Evidence for a Cluster of Genes on Chromosome 17Q11–Q21 Controlling Susceptibility to Tuberculosis and Leprosy in Brazilians

Total Page:16

File Type:pdf, Size:1020Kb

Evidence for a Cluster of Genes on Chromosome 17Q11–Q21 Controlling Susceptibility to Tuberculosis and Leprosy in Brazilians Genes and Immunity (2004) 5, 46–57 & 2004 Nature Publishing Group All rights reserved 1466-4879/04 $25.00 www.nature.com/gene Evidence for a cluster of genes on chromosome 17q11–q21 controlling susceptibility to tuberculosis and leprosy in Brazilians SE Jamieson1, EN Miller1, GF Black1, CS Peacock1, HJ Cordell1, JMM Howson1, M-A Shaw2, D Burgner3,6,WXu4, Z Lins-Lainson5, JJ Shaw5,7, F Ramos5, F Silveira5 and JM Blackwell1 1Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, UK; 2Department of Biology, University of Leeds, Leeds, UK; 3The Wellcome Trust Centre for Human Genetics, Oxford, UK; 4Wolfson Institute for Biomedical Research, Rayne Institute, University College, London, UK; 5Instituto Evandro Chagas, Belem, Brazil The region of conserved synteny on mouse chromosome 11/human 17q11–q21 is known to carry a susceptibility gene(s) for intramacrophage pathogens. The region is rich in candidates including NOS2A, CCL2/MCP-1, CCL3/MIP-1a, CCL4/MIP-1b, CCL5/RANTES, CCR7, STAT3 and STAT5A/5B. To examine the region in man, we studied 92 multicase tuberculosis (627 individuals) and 72 multicase leprosy (372 individuals) families from Brazil. Multipoint nonparametric analysis (ALLEGRO) using 16 microsatellites shows two peaks of linkage for leprosy at D17S250 (Zlr score 2.34; P¼0.01) and D17S1795 (Zlr 2.67; P¼0.004) and a single peak for tuberculosis at D17S250 (Zlr 2.04; P¼0.02). Combined analysis shows significant linkage (peak Zlr 3.38) at D17S250, equivalent to an allele sharing LOD score 2.48 (P¼0.0004). To determine whether one or multiple genes contribute, 49 informative single nucleotide polymorphisms were typed in candidate genes. Family-based allelic association testing that was robust to family clustering demonstrated significant associations with tuberculosis susceptibility at four loci separated by intervals (NOS2A–8.4 Mb–CCL18–32.3 kb–CCL4–6.04 Mb–STAT5B) up to several Mb. Stepwise conditional logistic regression analysis using a case/pseudo-control data set showed that the four genes contributed separate main effects, consistent with a cluster of susceptibility genes across 17q11.2. Genes and Immunity (2004) 5, 46–57. doi:10.1038/sj.gene.6364029 Keywords: chromosome 17q11-q22; tuberculosis; leprosy; genetic susceptibility Introduction (eg CCL2/SCYA2/MCP-1, CCL3/SCYA3/MIP-1a, CCL4/ SCYA4/MIP-1b, CCL5/SCYA5/RANTES), the gene Many studies support the hypothesis that susceptibility (CCR7) encoding the receptor for CCL19/CCL21 and to Mycobacterium tuberculosis and M. leprae is genetically genes for signal transducers and activators of transcrip- regulated in humans.1,2 One approach to identifying tion STAT3, STAT5A and STAT5B. candidate genes is to study genetic susceptibility in mice The shared intramacrophage niche of leishmanial and and examine the regions of conserved synteny in man. mycobacterial (M. tuberculosis and M. leprae) pathogens, We took this approach to analyse susceptibility to and the similar spectra of disease caused by the two leishmanial infections.3,4 This led to identification of a groups of organisms, means that any gene identified as a region on mouse chromosome 11, syntenic with human susceptibility gene for one group of pathogens becomes a chromosome 17q11.1–q12, which carries susceptibility candidate for the others. A well-characterized example of genes to cutaneous leishmaniasis.5,6 This region is rich in this is the gene (Nramp1/NRAMP1) located on mouse candidate susceptibility genes. These include the gene chromosome 1/human 2q35 that encodes the natural (NOS2A) encoding the inducible form of nitric oxide resistance associated macrophage protein 1, now rede- synthase (iNOS), genes encoding members of the family signated as solute carrier family 11a member 1 (Slc11a1 in of small inducible chemokine gene (designated, for mice; SLC11A1 in man). This gene controls innate example, chemokine, CC motif, ligand 1 or CCL1) cluster resistance to Leishmania donovani,7–10 M. bovis BCG,11 M. lepraemurium12,13 and M. intracellulare14 in the mouse, 15–22 Correspondence: Dr JM Blackwell, Cambridge Institute for Medical and contributes to susceptibility to M. tuberculosis, Research, Wellcome Trust/MRC Building, University of Cambridge School M. leprae23 and L. donovani24,25 in man. of Clinical Medicine, Addenbrookes Hospital, Hills Road, Cambridge CB2 The successful mouse-to-man precedent set with 2XY, UK. E-mail: [email protected] Slc11a1/SLC11A1 encouraged us to take a similar 6Present address: Princess Margaret Hospital, Subiaco, Western approach in studying the region of human chromosome Australia, Australia. 17q syntenic with murine chromosome 11 in relation to 7Present address: Department of Parasitology, Institute of Bio- medical Sciences, Sao Paulo University, 05508-900, Brazil. genetic susceptibility to leprosy and tuberculosis in man. Received 17 April 2003; revised 14 August 2003; accepted 19 August Here we present linkage and allelic association data for 2003 polymorphic loci at 17q11–q21 from 92 multicase families Genetic susceptibility to tuberculosis and leprosy SE Jamieson et al 47 Table 1 Family structures for the 92 tuberculosis and 72 leprosy families collected from Belem, Brazil Number Families structure TB Leprosy per se Leprosy LL subtype Leprosy TT subtype No. of families 92 72 72 72 No. of nuclear families 118 87 56a 48a Nuclear families with 1 affected sib 58 19 27 22 Nuclear families with 2 affected sibs 37 50 25 17 Nuclear families with 3 affected sibs 17 7 3 7 Nuclear families with 4 affected sibs 4 5 1 1 Nuclear families with 5 affected sibs 2 5 0 0 Nuclear families with 6 affected sibs 0 0 0 1 Nuclear families with 7 affected sibs 0 1 0 0 No. of affected offspring 209 192 90 87 No. of affected parents 71 41 25 10 Total no. of affected individualsb 280 208c 109 93 Total no. of individuals 627 372 372 372 Nuclear families with a single affected offspring were always part of an extended multicase pedigree. aIn all, 17 nuclear families contain both LL and TT affected individuals; therefore, the numbers of LL+TT nuclear families do not add up to leprosy per se. bDue to pedigree structure, some individuals are classed as both sibs and parents in different nuclear families; therefore, the total number of affected is not the sibs+parents. cSix leprosy per se individuals not classified as LL or TT; therefore, the numbers of affected children and parents for LL+TT do not add up to leprosy per se. of tuberculosis and 72 multicase families of leprosy from D17S33 27.9Mb NOS2A CCL2 Belem, Brazil that support the hypothesis that a cluster of 5.7cM XulNOS 28.6Mb TNFAIP1 CCL7 4.2cM 31.0Mb CRLF3 CCL11 genes in this region regulate mycobacterial infections in man. CCL8 3.7cM D17S798 CCL13 CCL1 5.8cM D17S1293 34.6Mb CCL Cluster 8.0cM } D17S927 36.4Mb Results 4.8cM D17S250 40.1Mb CSF3 D17S1814 THRA1 MMP28 41.1Mb Genetic and physical maps for chromosome 5.8cM D17S1299 CCR7 CCL5 42.5Mb STAT5A & 5B CCL16 CCL14 17q11.1–q21.31 7.8cM STAT3 3.8cM D17S930 44.9Mb CCL15 1.4cM CCL23 A total of 627 individuals from 98 multicase tuberculosis 2.3cM CCL18 CCL3 families and 372 individuals from 72 multicase leprosy 3.9cM CCL4 D17S2015 48.5Mb SP2 families (Table 1) were initially genotyped for 16 2.6cM D17S1785 49.6 4.2cM D17S1868 D17S1795 microsatellite markers across the chromosome 17q11.1– D17S1869 q21.31 region. Marker–marker linkage data from the 6.8cM D17S1877 combined set of families were used to generate a genetic D17S787 55.7Mb 26 27 map (Figure 1) using CRI-Map and MAP-O-MAT. Figure 1 Line diagram comparing genetic map (Kosambi cM) This is compared (Figure 1) to the current physical map generated using CRI-Map and MAP-O-MAT with physical map order for the same markers obtained from the UCSC (Mb) based on information from the November 2002 freeze of the Human Genome Project Working Draft, November 2002 working draft of the human genome at (Electronic Database freeze (NCBI Build 31). The only inconsistency between Information 5). the genetic and physical map order is for the marker D17S2015. This discrepancy in position could be due contributed by the TT leprosy families (Zlr 2.58; P¼0.005), either to assembly errors in the physical map or to a lack whereas LL families appear to make some contribution to of informative meioses in the genetic mapping. Since the the leprosy per se peak at D17S1975. Combined linkage genetic map provides a more accurate representation of analysis for all leprosy and tuberculosis families gave a recombination events across the region, we used the peak multipoint Zlr¼3.38 (P¼0.0004). Again, the TT leprosy genetic map generated from the Brazilian families in subtype appeared to account for all of the combined multipoint linkage analyses presented below. linkage (multipoint Zlr 3.56; P¼0.0002). No evidence for linkage is observed for either leprosy or tuberculosis at Linking disease susceptibility to chromosome 17 NOS2A, which lies immediately proximal to the region of Results of multipoint nonparametric linkage analyses linkage. Multipoint information content remained constant carried out in ALLEGRO are presented in Figure 2. For (mean7s.d.) across the region (tuberculosis families: leprosy per se, a broad region of linkage was observed, 0.5370.05; leprosy families: 0.5170.05; LL subtype: 7 7 with an initial peak (Zlr 2.34; P¼0.01) at D17S250 and a 0.71 0.05; TT subtype: 0.81 0.09). second peak (Zlr 2.67; P¼0.004) at D17S1795. For tuberculosis, a single peak (Zlr 2.04; P¼0.02) suggestive Family-based transmission disequilibrium testing of linkage was observed at D17S250, with no linkage at (TDT) D17S1795.
Recommended publications
  • Molecular Genetic Analysis of Two Loci (Ity2 and Ity3) Involved in The
    Copyright Ó 2007 by the Genetics Society of America DOI: 10.1534/genetics.107.075523 Molecular Genetic Analysis of Two Loci (Ity2 and Ity3) Involved in the Host Response to Infection With Salmonella Typhimurium Using Congenic Mice and Expression Profiling Vanessa Sancho-Shimizu,*,† Rabia Khan,*,† Serge Mostowy,† Line Larivie`re,† Rosalie Wilkinson,† Noe´mie Riendeau,† Marcel Behr† and Danielle Malo*,†,‡,1 *Department of Human Genetics, McGill University, Montreal, Quebec H3G 1A4, Canada and †Center for the Study of Host Resistance, McGill University Health Center, Montreal, Quebec H3G 1A4, Canada and ‡Department of Medicine, McGill University, Montreal, Quebec H3G 1A4, Canada Manuscript received May 4, 2007 Accepted for publication July 27, 2007 ABSTRACT Numerous genes have been identified to date that contribute to the host response to systemic Sal- monella Typhimurium infection in mice. We have previously identified two loci, Ity2 and Ity3, that control survival to Salmonella infection in the wild-derived inbred MOLF/Ei mouse using a (C57BL/6J 3 MOLF/ Ei)F2cross. We validated the existence of these two loci by creating congenic mice carrying each quan- titative trait locus (QTL) in isolation. Subcongenic mice generated for each locus allowed us to define the critical intervals underlying Ity2 and Ity3. Furthermore, expression profiling was carried out with the aim of identifying differentially expressed genes within the critical intervals as potential candidate genes. Genomewide expression arrays were used to interrogate expression differences in the Ity2 congenics, leading to the identification of a new candidate gene (Havcr2, hepatitis A virus cellular receptor 2). Interval-specific oligonucleotide arrays were created for Ity3, identifying one potential candidate gene (Chi3l1, chitinase 3-like 1) to be pursued further.
    [Show full text]
  • Cellular and Plasma Proteomic Determinants of COVID-19 and Non-COVID-19 Pulmonary Diseases Relative to Healthy Aging
    RESOURCE https://doi.org/10.1038/s43587-021-00067-x Cellular and plasma proteomic determinants of COVID-19 and non-COVID-19 pulmonary diseases relative to healthy aging Laura Arthur1,8, Ekaterina Esaulova 1,8, Denis A. Mogilenko 1, Petr Tsurinov1,2, Samantha Burdess1, Anwesha Laha1, Rachel Presti 3, Brian Goetz4, Mark A. Watson1, Charles W. Goss5, Christina A. Gurnett6, Philip A. Mudd 7, Courtney Beers4, Jane A. O’Halloran3 and Maxim N. Artyomov1 ✉ We examine the cellular and soluble determinants of coronavirus disease 2019 (COVID-19) relative to aging by performing mass cytometry in parallel with clinical blood testing and plasma proteomic profiling of ~4,700 proteins from 71 individuals with pul- monary disease and 148 healthy donors (25–80 years old). Distinct cell populations were associated with age (GZMK+CD8+ T cells and CD25low CD4+ T cells) and with COVID-19 (TBET−EOMES− CD4+ T cells, HLA-DR+CD38+ CD8+ T cells and CD27+CD38+ B cells). A unique population of TBET+EOMES+ CD4+ T cells was associated with individuals with COVID-19 who experienced moderate, rather than severe or lethal, disease. Disease severity correlated with blood creatinine and urea nitrogen levels. Proteomics revealed a major impact of age on the disease-associated plasma signatures and highlighted the divergent contri- bution of hepatocyte and muscle secretomes to COVID-19 plasma proteins. Aging plasma was enriched in matrisome proteins and heart/aorta smooth muscle cell-specific proteins. These findings reveal age-specific and disease-specific changes associ- ated with COVID-19, and potential soluble mediators of the physiological impact of COVID-19.
    [Show full text]
  • Anti-OX40 Antibody Directly Enhances the Function of Tumor-Reactive CD8+ T Cells
    Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Anti-OX40 antibody directly enhances the function of tumor-reactive CD8+ T cells and synergizes with PI3Kβ inhibition in PTEN loss melanoma Weiyi Peng1,5*, Leila J. Williams1, Chunyu Xu1,5, Brenda Melendez1, Jodi A. McKenzie1,6, Yuan Chen1, Heather Jackson2, Kui S. Voo3, Rina M. Mbofung1,7,, Sara E. Leahey1, Jian Wang4, Greg Lizee1, Hussein A. Tawbi1, Michael A. Davies1, Axel Hoos2, James Smothers2, Roopa Srinivasan2, Elaine Paul2, Niranjan Yanamandra2* and Patrick Hwu1* 1Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX. 2Oncology R&D, Immuno-Oncology and Combinations RU, GlaxoSmithKline, 1250 S. Collegeville Rd, Collegeville, PA 19426, United States 3Oncology Research for Biologics and Immunotherapy Translation Platform, The University of Texas MD Anderson Cancer Center, Houston, TX. 4Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX. 5Present address: Department of Biology and Biochemistry, University of Houston, Houston, TX. 6Present address: Eisai Inc., Woodcliff Lake, NJ. 7Present address: Merck Research Laboratories, Palo Alto, CA. Running Title: OX40 agonist-based cancer immunotherapy Keywords: OX40, PI3K, cancer immunotherapy Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 2 *Corresponding Authors: Patrick Hwu, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030.
    [Show full text]
  • Transcriptional Regulation of RKIP in Prostate Cancer Progression
    Health Science Campus FINAL APPROVAL OF DISSERTATION Doctor of Philosophy in Biomedical Sciences Transcriptional Regulation of RKIP in Prostate Cancer Progression Submitted by: Sandra Marie Beach In partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biomedical Sciences Examination Committee Major Advisor: Kam Yeung, Ph.D. Academic William Maltese, Ph.D. Advisory Committee: Sonia Najjar, Ph.D. Han-Fei Ding, M.D., Ph.D. Manohar Ratnam, Ph.D. Senior Associate Dean College of Graduate Studies Michael S. Bisesi, Ph.D. Date of Defense: May 16, 2007 Transcriptional Regulation of RKIP in Prostate Cancer Progression Sandra Beach University of Toledo ACKNOWLDEGMENTS I thank my major advisor, Dr. Kam Yeung, for the opportunity to pursue my degree in his laboratory. I am also indebted to my advisory committee members past and present, Drs. Sonia Najjar, Han-Fei Ding, Manohar Ratnam, James Trempe, and Douglas Pittman for generously and judiciously guiding my studies and sharing reagents and equipment. I owe extended thanks to Dr. William Maltese as a committee member and chairman of my department for supporting my degree progress. The entire Department of Biochemistry and Cancer Biology has been most kind and helpful to me. Drs. Roy Collaco and Hong-Juan Cui have shared their excellent technical and practical advice with me throughout my studies. I thank members of the Yeung laboratory, Dr. Sungdae Park, Hui Hui Tang, Miranda Yeung for their support and collegiality. The data mining studies herein would not have been possible without the helpful advice of Dr. Robert Trumbly. I am also grateful for the exceptional assistance and shared microarray data of Dr.
    [Show full text]
  • A Genome‐Wide Association Study Highlights a Regulatory Role for IFNG‐AS1
    bioRxiv preprint doi: https://doi.org/10.1101/2020.01.13.903989; this version posted January 14, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. A genome‐wide association study highlights a regulatory role for IFNG‐AS1 contributing to cutaneous leishmaniasis in Brazil Short title: A GWAS for cutaneous leishmaniasis in Brazil Léa C. Castellucci1,2,*, Lucas Almeida1,2,*, Svetlana Cherlin3,*, Michaela Fakiola4, Edgar Carvalho1, Amanda B. Figueiredo5, Clara M. Cavalcanti5, Natalia S. Alves5, Walderez O. Dutra1,6 , Kenneth J. Gollob1,5,7, Heather J. Cordell3 , and Jenefer M. Blackwell8,9 *Equal contributions 1National Institute of Science and Technology in Tropical Diseases, Brazil; 2Federal University of Bahia, Salvador, Brazil; 3Population Health Sciences Institute, Newcastle University, UK; 4INGM‐National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi" Milan, Milan, Italy; 5International Center for Research, AC Camargo Cancer Center, São Paulo, Brazil; 6Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; 7Núcleo de Ensino e Pesquisa, Instituto Mario Penna, Belo Horizonte, Brazil; 8Department of Pathology, University of Cambridge, UK; 9Telethon Kids Institute, The University of Western Australia, Western Australia Corresponding author: Jenefer M. Blackwell ([email protected] Correspondence to: Jenefer M. Blackwell, PO Box 855, West Perth, Western Australia 6872; Hospital Avenue, Nedlands, Western Australia 6009; Phone: +61 8 63191000. Funding British Medical Research Council (MRC) MR/N017390/1, FAPEMIG grant in cooperation with MRC/CONFAP (CBB-APQ-00883-16), National Institute of Science and Technology in Tropical Diseases, Brazil (N° 573839/2008–5), CNPq (K.J.G.
    [Show full text]
  • Systems Analysis Implicates WAVE2&Nbsp
    JACC: BASIC TO TRANSLATIONAL SCIENCE VOL.5,NO.4,2020 ª 2020 THE AUTHORS. PUBLISHED BY ELSEVIER ON BEHALF OF THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION. THIS IS AN OPEN ACCESS ARTICLE UNDER THE CC BY-NC-ND LICENSE (http://creativecommons.org/licenses/by-nc-nd/4.0/). PRECLINICAL RESEARCH Systems Analysis Implicates WAVE2 Complex in the Pathogenesis of Developmental Left-Sided Obstructive Heart Defects a b b b Jonathan J. Edwards, MD, Andrew D. Rouillard, PHD, Nicolas F. Fernandez, PHD, Zichen Wang, PHD, b c d d Alexander Lachmann, PHD, Sunita S. Shankaran, PHD, Brent W. Bisgrove, PHD, Bradley Demarest, MS, e f g h Nahid Turan, PHD, Deepak Srivastava, MD, Daniel Bernstein, MD, John Deanfield, MD, h i j k Alessandro Giardini, MD, PHD, George Porter, MD, PHD, Richard Kim, MD, Amy E. Roberts, MD, k l m m,n Jane W. Newburger, MD, MPH, Elizabeth Goldmuntz, MD, Martina Brueckner, MD, Richard P. Lifton, MD, PHD, o,p,q r,s t d Christine E. Seidman, MD, Wendy K. Chung, MD, PHD, Martin Tristani-Firouzi, MD, H. Joseph Yost, PHD, b u,v Avi Ma’ayan, PHD, Bruce D. Gelb, MD VISUAL ABSTRACT Edwards, J.J. et al. J Am Coll Cardiol Basic Trans Science. 2020;5(4):376–86. ISSN 2452-302X https://doi.org/10.1016/j.jacbts.2020.01.012 JACC: BASIC TO TRANSLATIONALSCIENCEVOL.5,NO.4,2020 Edwards et al. 377 APRIL 2020:376– 86 WAVE2 Complex in LVOTO HIGHLIGHTS ABBREVIATIONS AND ACRONYMS Combining CHD phenotype–driven gene set enrichment and CRISPR knockdown screening in zebrafish is an effective approach to identifying novel CHD genes.
    [Show full text]
  • Differential Expression of Interferon-Γ and Chemokine Genes
    Differential expression of interferon-γ and chemokine genes distinguishes Rasmussen encephalitis from cortical dysplasia and provides evidence for an early Th1 immune response Owens et al. Owens et al. Journal of Neuroinflammation 2013, 10:56 http://www.jneuroinflammation.com/content/10/1/56 Owens et al. Journal of Neuroinflammation 2013, 10:56 JOURNAL OF http://www.jneuroinflammation.com/content/10/1/56 NEUROINFLAMMATION RESEARCH Open Access Differential expression of interferon-γ and chemokine genes distinguishes Rasmussen encephalitis from cortical dysplasia and provides evidence for an early Th1 immune response Geoffrey C Owens1,7*, My N Huynh1, Julia W Chang1, David L McArthur1, Michelle J Hickey2, Harry V Vinters2,3, Gary W Mathern1,4,5,6† and Carol A Kruse1,6† Abstract Background: Rasmussen encephalitis (RE) is a rare complex inflammatory disease, primarily seen in young children, that is characterized by severe partial seizures and brain atrophy. Surgery is currently the only effective treatment option. To identify genes specifically associated with the immunopathology in RE, RNA transcripts of genes involved in inflammation and autoimmunity were measured in brain tissue from RE surgeries and compared with those in surgical specimens of cortical dysplasia (CD), a major cause of intractable pediatric epilepsy. Methods: Quantitative polymerase chain reactions measured the relative expression of 84 genes related to inflammation and autoimmunity in 12 RE specimens and in the reference group of 12 CD surgical specimens. Data were analyzed by consensus clustering using the entire dataset, and by pairwise comparison of gene expression levels between the RE and CD cohorts using the Harrell-Davis distribution-free quantile estimator method.
    [Show full text]
  • Inhibition of CCL7 Derived from Mo-Mdscs Prevents Metastatic
    Ren et al. Cell Death and Disease (2021) 12:484 https://doi.org/10.1038/s41419-021-03698-5 Cell Death & Disease ARTICLE Open Access Inhibition of CCL7 derived from Mo-MDSCs prevents metastatic progression from latency in colorectal cancer Xiaoli Ren1,2, Jianbiao Xiao1,3, Wanning Zhang1,3,FeifeiWang1,3, Yongrong Yan1,3,XuehuiWu 1,3, Zhicheng Zeng1,3, Yumei He4,WeiYang1,3, Wangjun Liao5,YanqingDing1,3 and Li Liang 1,3 Abstract In colorectal cancer (CRC), overt metastases often appear after years of latency. But the signals that cause micro- metastatic cells to remain indolent, thereby enabling them to survive for extended periods of time, are unclear. Immunofluorescence and co-immunoprecipitation assays were used to explore the co-localization of CCL7 and CCR2. Immunohistochemical (IHC) assays were employed to detect the characters of metastatic HT29 cells in mice liver. Flow cytometry assays were performed to detect the immune cells. Bruberin vivo MS FX Pro Imager was used to observe the liver metastasis of CRC in mice. Quantitative real-time PCR (qRT-PCR) and western blot were employed to detect the expressions of related proteins. Trace RNA sequencing was employed to identify differentially expressed genes in MDSCs from liver micro-M and macro-M of CRC in mice. Here, we firstly constructed the vitro dormant cell models and metastatic dormant animal models of colorectal cancer. Then we found that myeloid-derived suppressor cells (MDSCs) were increased significantly from liver micro-metastases to macro-metastases of CRC in mice. Moreover, monocytic MDSCs (Mo-MDSC) significantly promoted the dormant activation of micro-metastatic cells compared to 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; polymorphonuclear MDSCs (PMN-MDSC).
    [Show full text]
  • Genomic and Expression Profiling of Chromosome 17 in Breast Cancer Reveals Complex Patterns of Alterations and Novel Candidate Genes
    [CANCER RESEARCH 64, 6453–6460, September 15, 2004] Genomic and Expression Profiling of Chromosome 17 in Breast Cancer Reveals Complex Patterns of Alterations and Novel Candidate Genes Be´atrice Orsetti,1 Me´lanie Nugoli,1 Nathalie Cervera,1 Laurence Lasorsa,1 Paul Chuchana,1 Lisa Ursule,1 Catherine Nguyen,2 Richard Redon,3 Stanislas du Manoir,3 Carmen Rodriguez,1 and Charles Theillet1 1Ge´notypes et Phe´notypes Tumoraux, EMI229 INSERM/Universite´ Montpellier I, Montpellier, France; 2ERM 206 INSERM/Universite´ Aix-Marseille 2, Parc Scientifique de Luminy, Marseille cedex, France; and 3IGBMC, U596 INSERM/Universite´Louis Pasteur, Parc d’Innovation, Illkirch cedex, France ABSTRACT 17q12-q21 corresponding to the amplification of ERBB2 and collinear genes, and a large region at 17q23 (5, 6). A number of new candidate Chromosome 17 is severely rearranged in breast cancer. Whereas the oncogenes have been identified, among which GRB7 and TOP2A at short arm undergoes frequent losses, the long arm harbors complex 17q21 or RP6SKB1, TBX2, PPM1D, and MUL at 17q23 have drawn combinations of gains and losses. In this work we present a comprehensive study of quantitative anomalies at chromosome 17 by genomic array- most attention (6–10). Furthermore, DNA microarray studies have comparative genomic hybridization and of associated RNA expression revealed additional candidates, with some located outside current changes by cDNA arrays. We built a genomic array covering the entire regions of gains, thus suggesting the existence of additional amplicons chromosome at an average density of 1 clone per 0.5 Mb, and patterns of on 17q (8, 9). gains and losses were characterized in 30 breast cancer cell lines and 22 Our previous loss of heterozygosity mapping data pointed to the primary tumors.
    [Show full text]
  • Neutrophil Chemoattractant Receptors in Health and Disease: Double-Edged Swords
    Cellular & Molecular Immunology www.nature.com/cmi REVIEW ARTICLE Neutrophil chemoattractant receptors in health and disease: double-edged swords Mieke Metzemaekers1, Mieke Gouwy1 and Paul Proost 1 Neutrophils are frontline cells of the innate immune system. These effector leukocytes are equipped with intriguing antimicrobial machinery and consequently display high cytotoxic potential. Accurate neutrophil recruitment is essential to combat microbes and to restore homeostasis, for inflammation modulation and resolution, wound healing and tissue repair. After fulfilling the appropriate effector functions, however, dampening neutrophil activation and infiltration is crucial to prevent damage to the host. In humans, chemoattractant molecules can be categorized into four biochemical families, i.e., chemotactic lipids, formyl peptides, complement anaphylatoxins and chemokines. They are critically involved in the tight regulation of neutrophil bone marrow storage and egress and in spatial and temporal neutrophil trafficking between organs. Chemoattractants function by activating dedicated heptahelical G protein-coupled receptors (GPCRs). In addition, emerging evidence suggests an important role for atypical chemoattractant receptors (ACKRs) that do not couple to G proteins in fine-tuning neutrophil migratory and functional responses. The expression levels of chemoattractant receptors are dependent on the level of neutrophil maturation and state of activation, with a pivotal modulatory role for the (inflammatory) environment. Here, we provide an overview
    [Show full text]
  • Multi-Targeted Mechanisms Underlying the Endothelial Protective Effects of the Diabetic-Safe Sweetener Erythritol
    Multi-Targeted Mechanisms Underlying the Endothelial Protective Effects of the Diabetic-Safe Sweetener Erythritol Danie¨lle M. P. H. J. Boesten1*., Alvin Berger2.¤, Peter de Cock3, Hua Dong4, Bruce D. Hammock4, Gertjan J. M. den Hartog1, Aalt Bast1 1 Department of Toxicology, Maastricht University, Maastricht, The Netherlands, 2 Global Food Research, Cargill, Wayzata, Minnesota, United States of America, 3 Cargill RandD Center Europe, Vilvoorde, Belgium, 4 Department of Entomology and UCD Comprehensive Cancer Center, University of California Davis, Davis, California, United States of America Abstract Diabetes is characterized by hyperglycemia and development of vascular pathology. Endothelial cell dysfunction is a starting point for pathogenesis of vascular complications in diabetes. We previously showed the polyol erythritol to be a hydroxyl radical scavenger preventing endothelial cell dysfunction onset in diabetic rats. To unravel mechanisms, other than scavenging of radicals, by which erythritol mediates this protective effect, we evaluated effects of erythritol in endothelial cells exposed to normal (7 mM) and high glucose (30 mM) or diabetic stressors (e.g. SIN-1) using targeted and transcriptomic approaches. This study demonstrates that erythritol (i.e. under non-diabetic conditions) has minimal effects on endothelial cells. However, under hyperglycemic conditions erythritol protected endothelial cells against cell death induced by diabetic stressors (i.e. high glucose and peroxynitrite). Also a number of harmful effects caused by high glucose, e.g. increased nitric oxide release, are reversed. Additionally, total transcriptome analysis indicated that biological processes which are differentially regulated due to high glucose are corrected by erythritol. We conclude that erythritol protects endothelial cells during high glucose conditions via effects on multiple targets.
    [Show full text]
  • Nutrient Health and EROEN Compounds Resegsterixsteextics: Production * Gets Cartrai, Agairaxxxix
    US 2011 O153221A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2011/0153221 A1 Stefanon et al. (43) Pub. Date: Jun. 23, 2011 (54) DIAGNOSTIC SYSTEM FOR SELECTING (52) U.S. Cl. .......................................................... 702/19 NUTRITION AND PHARMACOLOGICAL PRODUCTS FOR ANIMALS (57) ABSTRACT (76) Inventors: Bruno Stefanon, Martignacco (IT): W.Year Jean Dodds.Odds, Santa Monica,IVIon1ca, CA An analysis of the profile of a non-human animal comprises: a) providing a genotypic database to the species of the non (21) Appl. No.: 12/927,769 human animal Subject or a selected group of the species; b obtaining animal data; c) correlating the database of a) with (22) Filed:1-1. Nov. 19, 2010 the data ofb) to determinea relationshipp between the database of a) and the data of b); c) determining the profile of the Related U.S. Application Data animal based on the correlating step; and d) determining a (63)63) ContinuationConti offaroplication application No. 12/316.824,:4'. filed'O geneticmolecular profile dietary based signature on the being molecular a variation dietary of signature, expression the of Dec. 16, 2008, now Pat. No. 7,873,482. a set of genes which may differ for the genotype of each O O animal or a group of animals Nutrition and pharmalogical Publication Classification assessments are made. Reporting the determination is by the (51) Int. Cl. Internet, and payment for the report is obtained through the G06F 9/00 (2011.01) Internet. 38s. 4 gas registics, $88's *.icisixxxiiserscies: 8 texrigixi exsix * processire statisy • Essex: 88& goEffect onXXXXWWYYX Nutrient health and EROEN Compounds resegsterixsteextics: production * gets cartrai, agairaxxxix.
    [Show full text]