1
|
Rothfuß C, Baumann T, Donakonda S, Brauchle B, Marcinek A, Urban C, Mergner J, Pedde AM, Hirschberger A, Krupka C, Neumann AS, Hänel G, Merten C, Öllinger R, Hecker JS, Bauer T, Schmid C, Götze KS, Altomonte J, Bücklein V, Jacobs R, Rad R, Dawid C, Simeoni L, Schraven B, Pichlmair A, Subklewe M, Knolle PA, Böttcher JP, Höchst B. Two-layered immune escape in AML is overcome by Fcγ receptor activation and inhibition of PGE2 signaling in NK cells. Blood 2025; 145:1395-1406. [PMID: 39840945 DOI: 10.1182/blood.2024025706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/13/2024] [Accepted: 11/29/2024] [Indexed: 01/23/2025] Open
Abstract
ABSTRACT Loss of anticancer natural killer (NK) cell function in patients with acute myeloid leukemia (AML) is associated with fatal disease progression and remains poorly understood. Here, we demonstrate that AML blasts isolated from patients rapidly inhibit NK cell function and escape NK cell-mediated killing. Transcriptome analysis of NK cells exposed to AML blasts revealed increased CREM expression and transcriptional activity, indicating enhanced cyclic adenosine monophosphate (cAMP) signaling, confirmed by uniform production of the cAMP-inducing prostanoid prostaglandin E2 (PGE2) by all AML-blast isolates from patients. Phosphoproteome analysis disclosed that PGE2 induced a blockade of lymphocyte-specific protein tyrosine kinase (LCK)-extracellular signal-regulated kinase signaling that is crucial for NK cell activation, indicating a 2-layered escape of AML blasts with low expression of NK cell-activating ligands and inhibition of NK cell signaling. To evaluate the therapeutic potential to target PGE2 inhibition, we combined Fcγ-receptor-mediated activation with the prevention of inhibitory PGE2 signaling. This rescued NK cell function and restored the killing of AML blasts. Thus, we identify the PGE2-LCK signaling axis as the key barrier for NK cell activation in 2-layered immune escape of AML blasts that can be targeted for immune therapy to reconstitute anticancer NK cell immunity in patients with AML.
Collapse
MESH Headings
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Dinoprostone/immunology
- Dinoprostone/metabolism
- Signal Transduction/immunology
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Tumor Escape/immunology
- Lymphocyte Activation/immunology
Collapse
Affiliation(s)
- Charlotte Rothfuß
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Tobias Baumann
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Sainitin Donakonda
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Bettina Brauchle
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anetta Marcinek
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Urban
- Institute of Virology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at Munich Institute of Robotics and Machine Intelligence, Technical University of Munich, Munich, Germany
| | - Anna-Marie Pedde
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Anna Hirschberger
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christina Krupka
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anne-Sophie Neumann
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gerulf Hänel
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Camilla Merten
- Institut of Molecular and Clinical Immunology, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Judith S Hecker
- Department of Medicine III, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Tanja Bauer
- Institute of Virology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christian Schmid
- Food Chemistry and Molecular Sensory Science, Technical University of Munich, Munich, Germany
| | - Katharina S Götze
- Department of Medicine III, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Jennifer Altomonte
- Department of Internal Medicine II, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Veit Bücklein
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Roland Jacobs
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, Hannover, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Corina Dawid
- Food Chemistry and Molecular Sensory Science, Technical University of Munich, Munich, Germany
| | - Luca Simeoni
- Institut of Molecular and Clinical Immunology, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Burkhart Schraven
- Institut of Molecular and Clinical Immunology, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Andreas Pichlmair
- Institute of Virology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Marion Subklewe
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Molecular Immunology, School of Life Science, Technical University of Munich, Munich, Germany
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Experimental Immunology, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Bastian Höchst
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
2
|
Ochayon DE, DeVore SB, Chang WC, Krishnamurthy D, Seelamneni H, Grashel B, Spagna D, Andorf S, Martin LJ, Biagini JM, Waggoner SN, Khurana Hershey GK. Progressive accumulation of hyperinflammatory NKG2D low NK cells in early childhood severe atopic dermatitis. Sci Immunol 2024; 9:eadd3085. [PMID: 38335270 PMCID: PMC11107477 DOI: 10.1126/sciimmunol.add3085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 12/21/2023] [Indexed: 02/12/2024]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease that often precedes the development of food allergy, asthma, and allergic rhinitis. The prevailing paradigm holds that a reduced frequency and function of natural killer (NK) cell contributes to AD pathogenesis, yet the underlying mechanisms and contributions of NK cells to allergic comorbidities remain ill-defined. Here, analysis of circulating NK cells in a longitudinal early life cohort of children with AD revealed a progressive accumulation of NK cells with low expression of the activating receptor NKG2D, which was linked to more severe AD and sensitivity to allergens. This was most notable in children co-sensitized to food and aeroallergens, a risk factor for development of asthma. Individual-level longitudinal analysis in a subset of children revealed coincident reduction of NKG2D on NK cells with acquired or persistent sensitization, and this was associated with impaired skin barrier function assessed by transepidermal water loss. Low expression of NKG2D on NK cells was paradoxically associated with depressed cytolytic function but exaggerated release of the proinflammatory cytokine tumor necrosis factor-α. These observations provide important insights into a potential mechanism underlying the development of allergic comorbidity in early life in children with AD, which involves altered NK cell functional responses, and define an endotype of severe AD.
Collapse
Affiliation(s)
- David E. Ochayon
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
| | - Stanley B. DeVore
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Cancer and Cell Biology Program, University of Cincinnati College of Medicine
| | - Wan-Chi Chang
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
| | - Durga Krishnamurthy
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center
| | - Harsha Seelamneni
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center
| | - Brittany Grashel
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
| | - Daniel Spagna
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
| | - Sandra Andorf
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Lisa J. Martin
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Jocelyn M. Biagini
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Stephen N. Waggoner
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Gurjit K. Khurana Hershey
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Cancer and Cell Biology Program, University of Cincinnati College of Medicine
- Department of Pediatrics, University of Cincinnati College of Medicine
| |
Collapse
|
3
|
Jost S, Ahn J, Chen S, Yoder T, Gikundiro KE, Lee E, Gressens SB, Kroll K, Craemer M, Kaynor GC, Lifton M, Tan CS. Upregulation of the NKG2D Ligand ULBP2 by JC Polyomavirus Infection Promotes Immune Recognition by Natural Killer Cells. J Infect Dis 2024; 229:1836-1844. [PMID: 37774496 PMCID: PMC11175686 DOI: 10.1093/infdis/jiad424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND JC polyomavirus (JCPyV) causes progressive multifocal leukoencephalopathy (PML), a potentially fatal complication of severe immune suppression with no effective treatment. Natural killer (NK) cells play critical roles in defense against viral infections; however, NK-cell response to JCPyV infection remains unexplored. METHODS NK- and T-cell responses against the JCPyV VP1 were compared using intracellular cytokine staining upon stimulation with peptide pools. A novel flow cytometry-based assay was developed to determine NK-cell killing efficiency of JCPyV-infected astrocyte-derived SVG-A cells. Blocking antibodies were used to evaluate the contribution of NK-cell receptors in immune recognition of JCPyV-infected cells. RESULTS In about 40% of healthy donors, we detected robust CD107a upregulation and IFN-γ production by NK cells, extending beyond T-cell responses. Next, using the NK-cell-mediated killing assay, we showed that coculture of NK cells and JCPyV-infected SVG-A cells leads to a 60% reduction in infection, on average. JCPyV-infected cells had enhanced expression of ULBP2-a ligand for the activating NK-cell receptor NKG2D, and addition of NKG2D blocking antibodies decreased NK-cell degranulation. CONCLUSIONS NKG2D-mediated activation of NK cells plays a key role in controlling JCPyV replication and may be a promising immunotherapeutic target to boost NK-cell anti-JCPyV activity.
Collapse
Affiliation(s)
- Stephanie Jost
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jenny Ahn
- Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah Chen
- Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Taylor Yoder
- Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kayitare Eunice Gikundiro
- Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Division of Infectious Diseases, Department of Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Esther Lee
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Simon B Gressens
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kyle Kroll
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Melissa Craemer
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Michelle Lifton
- Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - C Sabrina Tan
- Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Division of Infectious Diseases, Department of Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Division of Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Li S, Kang Y, Zeng Y. Targeting tumor and bone microenvironment: Novel therapeutic opportunities for castration-resistant prostate cancer patients with bone metastasis. Biochim Biophys Acta Rev Cancer 2024; 1879:189033. [PMID: 38040267 DOI: 10.1016/j.bbcan.2023.189033] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/22/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
Despite standard hormonal therapy that targets the androgen receptor (AR) attenuates prostate cancer (PCa) effectively in the initial stage, the tumor ultimately converts to castration-resistant prostate cancer (CRPC), and the acquired resistance is still a great challenge for the management of advanced prostate cancer patients. The tumor microenvironment (TME) consists of multiple cellular and noncellular agents is well known as a vital role during the development and progression of CRPC by establishing communication between TME and tumor cells. Additionally, as primary prostate cancer progresses towards metastasis, and CRPC always experiences bone metastasis, the TME is conducive to the spread of tumors to the distant sits, particularly in bone. In addition, the bone microenvironment (BME) is also closely related to the survival, growth and colonization of metastatic tumor cells. The present review summarized the recent studies which mainly focused on the role of TME or BME in the CRPC patients with bone metastasis, and discussed the underlying mechanisms, as well as the potential therapeutic values of targeting TME and BME in the management of metastatic CRPC patients.
Collapse
Affiliation(s)
- Shenglong Li
- Second ward of Bone and Soft Tissue Tumor Surgery,Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China; The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, Shenyang, China
| | - Yue Kang
- Department of Breast Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yu Zeng
- Department of Urology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China.
| |
Collapse
|
5
|
Agrez M, Chandler C, Thurecht KJ, Fletcher NL, Liu F, Subramaniam G, Howard CB, Blyth B, Parker S, Turner D, Rzepecka J, Knox G, Nika A, Hall AM, Gooding H, Gallagher L. An immunomodulating peptide with potential to suppress tumour growth and autoimmunity. Sci Rep 2023; 13:19741. [PMID: 37957274 PMCID: PMC10643673 DOI: 10.1038/s41598-023-47229-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/10/2023] [Indexed: 11/15/2023] Open
Abstract
Cancers and autoimmune diseases commonly co-exist and immune checkpoint inhibitor therapy (ICI) exacerbates autoimmune pathologies. We recently described a lipidic peptide, designated IK14004, that promotes expansion of immunosuppressive T regulatory (Treg) cells and uncouples interleukin-2 from interferon-gamma production while activating CD8+ T cells. Herein, we report IK14004-mediated inhibition of Lewis lung cancer (LLC) growth and re-invigoration of splenocyte-derived exhausted CD4+ T cells. In human immune cells from healthy donors, IK14004 modulates expression of the T cell receptor α/β subunits, induces Type I IFN expression, stimulates natural killer (NK) cells to express NKG2D/NKp44 receptors and enhances K562 cytotoxicity. In both T and NK cells, IK14004 alters the IL-12 receptor β1/β2 chain ratio to favour IL-12p70 binding. Taken together, this novel peptide offers an opportunity to gain further insight into the complexity of ICI immunotherapy so that autoimmune responses may be minimised without promoting tumour evasion from the immune system.
Collapse
Affiliation(s)
- Michael Agrez
- InterK Peptide Therapeutics Limited, New South Wales, Australia.
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia.
| | | | - Kristofer J Thurecht
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Feifei Liu
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Gayathri Subramaniam
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Christopher B Howard
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Benjamin Blyth
- Department of Oncology,, Peter MacCallum Cancer Centre and Sir Peter MacCallum, University of Melbourne, Melbourne, Australia
| | - Stephen Parker
- InterK Peptide Therapeutics Limited, New South Wales, Australia
| | | | | | - Gavin Knox
- Concept Life Sciences, Edinburgh, Scotland
| | | | | | | | | |
Collapse
|
6
|
Jin H, Zhu J, Xuan R, Zhou Y, Xue B, Yang D, Gao J, Zang Y, Xu L. A Crosstalk Between Castration-Resistant Prostate Cancer Cells, M2 Macrophages, and NK Cells: Role of the ATM-PI3K/AKT-PD-L1 Pathway. Immunol Invest 2023; 52:941-965. [PMID: 37732622 DOI: 10.1080/08820139.2023.2258930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Castration-resistant prostate cancer (CRPC) in males is associated with a poor prognosis and a higher risk of treatment-related adverse effects, with high mortality among cancers globally. It is thus imperative to explore novel potential molecules with dual therapeutic and biomarker functions. Based on the recent research findings, the expression levels of ataxia telangiectasia mutant kinase (ATM) in prostate cancer (PC) tissues collected from CRPC patients were higher than hormone-dependent PC patients. Using CRPC cell lines (C4-2 and CWR22Rv1), the transwell chamber experiments revealed ATM promoted macrophage recruitment in CRPC cells in vitro via C-X-C motif chemokine ligand 12 (CXCL12). Further in vitro investigations demonstrated that polarized macrophages prevented NK cell recruitment and reduced the immunocidal activity of NK cells against CRPC cell lines. Moreover, ATM boosted programmed death receptor ligand 1 (PD-L1) expression while inhibiting NK group 2D (NKG2D) ligand expression in selected cell lines via PI3K/AKT signaling pathway. The in vivo investigations revealed ATM induced proliferation of CRPC and macrophage recruitment, while the NK cell recruitment was found to suppress ATM expression and CRPC proliferation. In conclusion, it could be demonstrated that inhibiting ATM increased the susceptibility of CRPC to NK cell inhibitors by dampening the CXCL12 and PI3K/AKT-PD-L1 pathways, thereby offering a novel and individualized treatment protocol for treating CRPC.
Collapse
Affiliation(s)
- Hongliang Jin
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jin Zhu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Xuan
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yibin Zhou
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Boxin Xue
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dongrong Yang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jie Gao
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yachen Zang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lijun Xu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
7
|
Ochayon DE, DeVore SB, Chang WC, Krishnamurthy D, Seelamneni H, Grashel B, Spagna D, Andorf S, Martin LJ, Biagini JM, Waggoner S, Hershey GKK. Progressive accumulation of hyperinflammatory NKG2D low NK cells in early childhood severe atopic dermatitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.02.23290884. [PMID: 37333102 PMCID: PMC10274972 DOI: 10.1101/2023.06.02.23290884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease that often precedes the development of food allergy, asthma, and allergic rhinitis. The prevailing paradigm holds that a reduced frequency and function of natural killer (NK) cell contributes to AD pathogenesis, yet the underlying mechanisms and contributions of NK cells to allergic co-morbidities remain ill-defined. Herein, analysis of circulating NK cells in a longitudinal early life cohort of children with AD revealed a progressive accumulation of NK cells with low expression of the activating receptor NKG2D, which was linked to more severe AD and sensitivity to allergens. This was most notable in children co-sensitized to food and aero allergens, a risk factor for development of asthma. Individual-level longitudinal analysis in a subset of children revealed co-incident reduction of NKG2D on NK cells with acquired or persistent sensitization, and this was associated with impaired skin barrier function assessed by transepidermal water loss. Low expression of NKG2D on NK cells was paradoxically associated with depressed cytolytic function but exaggerated release of the proinflammatory cytokine TNF-α. These observations provide important insights into a potential mechanism underlying the development of allergic co-morbidity in early life in children with AD which involves altered NK-cell functional responses, and define an endotype of severe AD.
Collapse
|
8
|
A functional mammalian display screen identifies rare antibodies that stimulate NK cell-mediated cytotoxicity. Proc Natl Acad Sci U S A 2021; 118:2104099118. [PMID: 34330834 DOI: 10.1073/pnas.2104099118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Therapies that boost the antitumor immune response have shown a great deal of success. Although most of these therapies have focused on enhancing T cell functions, there is a growing interest in developing therapies that can target other immune cell subsets. Like T cells, natural killer (NK) cells are cytotoxic effector cells that play a key role in the antitumor response. To advance the development of NK-based therapies, we developed a functional screen to rapidly identify antibodies that can activate NK cells. We displayed antibodies on a mammalian target cell line and probed their ability to stimulate NK cell-mediated cytotoxicity. From this screen, we identified five antibodies that bound with high affinity to NK cells and stimulated NK cell-mediated cytotoxicity and interferon-γ (IFN-γ) secretion. We demonstrate that these antibodies can be further developed into bispecific antibodies to redirect NK cell-mediated cytotoxicity toward CD20+ B cell lymphoma cells and HER2+ breast cancer cells. While antibodies to two of the receptors, CD16 and NCR1, have previously been targeted as bispecific antibodies to redirect NK cell-mediated cytotoxicity, we demonstrate that bispecific antibodies targeting NCR3 can also potently activate NK cells. These results show that this screen can be used to directly identify antibodies that can enhance antitumor immune responses.
Collapse
|
9
|
Wijaya RS, Read SA, Schibeci S, Han S, Azardaryany MK, van der Poorten D, Lin R, Yuen L, Lam V, Douglas MW, George J, Ahlenstiel G. Expansion of dysfunctional CD56-CD16+ NK cells in chronic hepatitis B patients. Liver Int 2021; 41:969-981. [PMID: 33411395 DOI: 10.1111/liv.14784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 11/12/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Natural killer (NK) cells are primary innate effector cells that play an important role in the control of human viral infections. During chronic viral infection, NK cells undergo significant changes in phenotype, function and subset distribution, including the appearance of CD56-CD16+ (CD56-) NK cells, previously identified in chronic human immunodeficiency virus (HIV) and hepatitis C virus infection. However, the presence of CD56- NK cells in the pathogenesis of chronic hepatitis B (CHB) remains unknown. METHODS Phenotype and function of CD56- NK cells from patients with CHB (n = 28) were assessed using flow cytometry and in vitro stimulation with HBV antigen. RESULTS CHB patients had a higher frequency of CD56- NK cells compared to healthy controls in peripheral blood (6.2% vs 1.4%, P < .0001). Compared to CD56+ NK cells, CD56- NK cells had increased expression of inhibitory receptors, and reduced expression of activating receptors, as measured by MFI and qPCR. CD56- NK cells were less responsive to target cell and cytokine stimulation compared to their CD56+ counterparts. In addition, CD56- NK cells demonstrated defective dendritic cells (DCs) interactions resulting in reduced DCs maturation, lower expression of NK CD69 and impaired capacity of NK cells to eliminate immature DCs in co-culture studies. Finally, frequency of CD56- NK cells was positively correlated with serum HBV DNA levels. CONCLUSION Chronic HBV infection induces the expansion of highly dysfunctional of CD56- NK cells that likely contribute to inefficient innate and adaptive antiviral immune response in chronic HBV infection.
Collapse
Affiliation(s)
- Ratna S Wijaya
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, Australia.,Faculty of Medicine, Pelita Harapan University, Tangerang, Indonesia
| | - Scott A Read
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, Australia.,Blacktown Clinical School, Western Sydney University, Blacktown, NSW, Australia.,Blacktown Hospital, Blacktown, NSW, Australia
| | - Stephen Schibeci
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, Australia
| | - Shuanglin Han
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, Australia
| | - Mahmoud K Azardaryany
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, Australia
| | | | - Rita Lin
- Westmead Hospital, University of Sydney, Westmead, NSW, Australia
| | - Lawrence Yuen
- Westmead Hospital, University of Sydney, Westmead, NSW, Australia.,Discipline of Surgery, University of Sydney, Westmead, NSW, Australia
| | - Vincent Lam
- Westmead Hospital, University of Sydney, Westmead, NSW, Australia.,Discipline of Surgery, University of Sydney, Westmead, NSW, Australia
| | - Mark W Douglas
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, Australia.,Westmead Hospital, University of Sydney, Westmead, NSW, Australia.,Centre for Infectious Diseases and Microbiology, Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney at Westmead Hospital, Westmead, NSW, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, Australia.,Westmead Hospital, University of Sydney, Westmead, NSW, Australia
| | - Golo Ahlenstiel
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, Australia.,Blacktown Clinical School, Western Sydney University, Blacktown, NSW, Australia.,Blacktown Hospital, Blacktown, NSW, Australia
| |
Collapse
|
10
|
Constitutive expression of ULBP-4 on monocytes regulates NK cell NKG2D expression. Blood Adv 2020; 3:1563-1567. [PMID: 31097432 DOI: 10.1182/bloodadvances.2018025742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 04/15/2019] [Indexed: 12/17/2022] Open
Abstract
Key Points
The NKG2D ligand ULBP-4 is expressed on healthy monocytes. Monocyte ULBP-4 expression regulates NKG2D expression by NK cells.
Collapse
|
11
|
Pestaloamides A and B, two spiro-heterocyclic alkaloid epimers from the plant endophytic fungus Pestalotiopsis sp. HS30. Sci China Chem 2020. [DOI: 10.1007/s11426-020-9762-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
12
|
Natural Killer Cell Hypo-responsiveness in Chronic Lymphocytic Leukemia can be Circumvented In Vitro by Adequate Activating Signaling. Hemasphere 2019; 3:e308. [PMID: 31976482 PMCID: PMC6924557 DOI: 10.1097/hs9.0000000000000308] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by an acquired immune dysfunction, which may underlie the hampered efficacy of cellular immunotherapy. Most data on dampened immune responses in CLL come from studies investigating CLL and T cell interactions. Natural killer (NK) cells may be an attractive alternative source of effector cells in immunotherapy in CLL, provided that functionality is retained within the CLL micro-environment. Despite their important role in anti-tumor responses, NK cells are not extensively characterized in CLL. Here, we studied the expression of activating and inhibitory receptors on CLL-derived and healthy control (HC) NK cells, and their functional response towards several stimuli. NK cells from CLL patients have an increased maturation stage, with an expansion of NKG2C+ NK cells in CMV seropositive individuals. The cytotoxicity receptor NKG2D is downregulated, and the killing capacity through this receptor was markedly reduced in CLL-derived NK cells. In contrast, activation via CD16 (FCγRIII) led to adequate activation and functional responses in CLL-derived NK cells. These findings indicate that NK cells in CLL are not intrinsically defect and still perform effector functions upon adequate activating signaling. Clinical relevance of this finding was shown by treatment with novel nanobody-Fc constructs, which induced cytotoxic responses in both CLL- and HC-derived NK cells via CD16. Our results show that NK cells, in contrast to the T cell compartment, retain their function within the CLL micro-environment, provided that they receive an adequate activating signal. These findings warrant future studies on NK cell mediated immunotherapeutic strategies in CLL.
Collapse
|
13
|
Wijaya RS, Read SA, Schibeci S, Eslam M, Azardaryany MK, El-Khobar K, van der Poorten D, Lin R, Yuen L, Lam V, George J, Douglas MW, Ahlenstiel G. KLRG1+ natural killer cells exert a novel antifibrotic function in chronic hepatitis B. J Hepatol 2019; 71:252-264. [PMID: 30905683 DOI: 10.1016/j.jhep.2019.03.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Natural killer (NK) cells are known to exert strong antiviral activity. Killer cell lectin-like receptor subfamily G member 1 (KLRG1) is expressed by terminally differentiated NK cells and KLRG1-expressing lymphocytes are known to expand following chronic viral infections. We aimed to elucidate the previously unknown role of KLRG1 in the pathogenesis of chronic hepatitis B (CHB). METHODS KLRG1+ NK cells were taken from the blood and liver of healthy individuals and patients with CHB. The phenotype and function of these cells was assessed using flow cytometry and in vitro stimulation. RESULTS Patients with CHB had a higher frequency of KLRG1+ NK cells compared to healthy controls (blood 13.4 vs. 2.3%, p <0.0001 and liver 23.4 vs. 2.6%, p <0.01). KLRG1+ NK cells were less responsive to K562 and cytokine stimulation, but demonstrated enhanced cytotoxicity (9.0 vs. 4.8%, p <0.05) and IFN-γ release (8.0 vs. 1.5%, p <0.05) via antibody dependent cellular cytotoxicity compared to their KLRG1- counterparts. KLRG1+ NK cells possessed a mature phenotype, demonstrating stronger cytolytic activity and IFN-γ secretion against hepatic stellate cells (HSCs) than KLRG1- NK cells. Moreover, KLRG1+ NK cells more effectively induced primary HSC apoptosis in a TRAIL-dependent manner. Increased KLRG1+ NK cell frequency in the liver and blood was associated with lower fibrosis stage (F0/F1) in patients with CHB. Finally, the expression of CD44, degranulation and IFN-γ production were all increased in KLRG1+ NK cells following stimulation with osteopontin, the CD44 ligand, suggesting that HSC-derived osteopontin may cause KLRG1+ NK cell activation. CONCLUSIONS KLRG1+ NK cells likely play an antifibrotic role during the natural course of CHB infection. Harnessing this antifibrotic function may provide a novel therapeutic approach to treat liver fibrosis in patients with CHB. LAY SUMMARY Individuals that are chronically infected with hepatitis B virus (HBV) possess an increased number of immune cells, called natural killer (NK) cells expressing the surface marker KLRG1 in the blood and liver. Here, we demonstrate that these specific NK cells are able to kill activated stellate cells in the liver. Because activated stellate cells contribute to liver scarring, i.e. fibrosis, and subsequent liver dysfunction in individuals with chronic HBV infection, KLRG1+ NK cells are a novel immune cell type that can limit liver scarring.
Collapse
Affiliation(s)
- Ratna S Wijaya
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia; Faculty of Medicine, Pelita Harapan University, Tangerang, Indonesia
| | - Scott A Read
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia; Blacktown Clinical School, Western Sydney University, Blacktown, NSW 2148, Australia
| | - Stephen Schibeci
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Mohammed Eslam
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Mahmoud K Azardaryany
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Korri El-Khobar
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | | | - Rita Lin
- Westmead Hospital, University of Sydney, NSW, Australia
| | - Lawrence Yuen
- Westmead Hospital, University of Sydney, NSW, Australia; Discipline of Surgery, University of Sydney, Australia
| | - Vincent Lam
- Westmead Hospital, University of Sydney, NSW, Australia; Discipline of Surgery, University of Sydney, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia; Westmead Hospital, University of Sydney, NSW, Australia
| | - Mark W Douglas
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia; Westmead Hospital, University of Sydney, NSW, Australia; Centre for Infectious Diseases and Microbiology, Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney at Westmead Hospital, Westmead NSW 2145, Australia
| | - Golo Ahlenstiel
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia; Blacktown Clinical School, Western Sydney University, Blacktown, NSW 2148, Australia; Blacktown Hospital, Blacktown, NSW 2148, Australia.
| |
Collapse
|
14
|
Parvifoline AA Promotes Susceptibility of Hepatocarcinoma to Natural Killer Cell-Mediated Cytolysis by Targeting Peroxiredoxin. Cell Chem Biol 2019; 26:1122-1132.e6. [DOI: 10.1016/j.chembiol.2019.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/24/2019] [Accepted: 04/02/2019] [Indexed: 12/23/2022]
|
15
|
Luo D, Dong XW, Yan B, Liu M, Xue TH, Liu H, You JH, Li F, Wang ZL, Chen ZN. MG132 selectively upregulates MICB through the DNA damage response pathway in A549 cells. Mol Med Rep 2018; 19:213-220. [PMID: 30483783 PMCID: PMC6297755 DOI: 10.3892/mmr.2018.9676] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/23/2018] [Indexed: 12/26/2022] Open
Abstract
Natural killer (NK) cells recognize stress-activated NK group 2, member D (NKG2D) ligands in tumors. In the present study, the expression levels of NKG2D ligands were examined in four lung cancer cell lines (A549, PLA801D, NCI-H157 and NCI-H520). In the A549 cells, the expression of MHC class I polypeptiderelated sequence (MIC)A/B and UL16 binding protein (ULBP)1 was weak, the expression of ULBP2 was typical, and neither ULBP3 nor ULBP4 were expressed. The mechanism underlying the regulatory effect of a cancer treatment agent on the expression of NKG2D ligands was investigated using the proteasome inhibitor MG132. Following treatment for 8 h with MG132, the transcription levels of MICB and ULBP1 were upregulated 10.62- and 11.09-fold, respectively, and the expression levels of MICB and ULBP1 were increased by 68.18 and 23.65%, respectively. Notably, MICB exhibited significant time-dependent change. MG132 increased the transcription of MICB by acting at a site in the 480-bp MICB upstream promoter. The activity of the MICB promoter was upregulated 1.77-fold following treatment with MG132. MG132 treatment improved the cytotoxicity of NK cells, which was partially blocked by an antibody targeting NKG2D, and more specifically the MICB molecule. The expression of MICB induced by MG132 was inhibited by KU-55933 [ataxia telangiectasia mutated (ATM) kinase inhibitor], wortmannin (phosphoinositide 3 kinase inhibitor) and caffeine (ATM/ATM-Rad3-related inhibitor). The phosphorylation of checkpoint kinase 2 (Chk2), an event associated with DNA damage, was observed following treatment with MG132. These results indicated that MG132 selectively upregulates the expression of MICB in A549 cells, and increases the NKG2D-mediated cytotoxicity of NK cells. The regulatory effect of MG132 may be associated with the activation of Chk2, an event associated with DNA damage. The combination of MG132 with NK cell immunotherapy may have a synergistic effect that improves the therapeutic effect of lung cancer treatment.
Collapse
Affiliation(s)
- Dan Luo
- College of Life Science and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Xi-Wen Dong
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Bing Yan
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Mei Liu
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Tian-Hui Xue
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Hui Liu
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Jun-Hao You
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Fang Li
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Zi-Ling Wang
- College of Life Science and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Zhi-Nan Chen
- College of Life Science and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| |
Collapse
|
16
|
Zhu HF, Li Y. Small-Molecule Targets in Tumor Immunotherapy. NATURAL PRODUCTS AND BIOPROSPECTING 2018; 8:297-301. [PMID: 29974338 PMCID: PMC6102179 DOI: 10.1007/s13659-018-0177-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/02/2018] [Indexed: 06/08/2023]
Abstract
Cancer immunotherapy has been widely recognized as a powerful approach to fight cancers. To date, over 50 phase III trials in cancer immunotherapy are in progress. Among the many immunotherapy approaches, immune checkpoint therapy has attracted considerable attention. The reported clinical success of targeting the T cell immune checkpoint receptors PD-1 or CTLA4 by antibodies blockade in advanced stages of cancers has demonstrated the importance of immune modulation. But antibodies-based immunotherapy confronted with some disadvantages, such as immunogenicity, stability, membrane permeability, and production cost. Therefore, alternative approaches including small-molecule-regulated immune response are being introduced. In this review, we focused on some of the key intracellular pathways where small-molecule therapeutic is potential and attractive, which highlights the great potential of natural products in this field.
Collapse
Affiliation(s)
- Hui-Fang Zhu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132# Lanhei Road, Kunming, 650201, Yunnan, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yan Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132# Lanhei Road, Kunming, 650201, Yunnan, People's Republic of China.
| |
Collapse
|
17
|
Shen M, Chen Y, Xu L, Zhu R, Xue X, Tsai Y, Keng PC, Lee SO, Chen Y. Increased infiltration of macrophages to radioresistant lung cancer cells contributes to the development of the additional resistance of tumor cells to the cytotoxic effects of NK cells. Int J Oncol 2018; 53:317-328. [PMID: 29750425 DOI: 10.3892/ijo.2018.4394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 04/02/2018] [Indexed: 11/06/2022] Open
Abstract
In this study, in order to investigate the effects of increased macrophage infiltration to radioresistant lung tumors in regulating natural killer (NK) cell-mediated immunity, we examined whether the treatment of radioresistant cells with conditioned medium (CM) from phorbol myristate acetate (PMA)/interleukin (IL)-4 treated THP-1 cells (used as a tumor-associated macrophage source) leads to the development of the additional resistance of tumor cells to NK cell cytotoxicity. We found that the susceptibility of THP-1 CM-treated radioresistant cells to NK cell cytotoxicity was decreased compared to the non-treated cells. In addition, it was found that such a decreased susceptibility was associated with increased programmed death receptor ligand 1 (PD-L1) and decreased natural killer group 2D (NKG2D) ligand levels in tumor cells. We further discovered that the THP-1 cells secreted a high level of IL-6, and that blocking IL-6 action by the addition of a neutralizing antibody (Ab) for IL-6 into the THP-1 CM decreased the resistance of THP-1 CM-treated radioresistant cells to NK cell cytotoxicity. Moreover, we discovered that MEK/Erk was the most critical IL-6 downstream signaling pathway in triggering the THP-1 CM effect; thus, the addition of MEK/Erk inhibitor to THP-1 CM enhanced the susceptibility of the THP-1 CM-treated radioresistant cells to NK cell cytotoxicity. On the whole, the findings of this study suggest the existence of a malignant loop characterized by increased macrophage infiltration into radioresistant cells which, in turn, promotes the development of the additional resistance of these cells to NK cell cytotoxicity.
Collapse
Affiliation(s)
- Mingjing Shen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yongbing Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Lijun Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Rongying Zhu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xiang Xue
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Ying Tsai
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Peter C Keng
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Soo Ok Lee
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
18
|
Xu L, Shen M, Chen X, Zhu R, Yang DR, Tsai Y, Keng PC, Chen Y, Lee SO. Adipocytes affect castration-resistant prostate cancer cells to develop the resistance to cytotoxic action of NK cells with alterations of PD-L1/NKG2D ligand levels in tumor cells. Prostate 2018; 78:353-364. [PMID: 29330929 DOI: 10.1002/pros.23479] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/14/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Obesity affects prostate cancer (PCa) progression, and the periprostatic adipose tissue adjacent to the prostate is considered a driving force of disease progression. Adipocytes are the main cell population in adipose tissues and their paracrine role contributes to PCa progression, however its implication in modulating immune reactions remains largely unknown. We investigated the adipocyte role in controlling the susceptibility of castration-resistant PCa (CRPC) cells to the cytotoxic action of natural killer (NK) cells. METHODS Using primary NK cells as the NK cell source, NK cell cytotoxicities to CRPC cells, either control media treated or adipocyte-conditioned media (CM) treated, were tested in lactate dehydrogenase (LDH) release-based assays. The levels of programmed death receptor ligand (PD-L1) and NK group 2D (NKG2D) ligands in adipocyte CM-treated CRPC cells were analyzed in qPCR analyses. Effects of blocking adipocyte action on altering PD-L1/NKG2D ligand levels and the susceptibility of CRPC cells to NK cell cytotoxicity were investigated. RESULTS We found NK cell cytotoxicity to CRPC cells decreases when tumor cells are treated with adipocyte CM associated with PD-L1 and NKG2D ligand level alterations. Further, we discovered that the JAK/Stat3 signaling pathway was responsible for the adipocyte CM effect. Two adipokine molecules, IL-6 and leptin, were shown to be important in activation of the JAK/Stat3 signaling in CRPC cells to modulate the PD-L1/NKG2D ligand level alteration. Adding the inhibitors of JAK/Stat3 signaling or neutralizing antibodies of IL-6 or leptin increased the susceptibility of CRPC cells to NK cell action. CONCLUSIONS Blocking the adipocyte effect by inhibiting the IL-6/leptin-JAK/Stat3 signaling axis may enhance NK cell mediated immunity to CRPC cells and this strategy may help to develop future therapeutics to treat obese PCa patients.
Collapse
Affiliation(s)
- Lijun Xu
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
- Dep, artment of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Mingjing Shen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
- Dep, artment of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Xiaodong Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Rongying Zhu
- Dep, artment of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Dong-Rong Yang
- Dep, artment of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Ying Tsai
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Peter C Keng
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Soo Ok Lee
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| |
Collapse
|
19
|
Xu L, Shen M, Chen X, Yang DR, Tsai Y, Keng PC, Lee SO, Chen Y. In vitro-induced M2 type macrophages induces the resistance of prostate cancer cells to cytotoxic action of NK cells. Exp Cell Res 2018; 364:113-123. [PMID: 29408565 DOI: 10.1016/j.yexcr.2018.01.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 01/02/2023]
Abstract
Previous reports, including our experimental results, showed that macrophages migrate to prostate cancer (PCa) cells. We tested whether the migrated macrophages affect the susceptibility of castration-resistant PCa (CRPC) cells to cytotoxic actions of natural killer (NK) cells. We found treatment of tumor cells with the conditioned media (CM) of the PMA/IL-4 treated THP-1 cells (M2 type macrophages) (THP-1 CM) decreased the susceptibility of tumor cells to NK cell cytotoxicity, as a result of increased programmed death receptor ligand 1 (PD-L1) and decreased NK group 2D (NKG2D) ligands in CRPC cells. Meanwhile, the decreased susceptibility of tumor cells was also detected when NK cells were treated with THP-1 CM and used in NK cell cytotoxicity tests. Therefore, we observed higher resistance of CRPC cells when both tumor and NK cells were treated with THP-1 CM than when tumor cells or NK cells were individually treated. We further discovered that the PMA/IL-4 treated THP-1 cells secrete a high level of IL-6, so blocking the IL-6 action significantly decreased the PD-L1 level while recovering the NKG2D ligands, thus increasing the susceptibility of CRPC cells to NK cell action. Moreover, we discovered that JAK-Stat3 is the most critical IL-6 downstream signaling in triggering the THP-1 CM effect. Consequently, we found the susceptibility of CRPC cells to NK cells was increased when either JAK or Stat 3 inhibitor was added when tumor cells were treated with THP-1 CM, and that the best effect was observed when the JAK inhibitor and PD-L1 Ab were added together.
Collapse
Affiliation(s)
- Lijun Xu
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Mingjing Shen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Xiaodong Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Dong-Rong Yang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Ying Tsai
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Peter C Keng
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Soo Ok Lee
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
20
|
Xu L, Chen X, Shen M, Yang DR, Fang L, Weng G, Tsai Y, Keng PC, Chen Y, Lee SO. Inhibition of IL-6-JAK/Stat3 signaling in castration-resistant prostate cancer cells enhances the NK cell-mediated cytotoxicity via alteration of PD-L1/NKG2D ligand levels. Mol Oncol 2018; 12:269-286. [PMID: 28865178 PMCID: PMC5830627 DOI: 10.1002/1878-0261.12135] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 08/04/2017] [Accepted: 08/19/2017] [Indexed: 12/18/2022] Open
Abstract
To investigate whether IL‐6 signaling affects the susceptibility of castration‐resistant prostate cancer (CRPC) cells to cytotoxic action of natural killer (NK) cells, CRPC cell lines (having different IL‐6 levels) were developed by lentiviral transduction. While observing no secreted IL‐6 level in parental C4‐2 and CWR22Rv1 cells, we found the IL‐6 expression/secretion in these cells was induced after the transduction process and the IL‐6 level difference in C4‐2siIL‐6/sc and CWR22siIL‐6/sc cell CRPC cell sets could be detected. We then found that IL‐6‐knockdown cells were more susceptible to NK cell cytotoxicity than control cells due to lowered programmed death receptor ligand 1 (PD‐L1) and increased NK group 2D (NKG2D) ligand levels. In animal studies, to concur with the in vitro results, we found that IL‐6‐expressing cell‐derived tumors were more resistant to NK cell action than the tumors of IL‐6‐knockdown cells. Further, we discovered that JAK‐Stat3 is the most critical IL‐6 downstream signaling that modulates PD‐L1/NKG2D ligand levels in CRPC cells. Furthermore, inhibition of the JAK or Stat3 signaling effectively increased the susceptibility of C4‐2sc and CWRsc cells to NK cell cytotoxicity. We observed the most effective cytotoxicity when the PD‐L1 Ab and JAK inhibitor (or Stat 3 inhibitor) were used together. These results suggest that the strategy of targeting IL‐6 signaling (or its downstream signaling) may enhance the NK cell‐mediated immune action to CRPC tumors, thus yielding clinical implications in developing future immunotherapeutics of exploiting this strategy to treat patients with CRPC.
Collapse
Affiliation(s)
- LiJun Xu
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA.,Department of Urology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - XiaoDong Chen
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA.,Department of Urology, Ningbo Urology and Nephrology Hospital, China
| | - MingJing Shen
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA.,Department of Urology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Dong-Rong Yang
- Department of Urology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Laifu Fang
- Department of Pathology, Ningbo Yin Zhou Hospital, China
| | - Guobin Weng
- Department of Urology, Ningbo Urology and Nephrology Hospital, China
| | - Ying Tsai
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA
| | - Peter C Keng
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA
| | - Yuhchyau Chen
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA
| | - Soo Ok Lee
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA
| |
Collapse
|
21
|
|
22
|
Nakajima T, Okayama H, Ashizawa M, Noda M, Aoto K, Saito M, Monma T, Ohki S, Shibata M, Takenoshita S, Kono K. Augmentation of antibody-dependent cellular cytotoxicity with defucosylated monoclonal antibodies in patients with GI-tract cancer. Oncol Lett 2017; 15:2604-2610. [PMID: 29434980 DOI: 10.3892/ol.2017.7556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/06/2017] [Indexed: 12/27/2022] Open
Abstract
Enhancement of antibody-dependent cellular cytotoxicity (ADCC) with some modalities may be a promising approach to enhance the efficacy of therapeutic monoclonal antibodies (mAbs). It has previously been demonstrated that the removal of fucose from antibody oligosaccharides (defucosylation) leads to augmentation of ADCC activity. To establish clinically relevant evidence of this procedure, the present study evaluated trastuzumab- and cetuximab-mediated ADCC by comparing defucosylated mAbs with conventional mAbs using peripheral blood mononuclear cells (PBMCs). PBMCs were isolated from 20 patients with gastrointestinal tract cancer and 10 healthy volunteers. ADCCs were measured using PBMCs as effector cells and two gastric cancer cell lines as target cells. ADCCs were significantly enhanced with defucosylated mAbs compared with conventional mAbs using PBMC from the healthy donors and patients with cancer. The results confirmed that the cetuximab- and trastuzumab-mediated ADCCs in advanced disease were impaired in comparison to those in early disease or healthy individuals. However, when the defucosylated mAbs were used instead of the conventional mAbs, the ADCC activities in the advanced cases were almost comparable with those in early disease or healthy individuals. Furthermore, the expression of ADCC associated molecules were modified toward immunosuppressive status with a mitogen-activated protein kinase inhibitor in vitro, the conventional cetuximab- and trastuzumab-mediated ADCC was downregulated, and the defucosylated mAbs overcome the downregulation of ADCC. In conclusion, defucosylated therapeutic mAbs may enhance ADCC activities in patients with cancer, which may lead to more effective anti-cancer treatments.
Collapse
Affiliation(s)
- Takahiro Nakajima
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hirokazu Okayama
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Mai Ashizawa
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Masaru Noda
- Department of Breast Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Keita Aoto
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Motonobu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Tomoyuki Monma
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Shinji Ohki
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Masahiko Shibata
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Seiichi Takenoshita
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
23
|
Evaluation of the Functional Capacity of NK Cells of Melanoma Patients in an In Vitro Model of NK Cell Contact with K562 and FemX Tumor Cell Lines. J Membr Biol 2017; 250:507-516. [DOI: 10.1007/s00232-017-9977-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/08/2017] [Indexed: 12/31/2022]
|
24
|
Dubois S, Conlon KC, Müller JR, Hsu-Albert J, Beltran N, Bryant BR, Waldmann TA. IL15 Infusion of Cancer Patients Expands the Subpopulation of Cytotoxic CD56 bright NK Cells and Increases NK-Cell Cytokine Release Capabilities. Cancer Immunol Res 2017; 5:929-938. [PMID: 28842470 DOI: 10.1158/2326-6066.cir-17-0279] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 06/28/2017] [Accepted: 08/21/2017] [Indexed: 11/16/2022]
Abstract
The cytokine IL15 is required for survival and activation of natural killer (NK) cells as well as expansion of NK-cell populations. Here, we compare the effects of continuous IL15 infusions on NK-cell subpopulations in cancer patients. Infusions affected the CD56bright NK-cell subpopulation in that the expansion rates exceeded those of CD56dim NK-cell populations with a 350-fold increase in their total cell numbers compared with 20-fold expansion for the CD56dim subset. CD56bright NK cells responded with increased cytokine release to various stimuli, as expected given their immunoregulatory functions. Moreover, CD56bright NK cells gained the ability to kill various target cells at levels that are typical for CD56dim NK cells. Some increased cytotoxic activities were also observed for CD56dim NK cells. IL15 infusions induced expression changes on the surface of both NK-cell subsets, resulting in a previously undescribed and similar phenotype. These data suggest that IL15 infusions expand and arm CD56bright NK cells that alone or in combination with tumor-targeting antibodies may be useful in the treatment of cancer. Cancer Immunol Res; 5(10); 929-38. ©2017 AACR.
Collapse
Affiliation(s)
- Sigrid Dubois
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| | - Kevin C Conlon
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jürgen R Müller
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jennifer Hsu-Albert
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Nancy Beltran
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Bonita R Bryant
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| |
Collapse
|
25
|
Enhancing NK cell-mediated cytotoxicity to cisplatin-resistant lung cancer cells via MEK/Erk signaling inhibition. Sci Rep 2017; 7:7958. [PMID: 28801607 PMCID: PMC5554231 DOI: 10.1038/s41598-017-08483-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 07/11/2017] [Indexed: 01/15/2023] Open
Abstract
Major progress has been made clinically in inhibiting the programmed death receptor 1 (PD-1)/PD-L1 interaction to enhance T cell-mediated immune function, yet the effectiveness of anti-PD-L1/PD-1 agents in enhancing natural killer (NK) cell’s function remains largely unknown. Susceptibilities of cisplatin-resistant A549CisR and H157CisR cells vs. parental cells to the cytotoxic action of NK cells were examined. We found cisplatin-resistant cells more resistant to NK cell cytotoxicity than parental cells. There were constitutively higher expressions of PD-L1 in A549CisR and H157CisR cells than in parental cells in vitro, as well as in H157CisR cell-derived tumors than H157P cell-derived tumors. In contrast, we observed that the expression of PD-1 in NK cells was induced after co-culture with cisplatin-resistant cells. We also observed increased susceptibility of cisplatin-resistant cells to NK cell cytotoxicity when neutralizing antibody of PD-1 or PD-L1 was added. Further, we found that the NK group 2, member D (NKG2D) ligand levels were lower in A549CisR and H157CisR cells than in parental cells. Meanwhile, we discovered that the MEK/Erk signaling pathway played a significant role in this regulation, and the addition of a MEK/Erk pathway inhibitor significantly enhanced the PD-L1 Ab effect in enhancing NK cell cytotoxicity to cisplatin-resistant cells.
Collapse
|
26
|
Yin X, Lu X, Xiuwen Z, Min Z, Xiao R, Mao Z, Zhang Q. Role of NKG2D in cytokine-induced killer cells against lung cancer. Oncol Lett 2017; 13:3139-3143. [PMID: 28529563 DOI: 10.3892/ol.2017.5800] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 01/19/2017] [Indexed: 01/07/2023] Open
Abstract
It has been previously demonstrated that cytokine-induced killer (CIK) cells possess potent cytotoxicity against various cancer cells, including lung cancer cells. However, the mechanism by which CIK cells recognize lung cancer cells has not been understood. The interaction between killer cell lectin like receptor K1 (NKG2D) receptor and NKG2D ligands was demonstrated to serve an important role in target cell killing by natural killer cells. The present study investigated whether NKG2D receptor and NKG2D ligand interactions are involved in the CIK-directed killing of lung cancer cells. The expression of MICA and ULBP2 was detected in tumor and healthy tissue samples. The expression of MICA and ULBP2 in tumor tissue samples was higher compared with that in the healthy control tissue. The expression of NKG2D ligands was analyzed in A549 and Q56 cells through reverse transcription-quantitative polymerase chain reaction and flow cytometry. The results demonstrated that the lung cancer cell lines markedly expressed the NKG2D ligands. Furthermore, NKG2D ligand-expressing lung cancer cells were targeted by CIK cells, which was partially blocked by treating CIK cells with an antibody against NKG2D. The data of the current study has demonstrated that the NKG2D-NKG2D ligand interaction serves an essential role in mediating lung cancer cell killing by CIK cells.
Collapse
Affiliation(s)
- Xiaowei Yin
- Department of Respiratory Medicine, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Xuzhang Lu
- Department of Hematology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Zhang Xiuwen
- Department of Hematology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Zhou Min
- Department of Hematology, Changzhou No. 3 People's Hospital, Changzhou, Jiangsu 213000, P.R. China
| | - Rong Xiao
- Department of Hematology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Zhengdao Mao
- Department of Respiratory Medicine, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Qian Zhang
- Department of Respiratory Medicine, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
27
|
Shiraishi K, Mimura K, Kua LF, Koh V, Siang LK, Nakajima S, Fujii H, Shabbir A, Yong WP, So J, Takenoshita S, Kono K. Inhibition of MMP activity can restore NKG2D ligand expression in gastric cancer, leading to improved NK cell susceptibility. J Gastroenterol 2016; 51:1101-1111. [PMID: 27002316 DOI: 10.1007/s00535-016-1197-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 03/14/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND METHODS Natural killer (NK) cells can react with tumor cells through the balance of inhibitory and stimulatory signals between NK cell surface receptors and their ligands, such as MHC class I chain-related A (MICA), MHC class I chain-related B (MICB), and several UL16-binding proteins (ULBPs). In the present study, we evaluated the relationship between NKG2D ligand expression and matrix metalloproteinase (MMP) activity in in vitro culture systems of a panel of gastric cancer cell lines (n = 10) and clinical samples (n = 102). RESULTS First, the surface expression of NK group 2 member D (NKG2D) ligands (MICA, MICB, ULBP-2, and ULBP-3) on tumor cells was markedly downregulated on in vitro culture, in parallel to the upregulation of MMPs analyzed by gelatin zymography and gene expression microarray, whereas the transcript levels of NKG2D ligands remained unchanged on in vitro culture. Second, MMP-specific inhibitors could restore the downregulated expression of NKG2D ligands and functionally improve susceptibilities to NK cells in vitro. Third, the production of soluble NKG2D ligands was increased on in vitro culture and was inhibited by MMP-specific inhibitors. Finally, there was a significant inverse correlation between MMP-9 expression and NKG2D ligand expression as analyzed by immunohistochemistry in clinical tumor samples. CONCLUSION The present study is a comprehensive study demonstrating that upregulation of MMP activity can induce a downregulation of expression of NKG2D ligands in gastric cancer cells, leading to lower-level susceptibility to NK cells.
Collapse
Affiliation(s)
- Kensuke Shiraishi
- Department of Surgery, National University of Singapore, Singapore, Singapore
| | - Kousaku Mimura
- Department of Surgery, National University of Singapore, Singapore, Singapore
| | - Ley-Fang Kua
- Department of Hematology-Oncology, National University of Singapore, Singapore, Singapore
| | - Vivien Koh
- Department of Hematology-Oncology, National University of Singapore, Singapore, Singapore
| | - Lim Kee Siang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Shotaro Nakajima
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Hideki Fujii
- First Department of Surgery, University of Yamanashi, Kofu, Japan
| | - Asim Shabbir
- Department of Surgery, National University of Singapore, Singapore, Singapore
| | - Wei-Peng Yong
- Department of Hematology-Oncology, National University of Singapore, Singapore, Singapore
| | - Jimmy So
- Department of Surgery, National University of Singapore, Singapore, Singapore
| | - Seiichi Takenoshita
- Department of Advanced Cancer Immunotherapy, Fukushima Medical University, 1 Hikarigaoka, Fukushima City, 960-1295, Japan
| | - Koji Kono
- Department of Surgery, National University of Singapore, Singapore, Singapore. .,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,Department of Advanced Cancer Immunotherapy, Fukushima Medical University, 1 Hikarigaoka, Fukushima City, 960-1295, Japan. .,Department of Organ Regulatory Surgery, Fukushima Medical University, 1 Hikarigaoka, Fukushima City, 960-1295, Japan.
| |
Collapse
|
28
|
Ferreira-Teixeira M, Paiva-Oliveira D, Parada B, Alves V, Sousa V, Chijioke O, Münz C, Reis F, Rodrigues-Santos P, Gomes C. Natural killer cell-based adoptive immunotherapy eradicates and drives differentiation of chemoresistant bladder cancer stem-like cells. BMC Med 2016; 14:163. [PMID: 27769244 PMCID: PMC5075212 DOI: 10.1186/s12916-016-0715-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/06/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND High-grade non-muscle invasive bladder cancer (NMIBC) has a high risk of recurrence and progression to muscle-invasive forms, which seems to be largely related to the presence of tumorigenic stem-like cell populations that are refractory to conventional therapies. Here, we evaluated the therapeutic potential of Natural Killer (NK) cell-based adoptive immunotherapy against chemoresistant bladder cancer stem-like cells (CSCs) in a pre-clinical relevant model, using NK cells from healthy donors and NMIBC patients. METHODS Cytokine-activated NK cells from healthy donors and from high-grade NMIBC patients were phenotypically characterized and assayed in vitro against stem-like and bulk differentiated bladder cancer cells. Stem-like cells were isolated from two bladder cancer cell lines using the sphere-forming assay. The in vivo therapeutic efficacy was evaluated in mice bearing a CSC-induced orthotopic bladder cancer. Animals were treated by intravesical instillation of interleukin-activated NK cells. Tumor response was evaluated longitudinally by non-invasive bioluminescence imaging. RESULTS NK cells from healthy donors upon activation with IL-2 and IL-15 kills indiscriminately both stem-like and differentiated tumor cells via stress ligand recognition. In addition to cell killing, NK cells shifted CSCs towards a more differentiated phenotype, rendering them more susceptible to cisplatin, highlighting the benefits of a possible combined therapy. On the contrary, NK cells from NMIBC patients displayed a low density on NK cytotoxicity receptors, adhesion molecules and a more immature phenotype, losing their ability to kill and drive differentiation of CSCs. The local administration, via the transurethral route, of activated NK cells from healthy donors provides an efficient tumor infiltration and a subsequent robust tumoricidal activity against bladder cancer with high selective cytolytic activity against CSCs, leading to a dramatic reduction in tumor burden from 80 % to complete remission. CONCLUSION Although pre-clinical, our results strongly suggest that an immunotherapeutic strategy using allogeneic activated NK cells from healthy donors is effective and should be exploited as a complementary therapeutic strategy in high-risk NMIBC patients to prevent tumor recurrence and progression.
Collapse
Affiliation(s)
- Margarida Ferreira-Teixeira
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Daniela Paiva-Oliveira
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Belmiro Parada
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Urology and Renal Transplantation Department, Coimbra University Hospital Centre (CHUC), Coimbra, Portugal
| | - Vera Alves
- Institute of Immunology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Vitor Sousa
- Service of Anatomical Pathology, Coimbra University Hospital Centre (CHUC), Coimbra, Portugal.,Institute of Anatomical and Molecular Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Obinna Chijioke
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Flávio Reis
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Paulo Rodrigues-Santos
- Institute of Immunology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Immunology and Oncology Laboratory, Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Célia Gomes
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, Coimbra, Portugal. .,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
29
|
Mittica G, Capellero S, Genta S, Cagnazzo C, Aglietta M, Sangiolo D, Valabrega G. Adoptive immunotherapy against ovarian cancer. J Ovarian Res 2016; 9:30. [PMID: 27188274 PMCID: PMC4869278 DOI: 10.1186/s13048-016-0236-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 04/21/2016] [Indexed: 01/16/2023] Open
Abstract
The standard front-line therapy for epithelial ovarian cancer (EOC) is combination of debulking surgery and platinum-based chemotherapy. Nevertheless, the majority of patients experience disease recurrence. Although extensive efforts to find new therapeutic options, cancer cells invariably develop drug resistance and disease progression. New therapeutic strategies are needed to improve prognosis of patients with advanced EOC. Recently, several preclinical and clinical studies investigated feasibility and activity of adoptive immunotherapy in EOC. Our aim is to highlight prospective of adoptive immunotherapy in EOC, focusing on HLA-restricted Tumor Infiltrating Lymphocytes (TILs), and MHC-independent immune effectors such as natural killer (NK), and cytokine-induced killer (CIK). Adoptive cell therapy (ACT) has shown activity in several pre-clinical models. Available preclinical and clinical data suggest that adoptive cell therapy may provide the best benefit in settings of low tumor burden, minimal residual disease, or maintenance therapy. Further studies are needed to better define the optimal clinical setting.
Collapse
Affiliation(s)
- Gloria Mittica
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | - Sofia Genta
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | - Massimo Aglietta
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Dario Sangiolo
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Giorgio Valabrega
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy. .,Department of Oncology, University of Torino, Turin, Italy. .,Division of Medical Oncology-1, Candiolo Cancer Institute- FPO- IRCCS, Strada Provinciale 142 km 3.95, Candiolo, 10060, Turin, Italy.
| |
Collapse
|
30
|
Jia HY, Liu JL, Yuan MZ, Zhou CJ, Sun WD, Zhao JJ, Wang J, Liu L, Luan Y. Regulation Roles of MICA and NKG2D in Human Renal Cancer Cells. Asian Pac J Cancer Prev 2016; 16:3901-5. [PMID: 25987057 DOI: 10.7314/apjcp.2015.16.9.3901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Our aim was to investigation the roles of MHC class I chain-related gene A(MICA) and natural killer cell group 2D(NKG2D) in human renal cancer cells. MATERIALS AND METHODS The expression of membrane MICA (mMICA) on renal cells and NKG2D on NK cells were detected by flow cytometry (FCM); the content of sMICA were detected by enzyme linked immunosorbent assay (ELISA) and the distribution of mMICA on renal tumor tissues by immunohistochemistry; the interaction between MICA and NKG2D was observed by antibody closed method. RESULTS Our results showed that the expression of mMICA in renal cancer tissues was significantly higher than in controls, where the soluble MICA was not expressed. Cytotoxic activity of NK cells was significantly reduced after exposure to NKG2D and MICA antibodies (P<0.05), and serum containing sMICA can obviously lower the function of NKG2D (P<0.05). CONCLUSIONS The interaction of mMICA and NKG2D play important roles in mediation of cytotoxicity of NK cells in RCC. On the other hand, sMICA may mediate tumor immune escape through down- regulated NKG2D expression.
Collapse
Affiliation(s)
- Hong-Ying Jia
- Clinical Molecular Biology Laboratory, the Second Hospital of Shandong University, Jinan, Shandong Province, China E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Affandi JS, Montgomery J, Lee S, Price P. HIV patients stable on ART retain evidence of a high CMV load but changes to Natural Killer cell phenotypes reflect both HIV and CMV. AIDS Res Ther 2015; 12:41. [PMID: 26664457 PMCID: PMC4673723 DOI: 10.1186/s12981-015-0080-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/11/2015] [Indexed: 12/03/2022] Open
Abstract
Background Whilst ART corrects many effects of HIV disease, T cell populations retain features of accelerated immunological aging. Methods Here we analyse phenotypic changes to natural killer (NK) cells in HIV patients who began ART with <200 CD4 T-cells/µl and maintained virological control for 12–17 years, compared with CMV seropositive and seronegative healthy control donors. Results Humoral responses to CMV antigens (lysate, gB, IE-1) remain elevated in the patients (P < 0.0001) despite the long duration of ART. Patient’s NK cells responded poorly to K562 cells when assessed by CD107a and IFNγ, but this could not be attributed to CMV as responses were low in CMV-seronegative controls. Moreover HIV (and not CMV) increased expression of CD57 on CD56lo cells. Conclusions Comparisons with published studies suggest that CMV accelerates age-related increases in CD57 expression but levels plateau by 60–70 years of age, so the effect of CMV disappears. In HIV patients the plateau is higher and perhaps reached sooner. Electronic supplementary material The online version of this article (doi:10.1186/s12981-015-0080-9) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Kim JH, Lee JK. Sesamolin enhances NK cell lysis activity by increasing the expression of NKG2D ligands on Burkitt's lymphoma cells. Int Immunopharmacol 2015; 28:977-84. [PMID: 26298637 DOI: 10.1016/j.intimp.2015.08.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/01/2015] [Accepted: 08/11/2015] [Indexed: 12/28/2022]
Abstract
Sesamolin and sesamin are representative lignans found in sesame seed. The present study was designed to demonstrate the anti-cancer activity of sesamolin achieved by increasing the expression level of NKG2D ligands on Raji cells, which are derived from Burkitt's lymphoma. The anti-cancer activity of sesamolin was also compared with that of sesamin. The cytolysis activity of NK cells against Raji was elevated by the pretreatment of sesamolin on Raji, but not by sesamin. We found that higher NKG2D ligand expression increased the sensitivity of sesamolin-treated Raji to NK cell lysis, resulting from a more active ERK signaling pathway. Our results provide evidence that targeting the ERK signaling pathway may enhance the antitumor activity of lignans and that there is a potential immunotherapeutic value for cancer treatment.
Collapse
Affiliation(s)
- Jeong Hwa Kim
- Department of Biology Education, College of Education, Chungbuk National University, Chungbuk 361-763, Republic of Korea; College of Pharmacy, Chungbuk National University, Chungbuk 361-763, Republic of Korea
| | - Jae Kwon Lee
- Department of Biology Education, College of Education, Chungbuk National University, Chungbuk 361-763, Republic of Korea.
| |
Collapse
|
33
|
Mattiola I, Pesant M, Tentorio PF, Molgora M, Marcenaro E, Lugli E, Locati M, Mavilio D. Priming of Human Resting NK Cells by Autologous M1 Macrophages via the Engagement of IL-1β, IFN-β, and IL-15 Pathways. THE JOURNAL OF IMMUNOLOGY 2015; 195:2818-28. [PMID: 26276870 DOI: 10.4049/jimmunol.1500325] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/19/2015] [Indexed: 12/30/2022]
Abstract
The cross talk between NK cells and macrophages is emerging as a major line of defense against microbial infections and tumors. This study reveals a complex network of soluble mediators and cell-to-cell interactions allowing human classically activated (M1) macrophages, but not resting (M0) or alternatively activated (M2) macrophages, to prime resting autologous NK cells. In this article, we show that M1 increase NK cell cytotoxicity by IL-23 and IFN-β-dependent upregulation of NKG2D, IL-1β-dependent upregulation of NKp44, and trans-presentation of IL-15. Moreover, both IFN-β-dependent cis-presentation of IL-15 on NK cells and engagement of the 2B4-CD48 pathway are used by M1 to trigger NK cell production of IFN-γ. The disclosure of these synergic cellular mechanisms regulating the M1-NK cell cross talk provides novel insights to better understand the role of innate immune responses in the physiopathology of tumor biology and microbial infections.
Collapse
Affiliation(s)
- Irene Mattiola
- Leukocyte Biology Unit, Humanitas Clinical and Research Center, I-20089 Rozzano, Milan, Italy; Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, I-20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, I-20089 Rozzano, Milan, Italy; and
| | - Matthieu Pesant
- Leukocyte Biology Unit, Humanitas Clinical and Research Center, I-20089 Rozzano, Milan, Italy
| | - Paolo F Tentorio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, I-20089 Rozzano, Milan, Italy
| | - Martina Molgora
- Leukocyte Biology Unit, Humanitas Clinical and Research Center, I-20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, I-20089 Rozzano, Milan, Italy; and
| | - Emanuela Marcenaro
- Dipartimento di Medicina Sperimentale and Centro di Eccellenza per le Ricerche Biomediche, Università degli Studi di Genova, I-16132 Genoa, Italy
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, I-20089 Rozzano, Milan, Italy
| | - Massimo Locati
- Leukocyte Biology Unit, Humanitas Clinical and Research Center, I-20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, I-20089 Rozzano, Milan, Italy; and
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, I-20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, I-20089 Rozzano, Milan, Italy; and
| |
Collapse
|
34
|
Collery P, Mohsen A, Kermagoret A, Corre S, Bastian G, Tomas A, Wei M, Santoni F, Guerra N, Desmaële D, d’Angelo J. Antitumor activity of a rhenium (I)-diselenoether complex in experimental models of human breast cancer. Invest New Drugs 2015; 33:848-60. [PMID: 26108551 PMCID: PMC4491361 DOI: 10.1007/s10637-015-0265-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/17/2015] [Indexed: 12/24/2022]
Abstract
Rhenium (I)-diselenother (Re-diselenoether) is a water soluble metal-based compound, combining one atom of rhenium and two atoms of selenium. This compound has been reported to exhibit marked activities against several solid tumor cell lines. We now disclose an improved synthesis of this complex. The Re-diselenoether showed a potent inhibitory effect on MDA-MB231 cell division in vitro, which lasted when the complex was no longer present in the culture. Re-diselenoether induced a remarkable reduction of the volume of the primitive breast tumors and of the pulmonary metastases without clinical signs of toxicity, in mice-bearing a MDA-MB231 Luc+ tumor, orthotopically transplanted, after a daily oral administration at the dose of 10 mg/kg/d. Interestingly, an antagonism was observed when cisplatin was administered as a single i.p. injection 1 week after the end of the Re-diselenoether administration. In an effort to gain insight of the mechanisms of action of Re-diselenoether complex, interaction with 9-methylguanine as a nucleic acid base model was studied. We have shown that Re-diselenoether gave both mono- and bis-guanine Re adducts, the species assumed to be responsible for the DNA intrastrand lesions.
Collapse
Affiliation(s)
- Philippe Collery
- />Société de Coordination de Recherches Thérapeutiques, Algajola, France
| | - Ahmed Mohsen
- />Faculté de Pharmacie, Université Paris-Sud, Institut Galien, UMR CNRS 8612, Chatenay-Malabry, France
| | - Anthony Kermagoret
- />Faculté de Pharmacie, Université Paris-Sud, UMR CNRS 8076 BIOCIS, Chatenay-Malabry, France
| | - Samantha Corre
- />Department of Life Science, Imperial College of London, London, UK
| | - Gérard Bastian
- />Département de Pharmacologie, Centre Hospitalier Universitaire Pitié-Salpêtrière, Paris, France
| | - Alain Tomas
- />Laboratoire de Cristallographie et RMN, Faculté de Pharmacie, UMR CNRS 8015, Université Paris Descartes, Paris, France
| | - Ming Wei
- />Laboratoire Cellvax, Ecole Vétérinaire Nationale d’Alfort, Maisons Alfort, France
| | - François Santoni
- />Laboratoire de l’Office d’Equipement Hydraulique de Corse, Bastia, France
| | - Nadia Guerra
- />Department of Life Science, Imperial College of London, London, UK
| | - Didier Desmaële
- />Faculté de Pharmacie, Université Paris-Sud, Institut Galien, UMR CNRS 8612, Chatenay-Malabry, France
| | - Jean d’Angelo
- />Faculté de Pharmacie, Université Paris-Sud, UMR CNRS 8076 BIOCIS, Chatenay-Malabry, France
| |
Collapse
|
35
|
Naringenin enhances NK cell lysis activity by increasing the expression of NKG2D ligands on Burkitt's lymphoma cells. Arch Pharm Res 2015; 38:2042-8. [PMID: 26100136 DOI: 10.1007/s12272-015-0624-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/10/2015] [Indexed: 10/23/2022]
Abstract
Natural killer (NK) cells are capable of identifying and killing tumor cells as well as virus infected cells without pre-sensitization. NK cells express activating and inhibitory receptors, and can distinguish between normal and tumor cells. The present study was designed to demonstrate the importance of the expression level of NKG2D ligands on the Burkitt's lymphoma cell line, Raji, in enhancing NK cell cytolytic activity. Various flavonoids were used as stimulants to enhance the expression of NKG2D ligands. NK cell lysis activity against Raji was not changed by pre-treatment of Raji with luteolin, kaempferol, taxifolin and hesperetin. However, treatment of Raji with naringenin showed increased sensitivity to NK cell lysis than untreated control cells. The activity of naringenin was due to enhanced NKG2D ligand expression. These results provide evidence that narigenin's antitumor activity may be due to targeting of NKG2D ligand expression and suggests a possible immunotherapeutic role for cancer treatment.
Collapse
|
36
|
Bae DS, Lee JK. Development of NK cell expansion methods using feeder cells from human myelogenous leukemia cell line. Blood Res 2014; 49:154-61. [PMID: 25325034 PMCID: PMC4188780 DOI: 10.5045/br.2014.49.3.154] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/19/2014] [Accepted: 08/27/2014] [Indexed: 11/17/2022] Open
Abstract
Background Natural killer (NK) cells constantly survey surrounding tissues and remove newly generated cancer cells, independent of cancer antigen recognition. Although there have been a number of attempts to apply NK cells for cancer therapy, clinical application has been somewhat limited because of the difficulty in preparing a sufficient number of NK cells. Therefore, ex vivo NK cell expansion is one of the important steps for developing NK cell therapeutics. Methods CD3+ depleted lymphocytes were cocultured with IL-2 and with feeder cells (peripheral blood mononuclear cells [PBMCs], K562, and Jurkat) for 15 days. Expanded NK cells were tested for cytotoxicity against cancer cell lines. Results We compared feeder activities of three different cells-PBMC, K562, and Jurkat. K562 expanded NK cells by almost 20 fold and also showed powerful cytotoxic activity against cancer cells. K562-NK cells remarkably expressed the NK cell activation receptors, NKG2D, and DNAM-1. K562-NK cells exhibited more than two-fold production of cytotoxic granules compared with Jurkat-NK cells, producing more perforin and granzyme B than naïve NK cells. Conclusion Our findings suggest that K562 are more efficient feeder cells than Jurkat or PBMCs. K562 feeder cells expanded NK cells by almost 20 fold and showed powerful cytotoxic activity against cancer cells. We herein propose an intriguing approach for a design of NK cell expansion.
Collapse
Affiliation(s)
- Duk Seong Bae
- Department of Biology Education, College of Education, Chungbuk National University, Cheongju, Korea
| | - Jae Kwon Lee
- Department of Biology Education, College of Education, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
37
|
Robinet P, Baychelier F, Fontaine T, Picard C, Debré P, Vieillard V, Latgé JP, Elbim C. A polysaccharide virulence factor of a human fungal pathogen induces neutrophil apoptosis via NK cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:5332-42. [PMID: 24790151 DOI: 10.4049/jimmunol.1303180] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Aspergillus fumigatus is an opportunistic human fungal pathogen that sheds galactosaminogalactan (GG) into the environment. Polymorphonuclear neutrophils (PMNs) and NK cells are both part of the first line of defense against pathogens. We recently reported that GG induces PMN apoptosis. In this study, we show that PMN apoptosis occurs via a new NK cell-dependent mechanism. Reactive oxygen species, induced by the presence of GG, play an indispensable role in this apoptotic effect by increasing MHC class I chain-related molecule A expression at the PMN surface. This increased expression enables interaction between MHC class I chain-related molecule A and NKG2D, leading to NK cell activation, which in turn generates a Fas-dependent apoptosis-promoting signal in PMNs. Taken together, our results demonstrate that the crosstalk between PMNs and NK cells is essential to GG-induced PMN apoptosis. NK cells might thus play a role in the induction of PMN apoptosis in situations such as unexplained neutropenia or autoimmune diseases.
Collapse
Affiliation(s)
- Pauline Robinet
- Université Pierre et Marie Curie Paris 06, Unité Mixte de Recherche-S CR7, Sorbonne Université, F-75005 Paris, France; INSERM, Centre d'Immunologie et des Maladies Infectieuses, Unité Mixte de Recherche-S CR7, INSERM U1135, F-75013 Paris, France
| | - Florence Baychelier
- INSERM, Centre d'Immunologie et des Maladies Infectieuses, Unité Mixte de Recherche-S CR7, INSERM U1135, F-75013 Paris, France
| | | | - Capucine Picard
- Centre D'étude des Déficits Immunitaires, Assistance Publique-Hôpitaux de Paris, L'hôpital Necker - Enfants Malades, 75743 Paris, France; Paris Descartes University, Sorbonne Paris Cité, 75006 Paris, France; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U980, Necker Medical School, 75015 Paris, France; and
| | - Patrice Debré
- Université Pierre et Marie Curie Paris 06, Unité Mixte de Recherche-S CR7, Sorbonne Université, F-75005 Paris, France; INSERM, Centre d'Immunologie et des Maladies Infectieuses, Unité Mixte de Recherche-S CR7, INSERM U1135, F-75013 Paris, France; Département d'Immunologie, Assistance Publique - Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Vincent Vieillard
- INSERM, Centre d'Immunologie et des Maladies Infectieuses, Unité Mixte de Recherche-S CR7, INSERM U1135, F-75013 Paris, France
| | - Jean-Paul Latgé
- Institut Pasteur, Unité des Aspergillus, 75015 Paris, France
| | - Carole Elbim
- Université Pierre et Marie Curie Paris 06, Unité Mixte de Recherche-S CR7, Sorbonne Université, F-75005 Paris, France; INSERM, Centre d'Immunologie et des Maladies Infectieuses, Unité Mixte de Recherche-S CR7, INSERM U1135, F-75013 Paris, France;
| |
Collapse
|
38
|
Mimura K, Kamiya T, Shiraishi K, Kua LF, Shabbir A, So J, Yong WP, Suzuki Y, Yoshimoto Y, Nakano T, Fujii H, Campana D, Kono K. Therapeutic potential of highly cytotoxic natural killer cells for gastric cancer. Int J Cancer 2014; 135:1390-8. [PMID: 24615495 DOI: 10.1002/ijc.28780] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/16/2014] [Accepted: 01/27/2014] [Indexed: 01/12/2023]
Abstract
To develop more effective therapies for patients with advanced gastric cancer, we examined the potential of ex vivo expanded natural killer (NK) cells. We assessed the expression of ligands for NK Group 2 Member D (NKG2D, an important NK activation molecule) in primary tumors from 102 patients with gastric cancer by immunohistochemistry and determined their prognostic value. We then examined the in vitro and in vivo cytotoxicity of NK cells from healthy donors and patients with gastric cancer. The cytotoxicity of resting and of interleukin (IL)-2-activated NK cells was compared to that of NK cells expanded for 7 days by coculture with the K562-mb15-4.1BBL cell line. As a result, the expression of NKG2D ligands in primary tumors was correlated with favorable presenting features and outcomes, suggesting that gastric cancer may be sensitive to NK cell cytotoxicity. Although resting NK cells showed minimal cytotoxicity against gastric cancer cells, K562-mb15-4.1BBL-expanded NK cells were highly cytotoxic and significantly more powerful than IL-2-activated NK cells. Cytotoxicity was correlated with NKG2D ligand expression and could be modulated by mitogen-activated protein kinase and AKT-PI3 kinase inhibitors. The cytotoxicity of expanded NK cells against HER2-positive gastric cancer cells could be increased by Herceptin and further augmented by Lapatinib. Finally, expanded NK cells exhibited strong antitumor activity in immunodeficient mice engrafted with a gastric cancer cell line. In conclusion, gastric cancer tumors express NKG2D ligands and are highly susceptible to killing by NK cells stimulated by K562-mb15-4.1BBL. These results provide a strong rationale for clinical testing of these NK cells in patients and suggest their use to augment the effects of antibody therapy.
Collapse
Affiliation(s)
- Kousaku Mimura
- Department of Surgery, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hepatitis C virus impairs natural killer cell-mediated augmentation of complement synthesis. J Virol 2013; 88:2564-71. [PMID: 24352446 DOI: 10.1128/jvi.02988-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Natural killer (NK) cells and the complement system play critical roles in the first line of defense against pathogens. The synthesis of complement components C4 and C3 is transcriptionally downregulated by hepatitis C virus (HCV) core and NS5A proteins, and this negative regulation is apparent in chronically HCV-infected patients. In this study, we have examined the potential contribution of an NK cell line as a model in regulating complement synthesis. Coculture of NK cells (NK3.3) with human hepatoma cells (Huh7.5) expressing HCV core or NS5A protein led to a significant increase in C4 and C3 complement synthesis via enhanced specific transcription factors. Reestablishment of complement protein expression was found to be mediated by direct interaction between NKG2D on NK cells and the hepatocyte protein major histocompatibility complex class I-related chains A and B (MICA/B) and not to be associated with specific cytokine signaling events. On the other hand, C4 and C3 synthesis remained impaired in a coculture of NK cells and Huh7.5 cells infected with cell culture-grown HCV. The association between these two cell types through NKG2D and MICA/B was examined further, with MICA/B expression in HCV-infected hepatocytes found to remain inhibited during coculture. Further experiments revealed that the HCV NS2 and NS5B proteins are responsible for the HCV-associated decrease in MICA/B. These results suggest that HCV disables a key receptor ligand in infected hepatoma cells, thereby inhibiting the ability of infected cells to respond to stimuli from NK cells to positively regulate complement synthesis. IMPORTANCE The complement system contributes to the protection of the host from virus infection. However, the involvement of complement in viral hepatitis has not been well documented. Whether NK cells affect complement component expression in HCV-infected hepatocytes remains unknown. Here, we have shown how HCV subverts the ability of NK cells to positively mediate complement protein expression.
Collapse
|
40
|
Fernández L, Portugal R, Valentín J, Martín R, Maxwell H, González-Vicent M, Díaz MÁ, de Prada I, Pérez-Martínez A. In vitro Natural Killer Cell Immunotherapy for Medulloblastoma. Front Oncol 2013; 3:94. [PMID: 23626949 PMCID: PMC3630393 DOI: 10.3389/fonc.2013.00094] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 04/05/2013] [Indexed: 11/17/2022] Open
Abstract
How the immune system attacks medulloblastoma (MB) tumors effectively is unclear, although natural killer (NK) cells play an important role in immune defense against tumor cells. Interactions between receptors on NK cells and ligands expressed by tumor cells are critical for tumor control by immunotherapy. In this study, we analyzed tumor samples from 54 MB patients for expression of major histocompatibility complex class I-related chains A (MICA) and UL16 binding protein (ULPB-2), which are ligands for the NK group 2 member D activatory receptor (NKG2D). The percentage of MICA and ULBP-2 positive cells was higher than 25% in 68% and 6% of MB patients, respectively. A moderate-high intensity of MICA cytoplasmic staining was observed in 46% MB patients and weak ULBP-2 staining was observed in 8% MB patients. No correlation between MICA/ULBP-2 expression and patient outcome was found. We observed that HTB-186, a MB cell line, was moderately resistant to NK cell cytotoxicity in vitro. Blocking MICA/ULBP-2 on HTB-186, and NKG2D receptor on NK cells increased resistance to NK cell lysis in vitro. However, HLA class I blocking on HTB-186 and overnight incubation with IL-15 stimulated NK cells efficiently killed tumor cells in vitro. We conclude that although NKG2D/MICA-ULBP-2 interactions have a role in NK cell cytotoxicity against MB, high expression of HLA class I can protect MB from NK cell cytotoxicity. Even so, our in vitro data indicate that if NK cells are appropriately stimulated, they may have the potential to target MB in vivo.
Collapse
Affiliation(s)
- Lucia Fernández
- Department of Hemato-Oncology and Stem Cell Transplantation, Hospital Infantil Universitario Niño Jesús Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|