channel, subfamily M, member-3 (TRPM3, melastatin-2). Genome-wide linkage analysis mapped the ocular disease locus to the pericentric region of human chromosome 9. Whole exome and custom-target next-generation sequencing detected a heterozygous A-to-G transition in exon-3 of TRPM3 that co-segregated with disease. As a consequence of alternative splicing this missense mutation was predicted to result in the substitution of isoleucine-to-methionine at codon 65 (c.195A>G; p.I65 M) of TRPM3 transcript variant 9, and at codon 8 (c.24A>G; p.I8 M) of a novel TRPM3 transcript variant expressed in human lens. In both transcript variants the I-to-M substitution was predicted in silico to exert damaging effects on protein function. Furthermore, transient expression studies of a recombinant TRPM3-GFP reporter product predicted that the I-to-M substitution introduced an alternative translation start-site located 89 codons upstream from the native initiator methionine found in eight other TRPM3 transcript variants (1-8). Collectively, these studies have provided the first evidence that TRPM3 is associated with inherited ocular disease in humans, and further provide support for the important role of this cation channel in normal eye development.
Collapse
Affiliation(s)
- Thomas M. Bennett
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Donna S. Mackay
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Carla J. Siegfried
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Alan Shiels
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
221
|
Huang Q, Feng D, Liu K, Wang P, Xiao H, Wang Y, Zhang S, Liu Z. A medium-chain fatty acid receptor Gpr84 in zebrafish: expression pattern and roles in immune regulation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 45:252-258. [PMID: 24704214 DOI: 10.1016/j.dci.2014.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 03/24/2014] [Accepted: 03/25/2014] [Indexed: 06/03/2023]
Abstract
Gpr84 was recently identified as a receptor for medium-chain fatty acids, but its functions remain to be clarified. We reported the identification of a zebrafish Gpr84 homologue (zGpr84), which has a higher gene expression in the tissues of intestine, heart and liver. During embryogenesis, zGpr84 is maternally expressed and a significant increase is observed at segmentation period, and it is mainly restricted to the head region, pectoral fins, branchial arches, intestine and lateral line neuromast. Fasting or treatment with lipopolysaccharide (LPS) can induce significant up-regulation of zGpr84. We further demonstrated that zGpr84 is involved in the accumulation of lipid droplets in cells. Moreover, undecanoic acid (UA) can amplify LPS induced production of the proinflammatory cytokine IL-12 p40 through zGpr84, supporting the proposal that Gpr84 may play a role in directly linking fatty acid metabolism to immunological regulation. The resulting data in fish lay a foundation for a comprehensive exploration of the functions and evolution of Gpr84.
Collapse
Affiliation(s)
- Qiaoyan Huang
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Dong Feng
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Kai Liu
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Peng Wang
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Hongyan Xiao
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Ying Wang
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Shicui Zhang
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| | - Zhenhui Liu
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China.
| |
Collapse
|
222
|
Young GR, Mavrommatis B, Kassiotis G. Microarray analysis reveals global modulation of endogenous retroelement transcription by microbes. Retrovirology 2014; 11:59. [PMID: 25063042 PMCID: PMC4222864 DOI: 10.1186/1742-4690-11-59] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 07/15/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A substantial proportion of both the mouse and human genomes comprise of endogenous retroelements (REs), which include endogenous retroviruses. Over evolutionary time, REs accumulate inactivating mutations or deletions and thus lose the ability to replicate. Additionally, REs can be transcriptionally repressed by dedicated mechanisms of the host. Nevertheless, many of them still possess and express intact open reading frames, and their transcriptional activity has been associated with many physiological and pathological processes of the host. However, this association remains tenuous due to incomplete understanding of the mechanism by which RE transcription is regulated. Here, we use a bioinformatics tool to examine RE transcriptional activity, measured by microarrays, in murine and human immune cells responding to microbial stimulation. RESULTS Immune cell activation by microbial signals in vitro caused extensive changes in the transcription not only of the host genes involved in the immune response, but also of numerous REs. Modulated REs were frequently found near or embedded within similarly-modulated host genes. Focusing on probes reporting single-integration, intergenic REs, revealed extensive transcriptional responsiveness of these elements to microbial signals. Microbial stimulation modulated RE expression in a cell-intrinsic manner. In line with these results, the transcriptional activity of numerous REs followed characteristics in different tissues according to exposure to environmental microbes and was further heavily altered during viral infection or imbalances with intestinal microbiota, both in mice and humans. CONCLUSIONS Together, these results highlight the utility of improved methodologies in assessing RE transcription profiles in both archived and new microarray data sets. More importantly, application of this methodology suggests that immune activation, as a result of infection with pathogens or dysbiosis with commensal microbes, causes global modulation of RE transcription. RE responsiveness to external stimuli should, therefore, be considered in any association between RE transcription and disease.
Collapse
Affiliation(s)
| | | | - George Kassiotis
- Division of Immunoregulation, MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK.
| |
Collapse
|
223
|
Dittmar WJ, McIver L, Michalak P, Garner HR, Valdez G. EvoCor: a platform for predicting functionally related genes using phylogenetic and expression profiles. Nucleic Acids Res 2014; 42:W72-5. [PMID: 24848012 PMCID: PMC4086105 DOI: 10.1093/nar/gku442] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 05/03/2014] [Accepted: 05/07/2014] [Indexed: 01/18/2023] Open
Abstract
The wealth of publicly available gene expression and genomic data provides unique opportunities for computational inference to discover groups of genes that function to control specific cellular processes. Such genes are likely to have co-evolved and be expressed in the same tissues and cells. Unfortunately, the expertise and computational resources required to compare tens of genomes and gene expression data sets make this type of analysis difficult for the average end-user. Here, we describe the implementation of a web server that predicts genes involved in affecting specific cellular processes together with a gene of interest. We termed the server 'EvoCor', to denote that it detects functional relationships among genes through evolutionary analysis and gene expression correlation. This web server integrates profiles of sequence divergence derived by a Hidden Markov Model (HMM) and tissue-wide gene expression patterns to determine putative functional linkages between pairs of genes. This server is easy to use and freely available at http://pilot-hmm.vbi.vt.edu/.
Collapse
Affiliation(s)
- W James Dittmar
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Lauren McIver
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Pawel Michalak
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Harold R Garner
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Gregorio Valdez
- Virginia Tech Carilion Research Institute, Roanoke, VA 24016, USA; Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
224
|
Genomic and transcriptomic analyses match medulloblastoma mouse models to their human counterparts. Acta Neuropathol 2014; 128:123-36. [PMID: 24871706 DOI: 10.1007/s00401-014-1297-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 05/08/2014] [Accepted: 05/19/2014] [Indexed: 12/15/2022]
Abstract
Medulloblastoma is a malignant embryonal brain tumor with highly variable outcome. In order to study the biology of this tumor and to perform preclinical treatment studies, a lot of effort has been put into the generation of appropriate mouse models. The usage of these models, however, has become debatable with the advances in human medulloblastoma subgrouping. This study brings together multiple relevant mouse models and matches genetic alterations and gene expression data of 140 murine tumors with 423 human medulloblastomas in a global way. Using AGDEX analysis and k-means clustering, we show that the Blbp-cre::Ctnnb1(ex3)(Fl/+)Trp53 (Fl/Fl) mouse model fits well to human WNT medulloblastoma, and that, among various Myc- or Mycn-based mouse medulloblastomas, tumors in Glt1-tTA::TRE-MYCN/Luc mice proved to be most specific for human group 3 medulloblastoma. None of the analyzed models displayed a significant match to group 4 tumors. Intriguingly, mice with Ptch1 or Smo mutations selectively modeled SHH medulloblastomas of adulthood, although such mutations occur in all human age groups. We therefore suggest that the infantile or adult gene expression pattern of SHH MBs are not solely determined by specific mutations. This is supported by the observation that human medulloblastomas with PTCH1 mutations displayed more similarities to PTCH1 wild-type tumors of the same age group than to PTCH1-mutated tumors of the other age group. Together, we provide novel insights into previously unrecognized specificity of distinct models and suggest these findings as a solid basis to choose the appropriate model for preclinical studies on medulloblastoma.
Collapse
|
225
|
Quantitative epigenetic co-variation in CpG islands and co-regulation of developmental genes. Sci Rep 2014; 3:2576. [PMID: 23999385 PMCID: PMC6505400 DOI: 10.1038/srep02576] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/16/2013] [Indexed: 12/21/2022] Open
Abstract
The genome-wide variation of multiple epigenetic modifications in CpG islands (CGIs) and the interactions between them are of great interest. Here, we optimized an entropy-based strategy to quantify variation of epigenetic modifications and explored their interaction across mouse embryonic stem cells, neural precursor cells and brain. Our results showed that four epigenetic modifications (DNA methylation, H3K4me2, H3K4me3 and H3K27me3) of CGIs in the mouse genome undergo combinatorial variation during neuron differentiation. DNA methylation variation was positively correlated with H3K27me3 variation, and negatively correlated with H3K4me2/3 variation. We identified 5,194 CGIs differentially modified by epigenetic modifications (DEM-CGIs). Among them, the differentially DNA methylated CGIs overlapped significantly with the CGIs differentially modified by H3K27me3. Moreover, DEM-CGIs may contribute to co-regulation of related developmental genes including core transcription factors. Our entropy-based strategy provides an effective way of investigating dynamic cross-talk among epigenetic modifications in various biological processes at the macro scale.
Collapse
|
226
|
A systems biology approach to the analysis of subset-specific responses to lipopolysaccharide in dendritic cells. PLoS One 2014; 9:e100613. [PMID: 24949855 PMCID: PMC4065045 DOI: 10.1371/journal.pone.0100613] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 05/28/2014] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) are critical for regulating CD4 and CD8 T cell immunity, controlling Th1, Th2, and Th17 commitment, generating inducible Tregs, and mediating tolerance. It is believed that distinct DC subsets have evolved to control these different immune outcomes. However, how DC subsets mount different responses to inflammatory and/or tolerogenic signals in order to accomplish their divergent functions remains unclear. Lipopolysaccharide (LPS) provides an excellent model for investigating responses in closely related splenic DC subsets, as all subsets express the LPS receptor TLR4 and respond to LPS in vitro. However, previous studies of the LPS-induced DC transcriptome have been performed only on mixed DC populations. Moreover, comparisons of the in vivo response of two closely related DC subsets to LPS stimulation have not been reported in the literature to date. We compared the transcriptomes of murine splenic CD8 and CD11b DC subsets after in vivo LPS stimulation, using RNA-Seq and systems biology approaches. We identified subset-specific gene signatures, which included multiple functional immune mediators unique to each subset. To explain the observed subset-specific differences, we used a network analysis approach. While both DC subsets used a conserved set of transcription factors and major signalling pathways, the subsets showed differential regulation of sets of genes that ‘fine-tune’ the network Hubs expressed in common. We propose a model in which signalling through common pathway components is ‘fine-tuned’ by transcriptional control of subset-specific modulators, thus allowing for distinct functional outcomes in closely related DC subsets. We extend this analysis to comparable datasets from the literature and confirm that our model can account for cell subset-specific responses to LPS stimulation in multiple subpopulations in mouse and man.
Collapse
|
227
|
Gantier MP. Processing of Double-Stranded RNA in Mammalian Cells: A Direct Antiviral Role? J Interferon Cytokine Res 2014; 34:469-77. [DOI: 10.1089/jir.2014.0003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Michael P. Gantier
- Centre for Cancer Research, MIMR-PHI Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| |
Collapse
|
228
|
Mesner LD, Ray B, Hsu YH, Manichaikul A, Lum E, Bryda EC, Rich SS, Rosen CJ, Criqui MH, Allison M, Budoff MJ, Clemens TL, Farber CR. Bicc1 is a genetic determinant of osteoblastogenesis and bone mineral density. J Clin Invest 2014; 124:2736-49. [PMID: 24789909 PMCID: PMC4038574 DOI: 10.1172/jci73072] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Patient bone mineral density (BMD) predicts the likelihood of osteoporotic fracture. While substantial progress has been made toward elucidating the genetic determinants of BMD, our understanding of the factors involved remains incomplete. Here, using a systems genetics approach in the mouse, we predicted that bicaudal C homolog 1 (Bicc1), which encodes an RNA-binding protein, is responsible for a BMD quantitative trait locus (QTL) located on murine chromosome 10. Consistent with this prediction, mice heterozygous for a null allele of Bicc1 had low BMD. We used a coexpression network-based approach to determine how Bicc1 influences BMD. Based on this analysis, we inferred that Bicc1 was involved in osteoblast differentiation and that polycystic kidney disease 2 (Pkd2) was a downstream target of Bicc1. Knock down of Bicc1 and Pkd2 impaired osteoblastogenesis, and Bicc1 deficiency-dependent osteoblast defects were rescued by Pkd2 overexpression. Last, in 2 human BMD genome-wide association (GWAS) meta-analyses, we identified SNPs in BICC1 and PKD2 that were associated with BMD. These results, in both mice and humans, identify Bicc1 as a genetic determinant of osteoblastogenesis and BMD and suggest that it does so by regulating Pkd2 transcript levels.
Collapse
Affiliation(s)
- Larry D. Mesner
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Brianne Ray
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Yi-Hsiang Hsu
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Eric Lum
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth C. Bryda
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Clifford J. Rosen
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Michael H. Criqui
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Matthew Allison
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Matthew J. Budoff
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Thomas L. Clemens
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| | - Charles R. Farber
- Center for Public Health Genomics, University of Virginia,
Charlottesville, Virginia, USA. Hebrew SeniorLife Institute for Aging
Research and Harvard Medical School, Boston, Massachusetts, USA. Molecular
and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston,
Massachusetts, USA. Department of Veterinary Pathobiology, University of
Missouri, Columbia, Missouri, USA. Departments of Public Health Sciences and
Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA.
Maine Medical Center Research Institute, Scarborough, Maine, USA.
Division of Preventive Medicine, UCSD, La Jolla, California, USA.
Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,
Torrance, California, USA. Department of Orthopaedic Surgery, Johns Hopkins
School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
229
|
Identification of hematopoietic-specific regulatory elements from the CD45 gene and use for lentiviral tracking of transplanted cells. Exp Hematol 2014; 42:761-72.e1-10. [PMID: 24852660 DOI: 10.1016/j.exphem.2014.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/09/2014] [Accepted: 05/13/2014] [Indexed: 02/06/2023]
Abstract
The development of a hematopoietic reporter is crucial for determining the fate of lineages derived from cell-based therapies. A marking system will enable safer embryonic stem and induced pluripotent stem cell-based derivation of blood lineages and facilitate the development of efficient cellular reprogramming strategies based on direct fibroblast conversion. Here we report that the protein tyrosine phosphatase CD45 is an ideal candidate gene on which to base a hematopoietic reporter. CD45 regulatory elements were discovered by analyzing transcription factor chromatin occupancy (ChIP-seq) and promoter nuclease sensitivity (DNase-seq) to identify minimally sufficient sequences required for expression. After cloning the CD45 regulatory elements into an attenuated lentiviral backbone, we found that two transcriptional initiation regions were essential for high-level expression. Expressing CD45 promoters containing these regions and tethered to green fluorescent protein (GFP) in a primary B-cell differentiation assay and a transplantation model resulted in high levels of GFP in lymphoid, myeloid, and nucleated erythroid cells in mouse and human blood cell lineages. Moreover, GFP levels remained high 5 months after secondary transplantation, indicating persistence of the reporter. No CD45-driven GFP expression is observed after fibroblast or embryonic stem cell transduction. The GFP reporter is seen only after embryonic stem cells differentiate into hematopoietic cell progenitors and lineages, suggesting that this hematopoietic reporter system could be useful in validating potential autologous blood cell therapies.
Collapse
|
230
|
Ablasser A, Hemmerling I, Schmid-Burgk JL, Behrendt R, Roers A, Hornung V. TREX1 deficiency triggers cell-autonomous immunity in a cGAS-dependent manner. THE JOURNAL OF IMMUNOLOGY 2014; 192:5993-7. [PMID: 24813208 DOI: 10.4049/jimmunol.1400737] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cytosolic detection of DNA is crucial for the initiation of antiviral immunity but can also cause autoimmunity in the context of endogenous nucleic acids being sensed. Mutations in the human 3' repair exonuclease 1 (TREX1) have been linked to the type I IFN-associated autoimmune disease Aicardi-Goutières syndrome. The exact mechanisms driving unabated type I IFN responses in the absence of TREX1 are only partly understood, but it appears likely that accumulation of endogenous DNA species triggers a cell-autonomous immune response by activating a cytosolic DNA receptor. In this article, we demonstrate that knocking out the DNA sensor cyclic GMP-AMP synthase completely abrogates spontaneous induction of IFN-stimulated genes in TREX1-deficient cells. These findings indicate a key role of cyclic GMP-AMP synthase for the initiation of self-DNA-induced autoimmune disorders, thus providing important implications for novel therapeutic approaches.
Collapse
Affiliation(s)
- Andrea Ablasser
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany; and
| | - Inga Hemmerling
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany; and
| | - Jonathan L Schmid-Burgk
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany; and
| | - Rayk Behrendt
- Institute for Immunology, Medical Faculty Carl Gustav Carus, University of Technology, 01307 Dresden, Germany
| | - Axel Roers
- Institute for Immunology, Medical Faculty Carl Gustav Carus, University of Technology, 01307 Dresden, Germany
| | - Veit Hornung
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany; and
| |
Collapse
|
231
|
Wolf AR, Mootha VK. Functional genomic analysis of human mitochondrial RNA processing. Cell Rep 2014; 7:918-31. [PMID: 24746820 DOI: 10.1016/j.celrep.2014.03.035] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 02/14/2014] [Accepted: 03/11/2014] [Indexed: 11/16/2022] Open
Abstract
Both strands of human mtDNA are transcribed in continuous, multigenic units that are cleaved into the mature rRNAs, tRNAs, and mRNAs required for respiratory chain biogenesis. We sought to systematically identify nuclear-encoded proteins that contribute to processing of mtRNAs within the organelle. First, we devised and validated a multiplex MitoString assay that quantitates 27 mature and precursor mtDNA transcripts. Second, we applied MitoString profiling to evaluate the impact of silencing each of 107 mitochondrial-localized, predicted RNA-binding proteins. With the resulting data set, we rediscovered the roles of recently identified RNA-processing enzymes, detected unanticipated roles of known disease genes in RNA processing, and identified new regulatory factors. We demonstrate that one such factor, FASTKD4, modulates the half-lives of a subset of mt-mRNAs and associates with mtRNAs in vivo. MitoString profiling may be useful for diagnosing and deciphering the pathogenesis of mtDNA disorders.
Collapse
Affiliation(s)
- Ashley R Wolf
- Howard Hughes Medical Institute, Department of Molecular Biology, and Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02141, USA
| | - Vamsi K Mootha
- Howard Hughes Medical Institute, Department of Molecular Biology, and Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02141, USA.
| |
Collapse
|
232
|
Archbold JK, Martin JL, Sweet MJ. Towards selective lysophospholipid GPCR modulators. Trends Pharmacol Sci 2014; 35:219-26. [PMID: 24746475 DOI: 10.1016/j.tips.2014.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 03/13/2014] [Accepted: 03/14/2014] [Indexed: 01/08/2023]
Abstract
G-protein-coupled receptors (GPCRs) that recognize the lysophospholipids (LPLs) are grouped into two phylogenetically distinct families: the endothelial differentiation gene (Edg) and non-Edg GPCRs. Owing to their more recent identification, and hindered by a lack of selective pharmacological tools, our understanding of the functions and signaling pathways of the non-Edg GPCRs is still in its infancy. Targeting the non-conserved allosteric binding sites of the LPL GPCRs shows particular promise for the development of selective modulators by structure-based drug design. However, only one Edg GPCR (S1PR1) structure has been determined to date, and it has low sequence identity with the non-Edg GPCRs (<20%). Thus, a representative structure of a non-Edg GPCR remains a pressing objective for selective structure-based drug design. Obtaining selective modulators targeting the non-Edg receptors would help to unravel the biology behind these novel GPCRs and potentially will support therapeutic treatment of diseases such as cancer, inflammation, and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Julia K Archbold
- Division of Chemistry and Structural Biology, The University of Queensland, Institute for Molecular Bioscience, St Lucia, Brisbane, QLD 4072, Australia.
| | - Jennifer L Martin
- Division of Chemistry and Structural Biology, The University of Queensland, Institute for Molecular Bioscience, St Lucia, Brisbane, QLD 4072, Australia
| | - Matthew J Sweet
- Division of Molecular and Cell Biology, The University of Queensland, Institute for Molecular Bioscience, St Lucia, Brisbane, QLD 4072, Australia
| |
Collapse
|
233
|
Jain M, Ngoy S, Sheth SA, Swanson RA, Rhee EP, Liao R, Clish CB, Mootha VK, Nilsson R. A systematic survey of lipids across mouse tissues. Am J Physiol Endocrinol Metab 2014; 306:E854-68. [PMID: 24518676 PMCID: PMC3989739 DOI: 10.1152/ajpendo.00371.2013] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lipids are a diverse collection of macromolecules essential for normal physiology, but the tissue distribution and function for many individual lipid species remain unclear. Here, we report a mass spectrometry survey of lipid abundance across 18 mouse tissues, detecting ~1,000 mass spectrometry features, of which we identify 179 lipids from the glycerolipids, glycerophospholipids, lysophospholipids, acylcarnitines, sphingolipids, and cholesteryl ester classes. Our data reveal tissue-specific organization of lipids and can be used to generate testable hypotheses. For example, our data indicate that circulating triglycerides positively and negatively associated with future diabetes in humans are enriched in mouse adipose tissue and liver, respectively, raising hypotheses regarding the tissue origins of these diabetes-associated lipids. We also integrate our tissue lipid data with gene expression profiles to predict a number of substrates of lipid-metabolizing enzymes, highlighting choline phosphotransferases and sterol O-acyltransferases. Finally, we identify several tissue-specific lipids not present in plasma under normal conditions that may be of interest as biomarkers of tissue injury, and we show that two of these lipids are released into blood following ischemic brain injury in mice. This resource complements existing compendia of tissue gene expression and may be useful for integrative physiology and lipid biology.
Collapse
Affiliation(s)
- Mohit Jain
- Broad Institute, Cambridge, Massachusetts
| | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Transcriptional regulation and functional characterization of the oxysterol/EBI2 system in primary human macrophages. Biochem Biophys Res Commun 2014; 446:663-8. [DOI: 10.1016/j.bbrc.2014.01.069] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 01/17/2014] [Indexed: 01/12/2023]
|
235
|
Nishikata K, Cox RS, Shimoyama S, Yoshida Y, Matsui M, Makita Y, Toyoda T. Database construction for PromoterCAD: synthetic promoter design for mammals and plants. ACS Synth Biol 2014; 3:192-6. [PMID: 24364365 DOI: 10.1021/sb400178c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Synthetic promoters can control a gene's timing, location, and expression level. The PromoterCAD web server ( http://promotercad.org ) allows the design of synthetic promoters to control plant gene expression, by novel arrangement of cis-regulatory elements. Recently, we have expanded PromoterCAD's scope with additional plant and animal data: (1) PLACE (Plant Cis-acting Regulatory DNA Elements), including various sized sequence motifs; (2) PEDB (Mammalian Promoter/Enhancer Database), including gene expression data for mammalian tissues. The plant PromoterCAD data now contains 22 000 Arabidopsis thaliana genes, 2 200 000 microarray measurements in 20 growth conditions and 79 tissue organs and developmental stages, while the new mammalian PromoterCAD data contains 679 Mus musculus genes and 65 000 microarray measurements in 96 tissue organs and cell types ( http://promotercad.org/mammal/ ). This work presents step-by-step instructions for adding both regulatory motif and gene expression data to PromoterCAD, to illustrate how users can expand PromoterCAD functionality for their own applications and organisms.
Collapse
Affiliation(s)
- Koro Nishikata
- Integrated Database Unit, Advanced Center for Computing
and Communication (ACCC), RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Robert Sidney Cox
- Synthetic Genomics Research Team, Biomass Engineering
Program Cooperation Division, Center for Sustainable Resource Science
(CSRS), RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Sayoko Shimoyama
- Integrated Database Unit, Advanced Center for Computing
and Communication (ACCC), RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yuko Yoshida
- Integrated Database Unit, Advanced Center for Computing
and Communication (ACCC), RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Minami Matsui
- Synthetic Genomics Research Team, Biomass Engineering
Program Cooperation Division, Center for Sustainable Resource Science
(CSRS), RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Yuko Makita
- Integrated Database Unit, Advanced Center for Computing
and Communication (ACCC), RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Tetsuro Toyoda
- Integrated Database Unit, Advanced Center for Computing
and Communication (ACCC), RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
236
|
Yu CH, Nguyen TTK, Irvine KM, Sweet MJ, Frazer IH, Blumenthal A. Recombinant Wnt3a and Wnt5a elicit macrophage cytokine production and tolerization to microbial stimulation via Toll-like receptor 4. Eur J Immunol 2014; 44:1480-90. [PMID: 24643512 DOI: 10.1002/eji.201343959] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 12/17/2013] [Accepted: 01/24/2014] [Indexed: 01/10/2023]
Abstract
An increasing number of studies address the roles of Wnt proteins in shaping leukocyte functions. Recombinant Wnt3a and Wnt5a, prototypical activators of β-Catenin-dependent and -independent Wnt signaling, respectively, are widely used to investigate the effects of Wnt proteins on myeloid cell functions. Recent reports describe both proinflammatory and immunemodulatory effects of Wnt3a and Wnt5a on macrophages, DCs, and microglia. The underlying molecular mechanisms for this divergence are unclear. We show here that recombinant Wnt3a- and Wnt5a-induced cytokine production from murine C57BL/6 macrophages was dependent on TLR4 and inhibited by Polymyxin B. Similarly, impairment of TLR-induced cytokine production upon preexposure to Wnt proteins was TLR4 dependent. The extent of Wnt3a- and Wnt5a-induced inflammatory gene expression greatly varied between Wnt protein lots. We conclude that cytokine responses and TLR tolerization induced by recombinant Wnt proteins are likely explained by contaminating TLR4 agonists, although we cannot fully exclude that Wnt proteins have an intrinsic capacity to signal via TLR4. This study emphasizes the need for careful, independent verification of Wnt-mediated cellular responses.
Collapse
Affiliation(s)
- Chien-Hsiung Yu
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | | | | | | | | | | |
Collapse
|
237
|
Poon IKH, Goodall KJ, Phipps S, Chow JDY, Pagler EB, Andrews DM, Conlan CL, Ryan GF, White JA, Wong MKL, Horan C, Matthaei KI, Smyth MJ, Hulett MD. Mice deficient in heparanase exhibit impaired dendritic cell migration and reduced airway inflammation. Eur J Immunol 2014; 44:1016-30. [PMID: 24532362 DOI: 10.1002/eji.201343645] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 11/29/2013] [Accepted: 01/07/2014] [Indexed: 01/15/2023]
Abstract
Heparanase is a β-d-endoglucuronidase that cleaves heparan sulphate, a key component of the ECM and basement membrane. The remodelling of the ECM by heparanase has been proposed to regulate both normal physiological and pathological processes, including wound healing, inflammation, tumour angiogenesis and cell migration. Heparanase is also known to exhibit non-enzymatic functions by regulating cell adhesion, cell signalling and differentiation. In this study, constitutive heparanase-deficient (Hpse(-/-) ) mice were generated on a C57BL/6 background using the Cre/loxP recombination system, with a complete lack of heparanase mRNA, protein and activity. Although heparanase has been implicated in embryogenesis and development, Hpse(-/-) mice are anatomically normal and fertile. Interestingly, consistent with the suggested function of heparanase in cell migration, the trafficking of dendritic cells from the skin to the draining lymph nodes was markedly reduced in Hpse(-/-) mice. Furthermore, the ability of Hpse(-/-) mice to generate an allergic inflammatory response in the airways, a process that requires dendritic cell migration, was also impaired. These findings establish an important role for heparanase in immunity and identify the enzyme as a potential target for regulation of an immune response.
Collapse
Affiliation(s)
- Ivan K H Poon
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Han D, Moon S, Kim Y, Min H, Kim Y. Characterization of the membrane proteome and N-glycoproteome in BV-2 mouse microglia by liquid chromatography-tandem mass spectrometry. BMC Genomics 2014; 15:95. [PMID: 24495382 PMCID: PMC3938046 DOI: 10.1186/1471-2164-15-95] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 02/03/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Microglial cells are resident macrophages of the central nervous system and important cellular mediators of the immune response and neuroinflammatory processes. In particular, microglial activation and communication between microglia, astrocytes, and neurons are hallmarks of the pathogenesis of several neurodegenerative diseases. Membrane proteins and their N-linked glycosylation mediate this microglial activation and regulate many biological process including signal transduction, cell-cell communication, and the immune response. Although membrane proteins and N-glycosylation represent a valuable source of drug target and biomarker discovery, the knowledge of their expressed proteome in microglia is very limited. RESULTS To generate a large-scale repository, we constructed a membrane proteome and N-glycoproteome from BV-2 mouse microglia using a novel integrated approach, comprising of crude membrane fractionation, multienzyme-digestion FASP, N-glyco-FASP, and various mass spectrometry. We identified 6928 proteins including 2850 membrane proteins and 1450 distinct N-glycosylation sites on 760 N-glycoproteins, of which 556 were considered novel N-glycosylation sites. Especially, a total of 114 CD antigens are identified via MS-based analysis in normal conditions of microglia for the first time. Our bioinformatics analysis provides a rich proteomic resource for examining microglial function in, for example, cell-to-cell communication and immune responses. CONCLUSIONS Herein, we introduce a novel integrated proteomic approach for improved identification of membrane protein and N-glycosylation sites. To our knowledge, this workflow helped us to obtain the first and the largest membrane proteomic and N-glycoproteomic datesets for mouse microglia. Collectively, our proteomics and bioinformatics analysis significantly expands the knowledge of the membrane proteome and N-glycoproteome expressed in microglia within the brain and constitutes a foundation for ongoing proteomic studies and drug development for various neurological diseases.
Collapse
Affiliation(s)
| | | | | | | | - Youngsoo Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 28 Yongon-Dong, Seoul 110-799, Korea.
| |
Collapse
|
239
|
Tucker EJ, Wanschers BFJ, Szklarczyk R, Mountford HS, Wijeyeratne XW, van den Brand MAM, Leenders AM, Rodenburg RJ, Reljić B, Compton AG, Frazier AE, Bruno DL, Christodoulou J, Endo H, Ryan MT, Nijtmans LG, Huynen MA, Thorburn DR. Mutations in the UQCC1-interacting protein, UQCC2, cause human complex III deficiency associated with perturbed cytochrome b protein expression. PLoS Genet 2013; 9:e1004034. [PMID: 24385928 PMCID: PMC3873243 DOI: 10.1371/journal.pgen.1004034] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 10/29/2013] [Indexed: 12/01/2022] Open
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) is responsible for generating the majority of cellular ATP. Complex III (ubiquinol-cytochrome c oxidoreductase) is the third of five OXPHOS complexes. Complex III assembly relies on the coordinated expression of the mitochondrial and nuclear genomes, with 10 subunits encoded by nuclear DNA and one by mitochondrial DNA (mtDNA). Complex III deficiency is a debilitating and often fatal disorder that can arise from mutations in complex III subunit genes or one of three known complex III assembly factors. The molecular cause for complex III deficiency in about half of cases, however, is unknown and there are likely many complex III assembly factors yet to be identified. Here, we used Massively Parallel Sequencing to identify a homozygous splicing mutation in the gene encoding Ubiquinol-Cytochrome c Reductase Complex Assembly Factor 2 (UQCC2) in a consanguineous Lebanese patient displaying complex III deficiency, severe intrauterine growth retardation, neonatal lactic acidosis and renal tubular dysfunction. We prove causality of the mutation via lentiviral correction studies in patient fibroblasts. Sequence-profile based orthology prediction shows UQCC2 is an ortholog of the Saccharomyces cerevisiae complex III assembly factor, Cbp6p, although its sequence has diverged substantially. Co-purification studies show that UQCC2 interacts with UQCC1, the predicted ortholog of the Cbp6p binding partner, Cbp3p. Fibroblasts from the patient with UQCC2 mutations have deficiency of UQCC1, while UQCC1-depleted cells have reduced levels of UQCC2 and complex III. We show that UQCC1 binds the newly synthesized mtDNA-encoded cytochrome b subunit of complex III and that UQCC2 patient fibroblasts have specific defects in the synthesis or stability of cytochrome b. This work reveals a new cause for complex III deficiency that can assist future patient diagnosis, and provides insight into human complex III assembly by establishing that UQCC1 and UQCC2 are complex III assembly factors participating in cytochrome b biogenesis. Mitochondrial complex III deficiency is a devastating disorder that impairs energy generation, and leads to variable symptoms such as developmental regression, seizures, kidney dysfunction and frequently death. The genetic basis of complex III deficiency is not fully understood, with around half of cases having no known cause. This lack of genetic diagnosis is partly due to an incomplete understanding of the genes required for complex III assembly and function. We have identified two key proteins required for complex III, UQCC1 and UQCC2, and have elucidated the role of these inter-dependent proteins in the biogenesis of cytochrome b, the only complex III subunit that is encoded by mitochondrial DNA. We have shown that mutations in UQCC2 cause human complex III deficiency in a patient with neonatal lactic acidosis and renal tubulopathy. This work contributes to an improved understanding of complex III biogenesis, and will aid future molecular diagnoses of complex III deficiency.
Collapse
Affiliation(s)
- Elena J. Tucker
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Bas F. J. Wanschers
- Centre for Molecular and Biomolecular Informatics, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
- Nijmegen Center for Mitochondrial Disorders, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Radek Szklarczyk
- Centre for Molecular and Biomolecular Informatics, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Hayley S. Mountford
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Xiaonan W. Wijeyeratne
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Mariël A. M. van den Brand
- Nijmegen Center for Mitochondrial Disorders, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Anne M. Leenders
- Nijmegen Center for Mitochondrial Disorders, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Richard J. Rodenburg
- Nijmegen Center for Mitochondrial Disorders, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Boris Reljić
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Alison G. Compton
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Ann E. Frazier
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Damien L. Bruno
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - John Christodoulou
- Genetic Metabolic Disorders Research Unit, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Disciplines of Paediatrics & Child Health and Genetic Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Hitoshi Endo
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan
| | - Michael T. Ryan
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
- ARC Centre of Excellence for Coherent X-ray Science, La Trobe University, Melbourne, Australia
| | - Leo G. Nijtmans
- Nijmegen Center for Mitochondrial Disorders, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Martijn A. Huynen
- Centre for Molecular and Biomolecular Informatics, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
- * E-mail: (MAH); (DRT)
| | - David R. Thorburn
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, Victoria, Australia
- * E-mail: (MAH); (DRT)
| |
Collapse
|
240
|
Strittmatter L, Li Y, Nakatsuka NJ, Calvo SE, Grabarek Z, Mootha VK. CLYBL is a polymorphic human enzyme with malate synthase and β-methylmalate synthase activity. Hum Mol Genet 2013; 23:2313-23. [PMID: 24334609 DOI: 10.1093/hmg/ddt624] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CLYBL is a human mitochondrial enzyme of unknown function that is found in multiple eukaryotic taxa and conserved to bacteria. The protein is expressed in the mitochondria of all mammalian organs, with highest expression in brown fat and kidney. Approximately 5% of all humans harbor a premature stop polymorphism in CLYBL that has been associated with reduced levels of circulating vitamin B12. Using comparative genomics, we now show that CLYBL is strongly co-expressed with and co-evolved specifically with other components of the mitochondrial B12 pathway. We confirm that the premature stop polymorphism in CLYBL leads to a loss of protein expression. To elucidate the molecular function of CLYBL, we used comparative operon analysis, structural modeling and enzyme kinetics. We report that CLYBL encodes a malate/β-methylmalate synthase, converting glyoxylate and acetyl-CoA to malate, or glyoxylate and propionyl-CoA to β-methylmalate. Malate synthases are best known for their established role in the glyoxylate shunt of plants and lower organisms and are traditionally described as not occurring in humans. The broader role of a malate/β-methylmalate synthase in human physiology and its mechanistic link to vitamin B12 metabolism remain unknown.
Collapse
Affiliation(s)
- Laura Strittmatter
- Department of Molecular Biology, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | |
Collapse
|
241
|
Campeau PM, Kasperaviciute D, Lu JT, Burrage LC, Kim C, Hori M, Powell BR, Stewart F, Félix TM, van den Ende J, Wisniewska M, Kayserili H, Rump P, Nampoothiri S, Aftimos S, Mey A, Nair LDV, Begleiter ML, De Bie I, Meenakshi G, Murray ML, Repetto GM, Golabi M, Blair E, Male A, Giuliano F, Kariminejad A, Newman WG, Bhaskar SS, Dickerson JE, Kerr B, Banka S, Giltay JC, Wieczorek D, Tostevin A, Wiszniewska J, Cheung SW, Hennekam RC, Gibbs RA, Lee BH, Sisodiya SM. The genetic basis of DOORS syndrome: an exome-sequencing study. Lancet Neurol 2013; 13:44-58. [PMID: 24291220 PMCID: PMC3895324 DOI: 10.1016/s1474-4422(13)70265-5] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background Deafness, onychodystrophy, osteodystrophy, mental retardation, and seizures (DOORS) syndrome is a rare autosomal recessive disorder of unknown cause. We aimed to identify the genetic basis of this syndrome by sequencing most coding exons in affected individuals. Methods Through a search of available case studies and communication with collaborators, we identified families that included at least one individual with at least three of the five main features of the DOORS syndrome: deafness, onychodystrophy, osteodystrophy, intellectual disability, and seizures. Participants were recruited from 26 centres in 17 countries. Families described in this study were enrolled between Dec 1, 2010, and March 1, 2013. Collaborating physicians enrolling participants obtained clinical information and DNA samples from the affected child and both parents if possible. We did whole-exome sequencing in affected individuals as they were enrolled, until we identified a candidate gene, and Sanger sequencing to confirm mutations. We did expression studies in human fibroblasts from one individual by real-time PCR and western blot analysis, and in mouse tissues by immunohistochemistry and real-time PCR. Findings 26 families were included in the study. We did exome sequencing in the first 17 enrolled families; we screened for TBC1D24 by Sanger sequencing in subsequent families. We identified TBC1D24 mutations in 11 individuals from nine families (by exome sequencing in seven families, and Sanger sequencing in two families). 18 families had individuals with all five main features of DOORS syndrome, and TBC1D24 mutations were identified in half of these families. The seizure types in individuals with TBC1D24 mutations included generalised tonic-clonic, complex partial, focal clonic, and infantile spasms. Of the 18 individuals with DOORS syndrome from 17 families without TBC1D24 mutations, eight did not have seizures and three did not have deafness. In expression studies, some mutations abrogated TBC1D24 mRNA stability. We also detected Tbc1d24 expression in mouse phalangeal chondrocytes and calvaria, which suggests a role of TBC1D24 in skeletogenesis. Interpretation Our findings suggest that mutations in TBC1D24 seem to be an important cause of DOORS syndrome and can cause diverse phenotypes. Thus, individuals with DOORS syndrome without deafness and seizures but with the other features should still be screened for TBC1D24 mutations. More information is needed to understand the cellular roles of TBC1D24 and identify the genes responsible for DOORS phenotypes in individuals who do not have a mutation in TBC1D24. Funding US National Institutes of Health, the CIHR (Canada), the NIHR (UK), the Wellcome Trust, the Henry Smith Charity, and Action Medical Research.
Collapse
Affiliation(s)
- Philippe M Campeau
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Dalia Kasperaviciute
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| | - James T Lu
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA; Department of Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Choel Kim
- Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Mutsuki Hori
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Aichi, Japan
| | | | - Fiona Stewart
- Genetics Service, Belfast City Hospital, Belfast, Ireland
| | - Têmis Maria Félix
- Medical Genetics Service, Clinical Hospital of Porto Alegre, Porto Alegre, Brazil
| | - Jenneke van den Ende
- Department of Medical Genetics, University Hospital Antwerp, 2650 Antwerp, Belgium
| | - Marzena Wisniewska
- Department of Medical Genetics, Poznañ University of Medical Sciences, Poznañ, Poland
| | - Hülya Kayserili
- Medical Genetics Department, Istanbul Medical Faculty, Istanbul University, Turkey
| | - Patrick Rump
- Department of Genetics, University of Groningen, Groningen, Netherlands
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences and Research Centre, Kerala, India
| | - Salim Aftimos
- Genetic Health Service New Zealand-Northern Hub, Auckland City Hospital, Auckland, New Zealand
| | - Antje Mey
- Pediatric Neurology, Braunschweig Hospital, Braunschweig, Germany
| | - Lal D V Nair
- Department of Pediatrics, Saveetha Medical College and Hospital, Saveetha University, Chennai, Tamil Nadu, 600077, India
| | - Michael L Begleiter
- Division of Genetics, Children's Mercy Hospitals and Clinics and the University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Isabelle De Bie
- Department of Medical Genetics, Montreal Children's Hospital, McGill University Health Center, Quebec, Canada
| | - Girish Meenakshi
- Department of Pediatrics, NKP Salve Institute of Medical Sciences and Lata Mangeshkar Hospital, Maharashtra, India
| | - Mitzi L Murray
- University of Washington Medical Center, Seattle, WA, USA
| | - Gabriela M Repetto
- Center for Human Genetics, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Mahin Golabi
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Edward Blair
- Department of Clinical Genetics, Churchill Hospital, Oxford, UK
| | - Alison Male
- Clinical Genetics Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Fabienne Giuliano
- Centre Référence Anomalie Développement et Syndromes Malformatifs, Centre Hospitalier Universitaire de Nice, France
| | | | - William G Newman
- Manchester Centre for Genomic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; Manchester Centre for Genomic Centre for Genetic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; St Mary's Hospital, Manchester Academic Health Science Centre, Manchester, UK
| | - Sanjeev S Bhaskar
- Manchester Centre for Genomic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; Manchester Centre for Genomic Centre for Genetic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; St Mary's Hospital, Manchester Academic Health Science Centre, Manchester, UK
| | - Jonathan E Dickerson
- Manchester Centre for Genomic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; Manchester Centre for Genomic Centre for Genetic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; St Mary's Hospital, Manchester Academic Health Science Centre, Manchester, UK
| | - Bronwyn Kerr
- Manchester Centre for Genomic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; Manchester Centre for Genomic Centre for Genetic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; St Mary's Hospital, Manchester Academic Health Science Centre, Manchester, UK
| | - Siddharth Banka
- Manchester Centre for Genomic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; Manchester Centre for Genomic Centre for Genetic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK; St Mary's Hospital, Manchester Academic Health Science Centre, Manchester, UK
| | - Jacques C Giltay
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Dagmar Wieczorek
- Institut für Humangenetik, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Anna Tostevin
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| | - Joanna Wiszniewska
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Sau Wai Cheung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Raoul C Hennekam
- Department of Pediatrics and Translational Genetics, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Brendan H Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Howard Hughes Medical Institutes, Houston, TX, USA.
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK; Epilepsy Society, Buckinghamshire, UK.
| |
Collapse
|
242
|
Yurkovetskiy L, Burrows M, Khan AA, Graham L, Volchkov P, Becker L, Antonopoulos D, Umesaki Y, Chervonsky AV. Gender bias in autoimmunity is influenced by microbiota. Immunity 2013; 39:400-12. [PMID: 23973225 DOI: 10.1016/j.immuni.2013.08.013] [Citation(s) in RCA: 645] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 05/24/2013] [Indexed: 12/14/2022]
Abstract
Gender bias and the role of sex hormones in autoimmune diseases are well established. In specific pathogen-free nonobese diabetic (NOD) mice, females have 1.3-4.4 times higher incidence of type 1 diabetes (T1D). Germ-free (GF) mice lost the gender bias (female-to-male ratio 1.1-1.2). Gut microbiota differed in males and females, a trend reversed by male castration, confirming that androgens influence gut microbiota. Colonization of GF NOD mice with defined microbiota revealed that some, but not all, lineages overrepresented in male mice supported a gender bias in T1D. Although protection of males did not correlate with blood androgen concentration, hormone-supported expansion of selected microbial lineages may work as a positive-feedback mechanism contributing to the sexual dimorphism of autoimmune diseases. Gene-expression analysis suggested pathways involved in protection of males from T1D by microbiota. Our results favor a two-signal model of gender bias, in which hormones and microbes together trigger protective pathways.
Collapse
Affiliation(s)
- Leonid Yurkovetskiy
- Committee on Microbiology, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Sancak Y, Markhard AL, Kitami T, Kovács-Bogdán E, Kamer KJ, Udeshi ND, Carr SA, Chaudhuri D, Clapham DE, Li AA, Calvo SE, Goldberger O, Mootha VK. EMRE is an essential component of the mitochondrial calcium uniporter complex. Science 2013; 342:1379-82. [PMID: 24231807 DOI: 10.1126/science.1242993] [Citation(s) in RCA: 509] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The mitochondrial uniporter is a highly selective calcium channel in the organelle's inner membrane. Its molecular components include the EF-hand-containing calcium-binding proteins mitochondrial calcium uptake 1 (MICU1) and MICU2 and the pore-forming subunit mitochondrial calcium uniporter (MCU). We sought to achieve a full molecular characterization of the uniporter holocomplex (uniplex). Quantitative mass spectrometry of affinity-purified uniplex recovered MICU1 and MICU2, MCU and its paralog MCUb, and essential MCU regulator (EMRE), a previously uncharacterized protein. EMRE is a 10-kilodalton, metazoan-specific protein with a single transmembrane domain. In its absence, uniporter channel activity was lost despite intact MCU expression and oligomerization. EMRE was required for the interaction of MCU with MICU1 and MICU2. Hence, EMRE is essential for in vivo uniporter current and additionally bridges the calcium-sensing role of MICU1 and MICU2 with the calcium-conducting role of MCU.
Collapse
Affiliation(s)
- Yasemin Sancak
- Department of Molecular Biology, Massachusetts General Hospital, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Mango RL, Wu QP, West M, McCook EC, Serody JS, van Deventer HW. C-C chemokine receptor 5 on pulmonary mesenchymal cells promotes experimental metastasis via the induction of erythroid differentiation regulator 1. Mol Cancer Res 2013; 12:274-82. [PMID: 24197118 DOI: 10.1158/1541-7786.mcr-13-0164] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
UNLABELLED C-C Chemokine receptor 5 knockout (Ccr5(-/-)) mice develop fewer experimental pulmonary metastases than wild-type (WT) mice. This phenomenon was explored by applying gene expression profiling to the lungs of mice with these metastases. Consequently, erythroid differentiation regulator 1 (Erdr1) was identified as upregulated in the WT mice. Though commonly associated with bone marrow stroma, Erdr1 was differentially expressed in WT pulmonary mesenchymal cells (PMC) and murine embryonic fibroblasts (MEF). Moreover, the Ccr5 ligand Ccl4 increased its expression by 3.36 ± 0.14-fold. Ccr5 signaling was dependent on the mitogen-activated protein kinase kinase (Map2k) but not the phosphoinositide 3-kinase (Pi3k) pathway because treatment with U0126 inhibited upregulation of Erdr1, but treatment with LY294002 increased the expression by 3.44 ± 0.92-fold (P < 0.05). The effect Erdr1 on B16-F10 melanoma metastasis was verified by the adoptive transfer of WT MEFs into Ccr5(-/-) mice. In this model, MEFs that had been transduced with Erdr1 short hairpin RNA (shRNA) lowered metastasis by 33% compared with control transduced MEFs. The relevance of ERDR1 on human disease was assessed by coculturing chronic lymphocytic leukemia (CLL) cells with M2-10B4 stromal cells that had been transfected with shRNA or control plasmids. After 96 hours of coculture, the cell counts were higher with control cell lines than with Erdr1 knockdown lines [odds ratio (OR), 1.88 ± 0.27, 2.52 ± 0.66, respectively]. This increase was associated with a decrease in apoptotic cells (OR, 0.69 ± 0.18, 0.58 ± 0.12, respectively). IMPLICATIONS Therefore, ERDR1 is a stromal-derived factor that promotes cancer cell survival in vitro and in an experimental metastasis model.
Collapse
Affiliation(s)
- Robert L Mango
- University of North Carolina, CB 7305, 170 Manning Drive, Chapel Hill, NC 27599-7305.
| | | | | | | | | | | |
Collapse
|
245
|
Ashmole I, Bradding P. Ion channels regulating mast cell biology. Clin Exp Allergy 2013; 43:491-502. [PMID: 23600539 DOI: 10.1111/cea.12043] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 07/06/2012] [Accepted: 08/28/2012] [Indexed: 11/28/2022]
Abstract
Mast cells play a central role in the pathophysiology of asthma and related allergic conditions. Mast cell activation leads to the degranulation of preformed mediators such as histamine and the secretion of newly synthesised proinflammatory mediators such as leukotrienes and cytokines. Excess release of these mediators contributes to allergic disease states. An influx of extracellular Ca2+ is essential for mast cell mediator release. From the Ca2+ channels that mediate this influx, to the K+ , Cl- and transient receptor potential channels that set the cell membrane potential and regulate Ca2+ influx, ion channels play a critical role in mast cell biology. In this review we provide an overview of our current knowledge of ion channel expression and function in mast cells with an emphasis on how channels interact to regulate Ca2+ signalling.
Collapse
Affiliation(s)
- I Ashmole
- Department of Infection, Immunity and Inflammation, Institute for Lung Heath, Glenfield Hospital, University of Leicester, Leicester, LE3 9QP, UK
| | | |
Collapse
|
246
|
Franceschetti T, Kessler CB, Lee SK, Delany AM. miR-29 promotes murine osteoclastogenesis by regulating osteoclast commitment and migration. J Biol Chem 2013; 288:33347-60. [PMID: 24085298 DOI: 10.1074/jbc.m113.484568] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Osteoclast differentiation is regulated by transcriptional, post-transcriptional, and post-translational mechanisms. MicroRNAs are fundamental post-transcriptional regulators of gene expression. The function of the miR-29 (a/b/c) family in cells of the osteoclast lineage is not well understood. In primary cultures of mouse bone marrow-derived macrophages, inhibition of miR-29a, -29b, or -29c diminished formation of TRAP (tartrate-resistant acid phosphatase-positive) multinucleated osteoclasts, and the osteoclasts were smaller. Quantitative RT-PCR showed that all miR-29 family members increased during osteoclast differentiation, in concert with mRNAs for the osteoclast markers Trap (Acp5) and cathepsin K. Similar regulation was observed in the monocytic cell line RAW264.7. In stably transduced RAW264.7 cell lines expressing an inducible miR-29 competitive inhibitor (sponge construct), miR-29 knockdown impaired osteoclastic commitment and migration of pre-osteoclasts. However, miR-29 knockdown did not affect cell viability, actin ring formation, or apoptosis in mature osteoclasts. To better understand how miR-29 regulates osteoclast function, we validated miR-29 target genes using Luciferase 3'-UTR reporter assays and specific miR-29 inhibitors. We demonstrated that miR-29 negatively regulates RNAs critical for cytoskeletal organization, including Cdc42 (cell division control protein 42) and Srgap2 (SLIT-ROBO Rho GTPase-activating protein 2). Moreover, miR-29 targets RNAs associated with the macrophage lineage: Gpr85 (G protein-coupled receptor 85), Nfia (nuclear factor I/A), and Cd93. In addition, Calcr (calcitonin receptor), which regulates osteoclast survival and resorption, is a novel miR-29 target. Thus, miR-29 is a positive regulator of osteoclast formation and targets RNAs important for cytoskeletal organization, commitment, and osteoclast function. We hypothesize that miR-29 controls the tempo and amplitude of osteoclast differentiation.
Collapse
|
247
|
Mabbott NA, Baillie JK, Brown H, Freeman TC, Hume DA. An expression atlas of human primary cells: inference of gene function from coexpression networks. BMC Genomics 2013; 14:632. [PMID: 24053356 PMCID: PMC3849585 DOI: 10.1186/1471-2164-14-632] [Citation(s) in RCA: 276] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/25/2013] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The specialisation of mammalian cells in time and space requires genes associated with specific pathways and functions to be co-ordinately expressed. Here we have combined a large number of publically available microarray datasets derived from human primary cells and analysed large correlation graphs of these data. RESULTS Using the network analysis tool BioLayout Express3D we identify robust co-associations of genes expressed in a wide variety of cell lineages. We discuss the biological significance of a number of these associations, in particular the coexpression of key transcription factors with the genes that they are likely to control. CONCLUSIONS We consider the regulation of genes in human primary cells and specifically in the human mononuclear phagocyte system. Of particular note is the fact that these data do not support the identity of putative markers of antigen-presenting dendritic cells, nor classification of M1 and M2 activation states, a current subject of debate within immunological field. We have provided this data resource on the BioGPS web site (http://biogps.org/dataset/2429/primary-cell-atlas/) and on macrophages.com (http://www.macrophages.com/hu-cell-atlas).
Collapse
Affiliation(s)
- Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Edinburgh EH25 9RG, UK.
| | | | | | | | | |
Collapse
|
248
|
Pons T, Paramonov I, Boullosa C, Ibáñez K, Rojas AM, Valencia A. A common structural scaffold in CTD phosphatases that supports distinct catalytic mechanisms. Proteins 2013; 82:103-18. [PMID: 23900790 DOI: 10.1002/prot.24376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/28/2013] [Accepted: 07/12/2013] [Indexed: 01/01/2023]
Abstract
The phosphorylation and dephosphorylation of the carboxyl-terminal domain (CTD) of the largest RNA polymerase II (RNAPII) subunit is a critical regulatory checkpoint for transcription and mRNA processing. This CTD is unique to eukaryotic organisms and it contains multiple tandem-repeats with the consensus sequence Tyr(1) -Ser(2) -Pro(3) -Thr(4) -Ser(5) -Pro(6) -Ser(7) . Traditionally, CTD phosphatases that use metal-ion-independent (cysteine-based) and metal-ion-assisted (aspartate-based) catalytic mechanisms have been considered to belong to two independent groups. However, using structural comparisons we have identified a common structural scaffold in these two groups of CTD phosphatases. This common scaffold accommodates different catalytic processes with the same substrate specificity, in this case phospho-serine/threonine residues flanked by prolines. Furthermore, this scaffold provides a structural connection between two groups of protein tyrosine phosphatases (PTPs): Cys-based (classes I, II, and III) and Asp-based (class IV) PTPs. Redundancy in catalytic mechanisms is not infrequent and may arise in specific biological settings. To better understand the activity of the CTD phosphatases, we combined our structural analyses with data on CTD phosphatase expression in different human and mouse tissues. The results suggest that aspartate- and cysteine-based CTD-dephosphorylation acts in concert during cellular stress, when high levels of reactive oxygen species can inhibit the nucleophilic function of the catalytic cysteine, as occurs in mental and neurodegenerative disorders like schizophrenia, Alzheimer's and Parkinson's diseases. Moreover, these findings have significant implications for the study of the RNAPII-CTD dephosphorylation in eukaryotes.
Collapse
Affiliation(s)
- Tirso Pons
- Structural Biology and BioComputing Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | | | | | | |
Collapse
|
249
|
Das >UN. Lipoxins, resolvins, protectins, maresins and nitrolipids, and their clinical implications with specific reference to diabetes mellitus and other diseases: part II. ACTA ACUST UNITED AC 2013. [DOI: 10.2217/clp.13.32] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
250
|
Hohenhaus DM, Schaale K, Le Cao KA, Seow V, Iyer A, Fairlie DP, Sweet MJ. An mRNA atlas of G protein-coupled receptor expression during primary human monocyte/macrophage differentiation and lipopolysaccharide-mediated activation identifies targetable candidate regulators of inflammation. Immunobiology 2013; 218:1345-53. [PMID: 23948647 DOI: 10.1016/j.imbio.2013.07.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/04/2013] [Accepted: 07/06/2013] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptors (GPCRs) are among the most important targets in drug discovery. In this study, we used TaqMan Low Density Arrays to profile the full GPCR repertoire of primary human macrophages differentiated from monocytes using either colony stimulating factor-1 (CSF-1/M-CSF) (CSF-1 Mϕ) or granulocyte macrophage colony stimulating factor (GM-CSF) (GM-CSF Mϕ). The overall trend was a downregulation of GPCRs during monocyte to macrophage differentiation, but a core set of 10 genes (e.g. LGR4, MRGPRF and GPR143) encoding seven transmembrane proteins were upregulated, irrespective of the differentiating agent used. Several of these upregulated GPCRs have not previously been studied in the context of macrophage biology and/or inflammation. As expected, CSF-1 Mϕ and GM-CSF Mϕ exhibited differential inflammatory cytokine profiles in response to the Toll-like Receptor (TLR)4 agonist lipopolysaccharide (LPS). Moreover, 15 GPCRs were differentially expressed between these cell populations in the basal state. For example, EDG1 was expressed at elevated levels in CSF-1 Mϕ versus GM-CSF Mϕ, whereas the reverse was true for EDG6. 101 GPCRs showed differential regulation over an LPS time course, with 65 of these profiles being impacted by the basal differentiation state (e.g. GPRC5A, GPRC5B). Only 14 LPS-regulated GPCRs showed asynchronous behavior (divergent LPS regulation) with respect to differentiation status. Thus, the differentiation state primarily affects the magnitude of LPS-regulated expression, rather than causing major reprogramming of GPCR gene expression profiles. Several GPCRs showing differential profiles between CSF-1 Mϕ and GM-CSF Mϕ (e.g. P2RY8, GPR92, EMR3) have not been widely investigated in macrophage biology and inflammation. Strikingly, several closely related GPCRs displayed completely opposing patterns of regulation during differentiation and/or activation (e.g. EDG1 versus EDG6, LGR4 versus LGR7, GPRC5A versus GPRC5B). We propose that selective regulation of GPCR5A and GPCR5B in CSF-1 Mϕ contributes to skewing toward the M2 macrophage phenotype. Our analysis of the GPCR repertoire expressed during primary human monocyte to macrophage differentiation and TLR4-mediated activation provides a valuable new platform for conducting future functional analyses of individual GPCRs in human macrophage inflammatory pathways.
Collapse
Affiliation(s)
- Daniel M Hohenhaus
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld 4072, Australia; Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, Qld 4072, Australia
| | | | | | | | | | | | | |
Collapse
|