Nanosecond-Pulsed DBD Plasma-Generated Reactive Oxygen Species Trigger Immunogenic Cell Death in A549 Lung Carcinoma Cells Through Intracellular Oxidative Stress

Total Page:16

File Type:pdf, Size:1020Kb

Nanosecond-Pulsed DBD Plasma-Generated Reactive Oxygen Species Trigger Immunogenic Cell Death in A549 Lung Carcinoma Cells Through Intracellular Oxidative Stress International Journal of Molecular Sciences Article Nanosecond-Pulsed DBD Plasma-Generated Reactive Oxygen Species Trigger Immunogenic Cell Death in A549 Lung Carcinoma Cells through Intracellular Oxidative Stress Abraham Lin 1, Billy Truong 1, Sohil Patel 1, Nagendra Kaushik 2, Eun Ha Choi 2, Gregory Fridman 1, Alexander Fridman 1 and Vandana Miller 1,* 1 C. & J. Nyheim Plasma Institute, Drexel University, Philadelphia, PA 19104, USA; [email protected] (A.L.); [email protected] (B.T.); [email protected] (S.P.); [email protected] (G.F.); [email protected] (A.F.) 2 Plasma Bioscience Research Center, Kwangwoon University, Seoul 139791, Korea; [email protected] (N.K.); [email protected] (E.H.C.) * Correspondence: [email protected]; Tel.: +1-215-571-4074 Academic Editor: Hsueh-Wei Chang Received: 1 March 2017; Accepted: 28 April 2017; Published: 3 May 2017 Abstract: A novel application for non-thermal plasma is the induction of immunogenic cancer cell death for cancer immunotherapy. Cells undergoing immunogenic death emit danger signals which facilitate anti-tumor immune responses. Although pathways leading to immunogenic cell death are not fully understood; oxidative stress is considered to be part of the underlying mechanism. Here; we studied the interaction between dielectric barrier discharge plasma and cancer cells for oxidative stress-mediated immunogenic cell death. We assessed changes to the intracellular oxidative environment after plasma treatment and correlated it to emission of two danger signals: surface-exposed calreticulin and secreted adenosine triphosphate. Plasma-generated reactive oxygen and charged species were recognized as the major effectors of immunogenic cell death. Chemical attenuators of intracellular reactive oxygen species successfully abrogated oxidative stress following plasma treatment and modulated the emission of surface-exposed calreticulin. Secreted danger signals from cells undergoing immunogenic death enhanced the anti-tumor activity of macrophages. This study demonstrated that plasma triggers immunogenic cell death through oxidative stress pathways and highlights its potential development for cancer immunotherapy. Keywords: non-thermal plasma; cancer immunotherapy; immunogenic cell death; oxidative stress; calreticulin; adenosine triphosphate; plasma immunotherapy; nanosecond-pulsed dielectric barrier discharge 1. Introduction Methods to induce immunogenic cell death (ICD), where dying cells emit immuno-stimulatory signals, are being actively explored for cancer immunotherapy [1–5]. Cancerous cells undergoing ICD recruit and stimulate antigen presenting cells (APCs), key immune cells required for the initiation of an adaptive immune response [4–6]. This leads to the subsequent development and activation of tumor-specific effector T cells and memory T cells [7–9]. Thus, cancer is suppressed systemically and long-term protection against cancer recurrence is possible [8,9]. While mechanisms that promote ICD are under investigation, molecules characteristic of ICD known as damage-associated molecular patterns (DAMPs), have been identified [10,11]. These molecules are exposed on the cell surface or are secreted by cells undergoing autophagy, endoplasmic Int. J. Mol. Sci. 2017, 18, 966; doi:10.3390/ijms18050966 www.mdpi.com/journal/ijms Int.Int. J.J. Mol.Mol. Sci.Sci. 2017,, 18,, 966966 22 ofof 2424 reticulum (ER) stress, or oxidative stress from reactive oxygen species (ROS) [6,10–13]. DAMPs that reticulumare characteristically (ER) stress, or associated oxidative stresswith fromICD reactiveare: surface-exposed oxygen species calreticulin (ROS) [6,10 –(ecto-CRT),13]. DAMPs secreted that are characteristicallyadenosine triphosphate associated (ATP), with ICDreleased are: surface-exposedhigh mobility group calreticulin protein (ecto-CRT), B1 (HMGB1), secreted and adenosine surface- triphosphateexposed heat (ATP), shock released protein high90 (HSP90) mobility and group HSP70 protein [13–16]. B1 (HMGB1), Of these, andCRT surface-exposed and ATP are identified heat shock as proteinthe major 90 (HSP90)predictors and of HSP70 the [immuno-stimulatory13–16]. Of these, CRT capacity and ATP areof anti-cancer identified as therapeutics the major predictors [15–18]. ofRetrospective the immuno-stimulatory evaluation of capacityexperimental of anti-cancer and clinical therapeutics data has led [15 to– 18the]. identification Retrospective of evaluation several ICD of experimentalinducers, including and clinical some chemotherapeutics data has led to the identification(anthracyclines of and several oxaliplatin), ICD inducers, ionizing including radiation, some and chemotherapeuticsphotodynamic therapy (anthracyclines with hypericin and oxaliplatin),[1–3]. At the ionizing same time, radiation, new ICD and photodynamicinducers, such therapyas non- withthermal hypericin plasma, [1 are–3]. also At being the same investigated. time, new ICD inducers, such as non-thermal plasma, are also beingPlasma, investigated. known as the fourth state of matter, is an ionized gas composed of electrons, various ions,Plasma, electronically known excited as the fourthatoms stateand ofneutral matter, atoms is an and ionized molecules gas composed [19]. Two of electrons,major families various of ions,devices electronically for generating excited non-thermal atoms and neutralplasma atoms at at andmospheric molecules pressure [19]. Two for majorbiological families and of medical devices forapplications generating are non-thermal (Figure 1): plasma (1) plasma at atmospheric jets and pressure(2) direct for dielectric biological barrier and medical discharges applications (DBDs) are(employed (Figure1 in): (1)this plasma study) jets [20,21]. and (2)In directjets, most dielectric of the barrier plasma discharges is genera (DBDs)ted remotely, (employed and inplasma this study)products [20 are,21]. often In jets, delivered most of to the the plasma biological is generated target via remotely, a carrier andgas plasma[21]. Ionization products waves are often also deliveredproduce some to the plasma biological at targetthe treatment via a carrier surface gas [22] [21].. IonizationDBDs generate waves plasma also produce directly some at the plasma surface at thebeing treatment treated surface[20]. Due [22 ].to DBDs the electrode generate design, plasma directlyDBDs are at thetypically surface able being to treateda cover [ 20a ].much Due tolarger the electrodesurface area design, compared DBDs to are jets typically [23]. Furthermore, able to a cover the achemical much larger species surface deposited area compared on target surfaces, to jets [23 in]. Furthermore,particular, charged the chemical energetic species particles deposited and short-lived on target reactive surfaces, species, inparticular, are different charged between energetic the two particlessystems [20,21,24]. and short-lived reactive species, are different between the two systems [20,21,24]. FigureFigure 1.1. TwoTwo majormajor familiesfamilies ofof non-thermal,non-thermal, atmosphericatmospheric pressurepressure plasmaplasma devicesdevices forfor biomedicalbiomedical applicationsapplications areare jetsjets andand dielectricdielectric barrierbarrier dischargesdischarges (DBDs)(DBDs) ((AA)) JetsJets remotelyremotely generategenerate bulkbulk plasmaplasma andand plasmaplasma speciesspecies areare oftenoften transportedtransported toto thethe targettarget viavia aa carriercarrier gas;gas; ((BB)) DBDsDBDs plasmaplasma useuse thethe targettarget asas aa groundinggrounding electrodeelectrode toto generategenerate plasmaplasma directlydirectly ontoonto cells cells and and tissue. tissue. BesidesBesides thethe applied applied electric electric field field and plasmaand plasma temperature, temperature, a major determinanta major determinant of the composition of the ofcomposition plasma is theof plasma gas environment is the gas environment in which the in plasma whichis the generated plasma is [25 generated]. In the presence[25]. In the of presence oxygen, reactiveof oxygen, oxygen reactive species oxygen (ROS) species such (ROS) as atomic such oxygen as atomic (O), oxygen hydroxyl (O), radicals hydroxyl (•OH), radicals superoxide (•OH), superoxide− (O2•−), singlet1 oxygen (1O2), and hydrogen peroxide (H2O2) are produced [26–29]. When (O2• ), singlet oxygen ( O2), and hydrogen peroxide (H2O2) are produced [26–29]. When reactive nitrogenreactive nitrogen species (RNS)species are (RNS) desired are desired for wound for wound healing healing or other or specificother specific applications, applications, nitrogen nitrogen may bemay introduced be introduced into the into discharge the discharge environment environmen [30]. Majort [30]. measured Major measured species include species atomic include nitrogen atomic − (N),nitrogen peroxynitrite (N), peroxynitrite (ONOO− ),(ONOO and nitric), and oxide nitric (NO) oxide [31 (NO)]. Therefore, [31]. Theref it isore, plausible it is plausi to optimizeble to optimize plasma treatmentplasma treatment parameters parameters to produce to produce various chemicalvarious ch effectorsemical effectors for desired for biologicaldesired biological outcomes. outcomes. TheThe highlyhighly reactivereactive speciesspecies areare knownknown toto influenceinfluence biologicalbiological processesprocesses andand theirtheir rolerole inin cancercancer cellcell killingkilling hashas beenbeen extensivelyextensively researchedresearched [[32,33].32,33]. InIn fact,fact, chemotherapieschemotherapies andand radiationradiation therapytherapy oftenoften relyrely
Recommended publications
  • Killing Cancer Cells, Twice with One Shot
    Cell Death and Differentiation (2014) 21, 1–2 & 2014 Macmillan Publishers Limited All rights reserved 1350-9047/14 www.nature.com/cdd Editorial Killing cancer cells, twice with one shot ME Bianchi*,1 Cell Death and Differentiation (2014) 21, 1–2; doi:10.1038/cdd.2013.147 I can still remember lecturing that chemotherapy and radio- shows that transglutaminase (TG) 2, a protein with several therapy are effective because they kill cancer cells. That was complex activities, can prevent calreticulin exposure on the simple enough, and when I wanted to make it a bit more cell surface, and therefore TG2-overexpressing cancer cells complex, I ventured into explaining how they mainly killed can escape activation of the immune system.9 cancer cells because they divided much more than other cells, Curiously, calreticulin is also surface exposed by spermato- and how the killing involved apoptosis. Things have moved on zoa, both in mammals and nematodes, to facilitate fertiliza- quite a bit since then. Evidence accumulated in the past few tion. Following the phylogenetic trail to its widest extent, years indicates that some anti-cancer drugs do not kill cancer Sukkurwala et al.10 show that yeast calreticulin, CNE1 protein, cells, but merely push them into senescence, some do kill is surface exposed following ER stress, and facilitates yeast cells but not via apoptosis, and some do kill cells via apoptosis mating too.10 The binding of yeast pheromone a-factor to its G but that is not the main ingredient in their efficacy. protein-coupled receptor (GPCR) induces CNE1 surface The main theme of this issue is that dying cancer cells presentation.
    [Show full text]
  • Stress-Induced Inflammation Evoked by Immunogenic Cell Death Is
    www.nature.com/cdd ARTICLE OPEN Stress-induced inflammation evoked by immunogenic cell death is blunted by the IRE1α kinase inhibitor KIRA6 through HSP60 targeting Nicole Rufo1,2, Dimitris Korovesis3,Sofie Van Eygen1,2, Rita Derua4, Abhishek D. Garg 1, Francesca Finotello5, Monica Vara-Perez1,2, ž 6,7 2,8 9 7,10 11 6 Jan Ro anc , Michael Dewaele , Peter A. de Witte , Leonidas G. Alexopoulos✉ , Sophie Janssens , Lasse Sinkkonen , Thomas Sauter6, Steven H. L. Verhelst 3,12 and Patrizia Agostinis 1,2 © The Author(s) 2021 Mounting evidence indicates that immunogenic therapies engaging the unfolded protein response (UPR) following endoplasmic reticulum (ER) stress favor proficient cancer cell-immune interactions, by stimulating the release of immunomodulatory/ proinflammatory factors by stressed or dying cancer cells. UPR-driven transcription of proinflammatory cytokines/chemokines exert beneficial or detrimental effects on tumor growth and antitumor immunity, but the cell-autonomous machinery governing the cancer cell inflammatory output in response to immunogenic therapies remains poorly defined. Here, we profiled the transcriptome of cancer cells responding to immunogenic or weakly immunogenic treatments. Bioinformatics-driven pathway analysis indicated that immunogenic treatments instigated a NF-κB/AP-1-inflammatory stress response, which dissociated from both cell death and UPR. This stress-induced inflammation was specifically abolished by the IRE1α-kinase inhibitor KIRA6. Supernatants from immunogenic chemotherapy and KIRA6 co-treated cancer cells were deprived of proinflammatory/chemoattractant factors and failed to mobilize neutrophils and induce dendritic cell maturation. Furthermore, KIRA6 significantly reduced the in vivo vaccination potential of dying cancer cells responding to immunogenic chemotherapy. Mechanistically, we found that the anti-inflammatory effect of KIRA6 was still effective in IRE1α-deficient cells, indicating a hitherto unknown off-target effector of this IRE1α-kinase inhibitor.
    [Show full text]
  • Pathogen Response-Like Recruitment and Activation of Neutrophils by Sterile Immunogenic Dying Cells Drives Neutrophil-Mediated Residual Cell Killing
    Cell Death and Differentiation (2017) 24, 832–843 & 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved 1350-9047/17 www.nature.com/cdd Pathogen response-like recruitment and activation of neutrophils by sterile immunogenic dying cells drives neutrophil-mediated residual cell killing Abhishek D Garg*,1,2, Lien Vandenberk3, Shentong Fang4, Tekele Fasche4, Sofie Van Eygen1, Jan Maes5, Matthias Van Woensel6,7, Carolien Koks3, Niels Vanthillo5, Norbert Graf8, Peter de Witte5, Stefaan Van Gool9, Petri Salven4 and Patrizia Agostinis*,1 Innate immune sensing of dying cells is modulated by several signals. Inflammatory chemokines-guided early recruitment, and pathogen-associated molecular patterns-triggered activation, of major anti-pathogenic innate immune cells like neutrophils distinguishes pathogen-infected stressed/dying cells from sterile dying cells. However, whether certain sterile dying cells stimulate innate immunity by partially mimicking pathogen response-like recruitment/activation of neutrophils remains poorly understood. We reveal that sterile immunogenic dying cancer cells trigger (a cell autonomous) pathogen response-like chemokine (PARC) signature, hallmarked by co-release of CXCL1, CCL2 and CXCL10 (similar to cells infected with bacteria or viruses). This PARC signature recruits preferentially neutrophils as first innate immune responders in vivo (in a cross-species, evolutionarily conserved manner; in mice and zebrafish). Furthermore, key danger signals emanating from these dying cells, that is, surface calreticulin, ATP and nucleic acids stimulate phagocytosis, purinergic receptors and toll-like receptors (TLR) i.e. TLR7/8/9-MyD88 signaling on neutrophil level, respectively. Engagement of purinergic receptors and TLR7/8/9-MyD88 signaling evokes neutrophil activation, which culminates into H2O2 and NO-driven respiratory burst-mediated killing of viable residual cancer cells.
    [Show full text]
  • Α-TEA's Tumor Toxicity May Be Attributed to Its Capability of Inducing Oxidative Damage in the Endoplasmic Reticulum
    Portland State University PDXScholar University Honors Theses University Honors College 2015 α-TEA's Tumor Toxicity may be Attributed to its Capability of Inducing Oxidative Damage in the Endoplasmic Reticulum Diego Mathias Barragan Echenique Portland State University Follow this and additional works at: https://pdxscholar.library.pdx.edu/honorstheses Let us know how access to this document benefits ou.y Recommended Citation Barragan Echenique, Diego Mathias, "α-TEA's Tumor Toxicity may be Attributed to its Capability of Inducing Oxidative Damage in the Endoplasmic Reticulum" (2015). University Honors Theses. Paper 124. https://doi.org/10.15760/honors.172 This Thesis is brought to you for free and open access. It has been accepted for inclusion in University Honors Theses by an authorized administrator of PDXScholar. Please contact us if we can make this document more accessible: [email protected]. α-TEA’s Tumor Toxicity may be Attributed to its Capability of Inducing Oxidative Damage in the Endoplasmic Reticulum by Diego Mathias Barragan Echenique An undergraduate honors thesis submitted in partial fulfillment of the requirements for the degree of Bachelor of Science in University Honors and Biology Thesis Adviser William L. Redmond, PhD Portland State University 2015 Abstract α-tocopherol ether linked acetic acid (α-TEA) is a Vitamin E derivative with antineoplastic and anti-metastatic properties, tumor specificity, and immunogenic characteristics. The compound is structurally similar to vitamin E, however a key difference is that it doesn’t retain any antioxidant properties. Interestingly, orally administered α-TEA appears to stimulate anti-tumor immunity, which, along with its anti-metastatic and antineoplastic properties, makes the molecule’s mechanism of action worth investigating.
    [Show full text]
  • Reactive Oxygen Species and Antitumor Immunity—From Surveillance to Evasion
    cancers Review Reactive Oxygen Species and Antitumor Immunity—From Surveillance to Evasion Andromachi Kotsafti 1 , Marco Scarpa 2 , Ignazio Castagliuolo 3 and Melania Scarpa 1,* 1 Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; [email protected] 2 General Surgery Unit, Azienda Ospedaliera di Padova, 35128 Padua, Italy; [email protected] 3 Department of Molecular Medicine DMM, University of Padua, 35121 Padua, Italy; [email protected] * Correspondence: [email protected] Received: 8 June 2020; Accepted: 28 June 2020; Published: 1 July 2020 Abstract: The immune system is a crucial regulator of tumor biology with the capacity to support or inhibit cancer development, growth, invasion and metastasis. Emerging evidence show that reactive oxygen species (ROS) are not only mediators of oxidative stress but also players of immune regulation in tumor development. This review intends to discuss the mechanism by which ROS can affect the anti-tumor immune response, with particular emphasis on their role on cancer antigenicity, immunogenicity and shaping of the tumor immune microenvironment. Given the complex role that ROS play in the dynamics of cancer-immune cell interaction, further investigation is needed for the development of effective strategies combining ROS manipulation and immunotherapies for cancer treatment. Keywords: reactive oxygen species; oxidative stress; immunity; inflammation; cancer 1. Introduction Reactive oxygen species (ROS) are defined as chemically reactive derivatives of oxygen that elicits both harmful and beneficial effects in cells depending on their concentration. Oxidative stress occurs when ROS production overcomes the scavenging potential of cells or when the antioxidant response is severely impaired; as a consequence nonradical and free radical ROS such as hydrogen peroxide (H2O2), the superoxide radical (O2•) or the hydroxyl radical (OH•) accumulate [1].
    [Show full text]
  • Immunogenic Cell Death by the Novel Topoisomerase I Inhibitor TLC388 Enhances the Therapeutic Efficacy of Radiotherapy
    cancers Article Immunogenic Cell Death by the Novel Topoisomerase I Inhibitor TLC388 Enhances the Therapeutic Efficacy of Radiotherapy Kevin Chih-Yang Huang 1,2 , Shu-Fen Chiang 3,4, Pei-Chen Yang 4, Tao-Wei Ke 5,6, Tsung-Wei Chen 7,8, Ching-Han Hu 4, Yi-Wen Huang 4, Hsin-Yu Chang 4, William Tzu-Liang Chen 5,9,10,* and K. S. Clifford Chao 4,8,11,* 1 Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung 40402, Taiwan; [email protected] 2 Translation Research Core, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan 3 Lab of Precision Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Taichung 42055, Taiwan; [email protected] 4 Cancer Center, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan; [email protected] (P.-C.Y.); [email protected] (C.-H.H.); [email protected] (Y.-W.H.); [email protected] (H.-Y.C.) 5 Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan; [email protected] 6 School of Chinese Medicine & Graduate Institute of Chinese Medicine, China Medical University, Taichung 40402, Taiwan Citation: Huang, K.C.-Y.; 7 Department of Pathology, Asia University Hospital, Asia University, Taichung 41354, Taiwan; Chiang, S.-F.; Yang, P.-C.; Ke, T.-W.; [email protected] Chen, T.-W.; Hu, C.-H.; Huang, Y.-W.; 8 Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan Chang, H.-Y.; Chen, W.T.-L.; 9 Department of Surgery, School of Medicine, China Medical University, Taichung 40402, Taiwan Chao, K.S.C.
    [Show full text]
  • The Role of Immunogenic Cell Death
    cancers Review Current Approaches for Combination Therapy of Cancer: The Role of Immunogenic Cell Death 1, 2, 1 3 4 Zahra Asadzadeh y, Elham Safarzadeh y, Sahar Safaei , Ali Baradaran , Ali Mohammadi , Khalil Hajiasgharzadeh 1 , Afshin Derakhshani 1 , Antonella Argentiero 5, Nicola Silvestris 5,6,* and Behzad Baradaran 1,7,* 1 Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; [email protected] (Z.A.); [email protected] (S.S.); [email protected] (K.H.); [email protected] (A.D.) 2 Department of Immunology and Microbiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil 5618985991, Iran; [email protected] 3 Research & Development Lab, BSD Robotics, 4500 Brisbane, Australia; [email protected] 4 Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark; [email protected] 5 IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; [email protected] 6 Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy 7 Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran * Correspondence: [email protected] (N.S.); [email protected] (B.B.); Tel.: +98-413-337-1440 (B.B.) The first two authors contributed equally to this work. y Received: 12 March 2020; Accepted: 17 April 2020; Published: 23 April 2020 Abstract: Cell death resistance is a key feature of tumor cells. One of the main anticancer therapies is increasing the susceptibility of cells to death. Cancer cells have developed a capability of tumor immune escape.
    [Show full text]
  • RIG-I-Like Helicases Induce Immunogenic Cell Death of Pancreatic Cancer Cells and Sensitize Tumors Toward Killing by CD8 Þ T Cells
    Cell Death and Differentiation (2014) 21, 1825–1837 & 2014 Macmillan Publishers Limited All rights reserved 1350-9047/14 www.nature.com/cdd RIG-I-like helicases induce immunogenic cell death of pancreatic cancer cells and sensitize tumors toward killing by CD8 þ T cells This article has been corrected since Online Publication and a corrigendum appears in this issue P Duewell1,2, A Steger1, H Lohr1, H Bourhis1, H Hoelz1, SV Kirchleitner1, MR Stieg1, S Grassmann1, S Kobold1, JT Siveke3, S Endres1 and M Schnurr*,1,2 Pancreatic cancer is characterized by a microenvironment suppressing immune responses. RIG-I-like helicases (RLH) are immunoreceptors for viral RNA that induce an antiviral response program via the production of type I interferons (IFN) and apoptosis in susceptible cells. We recently identified RLH as therapeutic targets of pancreatic cancer for counteracting immunosuppressive mechanisms and apoptosis induction. Here, we investigated immunogenic consequences of RLH-induced tumor cell death. Treatment of murine pancreatic cancer cell lines with RLH ligands induced production of type I IFN and proinflammatory cytokines. In addition, tumor cells died via intrinsic apoptosis and displayed features of immunogenic cell death, such as release of HMGB1 and translocation of calreticulin to the outer cell membrane. RLH-activated tumor cells led to activation of dendritic cells (DCs), which was mediated by tumor-derived type I IFN, whereas TLR, RAGE or inflammasome signaling was dispensable. Importantly, CD8a þ DCs effectively engulfed apoptotic tumor material and cross-presented tumor- associated antigen to naive CD8 þ T cells. In comparison, tumor cell death mediated by oxaliplatin, staurosporine or mechanical disruption failed to induce DC activation and antigen presentation.
    [Show full text]
  • Profiling the Immunogenic Cell Death (ICD)
    Profiling the immunogenic cell death (ICD) mechanisms induced by Nano-Pulse Stimulation (NPS) treatment in mouse B16-F10 melanoma tumors using NanoString technology Amanda McDaniel, Snjezana Anand, Aman Alzubier, Juliette Berlin, Holly Hartman, Darrin Uecker and Richard Nuccitelli Pulse Biosciences, 3957 Point Eden Way, Hayward, CA 94545 Background Results Nano-Pulse Stimulation (NPS) is a non-thermal tumor therapy NPS Treatment Tumor cell releasing DAMPs binding to DC phagocytosing Mature T-cell Figures 1a-b. receptors on an immature DC tumor cell DC that delivers ultrashort electrical pulses (100-600ns) to tumor a. CD8+ cells. NPS opens nanopores in the membrane of the ER, NPS-induced T-cell immune 2+ allowing the efflux of Ca into the cytoplasm, causing ER response stress and the production of ROS. These effects induce an CD4+ immunogenic cell death (ICD)1 that both eliminates a primary T-cell tumor and inhibits the growth of a secondary re-challenge DAMPs Release – PRR Binding Adaptive Immune Response (AIR) Priming T cell Activation tumor in preclinical models2. To date, the primary mechanism b. Cell Death of action of most known ICD inducers is ER stress and ROS PERK Ire1 IP3R Casp12 CD86 Calr Ifng Lrp1 CD28 Il2 Il1b production leading to intrinsic mitochondrial apoptosis, and the Traf2 Ifna1 Casp8 Casp3 Ifnb1 eIF2α P2rx7 Nlrp3 release and translocation of damage associated molecular ASK1 CD80 Ifngr1 Casp1 Il1r1 4-6 Cd4 patterns (DAMPs) that bind to pattern recognition receptors Il6 Il6ra Atf4 JNK Casp7 Foxp3 Tlr4 Ifnar1 (PRRs) to prime the adaptive immune response. Here we Hmgb1 Tnf Il12a Il12rb1 Casp9 Ly96 Cd8a sought to profile the pathways involved in ER stress, apoptotic CHOP CytC Apaf1 Myd88 Il12b Il12rb2 Cd8b1 cell death and the immune response after NPS treatment, Figures 2a-d.
    [Show full text]
  • Insights Into Immunogenic Cell Death in Onco-Therapies
    cancers Review Restoring the Immunity in the Tumor Microenvironment: Insights into Immunogenic Cell Death in Onco-Therapies Ángela-Patricia Hernández 1 , Pablo Juanes-Velasco 1 , Alicia Landeira-Viñuela 1, Halin Bareke 1,2, Enrique Montalvillo 1, Rafael Góngora 1 and Manuel Fuentes 1,3,* 1 Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; [email protected] (Á.-P.H.); [email protected] (P.J.-V.); [email protected] (A.L.-V.); [email protected] (H.B.); [email protected] (E.M.); [email protected] (R.G.) 2 Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Institute of Health Sciences, Marmara University, 34722 Istanbul, Turkey 3 Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain * Correspondence: [email protected]; Tel.: +34-923-294-811 Simple Summary: Since the role of immune evasion was included as a hallmark in cancer, the idea of cancer as a single cell mass that replicate unlimitedly in isolation was dissolved. In this sense, cancer and tumorigenesis cannot be understood without taking into account the tumor microenvironment (TME) that plays a crucial role in drug resistance. Immune characteristics of TME can determine the success in treatment at the same time that antitumor therapies can reshape the immunity in TME. Here, we collect a variety of onco-therapies that have been demonstrated to induce an interesting Citation: Hernández, Á.-P.; immune response accompanying its pharmacological action that is named as “immunogenic cell Juanes-Velasco, P.; Landeira-Viñuela, death”. As this report shows, immunogenic cell death has been gaining importance in antitumor A.; Bareke, H.; Montalvillo, E.; therapy and should be studied in depth as well as taking into account other applications that may Góngora, R.; Fuentes, M.
    [Show full text]
  • Immunogenic Tumor Cell Death for Optimal Anticancer Therapy: the Calreticulin Exposure Pathway
    Published OnlineFirst April 26, 2010; DOI: 10.1158/1078-0432.CCR-09-2891 Published OnlineFirst on May 25, 2010 as 10.1158/1078-0432.CCR-09-2891 Molecular Pathways Clinical Cancer Research Immunogenic Tumor Cell Death for Optimal Anticancer Therapy: The Calreticulin Exposure Pathway Laurence Zitvogel1,2,3, Oliver Kepp2,3,4, Laura Senovilla2,3,4, Laurie Menger2,3,4, Nathalie Chaput1,2,3, and Guido Kroemer2,3,4 Abstract In response to some chemotherapeutic agents such as anthracyclines and oxaliplatin, cancer cells un- dergo immunogenic apoptosis, meaning that their corpses are engulfed by dendritic cells and that tu- mor cell antigens are presented to tumor-specific CD8+ T cells, which then control residual tumor cells. One of the peculiarities of immunogenic apoptosis is the early cell surface exposure of calreticulin (CRT), a protein that usually resides in the lumen of the endoplasmic reticulum (ER). When elicited by anthracyclines or oxaliplatin, the CRT exposure pathway is activated by pre-apoptotic ER stress and the phosphorylation of the eukaryotic translation initiation factor eIF2α by the kinase PERK, followed by caspase-8-mediated proteolysis of the ER-sessile protein BAP31, activation of the pro-apoptotic pro- teins Bax and Bak, anterograde transport of CRT from the ER to the Golgi apparatus and exocytosis of CRT-containing vesicles, finally resulting in CRT translocation onto the plasma membrane surface. In- terruption of this complex pathway abolishes CRT exposure, annihilates the immunogenicity of apo- ptosis, and reduces the immune response elicited by anticancer chemotherapies. We speculate that human cancers that are incapable of activating the CRT exposure pathway are refractory to the immune- mediated component of anticancer therapies.
    [Show full text]
  • Calreticulin in Phagocytosis and Cancer: Opposite Roles in Immune Response Outcomes
    Apoptosis (2019) 24:245–255 https://doi.org/10.1007/s10495-019-01532-0 REVIEW Calreticulin in phagocytosis and cancer: opposite roles in immune response outcomes Alejandro Schcolnik‑Cabrera1 · Bernardo Oldak3 · Mandy Juárez1 · Mayra Cruz‑Rivera2 · Ana Flisser2 · Fela Mendlovic2,3 Published online: 30 March 2019 © Springer Science+Business Media, LLC, part of Springer Nature 2019 Abstract Calreticulin (CRT) is a pleiotropic and highly conserved molecule that is mainly localized in the endoplasmic reticulum. Recently, CRT has gained special interest for its functions outside the endoplasmic reticulum where it has immunomodula- tory properties. CRT translocation to the cell membrane serves as an “eat me” signal and promotes efferocytosis of apoptotic cells and cancer cell removal with completely opposite outcomes. Efferocytosis results in a silenced immune response and homeostasis, while removal of dying cancer cells brought about by anthracycline treatment, ionizing-irradiation or photo- dynamic therapy results in immunogenic cell death with activation of the innate and adaptive immune responses. In addi- tion, CRT impacts phagocyte activation and cytokine production. The effects of CRT on cytokine production depend on its conformation, species specificity, degree of oligomerization and/or glycosylation, as well as its cellular localization and the molecular partners involved. The controversial roles of CRT in cancer progression and the possible role of the CALR gene mutations in myeloproliferative neoplasms are also addressed. The release of CRT and its influence on the different cells involved during efferocytosis and immunogenic cell death points to additional roles of CRT besides merely acting as an “eat me” signal during apoptosis. Understanding the contribution of CRT in physiological and pathological processes could give us some insight into the potential of CRT as a therapeutic target.
    [Show full text]