CD70 CD27 Ligation Between Neural Stem Cells and CD4+ T Cells

Total Page:16

File Type:pdf, Size:1020Kb

CD70 CD27 Ligation Between Neural Stem Cells and CD4+ T Cells Lee et al. Stem Cell Research & Therapy 2013, 4:56 http://stemcellres.com/content/4/3/56 RESEARCH Open Access CD70–CD27 ligation between neural stem cells and CD4+ T cells induces Fas–FasL-mediated T-cell death Eun Mi Lee1,2, Sunghoon Hurh1, Bumrae Cho1, Kook-Hwan Oh3, Seung U Kim4, Charles D Surh5, Jonathan Sprent6, Jaeseok Yang7, Jae Young Kim8 and Curie Ahn1,3,7* Abstract Introduction: Neural stem cells (NSCs) are among the most promising candidates for cell replacement therapy in neuronal injury and neurodegenerative diseases. One of the remaining obstacles for NSC therapy is to overcome the alloimmune response on NSCs by the host. Methods: To investigate the mechanisms of immune modulatory function derived from the interaction of human NSCs with allogeneic T cells, we examined the immune regulatory effects of human NSCs on allogeneic T cells in vitro. Results: Significantly, NSCs induced apoptosis of allogeneic T cells, in particular CD4+ T cells. Interaction of CD70 on NSCs and CD27 on CD4+ T cells mediated apoptosis of T cells. Thus, blocking CD70–CD27 interaction prevented NSC-mediated death of CD4+ T cells. Conclusions: We present a rational explanation of NSC-induced immune escape in two consecutive stages. First, CD70 constitutively expressed on NSCs engaged CD27 on CD4+ T cells, which induced Fas ligand expression on CD4+ T cells. Second, CD4+ T-cell apoptosis was followed by Fas–Fas ligand interaction in the CD4+ T cells. Keywords: Neural stem cells, Co-stimulatory molecules, Immune escape mechanism Introduction Transplantation of rodent embryonic stem cells (ESCs) Neural stem cells (NSCs) are among the most promising into allogeneic recipients indicated that they may have candidates for cell replacement therapy in neuronal in- immune-privileged properties. The injection of undiffer- jury and neurodegenerative diseases. Derivation methods entiated rat ESC-like cells into fully MHC-mismatched to produce neuronal cells with specific functions from rats led to the induction of donor-specific immuno- NSCs, such as dopaminergic neurons, have also been logical tolerance [6]. Moreover, undifferentiated allogen- established [1]. Therefore, it is highly likely that NSCs eic murine ESCs led to engraftment of these cells in the will become the first choice for stem cell treatment. thymus, spleen and liver, with establishment of mixed Most cellular and organ transplantation between two un- hematopoietic chimerism [7]. This also proved that ESCs related individuals results in graft rejection. However, it has represent a good research tool for possible therapeutic been often found that stem cell transplantation could es- applications in solid organ transplantation because of cape the graft rejection [2-4], presumably by inducing apop- their capability, when given alone, to induce tolerance to tosis of T cells via Fas–Fas ligand (FasL) interaction [5]. semi-allogeneic solid organ allograft [8]. NSC transplantation could be applied to the brain * Correspondence: [email protected] because this organ is subject to less surveillance by the 1 Transplantation Research Institute, Seoul National University Medical immune system than other parts of the body, and like brain Research Center, College of Medicine Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul 110-799, Korea cells NSCs may display characteristics of cells with immune 3Department of Internal Medicine, Seoul National University Hospital, 101 privilege. Consistent with this idea, T cells that infiltrate Daehak-ro, Jongno-gu, Seoul 110-744, Korea around transplanted NSCs in a stroke model exhibited little Full list of author information is available at the end of the article © 2013 Lee et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Lee et al. Stem Cell Research & Therapy 2013, 4:56 Page 2 of 10 http://stemcellres.com/content/4/3/56 alloimmune response [9], suggesting NSCs have a strong 5×104:1 × 105 cells/well on 96-well plates in well media. potential to protect cells of the central nervous system Adherent NSCs and nonadherent T cells were separately against harmful T-cell infiltration [10]. In a related study, harvested at the indicated times and washed in 1× PBS. the systemic injection of neuronal stem/precursor cells pro- vided a remarkable amelioration of the pathological features Analysis of T-cell apoptosis of experimentally induced autoimmune encephalomyelitis Apoptotic cell death was assessed using Annexin V staining and other preclinical models of neurological disorders coupled with propidium iodide (BD PharMingen, [11,12]. San Diego, CA, USA). T cells were cultured with HB1.F3 at In this paper, we have investigated the mechanisms re- a various densities or times. T cells were washed twice with sponsible for NSC-induced immune tolerance. Our studies 1× PBS. Then 2 × 105 to 5 × 105 Tcellswereaddedtoa12 indicate a two-stage model whereby human NSCs induce mmtesttubeandwashedtwicein1mlof1×binding apoptosis of allogeneic CD4+ T cells. First, CD70 expressed buffer. Cells were mixed gently with 5 μlAnnexin constitutively on NSCs engage CD27 on CD4+ Tcellsand V–fluorescein isothiocyanate and incubated at 4°C in the induce upregulation of FasL expression on the CD4+ dark for 5 minutes. After washing with binding buffer, 10 μl T cells. Second, T cells thereafter undergo apoptosis from of the 20 μg/ml propidium iodide solution (end fratricide; that is, through Fas–FasL interaction in the concentration 1 μg/ml) and 190 μl binding buffer were CD4+ T cells themselves. added to the cells. Cells were immediately analyzed by fluorescence-activated cell sorting (FACS). Materials and methods Cell culture Flow cytometry The immortalized human embryonic neural stem cell line, Adherent NSCs were trypsinized and resuspended in HB1.F3 was generated in a previous study by transfection staining buffer (0.1% BSA in PBS) to reach a final con- of primary cells from fetal human telencephalon tissue centration of 5 × 105 to 1 × 106 cells/ml. The cells were of 14 weeks gestation with amphotropic, replication- incubated for 20 minutes on ice with the following anti- incompetent retroviral vector containing v-myc [13]. bodies: anti-CD27 (O323), anti-CD70 (Ki-24), anti-Fas PrimaryNSCswereprovidedbyProfessorKim(Korea (APO-1), anti-FasL (NOK-1), anti-PD-1 (J105), anti-PD- University, Seoul, Korea) [14,15]. HB1.F3 cells and primary L1 (MIH1), anti-TR-1 (DJR1), anti-TR-2 (DJR2-4), and NSCs were cultured in DMEM with 10% heat-inactivated anti-TRAIL (RIK-2). After incubation, the cells were fetal bovine serum (Gibco, Carlsbad, CA, USA), and 1% washed twice with staining buffer and resuspended. The antibiotic–antimycotic solution (Gibco, Carlsbad, CA, stained cells were analyzed by FACS. USA). Human umbilical cord blood mesenchymal stem cells purchased from MEDIPOST Co., Ltd (Seoul, Korea) Semiquantitative RT-PCR were used. Human umbilical cord blood-derived mesen- Total RNA was prepared from CD4+ T cells using the RNA chymal stem cells were isolated and expanded as isolation kit (QIAGEN Inc, Valencia, CA, USA). Two described previously [16,17]. Cultures were maintained micrograms of RNA were reverse-transcribed. The reaction in a humidified incubator at 37°C containing 5% CO2. mixture consisted of 20 mM Tris–HCl (pH 8.4), 50 mM KCl, 5 mM MgCl2, 10 mM dithiothreitol, 40 U of + + CD4 and CD8 T-cell preparation RNaseOUT™,1mMdNTP,2.5μM of oligo(dT)20,200U Whole blood was collected from healthy volunteer donors of SuperScript™ III reverse transcriptase (Invitrogen, (n = 55, age: 24.85 ± 3.52, sex: male/female = 36/19). The Carlsbad, CA, USA) and the remaining volume was institutional review board of the Seoul National University diethylpyrocarbonate-treated water for a total volume of 20 Hospital approved this study, and all volunteers provided μl. The mixture was incubated at 50°C for 1 hour, and at written informed consent. Peripheral blood mononuclear 85°C for 5 minutes. For semiquantitative PCR, the reaction cells were isolated by Ficoll-Paque PLUS (Amersham mixture consisted of 10 mM Tris–HCl, 50 mM KCl, 0.1% Biosciences, Piscataway, NJ) density gradient centrifugation Triton X-100, 2 mM MgCl2,0.4mMdNTP,0.2pmolsense of peripheral venous blood [18]. The peripheral blood and antisense primer, 2.5 U Taq DNA polymerase and 3 μg mononuclear cells were separated into CD4+ and CD8+ cDNA. Primer sequences are provided in Additional file 1. T-cell populations by magnetic-activated cell separation (Miltenyi Biotec, Bergisch Gladbach, Germany). HLA-A, HLA-B and HLA-DR allele on HB1.F3 cells We examined HLA-A, HLA-B, and HLA-DR alleles on Co-culturing T cells with neural stem cells HB1.F3 cells using the PCR sequence-specific oligo- To determine NSC-dependent apoptosis, CD4+ or CD8+ T nucleotide method (Dynal RELI™ HLA typing kit, Dynal cells were cultured with HB1.F3 at a density of 5 × 104:5 × Biotech, Wirral, U.K.) [19]. The results are provided in 103,5×104:1 × 104,5×104:2.5 × 104,5×104:5 × 104,and Additional file 2. Lee et al. Stem Cell Research & Therapy 2013, 4:56 Page 3 of 10 http://stemcellres.com/content/4/3/56 Treatment of anti-human Fas ligand mAbs Combinatory treatment of anti-CD27, anti-PD-L1, or CD4+ T cells were incubated with HB1.F3 for 24 hours anti-CD40 mAb with a series of concentrations of neutralizing Fas ligand CD4+ T cells were incubated with HB1.F3 for 24 hours mAb (NOK-2): 0, 0.01, 0.1, 0.5, and 1 μg/ml. with 0.5 μg/ml of anti-CD27 (LG.7F9), anti-PD-L1 (MIH-1), or anti-CD40 mAb (5C3). Treatment of actinomycin D and cycloheximide Statistical analysis We treated a transcription inhibitor, actinomycin D + All results are expressed as the mean ± standard devi- (1 μg/ml; Sigma-Aldrich, St.
Recommended publications
  • A Fratricide-Resistant Allogeneic CAR T
    Investigation of ALLO-316: A Fratricide- Resistant Allogeneic CAR T Targeting CD70 As a Potential Therapy for the Treatment of AML Surabhi Srinivasan, Nguyen Tan, Hsin-Yuan Cheng, Yi Zhang, Silvia Tacheva-Grigorova, Tom Van Blarcom, Cesar Sommer, Duy Nguyen , Barbra Sasu, and Siler Panowski 1 Disclosures • Full-time employee of Allogene Therapeutics • Equity interest in Allogene Therapeutics ALLO-316 (CD70) utilizes TALEN® gene-editing technology pioneered and owned by Cellectis. Allogene has an exclusive license to the Cellectis technology for allogeneic products directed at this target and holds all global development and commercial rights for this investigational candidate. 22 CONFIDENTIAL Disclaimers This presentation is not intended for product promotion. All information is related to investigational therapies not available for commercial use. The safety and efficacy of the therapies have not been established for FDA approval. Forward-Looking Statements To the extent statements contained in this Presentation are not descriptions of historical facts regarding Allogene Therapeutics, Inc. (“Allogene,” “we,” “us,” or “our”), they are forward-looking statements reflecting management’s current beliefs and expectations. Forward-looking statements are subject to known and unknown risks, uncertainties, and other factors that may cause our or our industry’s actual results, levels or activity, performance, or achievements to be materially different from those anticipated by such statements. You can identify forward-looking statements by words such as “anticipate,” “believe,” “could,” “estimate,” “expect,” “intend,” “may,” “plan,” “potential,” “predict,” “project,” “should,” “will,” “would” or the negative of those terms, and similar expressions that convey uncertainty of future events or outcomes. Forward-looking statements contained in this Presentation include, but are not limited to, statements regarding: the ability to progress the clinical development of allogeneic CAR T (AlloCAR T™) therapies and the potential benefits of AlloCAR T™ therapy, including ALLO-316.
    [Show full text]
  • Multiple Interactions of the Cytosolic Polyproline Region of the CD95
    FEBS 25561 FEBS Letters 509 (2001) 255^262 View metadata, citation and similar papers at core.ac.uk brought to you by CORE Multiple interactions of the cytosolic polyproline region ofprovided the by CD95 Elsevier - Publisher Connector ligand: hints for the reverse signal transduction capacity of a death factor1 Jennifer Wenzela;2, Ralf Sanzenbachera;2, Markus Ghadimia, Marc Lewitzkyb, Qingchun Zhouc, David R. Kapland, Dieter Kabelitza, Stephan M. Fellerb, Ottmar Janssena;* aInstitute for Immunology, Christian-Albrechts-University, MichaelisstraMe 5, 24105 Kiel, Germany bCell Signalling Laboratory, Imperial Cancer Research Fund, University of Oxford, Institute of Molecular Medicine, John Radcli¡e Hospital, Headington, Oxford, UK cInstitute of Organic Synthesis, Center China Normal University, 430079 Wuhan, PR China dDepartment of Pathology, Case Western Reserve University, 2085 Adelbert Road, Cleveland, OH 44106, USA Received 19 September 2001; revised 7 November 2001; accepted 7 November 2001 First published online 20 November 2001 Edited by Giulio Superti-Furga regulate activation of CD4- and CD8-positive T cells in Abstract The CD95/Fas/Apo-1 ligand is expressed on activated lymphocytes, NK cells, platelets, certain immune-privileged cells vivo. Upon stimulation with T cell receptor (TCR) agonists and some tumor cells and induces apoptosis through the death in the presence of CD95, cell cycle progression of CD4-pos- receptor CD95/Fas/Apo-1. In murine T cells, membrane-bound itive cells was found to be inhibited [14^16], while CD8-pos- CD95L (Fas ligand) also acts as a costimulatory receptor to itive cells were activated to proliferate [13^16]. The molecular coordinate activation and function in vivo.
    [Show full text]
  • Human Melanoma-Reactive CD4+ and CD8+ CTL Clones Resist Fas
    Human Melanoma-Reactive CD4+ and CD8+ CTL Clones Resist Fas Ligand-Induced Apoptosis and Use Fas/Fas Ligand-Independent Mechanisms for Tumor Killing This information is current as of September 29, 2021. Licia Rivoltini, Marina Radrizzani, Paola Accornero, Paola Squarcina, Claudia Chiodoni, Arabella Mazzocchi, Chiara Castelli, Paolo Tarsini, Vincenzo Viggiano, Filiberto Belli, Mario P. Colombo and Giorgio Parmiani J Immunol 1998; 161:1220-1230; ; Downloaded from http://www.jimmunol.org/content/161/3/1220 References This article cites 60 articles, 32 of which you can access for free at: http://www.jimmunol.org/content/161/3/1220.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on September 29, 2021 • 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 © 1998 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Human Melanoma-Reactive CD41 and CD81 CTL Clones Resist Fas Ligand-Induced Apoptosis and Use Fas/Fas Ligand-Independent Mechanisms for Tumor Killing1 Licia Rivoltini,2* Marina Radrizzani,* Paola Accornero,* Paola Squarcina,* Claudia Chiodoni,* Arabella Mazzocchi,* Chiara Castelli,* Paolo Tarsini,* Vincenzo Viggiano,† Filiberto Belli,‡ Mario P.
    [Show full text]
  • Uva-DARE (Digital Academic Repository)
    UvA-DARE (Digital Academic Repository) Balancing effector lymphocyte formation via CD27-CD70 interactions Arens, R. Publication date 2003 Link to publication Citation for published version (APA): Arens, R. (2003). Balancing effector lymphocyte formation via CD27-CD70 interactions. General rights It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible. UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl) Download date:27 Sep 2021 Chapter 3 Constitutive CD27/CD70 interaction induces expansion of effector-type T cells and results in IFNy-mediated B cell depletion Ramon Arens*, Kiki Tesselaar*, Paul A. Baars, Gijs M.W. van Schijndel, Jenny Hendriks, Steven T. Pals, Paul Krimpenfort, Jannie Borst, Marinus H.J. van Oers, and René A.W. van Lier 'These authors contributed equally to this work Immunity 15, 801-812 (2001) Chapter 3 Constitutive CD27/CD70 interaction induces expansion of effector-type T cells and results in IFNy-mediated B cell depletion Ramon Arens123#, Kiki Tesselaar23", Paul A.
    [Show full text]
  • Increased Expression of CD154 and FAS in SLE Patients’ Lymphocytes Maria Elena Manea, Ruediger B
    Increased expression of CD154 and FAS in SLE patients’ lymphocytes Maria Elena Manea, Ruediger B. Mueller, Doru Dejica, Ahmed Sheriff, Georg Schett, Martin Herrmann, Peter Kern To cite this version: Maria Elena Manea, Ruediger B. Mueller, Doru Dejica, Ahmed Sheriff, Georg Schett, et al.. Increased expression of CD154 and FAS in SLE patients’ lymphocytes. Rheumatology International, Springer Verlag, 2009, 30 (2), pp.181-185. 10.1007/s00296-009-0933-4. hal-00568285 HAL Id: hal-00568285 https://hal.archives-ouvertes.fr/hal-00568285 Submitted on 23 Feb 2011 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Increased expression of CD154 and FAS in SLE patients’ lymphocytes Maria Elena Manea1‡, MD, Ruediger B. Mueller2,3‡, MD, Doru Dejica1, PhD, Ahmed Sheriff2, PhD, Georg Schett2, MD, Martin Herrmann2, PhD, Peter Kern4, MD 1 Department of Immunopathology. “Iuliu Hatieganu" University of Medicine and Pharmacy, Str Croitorilor no 19-21, 3400 Cluj-Napoca, Romania. 2 Department for Internal Medicine 3 and Institute for Clinical Immunology, University of Erlangen-Nürnberg, Germany 3 Departement of Rheumatologie, Kantonsspital St. Gallen, Switzerland 4 Franz von Prümmer Klinik, Bahnhofstraße 16, 97769 Bad Brückenau, Germany ‡ both authors equally contributed to the work Address correspondence and reprint requests to: Ruediger B.
    [Show full text]
  • CD95 Ligand - Death Factor and Costimulatory Molecule?
    Cell Death and Differentiation (2003) 10, 1215–1225 & 2003 Nature Publishing Group All rights reserved 1350-9047/03 $25.00 www.nature.com/cdd Review CD95 ligand - death factor and costimulatory molecule? O Janssen*,1, J Qian1, A Linkermann1 and D Kabelitz1 Tissue and Cellular Expression of CD95L 1 Institute for Immunology, Medical Center Schleswig-Holstein, Campus Kiel, Michaelisstrasse 5, D-24105 Kiel, Germany The CD95 ligand (CD95L, Apo-1L, FasL, CD178) is a 281- * Corresponding author: O Janssen. Tel: þ 49-431-5973377; Fax: þ 49-431- amino-acid-containing type II transmembrane protein of the 5973335; E-mail: [email protected] TNF family of death factors (Figure 1).1 Its death-inducing function is best documented in the context of activation- Received 24.4.03; revised 12.6.03; accepted 20.6.03; published online 1 August 2003 induced cell death (AICD) in T cells.2 CD95L is expressed as a Edited by T Ferguson death factor in cytotoxic T lymphocytes (CTL) to kill virally infected or transformed target cells and in natural killer (NK) cells, where it is upregulated by CD16 engagement and 3 Abstract cytokines including IL-2 and IL-12. Similarly, high levels of intracellular CD95L have been detected in monocytic cells The CD95 ligand is involved as a death factor in the with an inducible release upon activation.4 Under physiologi- regulation of activation-induced cell death, establishment cal conditions, CD95L is implicated in the control of erythroid of immune privilege and tumor cell survival. In addition, differentiation,5 angiogenesis in the eye6 and skin home- 7 CD95L may serve as a costimulatory molecule for T-cell ostasis.
    [Show full text]
  • Interaction Apoptosis Mediated by Fas/Fas Ligand Osteoclast Formation In
    Effect of IL-12 on TNF-α-Mediated Osteoclast Formation in Bone Marrow Cells: Apoptosis Mediated by Fas/Fas Ligand Interaction This information is current as of October 6, 2021. Hideki Kitaura, Noriko Nagata, Yuji Fujimura, Hitoshi Hotokezaka, Noriaki Yoshida and Koji Nakayama J Immunol 2002; 169:4732-4738; ; doi: 10.4049/jimmunol.169.9.4732 http://www.jimmunol.org/content/169/9/4732 Downloaded from References This article cites 54 articles, 29 of which you can access for free at: http://www.jimmunol.org/content/169/9/4732.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • 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 by guest on October 6, 2021 *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 © 2002 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Effect of IL-12 on TNF-␣-Mediated Osteoclast Formation in Bone Marrow Cells: Apoptosis Mediated by Fas/Fas Ligand Interaction1 Hideki Kitaura,2* Noriko Nagata,*† Yuji Fujimura,*† Hitoshi Hotokezaka,* Noriaki Yoshida,* and Koji Nakayama† Recently, it has been found that differentiation into osteoclasts is induced by TNF-␣.
    [Show full text]
  • Characterization of Murine CD70, the Ligand of the TNF Receptor Family Member CD 27
    UvA-DARE (Digital Academic Repository) Characterization of murine CD70, the ligand of the TNF receptor family member CD 27 Tesselaar, N.A.; Gravestein, L.A.; van Schijndel, G.; Borst, J.; van Lier, R.A.W. Publication date 1997 Published in The journal of immunology Link to publication Citation for published version (APA): Tesselaar, N. A., Gravestein, L. A., van Schijndel, G., Borst, J., & van Lier, R. A. W. (1997). Characterization of murine CD70, the ligand of the TNF receptor family member CD 27. The journal of immunology, 159, 4959-4965. General rights It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible. UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl) Download date:30 Sep 2021 Characterization of Murine CD70, the Ligand of the TNF Receptor Family Member CD27' Kiki Tesselaar,* Loes A.
    [Show full text]
  • Co-Expression of CD40L with CD70 Or OX40L Increases B-Cell Viability and Antitumor Efficacy
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 29 Research Paper Co-expression of CD40L with CD70 or OX40L increases B-cell viability and antitumor efficacy Chang-Ae Shin1, Hyun-Woo Cho1, A-Ri Shin2,3, Hyun-Jung Sohn2, Hyun-Il Cho2,3, Tai-Gyu Kim1,2,3 1Department of Microbiology and Immunology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea 2Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea 3Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea Correspondence to: Hyun-Il Cho, email: [email protected] Tai-Gyu Kim, email: [email protected] Keywords: cancer vaccine, B-cells, costimulatory ligand, anti-apoptosis, tumor immunity Received: February 24, 2016 Accepted: May 29, 2016 Published: June 15, 2016 ABSTRACT Activated B-cells are a promising alternative source of antigen-presenting cells. They can generally be obtained in sufficient numbers for clinical use, but in most instances produce weak immune responses and therapeutic effects that are suboptimal for use in therapeutic cancer vaccines. To improve the immunogenic potency and therapeutic efficacy of B-cell-based vaccines, ex vivo-activated B-cells were transduced with recombinant lentiviruses in order to express additional costimulatory ligands—CD40L, CD70, OX40L, or 4-1BBL—either individually or in pairs (CD70/CD40L, OX40L/CD40L, or 4-1BBL/CD40L). We observed that the expression of CD40L molecules on B-cells was crucial for T-cell priming and activation. Administration of B-cells co-expressing CD40L with the other costimulatory ligands provided substantial antigen-specific CD8 T-cell responses capable of provoking in vivo proliferation and potent cytolytic activities.
    [Show full text]
  • Signaling by the Epstein–Barr Virus LMP1 Protein Induces Potent
    Signaling by the Epstein–Barr virus LMP1 protein + + induces potent cytotoxic CD4 and CD8 T cell responses Il-Kyu Choia,b, Zhe Wanga,b, Qiang Kea,c, Min Honga,d, Yu Qiana,b, Xiujuan Zhaoa,e, Yuting Liuf, Hye-Jung Kimg, Jerome Ritza,b, Harvey Cantorg, Klaus Rajewskyh,1, Kai W. Wucherpfennigg, and Baochun Zhanga,b,g,1 aDepartment of Medical Oncology, Dana–Farber Cancer Institute, Boston, MA 02215; bDepartment of Medicine, Harvard Medical School, Boston, MA 02115; cDepartment of Diagnostics, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; dDepartment of Medical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; eDepartment of Cell Biology, Tianjin Medical University, Tianjin 300070, China; fProgram in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115; gDepartment of Cancer Immunology and Virology, Dana–Farber Cancer Institute, Boston, MA 02215; and hImmune Regulation and Cancer, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany Contributed by Klaus Rajewsky, December 9, 2017 (sent for review August 3, 2017; reviewed by Alan B. Rickinson and Susan L. Swain) The B-lymphotropic Epstein–Barr virus (EBV), pandemic in humans, pressive molecules that foster local immune privilege (9, 10). is rapidly controlled on initial infection by T cell surveillance; there- Apparently, host immune cells, particularly T cells, keep EBV- after, the virus establishes a lifelong latent infection in the host. If infected cells under constant surveillance, and EBV-driven ma- surveillance fails, fatal lymphoproliferation and lymphomagenesis + lignancies only arise when surveillance fails. ensue. The initial T cell response consists of predominantly CD8 + T cell responses against EBV-infected or transformed B cells cytotoxic T cells and a smaller expansion of CD4 cells.
    [Show full text]
  • The Role of Fasl/Fas Signaling Network in Infections and Tumorigenesis
    The Role of FasL/Fas Signaling Network in Infections and Tumorigenesis T Cell-Mediated Cytotoxicity: All multicellular organisms are susceptible to tumor formation as well as infections from pathogenic viruses and bacteria, which serve as threats to the host organism. The host in turn deploys an immune response to ward off infections and eliminate malignant cells. T cell-mediated cytotoxicity is a complex and coordinated process that is critical for efficient immunological control of pathogenic infections and tumors (Kagi et al., 1996). This host defense mechanism is mediated by cytotoxic lymphocytes, a class of cells that includes CD8+ cytotoxic T lymphocytes (CTLs). As important effectors of the adaptive immune system, CTLs scan tissues for cellular abnormalities and infections and have the ability to directly destroy malignant/infected cells by inducing apoptosis. Briefly, CTLs screen potential target cells for expression of foreign peptide fragments from cellular proteins and then destroy identified cells. The specificity of this process is mediated through cell surface T cell receptors on CTLs, which interact with antigen-derived peptides presented by the class I molecules of the major histocompatibility complex (MHC1) on the surface of target cells. Once the CTL identifies its target cell, it delivers the “lethal hit” by one of two distinct mechanisms described below (Takayama et al., 1995). The initially characterized pathway is a Figure 1 calcium-dependent granule exocytosis pathway. In this pathway, effector cells secrete cytolytic proteins such as perforin and granzyme that penetrate target cells, initiate cytosolic and nuclear changes which culminate in apoptosis, thereby eliminating potentially harmful cells. The existence of an alternate pathway was proposed based on the observations that effector T cells from perforin knockout mice retained cytotoxic activity and were capable of inducing apoptosis in target cells.
    [Show full text]
  • CD70 Knockout: a Novel Approach to Augment CAR-T Cell Function
    #1537 CD70 knockout: A novel approach to augment CAR-T cell function Mary-Lee Dequeant, Jason Sagert, Demetri Kalaitzidis, Hui Yu, Ashley Porras, Brigid McEwan, Zinkal Padalia, Paul Tetteh, Thao Nguyen, Andrew Dunn, Sarah Spencer, Nicolette Lee, Henia Dar, Daniel Henderson, Sushant Karnik, Pooja Keerthipati, Jonathan A. Terrett CRISPR Therapeutics, 610 Main Street, Cambridge, MA, USA 02139 Abstract KO of certain checkpoint genes is detrimental to CAR-T function CD70 and its ligand, CD27, have been described as both activating and suppressing for different cell types including B and T cells. There has been speculation that the CD70/CD27 axis can act in a checkpoint or co-stimulatory manner in certain Figure 8: CD70 KO confers superior cytotoxic activity than PD1 KO against an RCC Figure 10: KO of multiple checkpoint genes impairs CAR-T cell function immuno-oncology settings. Knockout (KO) of CD70 function scored highly in a CRISPR/Cas9 screen of candidate genes for enhanced T cell activity. Deletion of other checkpoint candidates such as PD1, TIM3, LAG3 and TIGIT scored much lower than CD70, both alone and in combination with each other. T cells with CD70 KO showed resistance to exhaustion upon repeated stimulation in culture. CAR-T cells with CD70 KO similarly showed exhaustion resistance, as well as a reduction in tumor cell line overexpressing PD-L1 A Input Bone Marrow (Day 100) 10000 apoptosis, increased proliferation, and improved target cell lysis upon sequential rechallenges. CTX130 is an investigational allogeneic CAR-T therapy currently being studied in patients with CD70-expressing tumors, including clear cell renal cell 100 2500 No RNP or AAV carcinoma and B and T cell malignancies.
    [Show full text]