CXCR4 Pathway Retards Muscle Atrophy During Cancer Cachexia

Total Page:16

File Type:pdf, Size:1020Kb

CXCR4 Pathway Retards Muscle Atrophy During Cancer Cachexia Oncogene (2016) 35, 6212–6222 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved 0950-9232/16 www.nature.com/onc ORIGINAL ARTICLE Activation of the SDF1/CXCR4 pathway retards muscle atrophy during cancer cachexia GB Martinelli1, D Olivari1, AD Re Cecconi1, L Talamini1, L Ottoboni2, SH Lecker3, C Stretch4, VE Baracos4, OF Bathe5, A Resovi6, R Giavazzi1, L Cervo7 and R Piccirillo1 Cancer cachexia is a life-threatening syndrome that affects most patients with advanced cancers and causes severe body weight loss, with rapid depletion of skeletal muscle. No treatment is available. We analyzed microarray data sets to identify a subset of genes whose expression is specifically altered in cachectic muscles of Yoshida hepatoma-bearing rodents but not in those with diabetes, disuse, uremia or fasting. Ingenuity Pathways Analysis indicated that three genes belonging to the C-X-C motif chemokine receptor 4 (CXCR4) pathway were downregulated only in muscles atrophying because of cancer: stromal cell-derived factor 1 (SDF1), adenylate cyclase 7 (ADCY7), and p21 protein-activated kinase 1 (PAK1). Notably, we found that, in the Rectus Abdominis muscle of cancer patients, the expression of SDF1 and CXCR4 was inversely correlated with that of two ubiquitin ligases induced in muscle wasting, atrogin-1 and MuRF1, suggesting a possible clinical relevance of this pathway. The expression of all main SDF1 isoforms (α, β, γ) also declined in Tibialis Anterior muscle from cachectic mice bearing murine colon adenocarcinoma or human renal cancer and drugs with anticachexia properties restored their expression. Overexpressing genes of this pathway (that is, SDF1 or CXCR4) in cachectic muscles increased the fiber area by 20%, protecting them from wasting. Similarly, atrophying myotubes treated with either SDF1α or SDF1β had greater total protein content, resulting from reduced degradation of overall long-lived proteins. However, inhibiting CXCR4 signaling with the antagonist AMD3100 did not affect protein homeostasis in atrophying myotubes, whereas normal myotubes treated with AMD3100 showed time- and dose-dependent reductions in diameter, until a plateau, and lower total protein content. This further confirms the involvement of a saturable pathway (that is, CXCR4). Overall, these findings support the idea that activating the CXCR4 pathway in muscle suppresses the deleterious wasting associated with cancer. Oncogene (2016) 35, 6212–6222; doi:10.1038/onc.2016.153; published online 23 May 2016 INTRODUCTION of cancer, diabetes, fasting, disuse or uremia led to the important Cancer cachexia is a highly debilitating syndrome, involving discovery of the atrogenes, in other words, genes that are severe body weight loss (BWL) due to depletion of skeletal muscle, differentially expressed in all kinds of muscle atrophy. Among with or without fat tissue loss.1 It cannot be corrected by them, Forkhead box-containing, subfamily O3 (FoxO3) is the nutritional supplementation2,3 and affects up to 80% of patients main transcription factor driving the expression of most of the with advanced cancers. The rapid loss of muscle mass is the main atrogenes, such as those implied in the lysosomal and proteaso- cause of functional impairment, fatigue and respiratory complica- mal pathways, which promote overall proteolysis.12,13 Two tions, ultimately leading to death in 20–48% of cases. To date, no muscle-restricted ubiquitin ligases, atrogin-1 and muscle RING treatment is available. finger protein 1 (MuRF1), are dramatically upregulated by FoxO3 in Various circulating inflammatory factors have been implicated all settings of muscle wasting, including cancer cachexia.14,15 in cancer cachexia, for example, tumor necrosis factor-α (TNFα) Molecules that block this activation of proteolysis or increase and interleukin 6 (IL6) that is increased in the serum of patients muscle protein synthesis might serve as pharmacological agents with advanced non-small-cell lung cancer,4 colon cancer,5 prostate to combat wasting. cancer6 and esophageal squamous cell cancer,7 but anticytokine Re-analysis of the same microarray datasets enabled us to therapies are ineffective.8,9 Moreover, these factors are common identify the genes specifically altered in rodent muscles because to other wasting syndromes, such as that caused by chronic of cancer and not because of diabetes, fasting, disuse or uremia. kidney diseases,10 and not restricted to cancer cachexia. Interestingly, three were drastically downregulated only in the To identify a genetic signature of muscle wasting typical of atrophying gastrocnemius of Yoshida hepatoma-bearing rats: cancer and not of other systemic (for example, uremia) or local (for stromal cell-derived factor 1 (SDF1) also named CXCL12, adenylate example, muscle disuse) dysfunctions, we re-analyzed previous cyclase 7 (ADCY7), and p21 protein-activated kinase 1 (PAK1). gene expression profiles generated by microarray.11 In that study, Ingenuity Pathway Analysis associated all of them to the C-X-C the gene expression comparison of muscles atrophying because chemokine receptor type 4 (CXCR4) pathway. 1Department of Oncology, IRCCS - Mario Negri Institute for Pharmacological Research, Milan, Italy; 2San Raffaele Scientific Institute, INSpe, Milan, Italy; 3Beth Israel Deaconess Center, Boston, MA, USA; 4Department of Oncology, University of Alberta, Edmonton, Alberta, Canada; 5Department of Surgery and Oncology, University of Calgary, Calgary, Alberta, Canada; 6Department of Oncology, Tumor Angiogenesis Unit, IRCCS - Mario Negri Institute for Pharmacological Research, Bergamo, Italy and 7Department of Neuroscience, IRCCS - Mario Negri Institute for Pharmacological Research, Milan, Italy. Correspondence: Dr R Piccirillo, Department of Oncology, IRCCS - Mario Negri Institute for Pharmacological Research, Via G La Masa 19, 20156 Milan, Italy. E-mail: [email protected] Received 3 September 2015; revised 5 February 2016; accepted 11 March 2016; published online 23 May 2016 CXCR4 pathway has a role in cancer cachexia GB Martinelli et al 6213 CXCR4 is a Gαi-coupled member of G protein-coupled receptors conditions of muscle atrophy tested (uremia, disuse, fasting, (GPCRs) whose ligand SDF1 has a role in angiogenesis by diabetes) (P40.05; Figure 1a). recruiting endothelial progenitor cells from the bone marrow We identified 52 cancer cachexia-specific genes or expressed through CXCR4.16 SDF1 also has strong chemotactic activity for sequence tags that were further examined by Ingenuity Pathway lymphocytes, and CXCR4 was initially identified as the receptor Analysis, showing that CXCR4 pathway had the most significant required for HIV cell entry17 and found mutated in a rare enrichment (P = 0.012, ratio 3/158). Three genes of this pathway immunodeficiency disorder in humans.18 Antagonists of the (SDF1, ADCY7 and PAK1) were consistently downregulated in CXCR4 pathway are on the market and are used to mobilize cachectic muscles only during cancer (Figures 1b and c). Each one hematopoietic progenitor cells from bone marrow or under encodes for a protein of a different subcellular compartment. 19 investigation to restrain HIV infection and tumor dissemination. SDF1 is the extracellular ligand of CXCR4, ADCY7 is a calcium- Various splicing variants of SDF1 exist (α, β, γ), but their different inhibitable membrane-bound enzyme, while PAK1 is a cytosolic 20 functional roles have not yet been fully elucidated. The SDF1/ kinase downstream of the CXCR4 pathway.27 They were down- 21 CXCR4 pathway has a role in muscle formation in zebrafish, in regulated to 34, 1 and 20% of controls, respectively (Figure 1c). Xenopus22 and also in muscle regeneration in rodents23 as well as in muscle differentiation in vitro.24 However, nothing is known SDF1 expression is attenuated in the cachectic Tibialis Anterior about its role in muscle atrophy induced by cancer. muscle of C26-bearing mice Combining genetic approaches in adult murine muscles with fl pharmacological strategies in cultured myotubes, we investigated The suppression of this in ammatory pathway in cachectic fl whether and how the SDF1/CXCR4 pathway could have a muscles was surprising, as cancer cachexia involves in amma- 28 fi fi functional role in muscle wasting during cancer cachexia. Finally, tion. Thus, we rst validated this nding in the cachectic tibialis as we25 and others26 have reported that some angiogenic anterior (TA) of another species (Mus musculus) with another inhibitors reversed or even prevented cachexia in vivo without tumor (C26 colon adenocarcinoma) injected by a different route affecting tumor growth, we also assessed whether upregulation of (subcutaneous). Eight-week-old female and male C26-bearing muscular and/or circulating SDF1 was among the mechanisms by mice undergo BWL about 13 days after injection of C26 cells in the which these drugs might counteract cachexia. dorsal subcutis (Figure 2a and data not shown, respectively). In accordance with institutional guidelines, animals were killed when at least four out of five signs of distress (loss of mobility, kyphosis, RESULTS ruffled fur, low body temperature, tremor) were present or when Genes belonging to the CXCR4 pathway are downregulated 420% of BW was lost in 72 h. All C26-bearing animals had to be specifically in the atrophying gastrocnemius muscle of Yoshida killed by 18 days from tumor injection, when the tumor reached hepatoma-bearing rats about 600 mm3 (Figures 2b and c). We analyzed a previously generated mouse microarray data set By contrast,
Recommended publications
  • Genome-Wide Association and Transcriptome Studies Identify Candidate Genes and Pathways for Feed Conversion Ratio in Pigs
    Miao et al. BMC Genomics (2021) 22:294 https://doi.org/10.1186/s12864-021-07570-w RESEARCH ARTICLE Open Access Genome-wide association and transcriptome studies identify candidate genes and pathways for feed conversion ratio in pigs Yuanxin Miao1,2,3, Quanshun Mei1,2, Chuanke Fu1,2, Mingxing Liao1,2,4, Yan Liu1,2, Xuewen Xu1,2, Xinyun Li1,2, Shuhong Zhao1,2 and Tao Xiang1,2* Abstract Background: The feed conversion ratio (FCR) is an important productive trait that greatly affects profits in the pig industry. Elucidating the genetic mechanisms underpinning FCR may promote more efficient improvement of FCR through artificial selection. In this study, we integrated a genome-wide association study (GWAS) with transcriptome analyses of different tissues in Yorkshire pigs (YY) with the aim of identifying key genes and signalling pathways associated with FCR. Results: A total of 61 significant single nucleotide polymorphisms (SNPs) were detected by GWAS in YY. All of these SNPs were located on porcine chromosome (SSC) 5, and the covered region was considered a quantitative trait locus (QTL) region for FCR. Some genes distributed around these significant SNPs were considered as candidates for regulating FCR, including TPH2, FAR2, IRAK3, YARS2, GRIP1, FRS2, CNOT2 and TRHDE. According to transcriptome analyses in the hypothalamus, TPH2 exhibits the potential to regulate intestinal motility through serotonergic synapse and oxytocin signalling pathways. In addition, GRIP1 may be involved in glutamatergic and GABAergic signalling pathways, which regulate FCR by affecting appetite in pigs. Moreover, GRIP1, FRS2, CNOT2,andTRHDE may regulate metabolism in various tissues through a thyroid hormone signalling pathway.
    [Show full text]
  • Bioinformatics Analysis Based on Gene Expression Omnibus
    ANTICANCER RESEARCH 39 : 1689-1698 (2019) doi:10.21873/anticanres.13274 Chemo-resistant Gastric Cancer Associated Gene Expression Signature: Bioinformatics Analysis Based on Gene Expression Omnibus JUN-BAO LIU 1* , TUNYU JIAN 2* , CHAO YUE 3, DAN CHEN 4, WEI CHEN 5, TING-TING BAO 6, HAI-XIA LIU 7, YUN CAO 8, WEI-BING LI 6, ZHIJIAN YANG 9, ROBERT M. HOFFMAN 9 and CHEN YU 6 1Traditional Chinese Medicine Department, People's Hospital of Henan Province, People's Hospital of Zhengzhou University, Zhengzhou, P.R. China; 2Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, P.R. China; 3Department of general surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China; 4Research Center of Clinical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China; 5Department of Head and Neck Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China; 6Department of Integrated TCM & Western Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China; 7Emergency Department, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, P.R. China; 8Master candidate of Oncology, Nanjing University of Chinese Medicine, Nanjing, P.R. China; 9AntiCancer, Inc., San Diego, CA, U.S.A. Abstract. Background/Aim: This study aimed to identify identified, including 13 up-regulated and 1,473 down-regulated biomarkers for predicting the prognosis of advanced gastric genes.
    [Show full text]
  • Epigenetic Modifications to Cytosine and Alzheimer's Disease
    University of Kentucky UKnowledge Theses and Dissertations--Chemistry Chemistry 2017 EPIGENETIC MODIFICATIONS TO CYTOSINE AND ALZHEIMER’S DISEASE: A QUANTITATIVE ANALYSIS OF POST-MORTEM TISSUE Elizabeth M. Ellison University of Kentucky, [email protected] Digital Object Identifier: https://doi.org/10.13023/ETD.2017.398 Right click to open a feedback form in a new tab to let us know how this document benefits ou.y Recommended Citation Ellison, Elizabeth M., "EPIGENETIC MODIFICATIONS TO CYTOSINE AND ALZHEIMER’S DISEASE: A QUANTITATIVE ANALYSIS OF POST-MORTEM TISSUE" (2017). Theses and Dissertations--Chemistry. 86. https://uknowledge.uky.edu/chemistry_etds/86 This Doctoral Dissertation is brought to you for free and open access by the Chemistry at UKnowledge. It has been accepted for inclusion in Theses and Dissertations--Chemistry by an authorized administrator of UKnowledge. For more information, please contact [email protected]. STUDENT AGREEMENT: I represent that my thesis or dissertation and abstract are my original work. Proper attribution has been given to all outside sources. I understand that I am solely responsible for obtaining any needed copyright permissions. I have obtained needed written permission statement(s) from the owner(s) of each third-party copyrighted matter to be included in my work, allowing electronic distribution (if such use is not permitted by the fair use doctrine) which will be submitted to UKnowledge as Additional File. I hereby grant to The University of Kentucky and its agents the irrevocable, non-exclusive, and royalty-free license to archive and make accessible my work in whole or in part in all forms of media, now or hereafter known.
    [Show full text]
  • Whole-Genome DNA Methylation and Hydroxymethylation Profiling for HBV-Related Hepatocellular Carcinoma
    INTERNATIONAL JOURNAL OF ONCOLOGY 49: 589-602, 2016 Whole-genome DNA methylation and hydroxymethylation profiling for HBV-related hepatocellular carcinoma CHAO YE*, RAN TAO*, QINGYI CAO, DANHUA ZHU, YINI WANG, JIE WANG, JUAN LU, ERMEI CHEN and LANJUAN LI State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China Received March 18, 2016; Accepted May 13, 2016 DOI: 10.3892/ijo.2016.3535 Abstract. Hepatocellular carcinoma (HCC) is a common tions between them. Taken together, in the present study we solid tumor worldwide with a poor prognosis. Accumulating conducted the first genome-wide mapping of DNA methyla- evidence has implicated important regulatory roles of epigen- tion combined with hydroxymethylation in HBV-related HCC etic modifications in the occurrence and progression of HCC. and provided a series of potential novel epigenetic biomarkers In the present study, we analyzed 5-methylcytosine (5-mC) for HCC. and 5-hydroxymethylcytosine (5-hmC) levels in the tumor tissues and paired adjacent peritumor tissues (APTs) from Introduction four individual HCC patients using a (hydroxy)methylated DNA immunoprecipitation approach combined with deep Hepatocellular carcinoma (HCC), a common solid tumor, is sequencing [(h)MeDIP-Seq]. Bioinformatics analysis revealed the third most frequent cause of cancer-related death in the that the 5-mC levels in the promoter regions of 2796 genes and world. Hepatitis B virus (HBV) infection is the main cause of the 5-hmC levels in 507 genes differed significantly between HCC in China (1).
    [Show full text]
  • Genetic Refinement and Physical Mapping of a Chromosome 16Q Candidate Region for Inflammatory Bowel Disease
    European Journal of Human Genetics (2001) 9, 731 ± 742 ã 2001 Nature Publishing Group All rights reserved 1018-4813/01 $15.00 www.nature.com/ejhg ARTICLE Genetic refinement and physical mapping of a chromosome 16q candidate region for inflammatory bowel disease Habib Zouali1,16, Mathias Chamaillard1,2,16, Suzane Lesage1, Jean-Pierre CeÂzard3, Jean-FreÂdeÂric Colombel4, Jacques Belaiche and the GETAID5, Sven Almer6, Curt Tysk7, Sean Montague8, Miquel Gassull9, Steen Christensen10, Yigael Finkel11, Corine Gower-Rousseau4, Robert Modigliani12, Jeanne Macry13, Hana Selinger-Leneman14, Gilles Thomas1,2,15 and Jean-Pierre Hugot*,1,2,3 1Fondation Jean Dausset CEPH, Paris, France; 2INSERM Unite 434, Paris, France; 3European Paediatric Working Group on the Genetics of IBD, Department of Paediatric Gastroenterology, Hopital Robert DebreÂ,Paris,France; 4Registre EPIMAD, HoÃpital Calmette, Lille, France; 5Department of Gastroenterology, CHU de LieÁge, Belgium; 6Division of Gastroenterology and Hepatology, IHM, LinkoÈpings universitet, LinkoÈping, Sweden; 7Department of Gastroenterology, OÈrebro Medical Center hospital, OÈrebro, Sweden; 8Department of Gastroenterology, Adelaide & Meath Hospital, Dublin, Ireland; 9Department of Gastroenterology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; 10Department of Gastroenterology Herlev Hospital, Herlev, Denmark; 11Department of Gastroenterology, Karolinska Children's Hospital, Stockholm, Sweden; 12Department of Gastroenterology, HoÃpital Saint Louis, Paris, France; 13INSERM Unite 458, Paris, France; 14INSERM Unite 535, Le Kremlin-BiceÃtre, France; 15Department of Surgery, HoÃpital Saint Antoine, Paris, France Crohn's disease (CD) is a complex genetic disorder for which a susceptibility gene, IBD1, has been mapped within the pericentromeric region of chromosome 16. In order to refine the location of IBD1, 77 multiplex CD families were genotyped for 26 microsatellite markers evenly spaced by approximately 1 cM.
    [Show full text]
  • Exploring the Genetic Architecture of Inflammatory Bowel Disease By
    bioRxiv preprint doi: https://doi.org/10.1101/058347; this version posted June 11, 2016. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Exploring the genetic architecture of inflammatory bowel disease by 2 whole genome sequencing identifies association at ADCY7 3 Yang Luo*1,2,3, Katrina M. de Lange*1, Luke Jostins4,5, Loukas Moutsianas1, Joshua Randall1, Nicholas A. 4 Kennedy6,7, Christopher A. Lamb8, Shane McCarthy1, Tariq Ahmad6,7, Cathryn Edwards9, Eva Goncalves 5 Serra1, Ailsa Hart10, Chris Hawkey11, John C. Mansfield12, Craig Mowat13, William G. Newman14,15, Sam 6 Nichols1, Martin Pollard1, Jack Satsangi16, Alison Simmons17,18, Mark Tremelling19, Holm Uhlig20, David C. 7 Wilson21,22, James C. Lee23, Natalie J. Prescott24, Charlie W. Lees16, Christopher G. Mathew24,25, Miles 8 Parkes23, Jeffrey C. Barrett*1, Carl A. Anderson*1 9 Abstract 10 In order to further resolve the genetic architecture of the inflammatory bowel diseases, ulcerative 11 colitis and Crohn’s disease, we sequenced the whole genomes of 4,280 patients at low coverage, 12 and compared them to 3,652 previously sequenced population controls across 73.5 million 13 variants. To increase power we imputed from these sequences into new and existing GWAS 14 cohorts, and tested for association at ~12 million variants in a total of 16,432 cases and 18,843 15 controls. We discovered a 0.6% frequency missense variant in ADCY7 that doubles risk of 16 ulcerative colitis, and offers insight into a new aspect of disease biology.
    [Show full text]
  • Protein Family Members. the GENE.FAMILY
    Table 3: Protein family members. The GENE.FAMILY col- umn shows the gene family name defined either by HGNC (superscript `H', http://www.genenames.org/cgi-bin/family_ search) or curated manually by us from Entrez IDs in the NCBI database (superscript `C' for `Custom') that we have identified as corresonding for each ENTITY.ID. The members of each gene fam- ily that are in at least one of our synaptic proteome datasets are shown in IN.SYNAPSE, whereas those not found in any datasets are in the column OUT.SYNAPSE. In some cases the intersection of two HGNC gene families are needed to specify the membership of our protein family; this is indicated by concatenation of the names with an ampersand. ENTITY.ID GENE.FAMILY IN.SYNAPSE OUT.SYNAPSE AC Adenylate cyclasesH ADCY1, ADCY2, ADCY10, ADCY4, ADCY3, ADCY5, ADCY7 ADCY6, ADCY8, ADCY9 actin ActinsH ACTA1, ACTA2, ACTB, ACTC1, ACTG1, ACTG2 ACTN ActininsH ACTN1, ACTN2, ACTN3, ACTN4 AKAP A-kinase anchoring ACBD3, AKAP1, AKAP11, AKAP14, proteinsH AKAP10, AKAP12, AKAP17A, AKAP17BP, AKAP13, AKAP2, AKAP3, AKAP4, AKAP5, AKAP6, AKAP8, CBFA2T3, AKAP7, AKAP9, RAB32 ARFGEF2, CMYA5, EZR, MAP2, MYO7A, MYRIP, NBEA, NF2, SPHKAP, SYNM, WASF1 CaM Endogenous ligands & CALM1, CALM2, EF-hand domain CALM3 containingH CaMKK calcium/calmodulin- CAMKK1, CAMKK2 dependent protein kinase kinaseC CB CalbindinC CALB1, CALB2 CK1 Casein kinase 1C CSNK1A1, CSNK1D, CSNK1E, CSNK1G1, CSNK1G2, CSNK1G3 CRHR Corticotropin releasing CRHR1, CRHR2 hormone receptorsH DAGL Diacylglycerol lipaseC DAGLA, DAGLB DGK Diacylglycerol kinasesH DGKB,
    [Show full text]
  • Program in Human Neutrophils Fails To
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 is online at: average * The Journal of Immunology Anaplasma phagocytophilum , 20 of which you can access for free at: 2005; 174:6364-6372; ; from submission to initial decision 4 weeks from acceptance to publication J Immunol doi: 10.4049/jimmunol.174.10.6364 http://www.jimmunol.org/content/174/10/6364 Insights into Pathogen Immune Evasion Mechanisms: Fails to Induce an Apoptosis Differentiation Program in Human Neutrophils Dori L. Borjesson, Scott D. Kobayashi, Adeline R. Whitney, Jovanka M. Voyich, Cynthia M. Argue and Frank R. DeLeo cites 28 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2005/05/03/174.10.6364.DC1 This article http://www.jimmunol.org/content/174/10/6364.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* • Why • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2005 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Insights into Pathogen Immune Evasion Mechanisms: Anaplasma phagocytophilum Fails to Induce an Apoptosis Differentiation Program in Human Neutrophils1 Dori L.
    [Show full text]
  • Supplemental Figures 04 12 2017
    Jung et al. 1 SUPPLEMENTAL FIGURES 2 3 Supplemental Figure 1. Clinical relevance of natural product methyltransferases (NPMTs) in brain disorders. (A) 4 Table summarizing characteristics of 11 NPMTs using data derived from the TCGA GBM and Rembrandt datasets for 5 relative expression levels and survival. In addition, published studies of the 11 NPMTs are summarized. (B) The 1 Jung et al. 6 expression levels of 10 NPMTs in glioblastoma versus non‐tumor brain are displayed in a heatmap, ranked by 7 significance and expression levels. *, p<0.05; **, p<0.01; ***, p<0.001. 8 2 Jung et al. 9 10 Supplemental Figure 2. Anatomical distribution of methyltransferase and metabolic signatures within 11 glioblastomas. The Ivy GAP dataset was downloaded and interrogated by histological structure for NNMT, NAMPT, 12 DNMT mRNA expression and selected gene expression signatures. The results are displayed on a heatmap. The 13 sample size of each histological region as indicated on the figure. 14 3 Jung et al. 15 16 Supplemental Figure 3. Altered expression of nicotinamide and nicotinate metabolism‐related enzymes in 17 glioblastoma. (A) Heatmap (fold change of expression) of whole 25 enzymes in the KEGG nicotinate and 18 nicotinamide metabolism gene set were analyzed in indicated glioblastoma expression datasets with Oncomine. 4 Jung et al. 19 Color bar intensity indicates percentile of fold change in glioblastoma relative to normal brain. (B) Nicotinamide and 20 nicotinate and methionine salvage pathways are displayed with the relative expression levels in glioblastoma 21 specimens in the TCGA GBM dataset indicated. 22 5 Jung et al. 23 24 Supplementary Figure 4.
    [Show full text]
  • Detection of H3k4me3 Identifies Neurohiv Signatures, Genomic
    viruses Article Detection of H3K4me3 Identifies NeuroHIV Signatures, Genomic Effects of Methamphetamine and Addiction Pathways in Postmortem HIV+ Brain Specimens that Are Not Amenable to Transcriptome Analysis Liana Basova 1, Alexander Lindsey 1, Anne Marie McGovern 1, Ronald J. Ellis 2 and Maria Cecilia Garibaldi Marcondes 1,* 1 San Diego Biomedical Research Institute, San Diego, CA 92121, USA; [email protected] (L.B.); [email protected] (A.L.); [email protected] (A.M.M.) 2 Departments of Neurosciences and Psychiatry, University of California San Diego, San Diego, CA 92103, USA; [email protected] * Correspondence: [email protected] Abstract: Human postmortem specimens are extremely valuable resources for investigating trans- lational hypotheses. Tissue repositories collect clinically assessed specimens from people with and without HIV, including age, viral load, treatments, substance use patterns and cognitive functions. One challenge is the limited number of specimens suitable for transcriptional studies, mainly due to poor RNA quality resulting from long postmortem intervals. We hypothesized that epigenomic Citation: Basova, L.; Lindsey, A.; signatures would be more stable than RNA for assessing global changes associated with outcomes McGovern, A.M.; Ellis, R.J.; of interest. We found that H3K27Ac or RNA Polymerase (Pol) were not consistently detected by Marcondes, M.C.G. Detection of H3K4me3 Identifies NeuroHIV Chromatin Immunoprecipitation (ChIP), while the enhancer H3K4me3 histone modification was Signatures, Genomic Effects of abundant and stable up to the 72 h postmortem. We tested our ability to use H3K4me3 in human Methamphetamine and Addiction prefrontal cortex from HIV+ individuals meeting criteria for methamphetamine use disorder or not Pathways in Postmortem HIV+ Brain (Meth +/−) which exhibited poor RNA quality and were not suitable for transcriptional profiling.
    [Show full text]
  • Sex Chromosome Complement Regulates Expression of Mood-Related Genes Marianne L Seney1,2, Kokomma I Ekong1, Ying Ding3,4, George C Tseng3,5 and Etienne Sibille1,2,6*
    Seney et al. Biology of Sex Differences 2013, 4:20 http://www.bsd-journal.com/content/4/1/20 RESEARCH Open Access Sex chromosome complement regulates expression of mood-related genes Marianne L Seney1,2, Kokomma I Ekong1, Ying Ding3,4, George C Tseng3,5 and Etienne Sibille1,2,6* Abstract Background: Studies on major depressive and anxiety disorders suggest dysfunctions in brain corticolimbic circuits, including altered gamma-aminobutyric acid (GABA) and modulatory (serotonin and dopamine) neurotransmission. Interestingly, sexual dimorphisms in GABA, serotonin, and dopamine systems are also reported. Understanding the mechanisms behind these sexual dimorphisms may help unravel the biological bases of the heightened female vulnerability to mood disorders. Here, we investigate the contribution of sex-related factors (sex chromosome complement, developmental gonadal sex, or adult circulating hormones) to frontal cortex expression of selected GABA-, serotonin-, and dopamine-related genes. Methods: As gonadal sex is determined by sex chromosome complement, the role of sex chromosomes cannot be investigated individually in humans. Therefore, we used the Four Core Genotypes (FCG) mouse model, in which sex chromosome complement and gonadal sex are artificially decoupled, to examine the expression of 13 GABA-related genes, 6 serotonin- and dopamine-related genes, and 8 associated signal transduction genes under chronic stress conditions. Results were analyzed by three-way ANOVA (sex chromosome complement × gonadal sex × circulating testosterone). A global perspective of gene expression changes was provided by heatmap representation and gene co-expression networks to identify patterns of transcriptional activities related to each main factor. Results: We show that under chronic stress conditions, sex chromosome complement influenced GABA/serotonin/ dopamine-related gene expression in the frontal cortex, with XY mice consistently having lower gene expression compared to XX mice.
    [Show full text]
  • Systems Biology Understanding of the Effects of Lithium on Cancer
    bioRxiv preprint doi: https://doi.org/10.1101/363077; this version posted August 24, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. Systems Biology Understanding of the Effects of Lithium on Cancer Weihao Ge1,2 and Eric Jakobsson1,2,3, * 1National Center for Supercomputing Applications, 2Center for Biophysics and Computational Biology, 3Department of Molecular and Integrative Physiology University of Illinois at Urbana-Champaign, Urbana, IL, USA Correspondence *Eric Jakobsson [email protected] Abstract Lithium has many widely varying biochemical and phenomenological effects, suggesting that a systems biology approach is required to understand its action. Multiple lines of evidence point to lithium as a significant factor in development of cancer, showing that understanding lithium action is of high importance. In this paper we undertake first steps towards a systems approach by analyzing mutual enrichment between the interactomes of lithium-sensitive enzymes and the pathways associated with cancer. This work integrates information from two important databases, STRING and KEGG pathways. We find that for the majority of cancer pathways the mutual enrichment is many times greater than chance, reinforcing previous lines of evidence that lithium is an important influence on cancer. bioRxiv preprint doi: https://doi.org/10.1101/363077; this version posted August 24, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]