Generation of Novel IL-10 Mesenchymal Stem/Stromal Cells

Total Page:16

File Type:pdf, Size:1020Kb

Generation of Novel IL-10 Mesenchymal Stem/Stromal Cells Mesenchymal Stem/Stromal Cells Induce the Generation of Novel IL-10 −Dependent Regulatory Dendritic Cells by SOCS3 Activation This information is current as of October 3, 2021. Xingxia Liu, Xuebin Qu, Yuan Chen, Lianming Liao, Kai Cheng, Changshun Shao, Martin Zenke, Armand Keating and Robert C. H. Zhao J Immunol 2012; 189:1182-1192; Prepublished online 2 July 2012; Downloaded from doi: 10.4049/jimmunol.1102996 http://www.jimmunol.org/content/189/3/1182 Supplementary http://www.jimmunol.org/content/suppl/2012/07/02/jimmunol.110299 http://www.jimmunol.org/ Material 6.DC1 References This article cites 50 articles, 18 of which you can access for free at: http://www.jimmunol.org/content/189/3/1182.full#ref-list-1 Why The JI? Submit online. by guest on October 3, 2021 • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2012 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Mesenchymal Stem/Stromal Cells Induce the Generation of Novel IL-10–Dependent Regulatory Dendritic Cells by SOCS3 Activation Xingxia Liu,*,1 Xuebin Qu,*,1 Yuan Chen,* Lianming Liao,† Kai Cheng,* Changshun Shao,‡ Martin Zenke,x Armand Keating,{,‖,# and Robert C. H. Zhao* Suppression of immune response by mesenchymal stem/stromal cells (MSCs) is well documented. However, their regulatory effects on immune cells, especially regulatory dendritic cells, are not fully understood. We have identified a novel Sca-1+Lin2CD1172 MSC population isolated from mouse embryonic fibroblasts (MEF) that suppressed lymphocyte proliferation in vitro. Moreover, the Sca-1+Lin2CD1172 MEF-MSCs induced hematopoietic stem/progenitor cells to differentiate into novel regulatory dendritic cells (DCs) (Sca-1+Lin2CD1172 MEF-MSC–induced DCs) when cocultured in the absence of exogenous cytokines. Small interfering RNA silencing showed that Sca-1+Lin2CD1172 MEF-MSCs induced the generation of Sca-1+Lin2CD1172 MEF-MSC–induced Downloaded from DCs via IL-10–activated SOCS3, whose expression was regulated by the JAK–STAT pathway. We observed a high degree of H3K4me3 modification mediated by MLL1 and a relatively low degree of H3K27me3 modification regulated by SUZ12 on the promoter of SOCS3 during SOCS3 activation. Importantly, infusion of Sca-1+CD1172Lin2 MEF-MSCs suppressed the inflam- matory response by increasing DCs with a regulatory phenotype. Thus, our results shed new light on the role of MSCs in modulating regulatory DC production and support the clinical application of MSCs to reduce the inflammatory response in numerous disease states. The Journal of Immunology, 2012, 189: 1182–1192. http://www.jimmunol.org/ esenchymal stem/stromal cells (MSCs), capable of DCs, especially regulatory DCs (regDCs), are key regulators of multilineage differentiation (1, 2), are weakly immu- immune responses. Several types of regDCs, with similar functions M nogenic and possess immune regulatory properties (3– but different phenotypes (13–18), can be induced by a cytokine 5). Previous studies have shown that bone marrow-derived MSCs mixture that includes GM-CSF, IL-10, and TGF-b in vitro; how- (BM-MSCs) can secrete inhibitory cytokines, suppress T cell ever, these induction conditions do not accurately mimic the mi- activation and proliferation, and regulate dendritic cell (DC) croenvironment in vivo, which involves a complex orchestration by guest on October 3, 2021 functions (6–9). In vivo, MSCs can prevent and treat graft-versus- and regulation of cytokine production and cell-cell interaction. host disease by suppressing Th1 lymphocyte function (10), at- Previous reports showed that BM-MSCs could influence DC dif- tenuate the pathological manifestations of experimental autoim- ferentiation (8, 9), and Cao’s group (19) showed that endothelial mune encephalomyelitis via regulatory T cell expansion and splenic stroma could induce the differentiation of CD11bhighIalow proinflammatory Th17 cell depression (11), and inhibit B cell regDCs from bone marrow hematopoietic stem/progenitor cells activation, proliferation, and IgG secretion in systemic lupus (BM-HPCs), but the induction of regDCs in vivo remains con- erythematosus (12). Although MSCs can regulate T and B immune troversial. cells via many mechanisms, relatively little is known about the Recent data support the concept that specific gene expression effects of MSCs on DC function. patterns that determine the behavior of individual cells are under *Institute of Basic Medical Sciences and School of Basic Medicine, Center of Excel- and Project of Institute of Basic Medical Sciences of Chinese Academy of Medical lence in Tissue Engineering, Chinese Academy of Medical Sciences and Peking Union Sciences Grant 2008PY09. Medical College, Beijing 100005, People’s Republic of China; †Academy of Integra- Address correspondence and reprint requests to Prof. Robert C.H. Zhao or Dr. tive Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350108, Armand Keating, Institute of Basic Medical Sciences and School of Basic Medicine, Fujian, People’s Republic of China; ‡Department of Genetics, Rutgers, The State x Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences University of New Jersey, Piscataway, NJ 08854; Department of Cell Biology, Insti- and Peking Union Medical College, 5 Dongdansantiao, Beijing 100005, People’s tute for Biomedical Engineering, Rhenish-Westphalian Technical University, Aachen { Republic of China (R.C.H.Z.) or Cell Therapy Program, Princess Margaret Hospital, University Medical School, 52074 Aachen, Germany; Cell Therapy Program, Prin- ‖ 610 University Avenue, Toronto, Ontario M5G 2M9, Canada (A.K.). E-mail cess Margaret Hospital, Toronto, Ontario M5G 2M9, Canada; Institute of Biomate- addresses: [email protected] (R.C.H.Z.) and [email protected] (A.K.) rials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5G 2M9, Canada; and #Institute of Medical Science, University of Toronto, Toronto, The online version of this article contains supplemental material. Ontario M5G 2M9, Canada Abbreviations used in this article: BM-HPC, bone marrow hematopoietic stem/ 1X.L. and X.Q. contributed equally to this work. progenitor cell; BM-MSC, bone marrow-derived mesenchymal stem/stromal cell; ChIP, chromatin immunoprecipitation; DC, dendritic cell; HPC, hematopoietic Received for publication October 17, 2011. Accepted for publication May 23, 2012. stem/progenitor cell; imDC, immature DC; maDC, mature DC; MEF, mouse embry- This work was supported by “863 Projects” of Ministry of Science and Technology of onic fibroblast; MSC, mesenchymal stem/stromal cell; NA, neutralizing Ab; NS, People’s Republic of China Grant 2006AA02A109; National Natural Science Foun- normal saline; PCA, principal components analysis; qRT-PCR, quantitative RT- 2 2 dation of China Grants 30830052, 30700321, 30800429, and 30911130363; Beijing PCR; regDC, regulatory DC; sDC, Sca-1+Lin CD117 MEF-MSC–induced DC; Ministry of Science and Technology Grant D07050701350701; Major National siRNA, small interfering RNA. Science and Technology Project Grants 2008ZX09101-044 and 2009ZX09503-025; National Key Scientific Program of China Grant 2011CB964900; Program for Copyright Ó 2012 by The American Association of Immunologists, Inc. 0022-1767/12/$16.00 Changjiang Scholars and Innovative Research Team in University Grant IRT0909; www.jimmunol.org/cgi/doi/10.4049/jimmunol.1102996 The Journal of Immunology 1183 the control of epigenetic alterations, including histone mod- Coculture experiment ifications. Methylation of the histone lysines, H3K4 and H3K27, HPCs were enriched from bone marrow cells by depleting lineage-specific mediated by distinct histone methyltransferase complexes, is cells. In brief, an EasySep mouse hematopoietic progenitor enrichment kit highly correlated with transcriptional activation or repression (20, (StemCell Technologies) was used to deplete lineage-specific cells. After + 2 2 21). A growing body of evidence has shown that epigenetic HPCs were purified, they were seeded onto Sca-1 CD117 Lin mono- 3 5 alterations are involved in the regulation of various genes layers at a density of 1 10 in 2 ml per well in 6-well plates. The ratio of MSC/HPC is 1:10. LPS was added on day 4. In some experiments, expressed during normal embryonic development, stem cell dif- a neutralizing Ab (NA) for IL-10 (10 mg/ml) was added every 2 d; 100 mM ferentiation, cancer growth, and autoimmune disease progress (22, NSC 74859 (a phospho-STAT3 inhibitor; Merck) or 10 mM CGP 41251 (a 23). Histone methylation has also been implicated in the devel- phospho-STAT5 inhibitor; Merck) was used for 24 or 48 h, respectively; opment of immune responses, especially in the production of IL- serum replacement (StemCell Technologies) was used to substitute for serum. After coculture on Sca-1+CD1172Lin2 feeder layers for 9
Recommended publications
  • IL-12 Abrogates Calcineurin-Dependent Immune Evasion During Leukemia Progression
    Author Manuscript Published OnlineFirst on May 29, 2019; DOI: 10.1158/0008-5472.CAN-18-3800 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. IL-12 abrogates calcineurin-dependent immune evasion during leukemia progression Jennifer L. Rabe,1* Lori Gardner,2* Rae Hunter,3 Jairo A. Fonseca,3 Jodi Dougan,3 Christy M. Gearheart,2 Michael S. Leibowitz,2 Cathy Lee-Miller,2 Dmitry Baturin,2 Susan P. Fosmire,2 Susan E. Zelasko,2 Courtney L. Jones,2 Jill E. Slansky,4 Manali Rupji,5 Bhakti Dwivedi,5 Curtis J. Henry,3,5,6 and Christopher C. Porter3,5,6 Affiliations: 1Molecular Biology Program, University of Colorado Denver, Aurora, CO. 2Department of Pediatrics, University of Colorado, Aurora, CO 3Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 4Integrated Department of Immunology, University of Colorado School of Medicine, Aurora, CO 5Winship Cancer Institute, Emory University, Atlanta, GA 6Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, GA JR and LAG contributed equally to this study. Corresponding Author: Christopher C. Porter, MD Emory University School of Medicine 1760 Haygood Drive, E370 Atlanta, GA 30322 Phone: 404-727-4881 Fax: 404-727-4455 [email protected] Running title: Leukemia-cell calcineurin drives immune evasion Conflict of interest statement The authors have declared that no conflicts of interest exist. Abstract: 199 words Main Text: 5797 words (excluding figure legends and references) Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on May 29, 2019; DOI: 10.1158/0008-5472.CAN-18-3800 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
    [Show full text]
  • Human IL12A / NKSF1 Protein (His Tag)
    Human IL12A / NKSF1 Protein (His Tag) Catalog Number: 10021-H08H General Information SDS-PAGE: Gene Name Synonym: CLMF; IL-12A; NFSK; NKSF1; P35 Protein Construction: A DNA sequence encoding the p35 subunit of human IL12, termed as IL12A (P29459) (Met 1-Ser 219) was expressed, fused with a polyhistidine tag at the C-terminus. Source: Human Expression Host: HEK293 Cells QC Testing Purity: > 92 % as determined by SDS-PAGE Bio Activity: Protein Description Measured by its ability to bind Human IL12B-his in functional ELISA. Interleukin-12 subunit alpha (IL12A/IL-12p35) is also known as Cytotoxic lymphocyte maturation factor 35 kDa subunit, cytotoxic lymphocyte Endotoxin: maturation factor 1, p35, NK cell stimulatory factor chain 1, and interleukin- 12 alpha chain. IL12A/IL-12p35 is a subunit of a cytokine that acts on T < 1.0 EU per μg of the protein as determined by the LAL method and natural killer cells, and has a broad array of biological activities. The cytokine is a disulfide-linked heterodimer composed of the 35-kD subunit Stability: encoded by this gene, and a 40-kD subunit that is a member of the Samples are stable for up to twelve months from date of receipt at -70 ℃ cytokine receptor family. IL12A/IL-12p35 is required for the T-cell- independent induction of IFN-gamma, and is important for the Predicted N terminal: Arg 23 differentiation of both Th1 and Th2 cells. The responses of lymphocytes to this cytokine are mediated by the activator of transcription protein STAT4. Molecular Mass: Nitric oxide synthase 2A (NOS2A/NOS2) is found to be required for the signaling process of this cytokine in innate immunity.
    [Show full text]
  • Recombinant Human IL12 Protein,HEK293 Expressed Catalog # AMS.IL2-H4210 for Research and Further Cell Culture Manufacturing Use
    Recombinant Human IL12 Protein,HEK293 Expressed Catalog # AMS.IL2-H4210 For Research and Further Cell Culture Manufacturing Use Description Source Recombinant Human IL12 Protein,HEK293 Expressed (Human IL12B & IL12A heterodimer) Ile 23 - Ser 328 (Accession # (AAH67499,IL12B) and Arg 23 - Ser 219 (P29459,IL12A)) was produced in human 293 cells (HEK293) Predicted N‐terminus Ile 23 (IL12B) and Arg 23 (IL12A) Molecular Human IL12B & IL12A heterodimer contains no "tag", and has a calculated MW of 36.6 kDa (IL12B) and 23.8 kDa (IL12A). The Characterization predicted N-terminus is Ile 23 (IL12B) and Arg 23 (IL12A). DTT-reduced Protein migrates as 33-47 kDa in SDS-PAGE due to glycosylation. Endotoxin Less than 1.0 EU per μg of the Human IL12B & IL12A heterodimer by the LAL method. Purity >95% as determined by SDS-PAGE. Bioactivity The bio-activity was determined by the dose-dependent release of IFN-gamma from the human NK92 cell line in presence of 20 ng/mL rhIL-2 (Catalog # IL2-H4216). The ED50 < 0.2 ng/mL, corresponding to a specific activity of >5,000,000 Unit/mg. Formulation and Storage Formulation Lyophilized from 0.22 μm filtered solution in PBS, pH7.4. Normally Mannitol or Trehalose are added as protectants before lyophilization. Contact us for customized product form or formulation. Reconstitution See Certificate of Analysis for reconstitution instructions and specific concentrations. Storage Lyophilized Protein should be stored at -20℃ or lower for long term storage. Upon reconstitution, working aliquots should be stored at -20℃ or -70℃. Avoid repeated freeze-thaw cycles. No activity loss was observed after storage at: ● 4-8℃ for 12 months in lyophilized state; ● -70℃ for 3 months under sterile conditions after reconstitution.
    [Show full text]
  • EBI3 Regulates the NK Cell Response to Mouse Cytomegalovirus Infection
    EBI3 regulates the NK cell response to mouse cytomegalovirus infection Helle Jensena, Shih-Yu Chenb, Lasse Folkersenc, Garry P. Nolanb, and Lewis L. Laniera,d,1 aDepartment of Microbiology and Immunology, University of California, San Francisco, CA 94143; bDepartment of Microbiology and Immunology, Stanford University, Palo Alto, CA 94304; cDepartment of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby DK-2800, Denmark; and dParker Institute for Cancer Immunotherapy, San Francisco, CA 94143 Contributed by Lewis L. Lanier, January 7, 2017 (sent for review November 1, 2016; reviewed by Michael A. Caligiuri and Daniel J. Cua) Natural killer (NK) cells are key mediators in the control of a role in shaping the subsequent adaptive immune responses. cytomegalovirus infection. Here, we show that Epstein–Barr virus- Crosstalk between NK cells and dendritic cells (DCs) early induced 3 (EBI3) is expressed by human NK cells after NKG2D or IL-12 during MCMV infection affects the outcome of the T-cell re- plus IL-18 stimulation and by mouse NK cells during mouse cytomeg- sponses. IL-10 secreted by various immune cells, including NK alovirus (MCMV) infection. The induction of EBI3 protein expression in cells, dampens the T-cell response by negatively affecting the mouse NK cells is a late activation event. Thus, early activation events maturation of DCs, and in the absence of IL-10 secretion of of NK cells, such as IFNγ production and CD69 expression, were not IFNγ and TNFα by NK cells enhances the maturation of DCs, −/− affected in EBI3-deficient (Ebi3 ) C57BL/6 (B6) mice during MCMV which boosts the T-cell response (11).
    [Show full text]
  • Evolutionary Divergence and Functions of the Human Interleukin (IL) Gene Family Chad Brocker,1 David Thompson,2 Akiko Matsumoto,1 Daniel W
    UPDATE ON GENE COMPLETIONS AND ANNOTATIONS Evolutionary divergence and functions of the human interleukin (IL) gene family Chad Brocker,1 David Thompson,2 Akiko Matsumoto,1 Daniel W. Nebert3* and Vasilis Vasiliou1 1Molecular Toxicology and Environmental Health Sciences Program, Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO 80045, USA 2Department of Clinical Pharmacy, University of Colorado Denver, Aurora, CO 80045, USA 3Department of Environmental Health and Center for Environmental Genetics (CEG), University of Cincinnati Medical Center, Cincinnati, OH 45267–0056, USA *Correspondence to: Tel: þ1 513 821 4664; Fax: þ1 513 558 0925; E-mail: [email protected]; [email protected] Date received (in revised form): 22nd September 2010 Abstract Cytokines play a very important role in nearly all aspects of inflammation and immunity. The term ‘interleukin’ (IL) has been used to describe a group of cytokines with complex immunomodulatory functions — including cell proliferation, maturation, migration and adhesion. These cytokines also play an important role in immune cell differentiation and activation. Determining the exact function of a particular cytokine is complicated by the influence of the producing cell type, the responding cell type and the phase of the immune response. ILs can also have pro- and anti-inflammatory effects, further complicating their characterisation. These molecules are under constant pressure to evolve due to continual competition between the host’s immune system and infecting organisms; as such, ILs have undergone significant evolution. This has resulted in little amino acid conservation between orthologous proteins, which further complicates the gene family organisation. Within the literature there are a number of overlapping nomenclature and classification systems derived from biological function, receptor-binding properties and originating cell type.
    [Show full text]
  • The Therapeutic Effects of Intratumoral Injection of IL12IL2GMCSF Fusion Protein on Canine Tumors
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.03.131904; this version posted June 4, 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. The therapeutic effects of intratumoral injection of IL12IL2GMCSF fusion protein on canine tumors Xiaobo Du1, Bin Zhang2, Fan Hu3, Chuantao Xie4, Haiyan Tian5, Qiang Gu6, Haohan Gong7, Xiaoguang Bai1, Jinyu Zhang1* Affiliations: 1 Beijing Kenuokefu Biotechnology Company, Beijing, 102200, China 2 Beijing Chongfuxing Animal Hospital, Beijing, 102209, China 3 Meilianzhonghe Veterinary Hospital Referral Center, Beijing, 100101, China 4 Beijing Puppy Town Animal Hospital, Beijing, 100102, China 5 Beijing Guanshang Animal Hospital, Beijing, 100088, China 6 Beijing Guanzhong Animal Hospital, Beijing, 100123, China 7 Loving Care International Pet Medical Center, Beijing, 100124, China *To whom correspondence should be addressed: Jinyu Zhang Beijing Kenuokefu Biotechnology Company Building 1, 29#, Shengmingyuan Road, Changping district Beijing, 102200, China Phone: 86-10-80765889 Email: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2020.06.03.131904; this version posted June 4, 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. Abstract Regulating the immune system through tumor immunotherapy to defeat tumors is currently one of the most popular methods of tumor treatment. We previously found that the combinations of IL12, IL2 and GMCSF has superior antitumor activities. In this study, IL12IL2GMCSF fusion protein was produced from 293 cells transduced by expression lentiviral vector.
    [Show full text]
  • Batf2 Differentially Regulates Tissue Immunopathology in Type 1 and Type 2 Diseases
    www.nature.com/mi ARTICLE OPEN Batf2 differentially regulates tissue immunopathology in Type 1 and Type 2 diseases Reto Guler1,2,3, Thabo Mpotje1,2, Mumin Ozturk1,2, Justin K. Nono1,2,5, Suraj P. Parihar1,2,3,4, Julius Ebua Chia1,2, Nada Abdel Aziz1,2,6, Lerato Hlaka1,2, Santosh Kumar1,2, Sugata Roy7, Adam Penn-Nicholson8, Willem A. Hanekom8, Daniel E. Zak9, Thomas J. Scriba8, Harukazu Suzuki7 and Frank Brombacher1,2,3 Basic leucine zipper transcription factor 2 (Batf2) activation is detrimental in Type 1-controlled infectious diseases, demonstrated during infection with Mycobacterium tuberculosis (Mtb) and Listeria monocytogenes Lm. In Batf2-deficient mice (Batf2−/−), infected with Mtb or Lm, mice survived and displayed reduced tissue pathology compared to infected control mice. Indeed, pulmonary inflammatory macrophage recruitment, pro-inflammatory cytokines and immune effectors were also decreased during tuberculosis. This explains that batf2 mRNA predictive early biomarker found in active TB patients is increased in peripheral blood. Similarly, Lm infection in human macrophages and mouse spleen and liver also increased Batf2 expression. In striking contrast, Type 2-controlled schistosomiasis exacerbates during infected Batf2−/− mice with increased intestinal fibro-granulomatous inflammation, pro-fibrotic immune cells, and elevated cytokine production leading to wasting disease and early death. Together, these data strongly indicate that Batf2 differentially regulates Type 1 and Type 2 immunity in infectious diseases. Mucosal Immunology (2019) 12:390–402; https://doi.org/10.1038/s41385-018-0108-2 INTRODUCTION elevated expression of BATF2 as an early correlate for Batf2 is a transcription factor that belongs to the basic leucine tuberculosis (TB) disease progression in adolescents with latent zipper transcription factor family, which includes Batf and Batf3.1,2 Mycobacterium tuberculosis (Mtb) infection.
    [Show full text]
  • Supplementary Material Supplementary Tables
    BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Immunother Cancer Supplementary Material Supplementary Tables Supplementary Table 1 Clinicopathological characteristics of the 49 individual patients analysed for immunotherapy response. Line of Patient systemic Hepatitis AFP Age ICB Treatment (target) Response status Stage* Grade# ID treatme status Level (years) nt Combination PD-1/PD-L1 + HCC 001 CTLA-4 1st Progressive Disease (PD) None 34206 70 C 3 HCC 002 PD-1/PD-L1 1st Partial Response (PR) HCV 2.6 60 C 2 Combination PD-1/PD-L1 + HCC 003 others 1st Stable Disease (SD) HBV 4.2 78 C 3 Combination PD-1/PD-L1 + HCC 004 1st Progressive Disease (PD) None 1384 76 C 3 CTLA-4 HCC 005 PD-1/PD-L1 1st Progressive Disease (PD) None 60500 73 C 2 HCC 006 PD-1/PD-L1 1st Progressive Disease (PD) HBV 6 52 C 2 Combination PD-1/PD-L1 + HCC 007 others 2nd Progressive Disease (PD) HBV 1138 62 B 3 HCC 008 PD-1/PD-L1 1st Stable Disease (SD) HBV 4 65 C 3 HCC 009 PD-1/PD-L1 1st Partial Response (PR) HCV 48392 65 C 3 Combination PD-1/PD-L1 + HCC 010 CTLA-4 1st Partial Response (PR) HBV 832 79 C 3 HCC 011 PD-1/PD-L1 2nd Progressive Disease (PD) None 20 67 B 2 HCC 012 PD-1/PD-L1 1st Partial Response (PR) None 11.5 81 C 2 HCC 013 PD-1/PD-L1 1st Progressive Disease (PD) None 16.8 71 C 2 HCC 014 PD-1/PD-L1 1st Stable Disease (SD) HBV 87 79 C 3 Ng HHM, et al.
    [Show full text]
  • KRAS Mutations Are Negatively Correlated with Immunity in Colon Cancer
    www.aging-us.com AGING 2021, Vol. 13, No. 1 Research Paper KRAS mutations are negatively correlated with immunity in colon cancer Xiaorui Fu1,2,*, Xinyi Wang1,2,*, Jinzhong Duanmu1, Taiyuan Li1, Qunguang Jiang1 1Department of Gastrointestinal Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China 2Queen Mary College, Medical Department, Nanchang University, Nanchang, Jiangxi, People's Republic of China *Equal contribution Correspondence to: Qunguang Jiang; email: [email protected] Keywords: KRAS mutations, immunity, colon cancer, tumor-infiltrating immune cells, inflammation Received: March 27, 2020 Accepted: October 8, 2020 Published: November 26, 2020 Copyright: © 2020 Fu et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT The heterogeneity of colon cancer tumors suggests that therapeutics targeting specific molecules may be effective in only a few patients. It is therefore necessary to explore gene mutations in colon cancer. In this study, we obtained colon cancer samples from The Cancer Genome Atlas, and the International Cancer Genome Consortium. We evaluated the landscape of somatic mutations in colon cancer and found that KRAS mutations, particularly rs121913529, were frequent and had prognostic value. Using ESTIMATE analysis, we observed that the KRAS-mutated group had higher tumor purity, lower immune score, and lower stromal score than the wild- type group. Through single-sample Gene Set Enrichment Analysis and Gene Set Enrichment Analysis, we found that KRAS mutations negatively correlated with enrichment levels of tumor infiltrating lymphocytes, inflammation, and cytolytic activities.
    [Show full text]
  • IL-4 Abrogates TH17 Cell-Mediated Inflammation by Selective Silencing of IL-23 in Antigen-Presenting Cells
    IL-4 abrogates TH17 cell-mediated inflammation by selective silencing of IL-23 in antigen-presenting cells Emmanuella Guenovaa,b,c,1,2, Yuliya Skabytskaa,1, Wolfram Hoetzeneckera,b,c,1, Günther Weindla,d, Karin Sauera, Manuela Thama, Kyu-Won Kime, Ji-Hyeon Parke, Ji Hae Seoe,f, Desislava Ignatovac, Antonio Cozzioc, Mitchell P. Levesquec, Thomas Volza,g, Martin Köberlea,g, Susanne Kaeslera, Peter Thomash, Reinhard Mailhammeri,3, Kamran Ghoreschia, Knut Schäkelj, Boyko Amarovk, Martin Eichnerl, Martin Schallera, Rachael A. Clarkb, Martin Röckena,2, and Tilo Biedermanna,g,2 aDepartment of Dermatology, Eberhard Karls University, 72076 Tübingen, Germany; dInstitute of Pharmacy, Department of Pharmacology and Toxicology, Freie Universität, 14195 Berlin, Germany; eNeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea; fDepartment of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, Republic of Korea; cDepartment of Dermatology, University Hospital of Zürich, 8091 Zürich, Switzerland; hDepartment of Dermatology and Allergology, Ludwig Maximilians University, 80337 Munich, Germany; iInstitute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany; jDepartment of Dermatology, Heidelberg University Hospital, 69115 Heidelberg, Germany; kInstitute
    [Show full text]
  • POGLUT1, the Putative Effector Gene Driven by Rs2293370 in Primary
    www.nature.com/scientificreports OPEN POGLUT1, the putative efector gene driven by rs2293370 in primary biliary cholangitis susceptibility Received: 6 June 2018 Accepted: 13 November 2018 locus chromosome 3q13.33 Published: xx xx xxxx Yuki Hitomi 1, Kazuko Ueno2,3, Yosuke Kawai1, Nao Nishida4, Kaname Kojima2,3, Minae Kawashima5, Yoshihiro Aiba6, Hitomi Nakamura6, Hiroshi Kouno7, Hirotaka Kouno7, Hajime Ohta7, Kazuhiro Sugi7, Toshiki Nikami7, Tsutomu Yamashita7, Shinji Katsushima 7, Toshiki Komeda7, Keisuke Ario7, Atsushi Naganuma7, Masaaki Shimada7, Noboru Hirashima7, Kaname Yoshizawa7, Fujio Makita7, Kiyoshi Furuta7, Masahiro Kikuchi7, Noriaki Naeshiro7, Hironao Takahashi7, Yutaka Mano7, Haruhiro Yamashita7, Kouki Matsushita7, Seiji Tsunematsu7, Iwao Yabuuchi7, Hideo Nishimura7, Yusuke Shimada7, Kazuhiko Yamauchi7, Tatsuji Komatsu7, Rie Sugimoto7, Hironori Sakai7, Eiji Mita7, Masaharu Koda7, Yoko Nakamura7, Hiroshi Kamitsukasa7, Takeaki Sato7, Makoto Nakamuta7, Naohiko Masaki 7, Hajime Takikawa8, Atsushi Tanaka 8, Hiromasa Ohira9, Mikio Zeniya10, Masanori Abe11, Shuichi Kaneko12, Masao Honda12, Kuniaki Arai12, Teruko Arinaga-Hino13, Etsuko Hashimoto14, Makiko Taniai14, Takeji Umemura 15, Satoru Joshita 15, Kazuhiko Nakao16, Tatsuki Ichikawa16, Hidetaka Shibata16, Akinobu Takaki17, Satoshi Yamagiwa18, Masataka Seike19, Shotaro Sakisaka20, Yasuaki Takeyama 20, Masaru Harada21, Michio Senju21, Osamu Yokosuka22, Tatsuo Kanda 22, Yoshiyuki Ueno 23, Hirotoshi Ebinuma24, Takashi Himoto25, Kazumoto Murata4, Shinji Shimoda26, Shinya Nagaoka6, Seigo Abiru6, Atsumasa Komori6,27, Kiyoshi Migita6,27, Masahiro Ito6,27, Hiroshi Yatsuhashi6,27, Yoshihiko Maehara28, Shinji Uemoto29, Norihiro Kokudo30, Masao Nagasaki2,3,31, Katsushi Tokunaga1 & Minoru Nakamura6,7,27,32 Primary biliary cholangitis (PBC) is a chronic and cholestatic autoimmune liver disease caused by the destruction of intrahepatic small bile ducts. Our previous genome-wide association study (GWAS) identifed six susceptibility loci for PBC.
    [Show full text]
  • Targeting Androgen Receptor (AR)!IL12A Signal Enhances Efficacy
    Published OnlineFirst March 3, 2016; DOI: 10.1158/1535-7163.MCT-15-0706 Cancer Biology and Signal Transduction Molecular Cancer Therapeutics Targeting Androgen Receptor (AR)!IL12A Signal Enhances Efficacy of Sorafenib plus NK Cells Immunotherapy to Better Suppress HCC Progression Liang Shi1,2, Hui Lin1, Gonghui Li1, Ren-An Jin1, Junjie Xu1,2, Yin Sun2, Wen-Lung Ma3, Shuyuan Yeh2, Xiujun Cai1, and Chawnshang Chang2,3 Abstract Gender disparity has long been considered as a key to fully matin immunoprecipitation assay were applied for mechanism understand hepatocellular carcinoma (HCC) development. At dissection. IHC was performed for sample staining. Our results the same time, immunotherapy related to IL12 still need more showed AR could suppress IL12A expression at the transcrip- investigation before being applied in clinical settings. The aim tional level via direct binding to the IL12A promoter region that of this study is to investigate the influence of the androgen resulted in repressing efficacy of NK cell cytotoxicity against receptor (AR) on natural killer (NK) cell–related innate HCC, and sorafenib treatment could enhance IL12A signals via immune surveillance in liver cancer, and provide a novel suppressing AR signals. These results not only help to explain therapeutic approach to suppress HCC via altering IL12A. By the AR roles in the gender disparity of HCC but also provide a using in vitro cell cytotoxicity test and in vivo liver orthotopic potential new therapy to better suppress HCC via combining xenograft mouse model, we identifiedtheroleofARinmod- sorafenib with NK cell–related immunotherapy. Mol Cancer Ther; ulating NK cell cytotoxicity. Luciferase report assay and chro- 15(4); 731–42.
    [Show full text]