1
|
Deng J, Yu Y, Yin Y, Hu G, Yang X, Chen C, Guo C, Tang Z. Revealing the effects of various immune cells in anorexia nervosa: Evidence from Mendelian randomization. Medicine (Baltimore) 2025; 104:e41817. [PMID: 40128032 PMCID: PMC11936630 DOI: 10.1097/md.0000000000041817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
The aim of this study was to assess the causal relationship between immune cells and anorexia nervosa (AN) by Mendelian randomization (MR). Data on immune cell phenotypes and AN were obtained from genome-wide association studies. Next, single nucleotide polymorphisms included in the MR analysis were screened according the basic assumptions. Furthermore, inverse variance weighted was used as the main method for MR analysis to evaluate the causal effect of immune cell phenotypes on AN. Finally, MR-Egger intercept, Cochran Q, and leave-one-out sensitivity analyses were used to assess horizontal pleiotropy, heterogeneity, and robustness, respectively. The MR analysis showed that NKT %lymphocyte (OR 1.201, 95% CI = 1.021-1.411, P = .027), NKT %T cell (OR 1.258, 95%CI 1.043-1.519, P = .017), double negative (DN) (CD4-CD8-) NKT %lymphocyte (OR 1.235, 95%CI 1.074-1.420, P = .003), DN (CD4-CD8-) NKT %T cell (OR 1.222, 95%CI 1.060-1.410, P = .006), CD8dim NKT absolute count (OR 1.225, 95%CI 1.045-1.436, P = .012), CD8dim NKT %lymphocyte (OR 1.214, 95%CI 1.043-1.414, P = .012), CD8dim NKT %T cell (OR 1.215, 95%CI 1.035-1.425, P = .017), CD16-CD56 on NKT (OR 1.193, 95%CI 1.014-1.402, P = .033), CD28 + CD45RA + CD8br %T cell (OR 1.020, 95%CI 1.002-1.037, P = .025) were associated with increased genetic susceptibility to AN. MR-Egger showed no horizontal pleiotropy (P ≥ .05). Cochran Q and sensitivity analysis showed that the results were not heterogeneous and were robust. This MR analysis revealed 9 immune cell phenotypes related to increased genetic susceptibility to AN, emphasizing the importance of NKT and CD8 in AN. This finding provides new insights for understanding the pathogenesis of AN and developing immune-targeted drugs.
Collapse
Affiliation(s)
- Juan Deng
- Department of Anesthesiology, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Yunfeng Yu
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuman Yin
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gang Hu
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinyu Yang
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Cong Chen
- School of Basic Medicine, Guizhou University of Traditional, Chinese Medicine, Guiyang, Guizhou, China
| | - Chenlu Guo
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhengguo Tang
- Department of Anesthesiology, The Third Hospital of Changsha, Changsha, Hunan, China
| |
Collapse
|
2
|
Cyndari KI, Scorza BM, Zacharias ZR, Strand L, Mahachi K, Oviedo JM, Gibbs L, Pessoa-Pereira D, Ausdal G, Hendricks D, Yahashiri R, Elkins JM, Gulbrandsen T, Peterson AR, Willey MC, Fairfax KC, Petersen CA. Resident Synovial Macrophages in Synovial Fluid: Implications for Immunoregulation in Infectious and Inflammatory Arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.29.560183. [PMID: 37873090 PMCID: PMC10592878 DOI: 10.1101/2023.09.29.560183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Objectives Resident synovial macrophages (RSM) provide immune sequestration of the joint space and are likely involved in initiation and perpetuation of the joint-specific immune response. We sought to identify RSM in synovial fluid (SF) and demonstrate migratory ability, in additional to functional changes that may perpetuate a chronic inflammatory response within joint spaces. Methods We recruited human patients presenting with undifferentiated arthritis in multiple clinical settings. We used flow cytometry to identify mononuclear cells in peripheral blood and SF. We used a novel transwell migration assay with human ex-vivo synovium obtained intra-operatively to validate flow cytometry findings. We used single cell RNA-sequencing (scRNA-seq) to further identify macrophage/monocyte subsets. ELISA was used to evaluate the bone-resorption potential of SF. Results We were able to identify a rare population of CD14dim, OPG+, ZO-1+ cells consistent with RSM in SF via flow cytometry. These cells were relatively enriched in the SF during infectious processes, but absolutely decreased compared to healthy controls. Similar putative RSM were identified using ex vivo migration assays when MCP-1 and LPS were used as migratory stimulus. scRNA-seq revealed a population consistent with RSM transcriptionally related to CD56+ cytotoxic dendritic cells and IDO+ M2 macrophages. Conclusion We identified a rare cell population consistent with RSM, indicating these cells are likely migratory and able to initiate or coordinate both acute (septic) or chronic (autoimmune or inflammatory) arthritis. RSM analysis via scRNA-seq indicated these cells are M2 skewed, capable of antigen presentation, and have consistent functions in both septic and inflammatory arthritis.
Collapse
Affiliation(s)
- Karen I Cyndari
- Department of Emergency Medicine, University of Iowa, Iowa City, IA
- Center for Emerging Infectious Diseases
| | - Breanna M Scorza
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| | - Zeb R Zacharias
- Human Immunology Core, University of Iowa, Iowa City, IA
- Holden Comprehensive Cancer Center, Iowa City, IA
| | | | - Kurayi Mahachi
- Research and Analytics, Enterprise Analytics, Sentara Health
| | | | - Lisa Gibbs
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Danielle Pessoa-Pereira
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| | - Graham Ausdal
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| | - Dylan Hendricks
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| | | | - Jacob M Elkins
- Department of Orthopedics, University of Iowa, Iowa City, IA
| | | | | | | | - Keke C Fairfax
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Christine A Petersen
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| |
Collapse
|
3
|
van Halteren AGS, Suwandi JS, Tuit S, Borst J, Laban S, Tsonaka R, Struijk A, Wiekmeijer AS, van Pel M, Roep BO, Zwaginga JJ, Lankester AC, Schepers K, van Tol MJD, Fibbe WE. A unique immune signature in blood separates therapy-refractory from therapy-responsive acute graft-versus-host disease. Blood 2023; 141:1277-1292. [PMID: 36044666 PMCID: PMC10651784 DOI: 10.1182/blood.2022015734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) is an immune cell‒driven, potentially lethal complication of allogeneic hematopoietic stem cell transplantation affecting diverse organs, including the skin, liver, and gastrointestinal (GI) tract. We applied mass cytometry (CyTOF) to dissect circulating myeloid and lymphoid cells in children with severe (grade III-IV) aGVHD treated with immune suppressive drugs alone (first-line therapy) or in combination with mesenchymal stromal cells (MSCs; second-line therapy). These results were compared with CyTOF data generated in children who underwent transplantation with no aGVHD or age-matched healthy control participants. Onset of aGVHD was associated with the appearance of CD11b+CD163+ myeloid cells in the blood and accumulation in the skin and GI tract. Distinct T-cell populations, including TCRγδ+ cells, expressing activation markers and chemokine receptors guiding homing to the skin and GI tract were found in the same blood samples. CXCR3+ T cells released inflammation-promoting factors after overnight stimulation. These results indicate that lymphoid and myeloid compartments are triggered at aGVHD onset. Immunoglobulin M (IgM) presumably class switched, plasmablasts, and 2 distinct CD11b- dendritic cell subsets were other prominent immune populations found early during the course of aGVHD in patients refractory to both first- and second-line (MSC-based) therapy. In these nonresponding patients, effector and regulatory T cells with skin- or gut-homing receptors also remained proportionally high over time, whereas their frequencies declined in therapy responders. Our results underscore the additive value of high-dimensional immune cell profiling for clinical response evaluation, which may assist timely decision-making in the management of severe aGVHD.
Collapse
Affiliation(s)
- Astrid G. S. van Halteren
- Department of Pediatrics, Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jessica S. Suwandi
- Department of Internal Medicine and Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sander Tuit
- Department of Internal Medicine and Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jelske Borst
- Department of Internal Medicine and Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sandra Laban
- Department of Internal Medicine and Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Roula Tsonaka
- Department of Biomedical Data Sciences, Medical Statistics Section, Leiden University Medical Center, Leiden, The Netherlands
| | - Ada Struijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Melissa van Pel
- Department of Internal Medicine and Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bart O. Roep
- Department of Internal Medicine and Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Diabetes Immunology, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA
| | - Jaap Jan Zwaginga
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arjan C. Lankester
- Pediatric Stem Cell Transplantation Unit, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Koen Schepers
- Department of Internal Medicine and Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maarten J. D. van Tol
- Department of Pediatrics, Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Willem E. Fibbe
- Department of Internal Medicine and Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
4
|
Picard LK, Claus M, Fasbender F, Watzl C. Human NK cells responses are enhanced by CD56 engagement. Eur J Immunol 2022; 52:1441-1451. [PMID: 35775327 DOI: 10.1002/eji.202249868] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/03/2022] [Accepted: 06/29/2022] [Indexed: 11/09/2022]
Abstract
Natural Killer (NK) cells are important innate lymphocytes for effective immune responses against intracellular pathogens and tumors. CD56 is a well-known marker for human NK cells, but there is very limited information about a functional role of this surface receptor. Here we show that engagement of CD56 can induce NK cell activation resulting in degranulation, IFN-γ secretion and morphological changes, making CD56 a potential co-activating receptor in NK cells. Interestingly, this effect was only observed in cytokine pre-activated and not in freshly isolated human NK cells, demonstrating that NK cell reactivity upon CD56 engagement was dependent on cytokine stimulation. Inhibition of Syk, PI3K, Erk and src-family-kinases impaired CD56-mediated NK cell stimulation. Finally, we used CRISPR/Cas9 to delete CD56 from primary human NK cells. While this abolished the stimulatory effect of CD56 on pre-activated NK cells, the cytotoxic activity of NK cells against several tumor target cells was not affected by the absence of CD56. This demonstrates that the stimulating effect of CD56 on pre-activated NK cells does not have a major impact on their cytotoxic activity, but it may contribute to the function of CD56 as a fungal recognition receptor and in the NK cell developmental synapse. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Lea Katharina Picard
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| | - Maren Claus
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| | - Frank Fasbender
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| | - Carsten Watzl
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| |
Collapse
|
5
|
Versteven M, Flumens D, Campillo-Davó D, De Reu H, Van Bruggen L, Peeters S, Van Tendeloo V, Berneman Z, Dolstra H, Anguille S, Hobo W, Smits E, Lion E. Anti-Tumor Potency of Short-Term Interleukin-15 Dendritic Cells Is Potentiated by In Situ Silencing of Programmed-Death Ligands. Front Immunol 2022; 13:734256. [PMID: 35250967 PMCID: PMC8891487 DOI: 10.3389/fimmu.2022.734256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/27/2022] [Indexed: 11/24/2022] Open
Abstract
Dendritic cell (DC) vaccines have proven to be a valuable tool in cancer immune therapy. With several DC vaccines being currently tested in clinical trials, knowledge about their therapeutic value has been significantly increased in the past decade. Despite their established safety, it has become clear that objective clinical responses are not yet robust enough, requiring further optimization. Improvements of this advanced therapy medicinal product encompass, among others, regulating their immune stimulating capacity by in situ gene engineering, in addition to their implementation in combination therapy regimens. Previously, we have reported on a superior monocyte-derived DC preparation, including interleukin-15, pro-inflammatory cytokines and immunological danger signals in the culture process. These so-called IL-15 DCs have already proven to exhibit several favorable properties as cancer vaccine. Evolving research into mechanisms that could further modulate the immune response towards cancer, points to programmed death-1 as an important player that dampens anti-tumor immunity. Aiming at leveraging the immunogenicity of DC vaccines, we hypothesized that additional implementation of the inhibitory immune checkpoint molecules programmed death-ligand (PD-L)1 and PD-L2 in IL-15 DC vaccines would exhibit superior stimulatory potential. In this paper, we successfully implemented PD-L silencing at the monocyte stage in the 3-day IL-15 DC culture protocol resulting in substantial downregulation of both PD-L1 and PD-L2 to levels below 30%. Additionally, we validated that these DCs retain their specific characteristics, both at the level of phenotype and interferon gamma secretion. Evaluating their functional characteristics, we demonstrate that PD-L silencing does not affect the capacity to induce allogeneic proliferation. Ultimately designed to induce a durable tumor antigen-specific immune response, PD-L silenced IL-15 DCs were capable of surpassing PD-1-mediated inhibition by antigen-specific T cells. Further corroborating the superior potency of short-term IL-15 DCs, the combination of immune stimulatory components during DC differentiation and maturation with in situ checkpoint inhibition supports further clinical translation.
Collapse
Affiliation(s)
- Maarten Versteven
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Donovan Flumens
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Diana Campillo-Davó
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Laura Van Bruggen
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Stefanie Peeters
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Division of Hematology, Antwerp University Hospital, Edegem, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Harry Dolstra
- Department of Laboratory Medicine – Laboratory of Hematology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Division of Hematology, Antwerp University Hospital, Edegem, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Willemijn Hobo
- Department of Laboratory Medicine – Laboratory of Hematology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Evelien Smits
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
- *Correspondence: Eva Lion,
| |
Collapse
|
6
|
Dutt TS, LaVergne SM, Webb TL, Baxter BA, Stromberg S, McFann K, Berry K, Tipton M, Alnachoukati O, Zier L, Ebel G, Dunn J, Henao-Tamayo M, Ryan EP. Comprehensive Immune Profiling Reveals CD56 + Monocytes and CD31 + Endothelial Cells Are Increased in Severe COVID-19 Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:685-696. [PMID: 34987111 DOI: 10.4049/jimmunol.2100830] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/15/2021] [Indexed: 01/08/2023]
Abstract
Immune response dysregulation plays a key role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogenesis. In this study, we evaluated immune and endothelial blood cell profiles of patients with coronavirus disease 2019 (COVID-19) to determine critical differences between those with mild, moderate, or severe COVID-19 using spectral flow cytometry. We examined a suite of immune phenotypes, including monocytes, T cells, NK cells, B cells, endothelial cells, and neutrophils, alongside surface and intracellular markers of activation. Our results showed progressive lymphopenia and depletion of T cell subsets (CD3+, CD4+, and CD8+) in patients with severe disease and a significant increase in the CD56+CD14+Ki67+IFN-γ+ monocyte population in patients with moderate and severe COVID-19 that has not been previously described. Enhanced circulating endothelial cells (CD45-CD31+CD34+CD146+), circulating endothelial progenitors (CD45-CD31+CD34+/-CD146-), and neutrophils (CD11b+CD66b+) were coevaluated for COVID-19 severity. Spearman correlation analysis demonstrated the synergism among age, obesity, and hypertension with upregulated CD56+ monocytes, endothelial cells, and decreased T cells that lead to severe outcomes of SARS-CoV-2 infection. Circulating monocytes and endothelial cells may represent important cellular markers for monitoring postacute sequelae and impacts of SARS-CoV-2 infection during convalescence and for their role in immune host defense in high-risk adults after vaccination.
Collapse
Affiliation(s)
- Taru S Dutt
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO
| | - Stephanie M LaVergne
- Department of Environmental Radiological and Health Sciences, Colorado State University, Fort Collins, CO
| | - Tracy L Webb
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO
| | - Bridget A Baxter
- Department of Environmental Radiological and Health Sciences, Colorado State University, Fort Collins, CO
| | - Sophia Stromberg
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO
| | - Kim McFann
- University of Colorado Health, Medical Center of the Rockies, Loveland, CO
| | - Kailey Berry
- Department of Molecular, Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, CO
| | - Madison Tipton
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO; and
| | - Omar Alnachoukati
- University of Colorado Health, Medical Center of the Rockies, Loveland, CO
| | - Linda Zier
- University of Colorado Health, Medical Center of the Rockies, Loveland, CO
| | - Greg Ebel
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO
| | - Julie Dunn
- University of Colorado Health, Medical Center of the Rockies, Loveland, CO.,University of Colorado Anschutz School of Medicine, Aurora, CO
| | - Marcela Henao-Tamayo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO
| | - Elizabeth P Ryan
- Department of Environmental Radiological and Health Sciences, Colorado State University, Fort Collins, CO;
| |
Collapse
|
7
|
Watanabe A, Togi M, Koya T, Taniguchi M, Sakamoto T, Iwabuchi K, Kato T, Shimodaira S. Identification of CD56 dim subpopulation marked with high expression of GZMB/PRF1/PI-9 in CD56 + interferon-α-induced dendritic cells. Genes Cells 2021; 26:313-327. [PMID: 33662167 DOI: 10.1111/gtc.12844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/15/2021] [Accepted: 02/28/2021] [Indexed: 01/08/2023]
Abstract
As the sentinels of innate and adaptive immune system, dendritic cells (DCs) have been considered to hold a great promise for medical application. Among the diverse types of DCs, monocyte-derived DCs (mo-DCs) generated in vitro have been most commonly employed. We have been improving the culture protocol and devised a protocol to produce mature interferon-α-induced DCs (IFN-DCs), hereinafter called (mat)IFN-DCs. While exploring the relationship between the expression of CD56 and the cytotoxic activity of (mat)IFN-DCs, we unexpectedly found that sorting of (mat)IFN-DCs with CD56 antibody-coated microbeads (MB) resulted in fractionating cells with tumoricidal activity into the flow-through (FT) but not MB-bound fraction. We uncovered that the FT fraction contains cells expressing low but substantial level of CD56. Moreover, those cells express granzyme B (GrB), perforin (PFN), and serpin B9 at high levels. By employing a specific inhibitor of PFN, we confirmed that direct tumoricidal activity relies on the GrB/PFN pathway. We designated subpopulation in FT fraction as CD56dim and that in CD56 positively sorted fraction as CD56bright , respectively. This is the first time, to our knowledge, to identify subpopulations of CD56-positive IFN-DCs with distinct tumoricidal activity which is ascribed to high expression of the components of GrB/PFN pathway.
Collapse
Affiliation(s)
- Asuka Watanabe
- Department of Regenerative Medicine, School of Medicine, Kanazawa Medical University, Kahoku-gun, Japan
| | - Misa Togi
- Department of Regenerative Medicine, School of Medicine, Kanazawa Medical University, Kahoku-gun, Japan.,Division of Stem Cell Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Kahoku-gun, Japan
| | - Terutsugu Koya
- Department of Regenerative Medicine, School of Medicine, Kanazawa Medical University, Kahoku-gun, Japan
| | - Makoto Taniguchi
- Division of Genome Damage Response Research, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kahoku-gun, Japan
| | - Takuya Sakamoto
- Department of Regenerative Medicine, School of Medicine, Kanazawa Medical University, Kahoku-gun, Japan
| | - Kuniyoshi Iwabuchi
- Division of Genome Damage Response Research, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kahoku-gun, Japan.,Department of Biochemistry I, School of Medicine, Kanazawa Medical University, Kahoku-gun, Japan
| | - Tomohisa Kato
- Division of Stem Cell Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Kahoku-gun, Japan
| | - Shigetaka Shimodaira
- Department of Regenerative Medicine, School of Medicine, Kanazawa Medical University, Kahoku-gun, Japan.,Division of Stem Cell Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Kahoku-gun, Japan
| |
Collapse
|
8
|
Development and validation of an immunoassay for quantification of NCAM-1 in human plasma. J Pharm Biomed Anal 2021; 197:113981. [PMID: 33657526 DOI: 10.1016/j.jpba.2021.113981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Neural Cell Adhesion Molecule 1 (NCAM-1), a multifunctional member of the immunoglobulin superfamily, is expressed on the surface of neurons, glia, skeletal muscle, and natural killer cells. NCAM-1 has been implicated as having a role in cell-cell adhesion, involved in development of the nervous system, and for cells involved in the expansion of T cells and dendritic cells which play an important role in immune surveillance. Sensitive and specific methods to quantify non-surface bound NCAM-1 are not available. METHOD A sandwich ligand binding assay was developed for quantification of NCAM-1 in plasma and validated using an electro-chemiluminescent (ECL) technology. RESULTS The data presented here demonstrated that the validated method met all prespecified criteria for precision, linearity, and accuracy in the range of 62.5 ng/mL to 4000.0 ng/mL, the range believed to be most relevant for plasma. The bioanalytical validation of the assay established the inter-assay coefficient of variation <8 % for calibration points, <2 % for high quality control (HQC), <8 % for medium quality control (MQC) and <19 % for low quality control (LQC) samples. Purified NCAM-1 spike-recovery experiment in plasma was used to determine assay accuracy; nominal concentrations (%) of NCAM-1 ranged from 91 % to 112 % for high and low spike level, respectively. Assay performance was subsequently evaluated for parallelism, selectivity, interference, and stability. CONCLUSION NCAM-1 assay has been developed and validated in human plasma and met all assay validation parameters pre-determined during development. Clinical testing of human plasma samples indicated that NCAM-1 does not seem to be influenced by age and was slightly influenced by gender. NCAM-1 assay has potential to be used as a biomarker assay once the assay is subjected to appropriate clinical assessment and diagnostic thresholds are established.
Collapse
|
9
|
Nörenberg J, Jaksó P, Barakonyi A. Gamma/Delta T Cells in the Course of Healthy Human Pregnancy: Cytotoxic Potential and the Tendency of CD8 Expression Make CD56+ γδT Cells a Unique Lymphocyte Subset. Front Immunol 2021; 11:596489. [PMID: 33603738 PMCID: PMC7884463 DOI: 10.3389/fimmu.2020.596489] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/17/2020] [Indexed: 01/21/2023] Open
Abstract
To date, pregnancy is an immunological paradox. The semi-allogenic fetus must be accepted by the maternal immune system, while defense against pathogens and immune surveillance cannot be compromised. Gamma/delta T cells are believed to play an important role in this immunological puzzle. In this study, we analyzed peripheral blood CD56+ γδT cells from pregnant women (1st, 2nd, and 3rd trimester) and non-pregnant women by multicolor flow cytometry. Interestingly, γδT cells represent almost half of CD3+/CD56+ cells. Among γδT cells, the CD56+ population expands in the 2nd and 3rd trimester. CD56+ γδT cells maintained a predominantly CD4–/CD8– or CD8+ phenotype, while CD56– γδT cells were in similar rates CD4–/CD8– or CD4+ during pregnancy. Investigation of the lysosomal degranulation marker CD107a revealed a preserved elevated rate of potentially cytotoxic CD56+ γδT cells in pregnancy, while their cytotoxic strength was reduced. Furthermore, CD56+ γδT cells continuously showed a higher prevalence of PD-1 expression. CD56+ γδT cells’ rate of PD-1 increased in the 1st trimester and decreased hereafter back to normal level. We correlated the cytotoxic potential and the expression of the inhibitory immune checkpoint PD-1 and were able to demonstrate that highly cytotoxic cells within this CD56+ γδT population tend to express PD-1, which might allow the inhibition of these cells after binding its ligand in the placenta. These findings should support the understanding of the complex processes, which ensure the maintenance of pregnancy.
Collapse
Affiliation(s)
- Jasper Nörenberg
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Pécs, Hungary
| | - Pál Jaksó
- Department of Pathology, Medical School, University of Pécs, Pécs, Hungary
| | - Alíz Barakonyi
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Pécs, Hungary.,Janos Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
10
|
Gunesch JT, Dixon AL, Ebrahim TAM, Berrien-Elliott MM, Tatineni S, Kumar T, Hegewisch-Solloa E, Fehniger TA, Mace EM. CD56 regulates human NK cell cytotoxicity through Pyk2. eLife 2020; 9:e57346. [PMID: 32510326 PMCID: PMC7358009 DOI: 10.7554/elife.57346] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/07/2020] [Indexed: 12/12/2022] Open
Abstract
Human natural killer (NK) cells are defined as CD56+CD3-. Despite its ubiquitous expression on human NK cells the role of CD56 (NCAM) in human NK cell cytotoxic function has not been defined. In non-immune cells, NCAM can induce signaling, mediate adhesion, and promote exocytosis through interactions with focal adhesion kinase (FAK). Here we demonstrate that deletion of CD56 on the NK92 cell line leads to impaired cytotoxic function. CD56-knockout (KO) cells fail to polarize during immunological synapse (IS) formation and have severely impaired exocytosis of lytic granules. Phosphorylation of the FAK family member Pyk2 at tyrosine 402 is decreased in NK92 CD56-KO cells, demonstrating a functional link between CD56 and signaling in human NK cells. Cytotoxicity, lytic granule exocytosis, and the phosphorylation of Pyk2 are rescued by the reintroduction of CD56. These data highlight a novel functional role for CD56 in stimulating exocytosis and promoting cytotoxicity in human NK cells.
Collapse
Affiliation(s)
| | - Amera L Dixon
- Baylor College of MedicineHoustonUnited States
- Rice UniversityHoustonUnited States
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical CenterNew YorkUnited States
| | - Tasneem AM Ebrahim
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical CenterNew YorkUnited States
- Barnard CollegeNew YorkUnited States
| | | | | | | | - Everardo Hegewisch-Solloa
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical CenterNew YorkUnited States
| | - Todd A Fehniger
- Washington University School of MedicineSt. LouisUnited States
| | - Emily M Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical CenterNew YorkUnited States
| |
Collapse
|
11
|
Engineered triple inhibitory receptor resistance improves anti-tumor CAR-T cell performance via CD56. Nat Commun 2019; 10:4109. [PMID: 31511513 PMCID: PMC6739330 DOI: 10.1038/s41467-019-11893-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 07/25/2019] [Indexed: 12/25/2022] Open
Abstract
The inhibitory receptors PD-1, Tim-3, and Lag-3 are highly expressed on tumor-infiltrating lymphocytes and compromise their antitumor activity. For efficient cancer immunotherapy, it is important to prevent chimeric antigen receptor T (CAR-T)-cell exhaustion. Here we downregulate these three checkpoint receptors simultaneously on CAR-T cells and that show the resulting PTL-CAR-T cells undergo epigenetic modifications and better control tumor growth. Furthermore, we unexpectedly find increased tumor infiltration by PTL-CAR-T cells and their clustering between the living and necrotic tumor tissue. Mechanistically, PTL-CAR-T cells upregulate CD56 (NCAM), which is essential for their effector function. The homophilic interaction between intercellular CD56 molecules correlates with enhanced infiltration of CAR-T cells, increased secretion of interferon-γ, and the prolonged survival of CAR-T cells. Ectopically expressed CD56 promotes CAR-T cell survival and antitumor response. Our findings demonstrate that genetic blockade of three checkpoint inhibitory receptors and the resulting high expression of CD56 on CAR-T cells enhances the inhibition of tumor growth. The inhibitory receptors PD-1, Tim-3 and Lag-3 act as negative feedback regulators of T cell responses. Here the authors improve CAR T cell antitumor efficacy by triple knockdown of these receptors, show it requires CD56, and correlate CD56-mediated homophilic cell interactions with CAR T cell efficacy.
Collapse
|
12
|
Van Acker HH, Van Acker ZP, Versteven M, Ponsaerts P, Pende D, Berneman ZN, Anguille S, Van Tendeloo VF, Smits EL. CD56 Homodimerization and Participation in Anti-Tumor Immune Effector Cell Functioning: A Role for Interleukin-15. Cancers (Basel) 2019; 11:E1029. [PMID: 31336622 PMCID: PMC6678613 DOI: 10.3390/cancers11071029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 07/17/2019] [Indexed: 12/16/2022] Open
Abstract
A particularly interesting marker to identify anti-tumor immune cells is the neural cell adhesion molecule (NCAM), also known as cluster of differentiation (CD)56. Namely, hematopoietic expression of CD56 seems to be confined to powerful effector immune cells. Here, we sought to elucidate its role on various killer immune cells. First, we identified the high motility NCAM-120 molecule to be the main isoform expressed by immune cells. Next, through neutralization of surface CD56, we were able to (1) demonstrate the direct involvement of CD56 in tumor cell lysis exerted by CD56-expressing killer cells, such as natural killer cells, gamma delta (γδ) T cells, and interleukin (IL)-15-cultured dendritic cells (DCs), and (2) reveal a putative crosstalk mechanism between IL-15 DCs and CD8 T cells, suggesting CD56 as a co-stimulatory molecule in their cell-to-cell contact. Moreover, by means of a proximity ligation assay, we visualized the CD56 homophilic interaction among cancer cells and between immune cells and cancer cells. Finally, by blocking the mitogen-activated protein kinase (MAPK) pathway and the phosphoinositide 3-kinase (PI3K)-Akt pathway, we showed that IL-15 stimulation directly led to CD56 upregulation. In conclusion, these results underscore the previously neglected importance of CD56 expression on immune cells, benefiting current and future immune therapeutic options.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Zoë P Van Acker
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling, University of Antwerp, 2610 Antwerp, Belgium
- Laboratory of Membrane Trafficking, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Maarten Versteven
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Experimental Cell Transplantation Group (ECTG), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Daniela Pende
- Immunology Laboratory, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling, University of Antwerp, 2610 Antwerp, Belgium
- Division of Hematology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Division of Hematology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
13
|
Timmers M, Roex G, Wang Y, Campillo-Davo D, Van Tendeloo VFI, Chu Y, Berneman ZN, Luo F, Van Acker HH, Anguille S. Chimeric Antigen Receptor-Modified T Cell Therapy in Multiple Myeloma: Beyond B Cell Maturation Antigen. Front Immunol 2019; 10:1613. [PMID: 31379824 PMCID: PMC6646459 DOI: 10.3389/fimmu.2019.01613] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 06/28/2019] [Indexed: 12/27/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T cell therapy is a rapidly emerging immunotherapeutic approach that is revolutionizing cancer treatment. The impressive clinical results obtained with CAR-T cell therapy in patients with acute lymphoblastic leukemia and lymphoma have fueled the development of CAR-T cells targeting other malignancies, including multiple myeloma (MM). The field of CAR-T cell therapy for MM is still in its infancy, but remains promising. To date, most studies have been performed with B cell maturation antigen (BCMA)-targeted CARs, for which high response rates have been obtained in early-phase clinical trials. However, responses are usually temporary, and relapses have frequently been observed. One of the major reasons for relapse is the loss or downregulation of BCMA expression following CAR-T therapy. This has fostered a search for alternative target antigens that are expressed on the MM cell surface. In this review, we provide an overview of myeloma target antigens other than BCMA that are currently being evaluated in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Marijke Timmers
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Gils Roex
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Yuedi Wang
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Viggo F I Van Tendeloo
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Yiwei Chu
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Zwi N Berneman
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Feifei Luo
- Biotherapy Research Center, Fudan University, Shanghai, China.,Department of Digestive Diseases, Huashan Hospital of Fudan University, Shanghai, China
| | - Heleen H Van Acker
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Sébastien Anguille
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
14
|
CD56 expression in breast cancer induces sensitivity to natural killer-mediated cytotoxicity by enhancing the formation of cytotoxic immunological synapse. Sci Rep 2019; 9:8756. [PMID: 31217484 PMCID: PMC6584531 DOI: 10.1038/s41598-019-45377-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 05/28/2019] [Indexed: 02/07/2023] Open
Abstract
We examined the potential value of the natural killer (NK) cell line; NK-92, as immunotherapy tool for breast cancer (BC) treatment and searched for biomarker(s) of sensitivity to NK-92-mediated cytotoxicity. The cytotoxic activity of NK-92 cells towards one breast precancerous and nine BC cell lines was analyzed using calcein-AM and degranulation assays. The molecules associated with NK-92-responsiveness were determined by differential gene expression analysis using RNA-sequencing and validated by RT-PCR, immunostaining and flow cytometry. NK-target interactions and immunological synapse formation were assessed by fluorescence microscopy. Potential biomarker expression was determined by IHC in 99 patient-derived BC tissues and 10 normal mammary epithelial tissues. Most (8/9) BC cell lines were resistant while only one BC and the precancerous cell lines were effectively killed by NK-92 lymphocytes. NK-92-sensitive target cells specifically expressed CD56, which ectopic expression in CD56-negative BC cells induced their sensitivity to NK-92-mediated killing, suggesting that CD56 is not only a biomarker of responsiveness but actively regulates NK function. CD56 adhesion molecules which are also expressed on NK cells accumulate at the immunological synapse enhancing NK-target interactions, cytotoxic granzyme B transfer from NK-92 to CD56-expressing target cells and induction of caspase 3 activation in targets. Interestingly, CD56 expression was found to be reduced in breast tumor tissues (36%) with strong inter- and intratumoral heterogeneity in comparison to normal breast tissues (80%). CD56 is a potential predictive biomarker for BC responsiveness to NK-92-cell based immunotherapy and loss of CD56 expression might be a mechanism of escape from NK-immunity.
Collapse
|
15
|
Boesch M, Cosma A, Sopper S. Flow Cytometry: To Dump or Not To Dump. THE JOURNAL OF IMMUNOLOGY 2018; 201:1813-1815. [DOI: 10.4049/jimmunol.1801037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 12/20/2022]
|
16
|
GFRA1: A Novel Molecular Target for the Prevention of Osteosarcoma Chemoresistance. Int J Mol Sci 2018; 19:ijms19041078. [PMID: 29617307 PMCID: PMC5979596 DOI: 10.3390/ijms19041078] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/26/2018] [Accepted: 03/31/2018] [Indexed: 12/11/2022] Open
Abstract
The glycosylphosphatidylinositol-linked GDNF (glial cell derived neurotrophic factor) receptor alpha (GFRA), a coreceptor that recognizes the GDNF family of ligands, has a crucial role in the development and maintenance of the nervous system. Of the four identified GFRA isoforms, GFRA1 specifically recognizes GDNF and is involved in the regulation of proliferation, differentiation, and migration of neuronal cells. GFRA1 has also been implicated in cancer cell progression and metastasis. Recent findings show that GFRA1 can contribute to the development of chemoresistance in osteosarcoma. GFRA1 expression was induced following treatment of osteosarcoma cells with the popular anticancer drug, cisplatin and induction of GFRA1 expression significantly suppressed apoptosis mediated by cisplatin in osteosarcoma cells. GFRA1 expression promotes autophagy by activating the SRC-AMPK signaling axis following cisplatin treatment, resulting in enhanced osteosarcoma cell survival. GFRA1-induced autophagy promoted tumor growth in mouse xenograft models, suggesting a novel function of GFRA1 in osteosarcoma chemoresistance.
Collapse
|
17
|
Van Acker HH, Capsomidis A, Smits EL, Van Tendeloo VF. CD56 in the Immune System: More Than a Marker for Cytotoxicity? Front Immunol 2017; 8:892. [PMID: 28791027 PMCID: PMC5522883 DOI: 10.3389/fimmu.2017.00892] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 07/12/2017] [Indexed: 11/13/2022] Open
Abstract
Over the past years, the phenotypic and functional boundaries distinguishing the main cell subsets of the immune system have become increasingly blurred. In this respect, CD56 (also known as neural cell adhesion molecule) is a very good example. CD56 is the archetypal phenotypic marker of natural killer cells but can actually be expressed by many more immune cells, including alpha beta T cells, gamma delta T cells, dendritic cells, and monocytes. Common to all these CD56-expressing cell types are strong immunostimulatory effector functions, including T helper 1 cytokine production and an efficient cytotoxic capacity. Interestingly, both numerical and functional deficiencies and phenotypic alterations of the CD56+ immune cell fraction have been reported in patients with various infectious, autoimmune, or malignant diseases. In this review, we will discuss our current knowledge on the expression and function of CD56 in the hematopoietic system, both in health and disease.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Anna Capsomidis
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium.,Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
18
|
Van Acker HH, Anguille S, Willemen Y, Van den Bergh JM, Berneman ZN, Lion E, Smits EL, Van Tendeloo VF. Interleukin-15 enhances the proliferation, stimulatory phenotype, and antitumor effector functions of human gamma delta T cells. J Hematol Oncol 2016; 9:101. [PMID: 27686372 PMCID: PMC5041439 DOI: 10.1186/s13045-016-0329-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/16/2016] [Indexed: 12/12/2022] Open
Abstract
Background Adoptive immunotherapy is gaining momentum to fight malignancies, whereby γδ T cells have received recent attention as an alternative cell source as to natural killer cells and αβ T cells. The advent of γδ T cells is largely due to their ability to recognize and target tumor cells using both innate characteristic and T cell receptor (TCR)-mediated mechanisms, their capacity to enhance the generation of antigen-specific T cell responses, and their potential to be used in an autologous or allogeneic setting. Methods In this study, we explored the beneficial effect of the immunostimulatory cytokine interleukin (IL)-15 on purified γδ T cells and its use as a stimulatory signal in the ex vivo expansion of γδ T cells for adoptive transfer. The expansion protocol was validated both with immune cells of healthy individuals and acute myeloid leukemia patients. Results We report that the addition of IL-15 to γδ T cell cultures results in a more activated phenotype, a higher proliferative capacity, a more pronounced T helper 1 polarization, and an increased cytotoxic capacity of γδ T cells. Moreover γδ T cell expansion starting with peripheral blood mononuclear cells from healthy individuals and acute myeloid leukemia patients is boosted in the presence of IL-15, whereby the antitumor properties of the γδ T cells are strengthened as well. Conclusions Our results support the rationale to explore the use of IL-15 in clinical adoptive therapy protocols exploiting γδ T cells.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Yannick Willemen
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium
| | - Johan M Van den Bergh
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium.,Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium
| |
Collapse
|
19
|
The Prognostic Impact of NK/NKT Cell Density in Periampullary Adenocarcinoma Differs by Morphological Type and Adjuvant Treatment. PLoS One 2016; 11:e0156497. [PMID: 27275582 PMCID: PMC4898776 DOI: 10.1371/journal.pone.0156497] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/16/2016] [Indexed: 12/23/2022] Open
Abstract
Background Natural killer (NK) cells and NK T cells (NKT) are vital parts of tumour immunosurveillance. However, their impact on prognosis and chemotherapy response in periampullary adenocarcinoma, including pancreatic cancer, has not yet been described. Methods Immune cell-specific expression of CD56, CD3, CD68 and CD1a was analysed by immunohistochemistry on tissue microarrays with tumours from 175 consecutive cases of periampullary adenocarcinoma, 110 of pancreatobiliary type (PB-type) and 65 of intestinal type (I-type) morphology. Kaplan-Meier and Cox regression analysis were applied to determine the impact of CD56+ NK/NKT cells on 5-year overall survival (OS). Results High density of CD56+ NK/NKT cells correlated with low N-stage and lack of perineural, lymphatic vessel and peripancreatic fat invasion. High density of CD56+ NK/NKT cells was associated with prolonged OS in Kaplan-Meier analysis (p = 0.003), and in adjusted Cox regression analysis (HR = 0.49; 95% CI 0.29–0.86). The prognostic effect of high CD56+ NK/NKT cell infiltration was only evident in cases not receiving adjuvant chemotherapy in PB-type tumours (p for interaction = 0.014). Conclusion This study demonstrates that abundant infiltration of CD56+ NK/NKT cells is associated with a prolonged survival in periampullary adenocarcinoma. However, the negative interaction with adjuvant treatment is noteworthy. NK cell enhancing strategies may prove to be successful in the management of these cancers.
Collapse
|
20
|
Anguille S, Smits EL, Bryant C, Van Acker HH, Goossens H, Lion E, Fromm PD, Hart DN, Van Tendeloo VF, Berneman ZN. Dendritic Cells as Pharmacological Tools for Cancer Immunotherapy. Pharmacol Rev 2015; 67:731-53. [PMID: 26240218 DOI: 10.1124/pr.114.009456] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Although the earliest—rudimentary—attempts at exploiting the immune system for cancer therapy can be traced back to the late 18th Century, it was not until the past decade that cancer immunotherapeutics have truly entered mainstream clinical practice. Given their potential to stimulate both adaptive and innate antitumor immune responses, dendritic cells (DCs) have come under intense scrutiny in recent years as pharmacological tools for cancer immunotherapy. Conceptually, the clinical effectiveness of this form of active immunotherapy relies on the completion of three critical steps: 1) the DCs used as immunotherapeutic vehicles must properly activate the antitumor immune effector cells of the host, 2) these immune effector cells must be receptive to stimulation by the DCs and be competent to mediate their antitumor effects, which 3) requires overcoming the various immune-inhibitory mechanisms used by the tumor cells. In this review, following a brief overview of the pivotal milestones in the history of cancer immunotherapy, we will introduce the reader to the basic immunobiological and pharmacological principles of active cancer immunotherapy using DCs. We will then discuss how current research is trying to define the optimal parameters for each of the above steps to realize the full clinical potential of DC therapeutics. Given its high suitability for immune interventions, acute myeloid leukemia was chosen here to showcase the latest research trends driving the field of DC-based cancer immunotherapy.
Collapse
Affiliation(s)
- Sébastien Anguille
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Evelien L Smits
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Christian Bryant
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Heleen H Van Acker
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Herman Goossens
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Eva Lion
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Phillip D Fromm
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | | | - Viggo F Van Tendeloo
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Zwi N Berneman
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| |
Collapse
|
21
|
Anguille S, Van Acker HH, Van den Bergh J, Willemen Y, Goossens H, Van Tendeloo VF, Smits EL, Berneman ZN, Lion E. Interleukin-15 Dendritic Cells Harness NK Cell Cytotoxic Effector Function in a Contact- and IL-15-Dependent Manner. PLoS One 2015; 10:e0123340. [PMID: 25951230 PMCID: PMC4423923 DOI: 10.1371/journal.pone.0123340] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 03/02/2015] [Indexed: 01/02/2023] Open
Abstract
The contribution of natural killer (NK) cells to the treatment efficacy of dendritic cell (DC)-based cancer vaccines is being increasingly recognized. Much current efforts to optimize this form of immunotherapy are therefore geared towards harnessing the NK cell-stimulatory ability of DCs. In this study, we investigated whether generation of human monocyte-derived DCs with interleukin (IL)-15 followed by activation with a Toll-like receptor stimulus endows these DCs, commonly referred to as "IL-15 DCs", with the capacity to stimulate NK cells. In a head-to-head comparison with "IL-4 DCs" used routinely for clinical studies, IL-15 DCs were found to induce a more activated, cytotoxic effector phenotype in NK cells, in particular in the CD56bright NK cell subset. With the exception of GM-CSF, no significant enhancement of cytokine/chemokine secretion was observed following co-culture of NK cells with IL-15 DCs. IL-15 DCs, but not IL-4 DCs, promoted NK cell tumoricidal activity towards both NK-sensitive and NK-resistant targets. This effect was found to require cell-to-cell contact and to be mediated by DC surface-bound IL-15. This study shows that DCs can express a membrane-bound form of IL-15 through which they enhance NK cell cytotoxic function. The observed lack of membrane-bound IL-15 on "gold-standard" IL-4 DCs and their consequent inability to effectively promote NK cell cytotoxicity may have important implications for the future design of DC-based cancer vaccine studies.
Collapse
Affiliation(s)
- Sébastien Anguille
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
- Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Heleen H. Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Johan Van den Bergh
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Yannick Willemen
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Viggo F. Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Evelien L. Smits
- Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research, University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Zwi N. Berneman
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
- Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| |
Collapse
|
22
|
Smits ELJM, Anguille S, Berneman ZN. Interferon α may be back on track to treat acute myeloid leukemia. Oncoimmunology 2014; 2:e23619. [PMID: 23734314 PMCID: PMC3654584 DOI: 10.4161/onci.23619] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 01/14/2013] [Indexed: 11/19/2022] Open
Abstract
Our own experience and a thorough literature review suggest that interferon α (IFNα) should be reconsidered for the treatment of acute myeloid leukemia patients. Most likely, the success of such treatment depends on the achievement of high serum levels of IFNα for several months, which can be obtained by using pegylated IFNα.
Collapse
Affiliation(s)
- Evelien L J M Smits
- Tumor Immunology Group, Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium ; Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | | | | |
Collapse
|
23
|
Valgardsdottir R, Capitanio C, Texido G, Pende D, Cantoni C, Pesenti E, Rambaldi A, Golay J, Introna M. Direct involvement of CD56 in cytokine-induced killer-mediated lysis of CD56+ hematopoietic target cells. Exp Hematol 2014; 42:1013-21.e1. [PMID: 25201755 DOI: 10.1016/j.exphem.2014.08.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/19/2014] [Accepted: 08/23/2014] [Indexed: 11/24/2022]
Abstract
Cytokine-induced killer (CIK) cells are in-vitro-expanded T lymphocytes that represent a heterogeneous population. A large majority of CIK cells are CD3(+)CD56(+), and this population has been shown to confer a cytotoxic effect against tumor targets. The scope of this work was to study whether CD56 has a direct role in CIK-mediated cytotoxicity. Blocking of CD56 with the anti-CD56 monoclonal antibody GPR165 significantly reduced CIK-mediated lysis of three CD56(+) hematopoietic tumor cell lines (AML-NS8, NB4, and KCL22), whereas no effect was observed on three CD56(-) hematopoietic tumor cell lines (K562, REH, and MOLT-4). Knockdown of CD56 in CIK cells by short interfering RNA made the cells less cytotoxic against a CD56(+) target, and knockdown of CD56 in target cells with lentiviral short hairpin RNA significantly altered their susceptibility to CIK-mediated lysis. Our data suggest that homophilic interaction between CD56 molecules may occur in tumor-cell recognition, leading to CIK-mediated cell death.
Collapse
Affiliation(s)
- Rut Valgardsdottir
- USS Centre of Cellular Therapy "G.Lanzani", USC Hematology and Bone Marrow Transplantation Unit, A.O. Papa Giovanni XXIII, Bergamo, Italy
| | - Cristina Capitanio
- USS Centre of Cellular Therapy "G.Lanzani", USC Hematology and Bone Marrow Transplantation Unit, A.O. Papa Giovanni XXIII, Bergamo, Italy
| | | | | | - Claudia Cantoni
- Istituto Giannina Gaslini, Genoa, Italy; Dipartimento di Medicina Sperimentale e Centro di Eccellenza per la Ricerca Biomedica, Università di Genova, Genoa, Italy
| | | | - Alessandro Rambaldi
- USS Centre of Cellular Therapy "G.Lanzani", USC Hematology and Bone Marrow Transplantation Unit, A.O. Papa Giovanni XXIII, Bergamo, Italy
| | - Josée Golay
- USS Centre of Cellular Therapy "G.Lanzani", USC Hematology and Bone Marrow Transplantation Unit, A.O. Papa Giovanni XXIII, Bergamo, Italy
| | - Martino Introna
- USS Centre of Cellular Therapy "G.Lanzani", USC Hematology and Bone Marrow Transplantation Unit, A.O. Papa Giovanni XXIII, Bergamo, Italy.
| |
Collapse
|
24
|
Van den Bergh JMJ, Guerti K, Willemen Y, Lion E, Cools N, Goossens H, Vorsters A, Van Tendeloo VFI, Anguille S, Van Damme P, Smits ELJM. HPV vaccine stimulates cytotoxic activity of killer dendritic cells and natural killer cells against HPV-positive tumour cells. J Cell Mol Med 2014; 18:1372-80. [PMID: 24979331 PMCID: PMC4124021 DOI: 10.1111/jcmm.12284] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/17/2014] [Indexed: 01/09/2023] Open
Abstract
Cervarix™ is approved as a preventive vaccine against infection with the human papillomavirus (HPV) strains 16 and 18, which are causally related to the development of cervical cancer. We are the first to investigate in vitro the effects of this HPV vaccine on interleukin (IL)-15 dendritic cells (DC) as proxy of a naturally occurring subset of blood DC, and natural killer (NK) cells, two innate immune cell types that play an important role in antitumour immunity. Our results show that exposure of IL-15 DC to the HPV vaccine results in increased expression of phenotypic maturation markers, pro-inflammatory cytokine production and cytotoxic activity against HPV-positive tumour cells. These effects are mediated by the vaccine adjuvant, partly through Toll-like receptor 4 activation. Next, we demonstrate that vaccine-exposed IL-15 DC in turn induce phenotypic activation of NK cells, resulting in a synergistic cytotoxic action against HPV-infected tumour cells. Our study thus identifies a novel mode of action of the HPV vaccine in boosting innate immunity, including killing of HPV-infected cells by DC and NK cells.
Collapse
Affiliation(s)
- Johan M J Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bakdash G, Schreurs I, Schreibelt G, Tel J. Crosstalk between dendritic cell subsets and implications for dendritic cell-based anticancer immunotherapy. Expert Rev Clin Immunol 2014; 10:915-26. [PMID: 24758519 DOI: 10.1586/1744666x.2014.912561] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Dendritic cells (DCs) are a family of professional antigen-presenting cells that have an indispensable role in the initiation of innate and adaptive immune responses against pathogens and tumor cells. The DC family is very heterogeneous. Two main types of naturally occurring DCs circulate in peripheral blood, each with its unique phenotypic and functional characteristics: myeloid DCs and plasmacytoid. There is an ample number of studies that have focused on the bi-directional crosstalk between DCs and natural killer cells or T cells. However, the crosstalk among the different DC subsets, in the context of infectious diseases and cancer, has until now not received much attention. Here, we review all available literature that has dealt with the crosstalk between plasmacytoid and myeloid DCs and the potential mode of action. Emphasis will be given to the therapeutic potential of the combination of DC subsets for DC-based immunotherapy.
Collapse
Affiliation(s)
- Ghaith Bakdash
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
26
|
Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2013; 2:e25771. [PMID: 24286020 PMCID: PMC3841205 DOI: 10.4161/onci.25771] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 12/26/2022] Open
Abstract
Dendritic cells (DCs) occupy a privileged position at the interface between innate and adaptive immunity, orchestrating a large panel of responses to both physiological and pathological cues. In particular, whereas the presentation of antigens by immature DCs generally results in the development of immunological tolerance, mature DCs are capable of priming robust, and hence therapeutically relevant, adaptive immune responses. In line with this notion, functional defects in the DC compartment have been shown to etiologically contribute to pathological conditions including (but perhaps not limited to) infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. Thus, the possibility of harnessing the elevated immunological potential of DCs for anticancer therapy has attracted considerable interest from both researchers and clinicians over the last decade. Alongside, several methods have been developed not only to isolate DCs from cancer patients, expand them, load them with tumor-associated antigens and hence generate highly immunogenic clinical grade infusion products, but also to directly target DCs in vivo. This intense experimental effort has culminated in 2010 with the approval by the US FDA of a DC-based preparation (sipuleucel-T, Provenge®) for the treatment of asymptomatic or minimally symptomatic metastatic castration-refractory prostate cancer. As an update to the latest Trial Watch dealing with this exciting field of research (October 2012), here we summarize recent advances in DC-based anticancer regimens, covering both high-impact studies that have been published during the last 13 mo and clinical trials that have been launched in the same period to assess the antineoplastic potential of this variant of cellular immunotherapy.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Anguille S, Lion E, Van den Bergh J, Van Acker HH, Willemen Y, Smits EL, Van Tendeloo VF, Berneman ZN. Interleukin-15 dendritic cells as vaccine candidates for cancer immunotherapy. Hum Vaccin Immunother 2013; 9:1956-61. [PMID: 23778748 DOI: 10.4161/hv.25373] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Owing to their professional antigen-presenting capacity and unique potential to induce tumor antigen-specific T cell immunity, dendritic cells (DCs) have attracted much interest over the past decades for therapeutic vaccination against cancer. Clinical trials have shown that the use of tumor antigen-loaded DCs in cancer patients is safe and that it has the potential to induce anti-tumor immunity which, in some cases, culminates in striking clinical responses. Unfortunately, in a considerable number of patients, DC vaccination is unable to mount effective anti-tumor immune responses and, if it does so, the resultant immunity is often insufficient to translate into tangible clinical benefit. This underscores the necessity to re-design and optimize the current procedures for DC vaccine manufacturing. A new generation of DC vaccines with improved potency has now become available for clinical use as a result of extensive pre-clinical research. One of the promising next-generation DC vaccine candidates are interleukin (IL)-15-differentiated DCs. In this commentary, we will compile the research data that have been obtained by our group and other groups with these so-called IL-15 DCs and summarize the evidence supporting the implementation of IL-15 DCs in DC-based cancer vaccination regimens.
Collapse
Affiliation(s)
- Sébastien Anguille
- Vaccine & Infectious Disease Institute; Laboratory of Experimental Hematology; Tumor Immunology Group (TIGR); University of Antwerp; Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine; Antwerp University Hospital; Antwerp, Belgium
| | | | | | | | | | | | | | | |
Collapse
|