251
|
Li Z, Chi Z, Ang WX, Chen C, Tay JC, Ng YY, Xu X, Wang J, Zhu J, Wang S. Experimental treatment of colorectal cancer in mice with human T cells electroporated with NKG2D RNA CAR. Immunotherapy 2020; 12:733-748. [PMID: 32571133 DOI: 10.2217/imt-2019-0137] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aim: Peritoneal metastasis is often present in end-stage neoplastic diseases, including recurrent colorectal cancer and is associated with decreased overall survival. Novel methods are needed. Materials & methods: We constructed first-, second- and third-generation chimeric antigen receptors (CARs) specific for NKG2D ligands and modified human T cells with mRNA electroporation. Results: NKG2D CAR expression was detectable for at least 6 days postelectroporation and mediated efficient cytotoxicity against NKG2DL+ tumor cells, but not NKG2DL-cells. Multiple infusions of the first-generation CAR-T cells into immunodeficient mice bearing established peritoneal colorectal xenografts led to significantly reduced tumor burden. Conclusion: mRNA CAR is an economical way to test new CARs and potentiates controlling on-target/off-tumor toxicity and cytokine storms. The use of NKG2D RNA CARs to treat colorectal peritoneal metastasis warrants further investigation.
Collapse
Affiliation(s)
- Zhendong Li
- Department of Biological Sciences, National University of Singapore, 117543 Singapore
| | - Zhixia Chi
- Department of Biological Sciences, National University of Singapore, 117543 Singapore
| | - Wei-Xia Ang
- Department of Biological Sciences, National University of Singapore, 117543 Singapore.,Institute of Bioengineering & Nanotechnology, 138669 Singapore
| | - Can Chen
- Department of Biological Sciences, National University of Singapore, 117543 Singapore
| | - Johan Ck Tay
- Department of Biological Sciences, National University of Singapore, 117543 Singapore
| | - Yu-Yang Ng
- Department of Biological Sciences, National University of Singapore, 117543 Singapore
| | - Xuehu Xu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Junjian Wang
- Department of Gynaecological Oncology, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou 310022, PR China
| | - Jianqing Zhu
- Department of Gynaecological Oncology, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou 310022, PR China
| | - Shu Wang
- Department of Biological Sciences, National University of Singapore, 117543 Singapore.,Institute of Bioengineering & Nanotechnology, 138669 Singapore
| |
Collapse
|
252
|
Abstract
Engagement of activating receptor NKG2D to its ligand mediates natural killer (NK) cell activation and enhances cytotoxicity. NKG2D ligands (NKG2DLs) are frequently expressed on the tumor cell surface. However, the expression patterns of different NKG2DLs vary between tumor cells. Downregulation of certain ligand enables the tumor cells to escape NK cell-mediated immunosurveillance. By generating tumor cell lines with high expression of NKG2D ligand MULT1, we aimed to explore the function of NKG2DLs diversity on the activation and regulation of NKG2D signaling pathway. NK cells were potently activated by the "acquired" MULT1 expression on MOVCAR 5009 cells. Further, the progression of the tumor was significantly inhibited in mice inoculated with MULT1-expressing MOVCAR 5009 cells. Also, the pulmonary metastasis of MULT1-expressing B16-F0 cells was also significantly reduced in vivo. Our results implied that "acquired" NKG2D ligands enhance antitumor responses of NK cells, providing insights for designing novel therapeutic strategies and drugs to enhance NK cell surveillance over malignances.
Collapse
|
253
|
Pereira BI, De Maeyer RPH, Covre LP, Nehar-Belaid D, Lanna A, Ward S, Marches R, Chambers ES, Gomes DCO, Riddell NE, Maini MK, Teixeira VH, Janes SM, Gilroy DW, Larbi A, Mabbott NA, Ucar D, Kuchel GA, Henson SM, Strid J, Lee JH, Banchereau J, Akbar AN. Sestrins induce natural killer function in senescent-like CD8 + T cells. Nat Immunol 2020; 21:684-694. [PMID: 32231301 PMCID: PMC10249464 DOI: 10.1038/s41590-020-0643-3] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 02/26/2020] [Indexed: 12/29/2022]
Abstract
Aging is associated with remodeling of the immune system to enable the maintenance of life-long immunity. In the CD8+ T cell compartment, aging results in the expansion of highly differentiated cells that exhibit characteristics of cellular senescence. Here we found that CD27-CD28-CD8+ T cells lost the signaling activity of the T cell antigen receptor (TCR) and expressed a protein complex containing the agonistic natural killer (NK) receptor NKG2D and the NK adaptor molecule DAP12, which promoted cytotoxicity against cells that expressed NKG2D ligands. Immunoprecipitation and imaging cytometry indicated that the NKG2D-DAP12 complex was associated with sestrin 2. The genetic inhibition of sestrin 2 resulted in decreased expression of NKG2D and DAP12 and restored TCR signaling in senescent-like CD27-CD28-CD8+ T cells. Therefore, during aging, sestrins induce the reprogramming of non-proliferative senescent-like CD27-CD28-CD8+ T cells to acquire a broad-spectrum, innate-like killing activity.
Collapse
Affiliation(s)
- Branca I Pereira
- Division of Infection and Immunity, University College London, London, UK
| | - Roel P H De Maeyer
- Division of Infection and Immunity, University College London, London, UK
| | - Luciana P Covre
- Division of Infection and Immunity, University College London, London, UK
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória, Brazil
| | | | - Alessio Lanna
- Division of Infection and Immunity, University College London, London, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sophie Ward
- Department of Medicine, Imperial College London, London, UK
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Emma S Chambers
- Division of Infection and Immunity, University College London, London, UK
| | - Daniel C O Gomes
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Natalie E Riddell
- Division of Infection and Immunity, University College London, London, UK
- Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Mala K Maini
- Division of Infection and Immunity, University College London, London, UK
| | - Vitor H Teixeira
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Samuel M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Derek W Gilroy
- Division of Medicine, University College London, London, UK
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Neil A Mabbott
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - George A Kuchel
- University of Connecticut Center on Aging, University of Connecticut, Farmington, CT, USA
| | - Sian M Henson
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jessica Strid
- Department of Medicine, Imperial College London, London, UK
| | - Jun H Lee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | | | - Arne N Akbar
- Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|
254
|
Mariotti FR, Quatrini L, Munari E, Vacca P, Tumino N, Pietra G, Mingari MC, Moretta L. Inhibitory checkpoints in human natural killer cells: IUPHAR Review 28. Br J Pharmacol 2020; 177:2889-2903. [PMID: 32335915 PMCID: PMC7279970 DOI: 10.1111/bph.15081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy leading to exceptional success. However, there is still the need to improve their efficacy in non‐responder patients. Natural killer (NK) cells represent the first line of defence against tumours, due to their ability to release immunomodulatory cytokines and kill target cells that have undergone malignant transformation. Harnessing NK cell response will open new possibilities to improve control of tumour growth. In this respect inhibitory checkpoints expressed on these innate lymphocytes represents a promising target for next‐generation immunotherapy. In this review, we will summarize recent evidences on the expression of NK cells receptors in cancer, with a focus on the inhibitory checkpoint programmed cell death protein 1 (PD‐1). We will also highlight the strength and limitations of the blockade of PD‐1 inhibitory pathway and suggest new combination strategies that may help to unleash more efficiently NK cell anti‐tumour response.
Collapse
Affiliation(s)
- F R Mariotti
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - L Quatrini
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - E Munari
- Department of Pathology, Sacro Cuore Don Calabria, Negrar, Italy
| | - P Vacca
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - N Tumino
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - G Pietra
- Laboratory of Immunology, Department of Integrated Oncological Therapies, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Experimental Medicine (DIMES), Università di Genova, Genoa, Italy
| | - M C Mingari
- Laboratory of Immunology, Department of Integrated Oncological Therapies, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Experimental Medicine (DIMES), Center of Excellence for Biomedical Research, Università di Genova, Genoa, Italy
| | - L Moretta
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
255
|
Paving the Way toward Successful Multiple Myeloma Treatment: Chimeric Antigen Receptor T-Cell Therapy. Cells 2020; 9:cells9040983. [PMID: 32316105 PMCID: PMC7226998 DOI: 10.3390/cells9040983] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/19/2022] Open
Abstract
Despite the significant progress of modern anticancer therapies, multiple myeloma (MM) is still incurable for the majority of patients. Following almost three decades of development, chimeric antigen receptor (CAR) T-cell therapy now has the opportunity to revolutionize the treatment landscape and meet the unmet clinical need. However, there are still several major hurdles to overcome. Here we discuss the recent advances of CAR T-cell therapy for MM with an emphasis on future directions and possible risks. Currently, CAR T-cell therapy for MM is at the first stage of clinical studies, and most studies have focused on CAR T cells targeting B cell maturation antigen (BCMA), but other antigens such as cluster of differentiation 138 (CD138, syndecan-1) are also being evaluated. Although this therapy is associated with side effects, such as cytokine release syndrome and neurotoxicity, and relapses have been observed, the benefit–risk balance and huge potential drive the ongoing clinical progress. To fulfill the promise of recent clinical trial success and maximize the potential of CAR T, future efforts should focus on the reduction of side effects, novel targeted antigens, combinatorial uses of different types of CAR T, and development of CAR T cells targeting more than one antigen.
Collapse
|
256
|
Dell'Oste V, Biolatti M, Galitska G, Griffante G, Gugliesi F, Pasquero S, Zingoni A, Cerboni C, De Andrea M. Tuning the Orchestra: HCMV vs. Innate Immunity. Front Microbiol 2020; 11:661. [PMID: 32351486 PMCID: PMC7174589 DOI: 10.3389/fmicb.2020.00661] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
Understanding how the innate immune system keeps human cytomegalovirus (HCMV) in check has recently become a critical issue in light of the global clinical burden of HCMV infection in newborns and immunodeficient patients. Innate immunity constitutes the first line of host defense against HCMV as it involves a complex array of cooperating effectors – e.g., inflammatory cytokines, type I interferon (IFN-I), natural killer (NK) cells, professional antigen-presenting cells (APCs) and phagocytes – all capable of disrupting HCMV replication. These factors are known to trigger a highly efficient adaptive immune response, where cellular restriction factors (RFs) play a major gatekeeping role. Unlike other innate immunity components, RFs are constitutively expressed in many cell types, ready to act before pathogen exposure. Nonetheless, the existence of a positive regulatory feedback loop between RFs and IFNs is clear evidence of an intimate cooperation between intrinsic and innate immunity. In the course of virus-host coevolution, HCMV has, however, learned how to manipulate the functions of multiple cellular players of the host innate immune response to achieve latency and persistence. Thus, HCMV acts like an orchestra conductor able to piece together and rearrange parts of a musical score (i.e., innate immunity) to obtain the best live performance (i.e., viral fitness). It is therefore unquestionable that innovative therapeutic solutions able to prevent HCMV immune evasion in congenitally infected infants and immunocompromised individuals are urgently needed. Here, we provide an up-to-date review of the mechanisms regulating the interplay between HCMV and innate immunity, focusing on the various strategies of immune escape evolved by this virus to gain a fitness advantage.
Collapse
Affiliation(s)
- Valentina Dell'Oste
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Matteo Biolatti
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Ganna Galitska
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Gloria Griffante
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Francesca Gugliesi
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Selina Pasquero
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Alessandra Zingoni
- Department of Molecular Immunology and Immunopathology, "Sapienza" University of Rome, Rome, Italy
| | - Cristina Cerboni
- Department of Molecular Immunology and Immunopathology, "Sapienza" University of Rome, Rome, Italy
| | - Marco De Andrea
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy.,Center for Translational Research on Autoimmune and Allergic Disease - CAAD, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
257
|
Natural Killer Cell Responses in Hepatocellular Carcinoma: Implications for Novel Immunotherapeutic Approaches. Cancers (Basel) 2020; 12:cancers12040926. [PMID: 32283827 PMCID: PMC7226319 DOI: 10.3390/cancers12040926] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) still represents a significant complication of chronic liver disease, particularly when cirrhosis ensues. Current treatment options include surgery, loco-regional procedures and chemotherapy, according to specific clinical practice guidelines. Immunotherapy with check-point inhibitors, aimed at rescuing T-cells from exhaustion, has been applied as second-line therapy with limited and variable success. Natural killer (NK) cells are an essential component of innate immunity against cancer and changes in phenotype and function have been described in patients with HCC, who also show perturbations of NK activating receptor/ligand axes. Here we discuss the current status of NK cell treatment of HCC on the basis of existing evidence and ongoing clinical trials on adoptive transfer of autologous or allogeneic NK cells ex vivo or after activation with cytokines such as IL-15 and use of antibodies to target cell-expressed molecules to promote antibody-dependent cellular cytotoxicity (ADCC). To this end, bi-, tri- and tetra-specific killer cell engagers are being devised to improve NK cell recognition of tumor cells, circumventing tumor immune escape and efficiently targeting NK cells to tumors. Moreover, the exciting technique of chimeric antigen receptor (CAR)-engineered NK cells offers unique opportunities to create CAR-NK with multiple specificities along the experience gained with CAR-T cells with potentially less adverse effects.
Collapse
|
258
|
Tong DL, Kempsell KE, Szakmany T, Ball G. Development of a Bioinformatics Framework for Identification and Validation of Genomic Biomarkers and Key Immunopathology Processes and Controllers in Infectious and Non-infectious Severe Inflammatory Response Syndrome. Front Immunol 2020; 11:380. [PMID: 32318053 PMCID: PMC7147506 DOI: 10.3389/fimmu.2020.00380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Sepsis is defined as dysregulated host response caused by systemic infection, leading to organ failure. It is a life-threatening condition, often requiring admission to an intensive care unit (ICU). The causative agents and processes involved are multifactorial but are characterized by an overarching inflammatory response, sharing elements in common with severe inflammatory response syndrome (SIRS) of non-infectious origin. Sepsis presents with a range of pathophysiological and genetic features which make clinical differentiation from SIRS very challenging. This may reflect a poor understanding of the key gene inter-activities and/or pathway associations underlying these disease processes. Improved understanding is critical for early differential recognition of sepsis and SIRS and to improve patient management and clinical outcomes. Judicious selection of gene biomarkers suitable for development of diagnostic tests/testing could make differentiation of sepsis and SIRS feasible. Here we describe a methodologic framework for the identification and validation of biomarkers in SIRS, sepsis and septic shock patients, using a 2-tier gene screening, artificial neural network (ANN) data mining technique, using previously published gene expression datasets. Eight key hub markers have been identified which may delineate distinct, core disease processes and which show potential for informing underlying immunological and pathological processes and thus patient stratification and treatment. These do not show sufficient fold change differences between the different disease states to be useful as primary diagnostic biomarkers, but are instrumental in identifying candidate pathways and other associated biomarkers for further exploration.
Collapse
Affiliation(s)
- Dong Ling Tong
- Artificial Intelligence Laboratory, Faculty of Engineering and Computing, First City University College, Petaling Jaya, Malaysia.,School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Karen E Kempsell
- Public Health England, National Infection Service, Porton Down, Salisbury, United Kingdom
| | - Tamas Szakmany
- Department of Anaesthesia Intensive Care and Pain Medicine, Division of Population Medicine, Cardiff University, Cardiff, United Kingdom
| | - Graham Ball
- School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
259
|
Zingoni A, Vulpis E, Loconte L, Santoni A. NKG2D Ligand Shedding in Response to Stress: Role of ADAM10. Front Immunol 2020; 11:447. [PMID: 32269567 PMCID: PMC7109295 DOI: 10.3389/fimmu.2020.00447] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/26/2020] [Indexed: 12/18/2022] Open
Abstract
NKG2D is an activating receptor expressed by NK cells and some subsets of T cells and represents a major recognition receptor for detection and elimination of cancer cells. The ligands of NKG2D are stress-induced self-proteins that can be secreted as soluble molecules by protease-mediated cleavage. The release of NKG2D ligands in the extracellular milieu is considered a mode of finely controlling their surface expression levels and represents a relevant immune evasion mechanism employed by cancer cells to elude NKG2D-mediated immune surveillance. A disintegrin and metalloproteinase 10 (ADAM10), a catalytically active member of the ADAM family of proteases, is involved in the cleavage of some NKG2D ligands in various types of cancer cells either in steady state conditions and in response to an ample variety of stress stimuli. Appealing immunotherapeutic strategies devoted to promoting NK cell-mediated recognition and elimination of cancer cells are based on the upregulation of NK cell activating ligands. In particular, activation of DNA damage response (DDR) and the induction of cellular senescence by chemotherapeutic agents are associated with increased expression of NKG2D ligands on cancer cell surface. Herein, we will review advances on the protease-mediated cleavage of NKG2D ligands in response to chemotherapy-induced stress focusing on: (i) the role played by ADAM10 in this process and (ii) the implications of NKG2D ligand shedding in the course of cancer therapy and in senescent cells.
Collapse
Affiliation(s)
- Alessandra Zingoni
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Elisabetta Vulpis
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Luisa Loconte
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Angela Santoni
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
260
|
NK cells and CD38: Implication for (Immuno)Therapy in Plasma Cell Dyscrasias. Cells 2020; 9:cells9030768. [PMID: 32245149 PMCID: PMC7140687 DOI: 10.3390/cells9030768] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/14/2020] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy represents a promising new avenue for the treatment of multiple myeloma (MM) patients, particularly with the availability of Monoclonal Antibodies (mAbs) as anti-CD38 Daratumumab and Isatuximab and anti-SLAM-F7 Elotuzumab. Although a clear NK activation has been demonstrated for Elotuzumab, the effect of anti-CD38 mAbs on NK system is controversial. As a matter of fact, an initial reduction of NK cells number characterizes Daratumumab therapy, limiting the potential role of this subset on myeloma immunotherapy. In this paper we discuss the role of NK cells along with anti-CD38 therapy and their implication in plasma cell dyscrasias, showing that mechanisms triggered by anti-CD38 mAbs ultimately lead to the activation of the immune system against myeloma cell growth.
Collapse
|
261
|
Tangye SG. Genetic susceptibility to EBV infection: insights from inborn errors of immunity. Hum Genet 2020; 139:885-901. [PMID: 32152698 DOI: 10.1007/s00439-020-02145-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous human pathogen, infecting > 90% of the adult population. In the vast majority of healthy individuals, infection with EBV runs a relatively benign course. However, EBV is by no means a benign pathogen. Indeed, apart from being associated with at least seven different types of malignancies, EBV infection can cause severe and often fatal diseases-hemophagocytic lymphohistiocytosis, lymphoproliferative disease, B-cell lymphoma-in rare individuals with specific monogenic inborn errors of immunity. The discovery and detailed investigation of inborn errors of immunity characterized by heightened susceptibility to, or increased frequency of, EBV-induced disease have elegantly revealed cell types and signaling pathways that play critical and non-redundant roles in host-defense against EBV. These analyses have revealed not only mechanisms underlying EBV-induced disease in rare genetic conditions, but also identified molecules and pathways that could be targeted to treat severe EBV infection and pathological consequences in immunodeficient hosts, or even potentially enhance the efficacy of an EBV-specific vaccine.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia. .,St. Vincent's Clinical School, University of NSW Sydney, Darlinghurst, NSW, 2010, Australia. .,Clincial Immunogenomics Research Consortium Australasia (CIRCA), Darlinghurst, NSW, Australia.
| |
Collapse
|
262
|
Høgh RI, Droujinine A, Møller SH, Jepsen SD, Mellergaard M, Andresen L, Skov S. Fumarate Upregulates Surface Expression of ULBP2/ULBP5 by Scavenging Glutathione Antioxidant Capacity. THE JOURNAL OF IMMUNOLOGY 2020; 204:1746-1759. [PMID: 32144161 DOI: 10.4049/jimmunol.1900740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/26/2020] [Indexed: 12/15/2022]
Abstract
Fumarate is a tricarboxylic acid cycle metabolite whose intracellular accumulation is linked to inflammatory signaling and development of cancer. In this study, we demonstrate that endogenous fumarate accumulation upregulates surface expression of the immune stimulatory NK group 2, member D (NKG2D) ligands ULBP2 and ULBP5. In agreement with this, accumulation of fumarate by the therapeutic drug dimethyl fumarate (DMF) also promotes ULBP2/5 surface expression. Mechanistically, we found that the increased ULBP2/5 expression was dependent on oxidative stress and the antioxidants N-acetylcysteine and glutathione (GSH) abrogated ULBP2/5 upregulated by DMF. Fumarate can complex with GSH and thereby exhaust cells of functional GSH capacity. In line with this, inhibition of GSH reductase (GR), the enzyme responsible for GSH recycling, promoted ULBP2/5 surface expression. Loss of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) associates with a malignant form of renal cancer characterized by fumarate accumulation and increased production of reactive oxygen species, highlighting fumarate as an oncometabolite. Interestingly, FH-deficient renal cancer cells had low surface expression of ULBP2/5 and were unresponsive to DMF treatment, suggesting that the fumarate-stimulating ULBP2/5 pathway is abrogated in these cells as an immune-evasive strategy. Together, our data show that ULBP2/5 expression can be upregulated by accumulation of fumarate, likely by depleting cells of GSH antioxidant capacity. Given that DMF is an approved human therapeutic drug, our findings support a broader use of DMF in treatment of cancers and inflammatory conditions.
Collapse
Affiliation(s)
- Rikke Illum Høgh
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alec Droujinine
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sofie Hedlund Møller
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Stine Dam Jepsen
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maiken Mellergaard
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Lars Andresen
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Søren Skov
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
263
|
Davies AJ, Rinaldi S, Costigan M, Oh SB. Cytotoxic Immunity in Peripheral Nerve Injury and Pain. Front Neurosci 2020; 14:142. [PMID: 32153361 PMCID: PMC7047751 DOI: 10.3389/fnins.2020.00142] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
Cytotoxicity and consequent cell death pathways are a critical component of the immune response to infection, disease or injury. While numerous examples of inflammation causing neuronal sensitization and pain have been described, there is a growing appreciation of the role of cytotoxic immunity in response to painful nerve injury. In this review we highlight the functions of cytotoxic immune effector cells, focusing in particular on natural killer (NK) cells, and describe the consequent action of these cells in the injured nerve as well as other chronic pain conditions and peripheral neuropathies. We describe how targeted delivery of cytotoxic factors via the immune synapse operates alongside Wallerian degeneration to allow local axon degeneration in the absence of cell death and is well-placed to support the restoration of homeostasis within the nerve. We also summarize the evidence for the expression of endogenous ligands and receptors on injured nerve targets and infiltrating immune cells that facilitate direct neuro-immune interactions, as well as modulation of the surrounding immune milieu. A number of chronic pain and peripheral neuropathies appear comorbid with a loss of function of cellular cytotoxicity suggesting such mechanisms may actually help to resolve neuropathic pain. Thus while the immune response to peripheral nerve injury is a major driver of maladaptive pain, it is simultaneously capable of directing resolution of injury in part through the pathways of cellular cytotoxicity. Our growing knowledge in tuning immune function away from inflammation toward recovery from nerve injury therefore holds promise for interventions aimed at preventing the transition from acute to chronic pain.
Collapse
Affiliation(s)
- Alexander J. Davies
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Simon Rinaldi
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Michael Costigan
- Department of Anesthesia, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurobiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Seog Bae Oh
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Dental Research Institute and Department of Neurobiology & Physiology, School of Dentistry, Seoul National University, Seoul, South Korea
| |
Collapse
|
264
|
Prinz D, Klein K, List J, Knab VM, Menzl I, Leidenfrost N, Heller G, Polić B, Putz EM, Witalisz-Siepracka A, Sexl V, Gotthardt D. Loss of NKG2D in murine NK cells leads to increased perforin production upon long-term stimulation with IL-2. Eur J Immunol 2020; 50:880-890. [PMID: 32052406 PMCID: PMC7318224 DOI: 10.1002/eji.201948222] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 01/07/2020] [Accepted: 02/11/2020] [Indexed: 01/12/2023]
Abstract
NK cells are innate lymphocytes responsible for lysis of pathogen-infected and transformed cells. One of the major activating receptors required for target cell recognition is the NK group 2D (NKG2D) receptor. Numerous reports show the necessity of NKG2D for effective tumor immune surveillance. Further studies identified NKG2D as a key element allowing tumor immune escape. We here use a mouse model with restricted deletion of NKG2D in mature NKp46+ cells (NKG2DΔNK ). NKG2DΔNK NK cells develop normally, have an unaltered IFN-γ production but kill tumor cell lines expressing NKG2D ligands (NKG2DLs) less efficiently. However, upon long-term stimulation with IL-2, NKG2D-deficient NK cells show increased levels of the lytic molecule perforin. Thus, our findings demonstrate a dual function of NKG2D for NK cell cytotoxicity; while NKG2D is a crucial trigger for cytotoxicity of tumor cells expressing activating ligands it is also capable to limit perforin production in IL-2 activated NK cells.
Collapse
Affiliation(s)
- Daniela Prinz
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Klara Klein
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Julia List
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Vanessa M Knab
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Ingeborg Menzl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Nicoletta Leidenfrost
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Gerwin Heller
- Institute of Internal Medicine I, Medical University, Vienna, Austria
| | - Bojan Polić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Eva Maria Putz
- St. Anna Children's Cancer Research Institute (CCRI), Medical University of Vienna, Vienna, Austria
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Dagmar Gotthardt
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
265
|
Recent Advances in Molecular Mechanisms of the NKG2D Pathway in Hepatocellular Carcinoma. Biomolecules 2020; 10:biom10020301. [PMID: 32075046 PMCID: PMC7094213 DOI: 10.3390/biom10020301] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/16/2020] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma is a common malignant tumor with high mortality. Its malignant proliferation, invasion, and metastasis are closely related to the cellular immune function of the patients. NKG2D is a key activated and type II membrane protein molecule expressed on the surface of almost all NK cells. The human NKG2D gene is 270 kb long, located at 12p12.3-p13.1, and contains 10 exons and 9 introns. The three-dimensional structure of the NKG2D monomeric protein contains two alpha-helices, two beta-lamellae, and four disulfide bonds, and its' signal of activation is transmitted mainly by the adaptor protein (DAP). NKG2D ligands, including MICA, MICB, and ULBPs, can be widely expressed in hepatoma cells. After a combination of NKG2D and DAP10 in the form of homologous two polymers, the YxxM motif in the cytoplasm is phosphorylated and then signaling pathways are also gradually activated, such as PI3K, PLCγ2, JNK-cJunN, and others. Activated NK cells can enhance the sensitivity to hepatoma cells and specifically dissolve by releasing a variety of cytokines (TNF-α and IFN-γ), perforin, and high expression of FasL, CD16, and TRAIL. NK cells may specifically bind to the over-expressed MICA, MICB, and ULBPs of hepatocellular carcinoma cells through the surface activating receptor NKG2D, which can help to accurately identify hepatoma, play a critical role in anti-hepatoma via the pathway of cytotoxic effects, and obviously delay the poor progress of hepatocellular carcinoma.
Collapse
|
266
|
Baumeister SHC, Rambaldi B, Shapiro RM, Romee R. Key Aspects of the Immunobiology of Haploidentical Hematopoietic Cell Transplantation. Front Immunol 2020; 11:191. [PMID: 32117310 PMCID: PMC7033970 DOI: 10.3389/fimmu.2020.00191] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/24/2020] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cell transplantation from a haploidentical donor is increasingly used and has become a standard donor option for patients lacking an appropriately matched sibling or unrelated donor. Historically, prohibitive immunological barriers resulting from the high degree of HLA-mismatch included graft-vs.-host disease (GVHD) and graft failure. These were overcome with increasingly sophisticated strategies to manipulate the sensitive balance between donor and recipient immune cells. Three different approaches are currently in clinical use: (a) ex vivo T-cell depletion resulting in grafts with defined immune cell content (b) extensive immunosuppression with a T-cell replete graft consisting of G-CSF primed bone marrow and PBSC (GIAC) (c) T-cell replete grafts with post-transplant cyclophosphamide (PTCy). Intriguing studies have recently elucidated the immunologic mechanisms by which PTCy prevents GVHD. Each approach uniquely affects post-transplant immune reconstitution which is critical for the control of post-transplant infections and relapse. NK-cells play a key role in haplo-HCT since they do not mediate GVHD but can successfully mediate a graft-vs.-leukemia effect. This effect is in part regulated by KIR receptors that inhibit NK cell cytotoxic function when binding to the appropriate HLA-class I ligands. In the context of an HLA-class I mismatch in haplo-HCT, lack of inhibition can contribute to NK-cell alloreactivity leading to enhanced anti-leukemic effect. Emerging work reveals immune evasion phenomena such as copy-neutral loss of heterozygosity of the incompatible HLA alleles as one of the major mechanisms of relapse. Relapse and infectious complications remain the leading causes impacting overall survival and are central to scientific advances seeking to improve haplo-HCT. Given that haploidentical donors can typically be readily approached to collect additional stem- or immune cells for the recipient, haplo-HCT represents a unique platform for cell- and immune-based therapies aimed at further reducing relapse and infections. The rapid advancements in our understanding of the immunobiology of haplo-HCT are therefore poised to lead to iterative innovations resulting in further improvement of outcomes with this compelling transplant modality.
Collapse
Affiliation(s)
- Susanne H C Baumeister
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Benedetta Rambaldi
- Harvard Medical School, Boston, MA, United States.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, United States.,Bone Marrow Transplant Unit, Clinical and Experimental Sciences Department, ASST Spedali Civili, University of Pavia, Brescia, Italy
| | - Roman M Shapiro
- Harvard Medical School, Boston, MA, United States.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Rizwan Romee
- Harvard Medical School, Boston, MA, United States.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, United States
| |
Collapse
|
267
|
Delaune V, Toso C, Kahler-Quesada A, Slits F, Gex Q, Kaya G, Lavallard V, Orci LA, Peloso A, Lacotte S. Antibody-induced NKG2D blockade in a rat model of intraportal islet transplantation leads to a deleterious reaction. Transpl Int 2020; 33:675-688. [PMID: 32003082 DOI: 10.1111/tri.13589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/04/2019] [Accepted: 01/28/2020] [Indexed: 11/30/2022]
Abstract
Intraportal islet transplantation is plagued by an acute destruction of transplanted islets. Amongst the first responders, NK cells and macrophages harbour an activating receptor, NKG2D, recognizing ligands expressed by stressed cells. We aimed to determine whether islet NKG2D ligand expression increases with culture time, and to analyse the impact of antibody-induced NKG2D blockade in islet transplantation. NKG2D-ligand expression was analysed in rat and human islets. Syngeneic marginal mass intraportal islet transplantations were performed in rats: control group, recipients transplanted with NKG2D-recombinant-treated islets (recombinant group), and recipients treated with a mouse anti-rat anti-NKG2D antibody and transplanted with recombinant-treated islets (antibody-recombinant group). Islets demonstrated increased gene expression of NKG2D ligands with culture time. Blockade of NKG2D on NK cells decreased in vitro cytotoxicity against islets. Recipients from the control and recombinant groups showed similar metabolic results; conversely, treatment with the antibody resulted in lower diabetes reversal. The antibody depleted circulating and liver NK cells in recipients, who displayed increased macrophage infiltration of recipient origin around the transplanted islets. In vitro blockade of NKG2D ligands had no impact on early graft function. Systemic treatment of recipients with an anti-NKG2D antibody was deleterious to the islet graft, possibly through an antibody-dependent cell-mediated cytotoxicity reaction.
Collapse
Affiliation(s)
- Vaihere Delaune
- Divisions of Abdominal and Transplantation Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland.,Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christian Toso
- Divisions of Abdominal and Transplantation Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland.,Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Arianna Kahler-Quesada
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Florence Slits
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Quentin Gex
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gürkan Kaya
- Division of Dermatology and Venereology, Department of Internal Medicine Specialties, Geneva University Hospitals, Geneva, Switzerland
| | - Vanessa Lavallard
- Cell Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lorenzo Annibale Orci
- Divisions of Abdominal and Transplantation Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland.,Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Andrea Peloso
- Divisions of Abdominal and Transplantation Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland.,Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stéphanie Lacotte
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
268
|
Luo Q, Luo W, Zhu Q, Huang H, Peng H, Liu R, Xie M, Li S, Li M, Hu X, Zou Y. Tumor-Derived Soluble MICA Obstructs the NKG2D Pathway to Restrain NK Cytotoxicity. Aging Dis 2020; 11:118-128. [PMID: 32010486 PMCID: PMC6961768 DOI: 10.14336/ad.2019.1017] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/28/2019] [Indexed: 12/31/2022] Open
Abstract
The natural killer group 2D (NKG2D) receptor and its ligands play important roles in immune surveillance. In this study, we observed that the average serum soluble MICA (sMICA) concentration of 174 hepatocellular carcinoma (HCC) patients was significantly higher than that in 80 healthy subjects (602.17 ± 338.15 vs. 72.26 ± 87.88 pg/ml, t = 3.107, P=0.002). The levels of serum sMICA in 44 HCC patients with initial levels above 400 pg/ml declined significantly after surgical removal of the liver cancer tissue (P<0.001). Moreover, the mean survival time of HCC patients who had sMICA above 400 pg/ml was significantly shorter than that HCC patients with lower sMICA levels (P<0.001). Using the reporter cell line (NKG2D-2B4) in which activation of the NKG2D receptor pathway results in GFP expression based on the stimulation of immobilized rMICA, we showed that the number of GFP-expressing cells decreased sharply in presence of sMICA. Upon adding sMICA, the release of cytokines IFN-γ, TNF-α, and IL-8 by NK cell line (NKL) under stimulation of immobilized rMICA was blocked. Using MICA-expressing cells as the target cells, we observed that about 80% of target cells were killed by NKL at E:T of 10:1, but in presence of sMICAhigh serum of HCC patients, the dead target cells were reduced to 30.8%. Compared in presence of sMICAlow serum from HCC patients, there were 63.7% of target cells dead (p=0.043). Thus, our data suggested that sMICA obstructs the activation of NKG2D pathway to protect tumor cells from NK cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Qizhi Luo
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Weiguang Luo
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China.,2Department of Physiology, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Quan Zhu
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Hongjun Huang
- 3Cancer Hospital of Hunan, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Huiyun Peng
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Rongjiao Liu
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Min Xie
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Shili Li
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Ming Li
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Xiaocui Hu
- 3Cancer Hospital of Hunan, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Yizhou Zou
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| |
Collapse
|
269
|
Terrén I, Orrantia A, Mikelez-Alonso I, Vitallé J, Zenarruzabeitia O, Borrego F. NK Cell-Based Immunotherapy in Renal Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12020316. [PMID: 32013092 PMCID: PMC7072691 DOI: 10.3390/cancers12020316] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/14/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells are cytotoxic lymphocytes that are able to kill tumor cells without prior sensitization. It has been shown that NK cells play a pivotal role in a variety of cancers, highlighting their relevance in tumor immunosurveillance. NK cell infiltration has been reported in renal cell carcinoma (RCC), the most frequent kidney cancer in adults, and their presence has been associated with patients’ survival. However, the role of NK cells in this disease is not yet fully understood. In this review, we summarize the biology of NK cells and the mechanisms through which they are able to recognize and kill tumor cells. Furthermore, we discuss the role that NK cells play in renal cell carcinoma, and review current strategies that are being used to boost and exploit their cytotoxic capabilities.
Collapse
Affiliation(s)
- Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Ane Orrantia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Idoia Mikelez-Alonso
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
- CIC biomaGUNE, 20014 Donostia-San Sebastián, Spain
| | - Joana Vitallé
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Francisco Borrego
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
- Correspondence: ; Tel.: +34-94-600-6000 (ext. 7079)
| |
Collapse
|
270
|
Yang C, Li Y, Yang Y, Chen Z. Overview of Strategies to Improve Therapy against Tumors Using Natural Killer Cell. J Immunol Res 2020; 2020:8459496. [PMID: 32411806 PMCID: PMC7201677 DOI: 10.1155/2020/8459496] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/24/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022] Open
Abstract
NK cells are lymphocytes with antitumor properties and can directly lyse tumor cells in a non-MHC-restricted manner. However, the tumor microenvironment affects the immune function of NK cells, which leads to immune evasion. This may be related to the pathogenesis of some diseases. Therefore, great efforts have been made to improve the immunotherapy effect of natural killer cells. NK cells from different sources can meet different clinical needs, in order to minimize the inhibition of NK cells and maximize the response potential of NK cells, for example, modification of NK cells can increase the number of NK cells in tumor target area, change the direction of NK cells, and improve their targeting ability to malignant cells. Checkpoint blocking is also a promising strategy for NK cells to kill tumor cells. Combination therapy is another strategy for improving antitumor ability, especially in combination with oncolytic viruses and nanomaterials. In this paper, the mechanisms affecting the activity of NK cells were reviewed, and the therapeutic potential of different basic NK cell strategies in tumor therapy was focused on. The main strategies for improving the immune function of NK cells were described, and some new strategies were proposed.
Collapse
Affiliation(s)
- Chaopin Yang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Experimental Center, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510176, China
| | - Yue Li
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Experimental Center, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510176, China
| | - Yaozhang Yang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Experimental Center, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510176, China
| | - Zhiyi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Experimental Center, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510176, China
| |
Collapse
|
271
|
Hitting More Birds with a Stone: Impact of TGF-β on ILC Activity in Cancer. J Clin Med 2020; 9:jcm9010143. [PMID: 31948072 PMCID: PMC7019362 DOI: 10.3390/jcm9010143] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 12/20/2019] [Accepted: 01/03/2020] [Indexed: 12/17/2022] Open
Abstract
Transforming growth factor (TGF)-β is a central immunosuppressive cytokine within tumor microenvironment inhibiting the expansion and function of major cellular components of adaptive and innate immune system. Among them, compelling evidence has demonstrated that TGF-β is a key regulator of natural killer (NK) cells, innate lymphoid cells (ILCs) with a critical role in immunosurveillance against different kinds of cancer cells. A TGF-β rich tumor microenvironment blocks NK cell activity at multiple levels. This immunosuppressive factor exerts direct regulatory effects on NK cells including inhibition of cytokine production, alteration of activating/inhibitory receptor expression, and promotion of the conversion into non cytotoxic group I ILC (ILC1). Concomitantly, TGF-β can render tumor cells less susceptible to NK cell-mediated recognition and lysis. Indeed, accumulating evidence suggest that changes in levels of NKG2D ligands, mainly MICA, as well as an increase of immune checkpoint inhibitors (e.g., PD-L1) and other inhibitory ligands on cancer cells significantly contribute to TGF-β-mediated suppression of NK cell activity. Here, we will take into consideration two major mechanisms underlying the negative regulation of ILC function by TGF-β in cancer. First, we will address how TGF-β impacts the balance of signals governing NK cell activity. Second, we will review recent advances on the role of this cytokine in driving ILC plasticity in cancer. Finally, we will discuss how the development of therapeutic approaches blocking TGF-β may reverse the suppression of host immune surveillance and improve anti-tumor NK cell response in the clinic.
Collapse
|
272
|
Influence of major histocompatibility complex class I chain-related gene A polymorphisms on cytomegalovirus disease after allogeneic hematopoietic cell transplantation. Hematol Oncol Stem Cell Ther 2019; 13:32-39. [PMID: 31881183 DOI: 10.1016/j.hemonc.2019.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 09/18/2019] [Accepted: 10/27/2019] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE/BACKGROUND Cytomegalovirus (CMV) infection and disease are common infectious complications after allogeneic hematopoietic cell transplantation (alloHCT). Major histocompatibility complex (MHC) class I chain-related gene A (MICA) is a ligand of the natural killer (NKG2D) receptor on immune effector cells that helps mediate NK cell alloreactivity. We hypothesized that MICA polymorphisms may influence CMV infection and disease incidence after alloHCT. METHODS We conducted a retrospective analysis of 423 adults at the Cleveland Clinic with hematologic malignancies treated with a matched related or unrelated donor alloHCT. CMV cases analyzed included a compositive of instances of viral copy replication above detection limits as well as any biopsy-proven tissue invasive disease episodes. Genotypes at the MICA-129 position have been categorized as weak (valine/valine; V/V), intermediate (methionine/valine; M/V), or strong (methionine/methionine; M/M) receptor affinity. RESULTS In multivariable analysis, V/V donor MICA-129 genotype was associated with CMV infection and disease (hazard ratio [HR] = 1.40; 95% confidence interval [CI], 1.00-1.96; p = .05), but not MICA mismatch (HR = 1.38; 95% CI, 0.83-2.29; p = .22). There was no association of acute or chronic GVHD with MICA donor-recipient mismatch (HR = 1.05; 95% 95% CI, 0.66-1.68; p = .83 and HR = 0.94; 95% CI, 0.51-1.76; p = .85, respectively) or V/V donor MICA-129 genotypes (HR = 1.02; 95% CI, 0.79-1.31; p = .89 and HR = 0.89; 95% CI, 0.65-1.22; p = .47, respectively). CONCLUSION These findings suggest that the donor MICA-129 V/V genotype with weak NKG2D receptor binding affinity is associated with an increased risk of CMV infection and disease after alloHCT.
Collapse
|
273
|
The activation of bystander CD8 + T cells and their roles in viral infection. Exp Mol Med 2019; 51:1-9. [PMID: 31827070 PMCID: PMC6906361 DOI: 10.1038/s12276-019-0316-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/09/2019] [Accepted: 07/05/2019] [Indexed: 02/06/2023] Open
Abstract
During viral infections, significant numbers of T cells are activated in a T cell receptor-independent and cytokine-dependent manner, a phenomenon referred to as "bystander activation." Cytokines, including type I interferons, interleukin-18, and interleukin-15, are the most important factors that induce bystander activation of T cells, each of which plays a somewhat different role. Bystander T cells lack specificity for the pathogen, but can nevertheless impact the course of the immune response to the infection. For example, bystander-activated CD8+ T cells can participate in protective immunity by secreting cytokines, such as interferon-γ. They also mediate host injury by exerting cytotoxicity that is facilitated by natural killer cell-activating receptors, such as NKG2D, and cytolytic molecules, such as granzyme B. Interestingly, it has been recently reported that there is a strong association between the cytolytic function of bystander-activated CD8+ T cells and host tissue injury in patients with acute hepatitis A virus infection. The current review addresses the induction of bystander CD8+ T cells, their effector functions, and their potential roles in immunity to infection, immunopathology, and autoimmunity.
Collapse
|
274
|
Kim KH, Yu HT, Hwang I, Park S, Park SH, Kim S, Shin EC. Phenotypic and Functional Analysis of Human NK Cell Subpopulations According to the Expression of FcεRIγ and NKG2C. Front Immunol 2019; 10:2865. [PMID: 31867015 PMCID: PMC6908468 DOI: 10.3389/fimmu.2019.02865] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/21/2019] [Indexed: 01/05/2023] Open
Abstract
Human memory-like NK cells are commonly defined by either a lack of FcεRIγ or gain of NKG2C expression. Here, we investigated the heterogeneity of human CD56dim NK cell subpopulations according to the expression of FcεRIγ and NKG2C in a large cohort (n = 127). Although the frequency of FcεRIγ− and NKG2C+ NK cells positively correlated, the FcεRIγ− and NKG2C+ NK cell populations did not exactly overlap. The FcεRIγ+NKG2C+, FcεRIγ−NKG2C+, and FcεRIγ−NKG2C− NK cell populations were only evident after HCMV infection, but each had distinct characteristics. Among the subpopulations, FcεRIγ−NKG2C+ NK cells exhibited the most restricted killer immunoglobulin-like receptor repertoire, suggesting clonal expansion. Moreover, FcεRIγ−NKG2C+ NK cells exhibited the lowest Ki-67 and highest Bcl-2 expression, indicating the long-lived quiescent memory-like property. Functionally, FcεRIγ−NKG2C+ NK cells had weak natural effector function against K562 but strong effector functions by CD16 engagement, whereas FcεRIγ+NKG2C+ NK cells had strong effector functions in both settings. Anatomically, the FcεRIγ+NKG2C+, FcεRIγ−NKG2C+, and FcεRIγ−NKG2C− NK cell populations were present in multiple human peripheral organs. In conclusion, we demonstrate the heterogeneity of memory-like NK cells stratified by FcεRIγ and NKG2C and suggest both markers be utilized to better define these cells.
Collapse
Affiliation(s)
- Kyung Hwan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Hee Tae Yu
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.,Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Ilwoong Hwang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Sungha Park
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Sungjin Kim
- Department of Medical Microbiology and Immunology, Center for Comparative Medicine, University of California, Davis, Davis, CA, United States
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| |
Collapse
|
275
|
Martinović KM, Milićević M, Larsen AK, Džodić R, Jurišić V, Konjević G, Vuletić A. Effect of cytokines on NK cell activity and activating receptor expression in high-risk cutaneous melanoma patients. Eur Cytokine Netw 2019; 30:160-167. [PMID: 32096478 DOI: 10.1684/ecn.2019.0440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2019] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Stage II melanoma patients have high risk for regional and distant metastases and may benefit from novel therapeutic strategies. To clarify the role of NK cells in Stage II melanoma, we characterized the cytotoxic activity of NK cells and the expression of various activating and inhibitory receptors in high-risk cutaneous melanoma patients (Stages IIB and IIC) compared to low-risk patients (Stage IA). MATERIALS AND METHODS Native and cytokine-treated peripheral blood mononuclear cells were used for functional and phenotypical analyses. RESULTS Compared to Stage IA-B patients, Stage IIB-C patients showed significantly decreased NK cell activity, as well as decreased expression of the activating NKG2D and CD161 receptors, most likely due to increased serum levels of the immunosuppressive cytokine TGF-β1 in these patients. Interestingly, treatment of periperal blood mononuclear cells with IFN-α, IL-2, IL-12 or the combination of IL-12 and IL-18 significantly induced NK cell activity for both groups of melanoma patients. However, only low-risk patients had a significant increase in the expression of the NKG2D receptor after in vitro treatment with IFN-α, as well as an significant increase in the expression of CD161 after treatment with IFN-α or IL-12. Although IL-2 induced the expression of NKG2D in both groups of patients, this increase was significantly lower in high-risk melanoma. CONCLUSION NK cell parameters may be useful as biomarkers of disease progression in localized melanoma patients. Our results further suggest that the use of NK cell-activating cytokines in combination with inhibitors of immunosuppressive factors like TGF-β1 could be a therapeutic option for the treatment of high-risk cutaneous melanoma patients.
Collapse
Affiliation(s)
- Katarina Mirjačić Martinović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Milica Milićević
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Annette K Larsen
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine, INSERM U938 and Sorbonne University, Kourilsky building 1st floor, Hôpital Saint-Antoine, 184 rue du Faubourg Saint Antoine, 75571 PARIS Cédex 12 France
| | - Radan Džodić
- Surgical Oncology Clinic, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia, School of Medicine, University of Belgrade, Dr Subotića 8, 11000 Beograd, Serbia
| | - Vladimir Jurišić
- Faculty of Medical Sciences, University of Kragujevac, P.BOX 124, 34000 Kragujevac, Serbia
| | - Gordana Konjević
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia, School of Medicine, University of Belgrade, Dr Subotića 8, 11000 Beograd, Serbia
| | - Ana Vuletić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| |
Collapse
|
276
|
Tahrali I, Kucuksezer UC, Akdeniz N, Altintas A, Uygunoglu U, Aktas-Cetin E, Deniz G. CD3 -CD56 + NK cells display an inflammatory profile in RR-MS patients. Immunol Lett 2019; 216:63-69. [PMID: 31589897 DOI: 10.1016/j.imlet.2019.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/22/2019] [Accepted: 10/03/2019] [Indexed: 01/06/2023]
Abstract
Multiple Sclerosis (MS) is an immune-mediated and neurodegenerative disease of central nervous system. Relapsing-remitting (RR)-MS occurring with acute attacks and remissions, is the most common clinical type of MS. There are different strategies applied in first-line treatment of RR-MS patients such as interferon-beta (IFN-β) and glatiramer acetate. In this study, activating and inhibitory receptor expressions and interleukin (IL)-22 levels of NK cells were investigated in RR-MS patients with or without IFN-β therapy. Activating receptor expression and IL-22 levels of NK cells were increased in RR-MS patients under IFN-β therapy. Elevated NK cells with activating profile and increased IL-22 under IFN-β therapy suggest that IFN-β treatment might direct NK cells toward a pro-inflammatory status.
Collapse
Affiliation(s)
- Ilhan Tahrali
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Umut Can Kucuksezer
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Nilgun Akdeniz
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Ayse Altintas
- Koc University, Faculty of Medicine, Department of Neurology, Istanbul, Turkey; Istanbul University Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Ugur Uygunoglu
- Istanbul University Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Esin Aktas-Cetin
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Gunnur Deniz
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey.
| |
Collapse
|
277
|
Hu J, Xia X, Gorlick R, Li S. Induction of NKG2D ligand expression on tumor cells by CD8 + T-cell engagement-mediated activation of nuclear factor-kappa B and p300/CBP-associated factor. Oncogene 2019; 38:7433-7446. [PMID: 31427736 PMCID: PMC6895417 DOI: 10.1038/s41388-019-0960-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/29/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022]
Abstract
The ligands for the natural killer group 2 (NKG2D) protein render tumor cells susceptible to NKG2D-dependent immune cell attack. However, cancer cells escape from immune surveillance by downregulating NKG2D ligands. We previously discovered that engagement of activated CD8+ T cells and tumor cells induces NKG2D ligands on tumor cells, but the underlying mechanism remains to be defined. Both in vivo mouse tumor models and in vitro cell assays were performed to study the downstream signaling. Our results supported the notion that, upon engagement with the cognate receptors, CD137 ligand and CD40 initiates activation of nuclear factor-kappa B (NF-κB) signaling in tumor cells even in the absence of CD8+ T cells. Like tumor and CD8+ T cell contact-dependent NKG2D ligand induction, this CD137L/CD40-mediated signaling activation was associated with elevated levels of acetyltransferase P300/CBP-associated factor (PCAF), whereas inhibition of phosphorylated NF-κB abrogated PCAF induction. Although stimulation of CD137L/CD40-mediated signaling is vital, inflammatory cytokines, including interferon gamma (IFNγ) and TNFα, also facilitate NKG2D ligand-induced immune surveillance via both facilitating T-cell chemotaxis and CD137L/CD40 induced NF-κB/PCAF activation. Collectively, our results unveil a novel mechanism of NKG2D ligand upregulation involving reverse signaling of CD40 and CD137L on tumor cells which, along with inflammatory cytokines IFNγ and TNFα, stimulate downstream NF-κB and PCAF activation. Understanding this mechanism may help in development of induced NKG2D ligand-dependent T-cell therapy against cancers.
Collapse
Affiliation(s)
- Jiemiao Hu
- Division of Pediatrics, Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xueqing Xia
- Division of Pediatrics, Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard Gorlick
- Division of Pediatrics, Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shulin Li
- Division of Pediatrics, Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
278
|
Lazarova M, Steinle A. Impairment of NKG2D-Mediated Tumor Immunity by TGF-β. Front Immunol 2019; 10:2689. [PMID: 31803194 PMCID: PMC6873348 DOI: 10.3389/fimmu.2019.02689] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor-β (TGF-β) suppresses innate and adaptive immune responses via multiple mechanisms. TGF-β also importantly contributes to the formation of an immunosuppressive tumor microenvironment thereby promoting tumor growth. Amongst others, TGF-β impairs tumor recognition by cytotoxic lymphocytes via NKG2D. NKG2D is a homodimeric C-type lectin-like receptor expressed on virtually all human NK cells and cytotoxic T cells, and stimulates their effector functions upon engagement by NKG2D ligands (NKG2DL). While NKG2DL are mostly absent from healthy cells, their expression is induced by cellular stress and malignant transformation, and, accordingly, frequently detected on various tumor cells. Hence, the NKG2D axis is thought to play a decisive role in cancer immunosurveillance and, obviously, often is compromised in clinically apparent tumors. There is mounting evidence that TGF-β, produced by tumor cells and immune cells in the tumor microenvironment, plays a key role in blunting the NKG2D-mediated tumor surveillance. Here, we review the current knowledge on the impairment of NKG2D-mediated cancer immunity through TGF-β and discuss therapeutic approaches aiming at counteracting this major immune escape pathway. By reducing tumor-associated expression of NKG2DL and blinding cytotoxic lymphocytes through down-regulation of NKG2D, TGF-β is acting upon both sides of the NKG2D axis severely compromising NKG2D-mediated tumor rejection. Consequently, novel therapies targeting the TGF-β pathway are expected to reinvigorate NKG2D-mediated tumor elimination and thereby to improve the survival of cancer patients.
Collapse
Affiliation(s)
- Mariya Lazarova
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
279
|
Mekonnen YA, Gültas M, Effa K, Hanotte O, Schmitt AO. Identification of Candidate Signature Genes and Key Regulators Associated With Trypanotolerance in the Sheko Breed. Front Genet 2019; 10:1095. [PMID: 31803229 PMCID: PMC6872528 DOI: 10.3389/fgene.2019.01095] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/11/2019] [Indexed: 12/23/2022] Open
Abstract
African animal trypanosomiasis (AAT) is caused by a protozoan parasite that affects the health of livestock. Livestock production in Ethiopia is severely hampered by AAT and various controlling measures were not successful to eradicate the disease. AAT affects the indigenous breeds in varying degrees. However, the Sheko breed shows better trypanotolerance than other breeds. The tolerance attributes of Sheko are believed to be associated with its taurine genetic background but the genetic controls of these tolerance attributes of Sheko are not well understood. In order to investigate the level of taurine background in the genome, we compare the genome of Sheko with that of 11 other African breeds. We find that Sheko has an admixed genome composed of taurine and indicine ancestries. We apply three methods: (i) The integrated haplotype score (iHS), (ii) the standardized log ratio of integrated site specific extended haplotype homozygosity between populations (Rsb), and (iii) the composite likelihood ratio (CLR) method to discover selective sweeps in the Sheko genome. We identify 99 genomic regions harboring 364 signature genes in Sheko. Out of the signature genes, 15 genes are selected based on their biological importance described in the literature. We also identify 13 overrepresented pathways and 10 master regulators in Sheko using the TRANSPATH database in the geneXplain platform. Most of the pathways are related with oxidative stress responses indicating a possible selection response against the induction of oxidative stress following trypanosomiasis infection in Sheko. Furthermore, we present for the first time the importance of master regulators involved in trypanotolerance not only for the Sheko breed but also in the context of cattle genomics. Our finding shows that the master regulator Caspase is a key protease which plays a major role for the emergence of adaptive immunity in harmony with the other master regulators. These results suggest that designing and implementing genetic intervention strategies is necessary to improve the performance of susceptible animals. Moreover, the master regulatory analysis suggests potential candidate therapeutic targets for the development of new drugs for trypanosomiasis treatment.
Collapse
Affiliation(s)
- Yonatan Ayalew Mekonnen
- Breeding Informatics Group, Department of Animal Sciences, University of Göttingen, Göttingen, Germany
| | - Mehmet Gültas
- Breeding Informatics Group, Department of Animal Sciences, University of Göttingen, Göttingen, Germany.,Center for Integrated Breeding Research (CiBreed), University of Göttingen, Göttingen, Germany
| | - Kefena Effa
- Animal Biosciences, National Program Coordinator for African Dairy Genetic Gain, International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
| | - Olivier Hanotte
- Cells, Organisms amd Molecular Genetics, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom.,LiveGene, International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
| | - Armin O Schmitt
- Breeding Informatics Group, Department of Animal Sciences, University of Göttingen, Göttingen, Germany.,Center for Integrated Breeding Research (CiBreed), University of Göttingen, Göttingen, Germany
| |
Collapse
|
280
|
Molfetta R, Zingoni A, Santoni A, Paolini R. Post-translational Mechanisms Regulating NK Cell Activating Receptors and Their Ligands in Cancer: Potential Targets for Therapeutic Intervention. Front Immunol 2019; 10:2557. [PMID: 31736972 PMCID: PMC6836727 DOI: 10.3389/fimmu.2019.02557] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022] Open
Abstract
Efficient clearance of transformed cells by Natural Killer (NK) cells is regulated by several activating receptors, including NKG2D, NCRs, and DNAM-1. Expression of these receptors as well as their specific “induced self” ligands is finely regulated during malignant transformation through the integration of different mechanisms acting on transcriptional, post-transcriptional, and post-translational levels. Among post-translational mechanisms, the release of activating ligands in the extracellular milieu through protease-mediated cleavage or by extracellular vesicle secretion represents some relevant cancer immune escape processes. Moreover, covalent modifications including ubiquitination and SUMOylation also contribute to negative regulation of NKG2D and DNAM-1 ligand surface expression resulting either in ligand intracellular retention and/or ligand degradation. All these mechanisms greatly impact on NK cell mediated recognition and killing of cancer cells and may be targeted to potentiate NK cell surveillance against tumors. Our mini review summarizes the main post-translational mechanisms regulating the expression of activating receptors and their ligands with particular emphasis on the contribution of ligand shedding and of ubiquitin and ubiquitin-like modifications in reducing target cell susceptibility to NK cell-mediated killing. Strategies aimed at inhibiting shedding of activating ligands and their modifications in order to preserve ligand expression on cancer cells will be also discussed.
Collapse
Affiliation(s)
- Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
281
|
Berry R, Watson GM, Jonjic S, Degli-Esposti MA, Rossjohn J. Modulation of innate and adaptive immunity by cytomegaloviruses. Nat Rev Immunol 2019; 20:113-127. [PMID: 31666730 DOI: 10.1038/s41577-019-0225-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2019] [Indexed: 02/07/2023]
Abstract
The coordinated activities of innate and adaptive immunity are critical for effective protection against viruses. To counter this, some viruses have evolved sophisticated strategies to circumvent immune cell recognition. In particular, cytomegaloviruses encode large arsenals of molecules that seek to subvert T cell and natural killer cell function via a remarkable array of mechanisms. Consequently, these 'immunoevasins' play a fundamental role in shaping the nature of the immune system by driving the evolution of new immune receptors and recognition mechanisms. Here, we review the diverse strategies adopted by cytomegaloviruses to target immune pathways and outline the host's response.
Collapse
Affiliation(s)
- Richard Berry
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia. .,Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia.
| | - Gabrielle M Watson
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Mariapia A Degli-Esposti
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Centre for Experimental Immunology, Lions Eye Institute, Perth, Western Australia, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
282
|
Sheehan C, D'Souza-Schorey C. Tumor-derived extracellular vesicles: molecular parcels that enable regulation of the immune response in cancer. J Cell Sci 2019; 132:132/20/jcs235085. [PMID: 31615844 DOI: 10.1242/jcs.235085] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous collection of membrane-bound vesicles released by cells that contain bioactive cargoes including proteins, lipids and nucleic acids. Multiple subpopulations of EVs have now been recognized and these include exosomes and microvesicles. EVs have been thought to facilitate intercellular and distal communication to bring about various processes that enable tumor progression and metastases. Here, we describe the current knowledge of the functional cargo contained within EVs, with a focus on tumor microvesicles, and review the emerging theory of how EVs support immune suppression in cancer.
Collapse
Affiliation(s)
- Colin Sheehan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556-0369, USA
| | | |
Collapse
|
283
|
Della Chiesa M, De Maria A, Muccio L, Bozzano F, Sivori S, Moretta L. Human NK Cells and Herpesviruses: Mechanisms of Recognition, Response and Adaptation. Front Microbiol 2019; 10:2297. [PMID: 31636622 PMCID: PMC6788305 DOI: 10.3389/fmicb.2019.02297] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/20/2019] [Indexed: 12/01/2022] Open
Abstract
NK cells contribute to early defenses against viruses through their inborn abilities that include sensing of PAMPs and inflammatory signals such as cytokines or chemokines, recognition, and killing of infected cells through activating surface receptors engagement. Moreover, they support adaptive responses via Ab-dependent mechanisms, triggered by CD16, and DC editing. Their fundamental role in anti-viral responses has been unveiled in patients with NK cell deficiencies suffering from severe Herpesvirus infections. Notably, these infections, often occurring as primary infections early in life, can be efficiently cleared by NK, T, and B cells in healthy hosts. Herpesviruses however, generate a complicated balance with the host immune system through their latency cycle moving between immune control and viral reactivation. This lifelong challenge has contributed to the development of numerous evasion mechanisms by Herpesviruses, many of which devoted to elude NK cell surveillance from viral reactivations rather than primary infections. This delicate equilibrium can be altered in proportions of healthy individuals promoting virus reactivation and, more often, in immunocompromised subjects. However, the constant stimulus provided by virus-host interplay has also favored NK-cell adaptation to Herpesviruses. During anti-HCMV responses, NK cells can reshape their receptor repertoire and function, through epigenetic remodeling, and acquire adaptive traits such as longevity and clonal expansion abilities. The major mechanisms of recognition and effector responses employed by NK cells against Herpesviruses, related to their genomic organization will be addressed, including those allowing NK cells to generate memory-like responses. In addition, the mechanisms underlying virus reactivation or control will be discussed.
Collapse
Affiliation(s)
- Mariella Della Chiesa
- Department of Experimental Medicine (DIMES), School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Andrea De Maria
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.,Department of Health Sciences (DISSAL), School of Medical and Pharmaceutical Sciences University of Genoa, Genoa, Italy.,Clinica Malattie Infettive, Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Letizia Muccio
- Department of Experimental Medicine (DIMES), School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Federica Bozzano
- Laboratory of Tumor Immunology, Department of Immunology, IRCCS Ospedale Bambino Gesù, Rome, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Lorenzo Moretta
- Laboratory of Tumor Immunology, Department of Immunology, IRCCS Ospedale Bambino Gesù, Rome, Italy
| |
Collapse
|
284
|
Lai H, Zeng D, Liu C, Zhang Q, Wang X, Chen T. Selenium-containing ruthenium complex synergizes with natural killer cells to enhance immunotherapy against prostate cancer via activating TRAIL/FasL signaling. Biomaterials 2019; 219:119377. [DOI: 10.1016/j.biomaterials.2019.119377] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022]
|
285
|
NK Cells as Potential Targets for Immunotherapy in Endometriosis. J Clin Med 2019; 8:jcm8091468. [PMID: 31540116 PMCID: PMC6780982 DOI: 10.3390/jcm8091468] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 12/19/2022] Open
Abstract
Endometriosis is a common gynecological disease defined by the presence of endometrial-like tissue outside the uterus, most frequently on the pelvic viscera and ovaries, which is associated with pelvic pains and infertility. It is an inflammatory disorder with some features of autoimmunity. It is accepted that ectopic endometriotic tissue originates from endometrial cells exfoliated during menstruation and disseminating into the peritoneum by retrograde menstrual blood flow. It is assumed that the survival of endometriotic cells in the peritoneal cavity may be partially due to their abrogated elimination by natural killer (NK) cells. The decrease of NK cell cytotoxic activity in endometriosis is associated with an increased expression of some inhibitory NK cell receptors. It may be also related to the expression of human leukocyte antigen G (HLA-G), a ligand for inhibitory leukocyte immunoglobulin-like receptor subfamily B member 1 (LILRB1) receptors. The downregulated cytotoxic activity of NK cells may be due to inhibitory cytokines present in the peritoneal milieu of patients with endometriosis. The role of NK cell receptors and their ligands in endometriosis is also confirmed by genetic association studies. Thus, endometriosis may be a subject of immunotherapy by blocking NK cell negative control checkpoints including inhibitory NK cell receptors. Immunotherapies with genetically modified NK cells also cannot be excluded.
Collapse
|
286
|
Zare N, Haghjooy Javanmard SH, Mehrzad V, Eskandari N, Andalib AR. Effect of Plasma-Derived Exosomes of Refractory/Relapsed or Responsive Patients with Diffuse Large B-Cell Lymphoma on Natural Killer Cells Functions. CELL JOURNAL 2019; 22:40-54. [PMID: 31606965 PMCID: PMC6791076 DOI: 10.22074/cellj.2020.6550] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/20/2019] [Indexed: 12/14/2022]
Abstract
Objective The purpose of this study was to investigate effect of plasma-derived exosomes of refractory/relapsed or
responsive diffuse large B-cell lymphoma (DLBCL) patients on natural killer (NK) cell functions.
Materials and Methods In this cross-sectional and experimental study, NK cells were purified from responsive patients
(n=10) or refractory/relapsed patients (n=12) and healthy donors (n=12). NK cells were treated with plasma-derived
exosomes of responsive or refractory/relapsed patients. We examined the expression levels of hsa-miR-155-5p, hsa-
let-7g-5p, INPP5D (SHIP-1) and SOCS-1 in NK cells quantitative reverse transcription-polymerase chain reaction
(qRT-PCR). Percentages of NK cells expressing CD69, NKG2D and CD16, NK cell cytotoxicity and NK cell proliferation
(using flow-cytometry) as well as interferon-gamma (IFN-γ) level in the supernatant of NK cells using ELISA were also
investigated.
Results We observed an increased level of hsa-miR-155-5p and a decreased level of SOCS-1 in NK cells
treated with exosomes compared to untreated NK cell in healthy donors and DLBCL patients. An increase in
hsa-miR-155-5p level was associated with an increased level of IFN-γ in healthy donors. The decreased levels
of hsa-let-7g-5p were observed in NK cells treated with exosomes in comparison with untreated NK cells in
DLBCL patients (P<0.05). There was no significant difference in the percentage of CD69+NK cells and NKG2D+
NK cells in the absence or presence of exosomes of DLBCL patients in each group. Furthermore, we observed
significant reduction of NK cell proliferation in DLBCL patients and healthy donors in the presence of exosomes
of refractory/relapsed patients (P<0.05). A significant decrease was observed in cytotoxicity of NK cell in patients
with DLBCL treated with exosomes of responsive patients.
Conclusion Our findings demonstrated adverse effect of plasma-derived exosomes of DLBCL patients on some functions
of NK cell. It was also determined that low NK cell count might be associated with impaired response to R-CHOP and an
increased recurrence risk of cancer.
Collapse
Affiliation(s)
- Nasrin Zare
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - S Haghayegh Haghjooy Javanmard
- Department of Physiology, School of Medicine and Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Valiollah Mehrzad
- Department of Hematology and Medical Oncology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.Electronic Address:
| | - Ali Reza Andalib
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
287
|
Zhao NH, Qian Y, Wu CS, Wang JW, Fang Y, Fan XP, Gao S, Fan YC, Wang K. Diagnostic value of NKG2D promoter methylation in hepatitis B virus-associated hepatocellular carcinoma. Biomark Med 2019; 13:1093-1105. [PMID: 31411040 DOI: 10.2217/bmm-2019-0102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Aim: Natural killer cell receptor group 2D (NKG2D) plays an important role in the immune regulation of tumors. We speculate that DNA methylation are involved in the regulation of NKG2D gene. Methods: We investigated the methylation status of the NKG2D promoter in peripheral blood mononuclear cells of hepatocellular carcinoma (HCC) patients, chronic hepatitis B patients and healthy controls by methylation-specific PCR and the mRNA expression level was examined by real-time quantitative PCR. Results: The methylation frequency of NKG2D promoter in HCC patients was higher than that of chronic hepatitis B patients and healthy controls. NKG2D promoter methylation has a good predictive value for HCC diagnosis. Conclusion: NKG2D promoter methylation can be used as a noninvasive marker for detecting HCC.
Collapse
Affiliation(s)
- Ning-Hui Zhao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Yu Qian
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Chen-Si Wu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Jing-Wen Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Yu Fang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Xiao-Peng Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
- Institute of Hepatology, Shandong University, Jinan 250012, PR China
| | - Shuai Gao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
- Institute of Hepatology, Shandong University, Jinan 250012, PR China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
- Institute of Hepatology, Shandong University, Jinan 250012, PR China
- Shenzhen Research Institute of Shandong University, Shenzhen 518000, PR China
| |
Collapse
|
288
|
Xie X, Ma L, Zhou Y, Shen W, Xu D, Dou J, Shen B, Zhou C. Polysaccharide enhanced NK cell cytotoxicity against pancreatic cancer via TLR4/MAPKs/NF-κB pathway in vitro/vivo. Carbohydr Polym 2019; 225:115223. [PMID: 31521276 DOI: 10.1016/j.carbpol.2019.115223] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022]
Abstract
A polysaccharide isolated from Strongylocentrotus nudus eggs (SEP) reportedly displays immune activity in vivo. Here, its effect and underlying mechanism in the treatment of pancreatic cancer were investigated. SEP obviously inhibited pancreatic cancer growth by activating NK cells in vitro/vivo via TLR4/MAPKs/NF-κB signaling pathway, The tumor inhibitory rate achieved to 44.5% and 50.8% at a dose of 40 mg/kg in Bxpc-3 and SW1990 nude mice, respectively. Moreover, SEP obviously augmented the Gemcitabine (GEM) antitumor effect by upregulating NKG2D, which improved the sensitivity of NK cells targeting to its ligand MICA; meanwhile, the antitumor inhibitory rate was 68.6% in BxPC-3 tumor-bearing mice. Moreover, SEP reversed GEM-induced apoptosis and atrophy in both spleen and bone marrow via suppressing ROS secretion in vivo. These results suggested that pancreatic cancer was effectively inhibited by SEP-enhanced NK cytotoxicity mediated primarily through TLR4/MAPKs/NF-κB signaling pathway, representing a potential immunotherapy candidate for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Xin Xie
- School of Life Science and Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, PR China
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, PR China
| | - Yiran Zhou
- Department of General Surgery, Rui Jin Hospital, Research Institute of Pancreatic Diseases, School of Medicine, Shanghai JiaoTong University, Shanghai, 200025, PR China
| | - Wen Shen
- School of Life Science and Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, PR China
| | - Duiyue Xu
- School of Life Science and Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, PR China
| | - Jie Dou
- School of Life Science and Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, PR China
| | - Baiyong Shen
- Department of General Surgery, Rui Jin Hospital, Research Institute of Pancreatic Diseases, School of Medicine, Shanghai JiaoTong University, Shanghai, 200025, PR China.
| | - Changlin Zhou
- School of Life Science and Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, PR China.
| |
Collapse
|
289
|
Abstract
The C-type lectins are a superfamily of proteins that recognize a broad repertoire of ligands and that regulate a diverse range of physiological functions. Most research attention has focused on the ability of C-type lectins to function in innate and adaptive antimicrobial immune responses, but these proteins are increasingly being recognized to have a major role in autoimmune diseases and to contribute to many other aspects of multicellular existence. Defects in these molecules lead to developmental and physiological abnormalities, as well as altered susceptibility to infectious and non-infectious diseases. In this Review, we present an overview of the roles of C-type lectins in immunity and homeostasis, with an emphasis on the most exciting recent discoveries.
Collapse
|
290
|
Papillary Thyroid Carcinoma Variants are Characterized by Co-dysregulation of Immune and Cancer Associated Genes. Cancers (Basel) 2019; 11:cancers11081179. [PMID: 31443155 PMCID: PMC6721495 DOI: 10.3390/cancers11081179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/08/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) variants exhibit different prognosis, but critical characteristics of PTC variants that contribute to differences in pathogenesis are not well-known. This study aims to characterize dysregulated immune-associated and cancer-associated genes in three PTC subtypes to explore how the interplay between cancer and immune processes causes differential prognosis. RNA-sequencing data from The Cancer Genome Atlas (TCGA) were used to identify dysregulated genes in each variant. The dysregulation profiles of the subtypes were compared using functional pathways clustering and correlations to relevant clinical variables, genomic alterations, and microRNA regulation. We discovered that the dysregulation profiles of classical PTC (CPTC) and the tall cell variant (TCPTC) are similar and are distinct from that of the follicular variant (FVPTC). However, unique cancer or immune-associated genes are associated with clinical variables for each subtype. Cancer-related genes MUC1, FN1, and S100-family members were the most clinically relevant in CPTC, while APLN and IL16, both immune-related, were clinically relevant in FVPTC. RAET-family members, also immune-related, were clinically relevant in TCPTC. Collectively, our data suggest that dysregulation of both cancer and immune associated genes defines the gene expression landscapes of PTC variants, but different cancer or immune related genes may drive the phenotype of each variant.
Collapse
|
291
|
Chen X, Chen X, Gao J, Yang H, Duan Y, Feng Y, He X, Gong X, Wang H, Wu X, Chang J. Astragaloside III Enhances Anti-Tumor Response of NK Cells by Elevating NKG2D and IFN-γ. Front Pharmacol 2019; 10:898. [PMID: 31456687 PMCID: PMC6701288 DOI: 10.3389/fphar.2019.00898] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/16/2019] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells play an irreplaceable role in the development of colon cancer, in which antitumor function of NK cells was impaired. Astragaloside III is a natural compound from Astragalus that has been shown to have immunomodulatory effects in various systems. However, few studies have evaluated the antitumor effects of Astragaloside III through stimulating systemic immunity and regulating NK cells. In this study, flow cytometry, immunohistochemical analysis, and immunofunctional assays were performed to elucidate the functions of Astragaloside III in restoring antitumor function of NK cells. We demonstrated that Astragaloside III significantly elevated the expression of natural killer group 2D (NKG2D), Fas, and interferon-γ (IFN-γ) production in NK cells, leading to increased tumor-killing ability. Experiments in cell co-culture assays and CT26-bearing mice model further confirmed that Astragaloside III could effectively impede tumor growth by increasing infiltration of NK cells into tumor and upregulating the antitumor response of NK cells. We further revealed that Astragaloside III increased IFN-γ secretion of NK cells by enhancing the expression of transcription factor T-bet. In conclusion, the effective anti-tumor function of Astragaloside III was achieved through up-regulation of the immune response of NK cells and elevation of NKG2D, Fas, and IFN-γ production.
Collapse
Affiliation(s)
- Xingmeng Chen
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Xi Chen
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junxiao Gao
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Han Yang
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Yue Duan
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Yuxin Feng
- Tianjin Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin He
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoqun Gong
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Hanjie Wang
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Xiaoli Wu
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Jin Chang
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| |
Collapse
|
292
|
Guo C, Wang X, Zhang H, Zhi L, Lv T, Li M, Lu C, Zhu W. Structure-based rational design of a novel chimeric PD1-NKG2D receptor for natural killer cells. Mol Immunol 2019; 114:108-113. [PMID: 31351411 DOI: 10.1016/j.molimm.2019.07.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/08/2019] [Accepted: 07/11/2019] [Indexed: 02/09/2023]
Abstract
Chimeric antigen receptor (CAR)-engineered natural killer (NK) cells have the potential to provide the potential for the implementation of allogeneic "off-the-shelf" cellular therapy against cancers. Currently, most CARs are not optimized for NK cells, so new NK-tailored CARs are needed. Here, a major activating receptor of NK cells, NKG2D was harnessed to design different chimeric receptors that mediate strong NK cell signaling. In these NKG2D signaling-based chimeric receptors, the extracellular domain of inhibitory receptor PD-1 was employed to reverse the immune escape mediated by PD-1 ligands in the solid tumors. To achieve the rational design of chimeric PD1-NKG2D receptors, we developed a transmembrane protein tertiary structure prediction program (PredMP & I-TASSER) and optimized the conformation of the PD-1 ectodomain by genetically altering the sequences encoding the hinge and intracellular domain. Finally, we identified a chimeric PD1-NKG2D receptor containing NKG2D hinge region and 4-1BB co-stimulatory domain to exhibit stable surface expression and mediate in vitro cytotoxicity of NK92 cells against various tumor cells. This strategy now provides a promising approach for the computer-aided design (CAD) of potent NK cell-tailored chimeric receptors with NKG2D signaling.
Collapse
Affiliation(s)
- Changjiang Guo
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Xiaoyin Wang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Huiyong Zhang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Lingtong Zhi
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Tanyu Lv
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Mingfeng Li
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Chengui Lu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Wuling Zhu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China.
| |
Collapse
|
293
|
Timmers M, Roex G, Wang Y, Campillo-Davo D, Van Tendeloo VFI, Chu Y, Berneman ZN, Luo F, Van Acker HH, Anguille S. Chimeric Antigen Receptor-Modified T Cell Therapy in Multiple Myeloma: Beyond B Cell Maturation Antigen. Front Immunol 2019; 10:1613. [PMID: 31379824 PMCID: PMC6646459 DOI: 10.3389/fimmu.2019.01613] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 06/28/2019] [Indexed: 12/27/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T cell therapy is a rapidly emerging immunotherapeutic approach that is revolutionizing cancer treatment. The impressive clinical results obtained with CAR-T cell therapy in patients with acute lymphoblastic leukemia and lymphoma have fueled the development of CAR-T cells targeting other malignancies, including multiple myeloma (MM). The field of CAR-T cell therapy for MM is still in its infancy, but remains promising. To date, most studies have been performed with B cell maturation antigen (BCMA)-targeted CARs, for which high response rates have been obtained in early-phase clinical trials. However, responses are usually temporary, and relapses have frequently been observed. One of the major reasons for relapse is the loss or downregulation of BCMA expression following CAR-T therapy. This has fostered a search for alternative target antigens that are expressed on the MM cell surface. In this review, we provide an overview of myeloma target antigens other than BCMA that are currently being evaluated in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Marijke Timmers
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Gils Roex
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Yuedi Wang
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Viggo F I Van Tendeloo
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Yiwei Chu
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Zwi N Berneman
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Feifei Luo
- Biotherapy Research Center, Fudan University, Shanghai, China.,Department of Digestive Diseases, Huashan Hospital of Fudan University, Shanghai, China
| | - Heleen H Van Acker
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Sébastien Anguille
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
294
|
Vazquez-Gonzalez WG, Martinez-Alvarez JC, Arrazola-Garcia A, Perez-Rodriguez M. Haplotype block 1 variant (HB-1v) of the NKG2 family of receptors. Hum Immunol 2019; 80:842-847. [PMID: 31320124 DOI: 10.1016/j.humimm.2019.07.276] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 01/04/2023]
Abstract
The natural killer group 2 (NKG2) family of receptors, encoded within the NK complex gene region (NKC), modulate the cytotoxic activity of NK cells. Two haplotype blocks throughout the NKC, hb-1 and hb-2 have been associated with different levels of overall natural cytotoxicity. Here, we evaluated allelic and genotype frequencies at rs1049174, rs2617160, rs2617170, rs2617171, rs1983526 (hb-1 haplotype), and rs2255336 and rs2246809 (hb-2 haplotype) in 928 subjects examined from Mexico City. The most frequent alleles and genotypes were as follows: C, CG to rs1049174; G, GG to rs2255336; T, AT to rs2617160; G, GG to rs2246809; C, CT to rs2617170; G, CG to rs2617171; and G, CG to rs1983526. Linkage disequilibrium analysis revealed that rs1049174, rs2617160, rs2617170, and rs2617171 constituted the haplotype block-1 variant (hb-1v) (r2 ≥ 0.89). Two predominant haplotypes of hb-1v were identified based on the allele content and included CTCG and GATC. This study is the first to evaluate the allelic and genotype frequency distribution of rs1049174, rs2255336, rs2617160, rs2246809, rs2617170, rs2617171, and rs1983526 in the population of Mexico City.
Collapse
Affiliation(s)
- Wendy Guadalupe Vazquez-Gonzalez
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Cuauhtémoc 330, Col. Doctores, CP 06720 Ciudad de México, Mexico
| | - Julio Cesar Martinez-Alvarez
- Banco Central de Sangre, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Cuauhtémoc 330, Col. Doctores, CP 06720 Ciudad de México, Mexico
| | - Araceli Arrazola-Garcia
- Banco Central de Sangre, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Cuauhtémoc 330, Col. Doctores, CP 06720 Ciudad de México, Mexico
| | - Martha Perez-Rodriguez
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Cuauhtémoc 330, Col. Doctores, CP 06720 Ciudad de México, Mexico.
| |
Collapse
|
295
|
Silva DOB, Correia NAA, de Barros FT, de Lima LPO, Morais A, Hassan R, Dellalibera E, de Mendonça Cavalcanti MDS, Muniz MTC. 3' untranslated region A>C (rs3212227) polymorphism of Interleukin 12B gene as a potential risk factor for Hodgkin's lymphoma in Brazilian children and adolescents. Tumour Biol 2019; 41:1010428319860400. [PMID: 31277552 DOI: 10.1177/1010428319860400] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Interleukin 12 plays an important role in immunoregulation between the T helper 1/T helper 2 lymphocytes and in the antiviral and antitumor immune response. The aim of this study was to investigate the possible association between the interleukin 12B polymorphism rs3212227 and the risk to develop Hodgkin's lymphoma in childhood and adolescents. A total of 100 patients with Hodgkin's lymphoma and a group of 181 healthy controls were selected at random from a forensic laboratory of the University of Pernambuco. The AA genotype was detected in the controls (53.04%) and the AC genotype was found in the patients (54%). The AC genotype showed an association with the development of Hodgkin's lymphoma (odds ratio = 2.091, 95% confidence interval = 1.240-3.523, p = 0.007). When AC + CC genotypes were analyzed together, an increase in risk of 1.9 times more chances for HL development could be observed (odds ratio = 1.923, 95% confidence interval = 1.166-3.170, p = 0.014). However, there was no association between the AC and CC genotypes of the interleukin 12B polymorphism with the clinical risk group (p = 0.992, p = 0.648, respectively). Our results suggest that the presence of the C allele may be contributing to the development of Hodgkin's lymphoma in children and adolescents.
Collapse
Affiliation(s)
- de Oliveira Bárbara Silva
- 1 Laboratory of Molecular Biology, Pediatric Hematology Oncology Center, University of Pernambuco, Recife, Brazil.,2 Biological Sciences Institute, University of Pernambuco, Recife, Brazil
| | - Nunes Amanda A Correia
- 1 Laboratory of Molecular Biology, Pediatric Hematology Oncology Center, University of Pernambuco, Recife, Brazil.,2 Biological Sciences Institute, University of Pernambuco, Recife, Brazil
| | - Ferreira Thiago de Barros
- 1 Laboratory of Molecular Biology, Pediatric Hematology Oncology Center, University of Pernambuco, Recife, Brazil.,3 Faculty of Medical Sciences, University of Pernambuco, Recife, Brazil
| | - Luísa Priscilla Oliveira de Lima
- 1 Laboratory of Molecular Biology, Pediatric Hematology Oncology Center, University of Pernambuco, Recife, Brazil.,2 Biological Sciences Institute, University of Pernambuco, Recife, Brazil
| | - Adriana Morais
- 4 Pediatric Outpatient Clinic, Pediatric Hematology Oncology Center, University of Pernambuco, Recife, Brazil
| | - Rocio Hassan
- 5 Oncovirology Laboratory, Bone Marrow Transplantation Center, National Cancer Institute of Brazil (INCA), Rio de Janeiro, Brazil
| | - Edileine Dellalibera
- 1 Laboratory of Molecular Biology, Pediatric Hematology Oncology Center, University of Pernambuco, Recife, Brazil
| | | | - Maria Tereza Cartaxo Muniz
- 1 Laboratory of Molecular Biology, Pediatric Hematology Oncology Center, University of Pernambuco, Recife, Brazil.,2 Biological Sciences Institute, University of Pernambuco, Recife, Brazil
| |
Collapse
|
296
|
Zhu M, Li M, Zhou W, Yang Y, Li F, Zhang L, Ji G. Qianggan extract improved nonalcoholic steatohepatitis by modulating lncRNA/circRNA immune ceRNA networks. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:156. [PMID: 31269941 PMCID: PMC6609373 DOI: 10.1186/s12906-019-2577-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 06/25/2019] [Indexed: 01/01/2023]
Abstract
Background The traditional Chinese medicine prescription, Qianggan formula have been confirmed to be effective on non-alcoholic steatohepatitis (NASH), however, the underlying molecular mechanisms remain obscure. Methods Thirty-six male C57BL/6 mice were randomly divided into three groups: normal chow diet group; methionine-and-choline-deficient diet (MCD) group, and Qianggan extract (QG) intervention group (0.4 g/kg daily) that fed with MCD. The efficacy of QG was biochemically and histologically evaluated. The expression profiles of messenger ribonucleic acids (mRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) were examined using microarray and verified by RT-qPCR. Results QG significantly improved the phenotypic characteristics of NASH, as serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and lactate dehydrogenase (LDH) levels and liver inflammatory cytokines were significantly decreased. By the cutoff of a 1.5-fold change and P < 0.05, 6193 mRNAs, 5692 lncRNAs and 4843 circRNAs were identified as differentially expressed between the MCD and normal groups, and 514 mRNAs, 1182 lncRNAs and 443 circRNAs were identified as differentially expressed between the QG and MCD groups. The intersections (244 mRNAs, 259 lncRNAs and 98 circRNAs) among the three groups were chosen for analysis. Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment revealed that most overlapping mRNAs were related to immune functions such as natural-killer-cell-mediated cytotoxicity, intestinal immune network for IgA production, and T cell receptor signaling pathway. Pathway interactions, protein-protein interactions and molecular complex detection (MCODE) analysis identified numerous immune-related hub genes e.g. natural cytotoxicity triggering receptor 1(Ncr1), C-X-C motif chemokine ligand 9 (Cxcl9), Klra1, and Cd28. Finally, two lncRNAs (Sngh1 and Slc36a3os) and four circRNAs (circ_0009029, circ_0004572, circ_0009212 and circ_0009453) in competing endogenous RNA (ceRNA) networks were constructed by Cytoscape, and immune-related mRNAs (e.g., Cd28, Cd8a, Il15, and Klrk1) were involved in the ceRNA networks. Conclusions LncRNA and circRNA-associated immune ceRNA networks might be the targets of QG in alleviating NASH, and our work may provide valuable clues for exploring the mechanisms underlying the effect of QG. Electronic supplementary material The online version of this article (10.1186/s12906-019-2577-6) contains supplementary material, which is available to authorized users.
Collapse
|
297
|
Futas J, Oppelt J, Jelinek A, Elbers JP, Wijacki J, Knoll A, Burger PA, Horin P. Natural Killer Cell Receptor Genes in Camels: Another Mammalian Model. Front Genet 2019; 10:620. [PMID: 31312212 PMCID: PMC6614441 DOI: 10.3389/fgene.2019.00620] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/13/2019] [Indexed: 12/23/2022] Open
Abstract
Due to production of special homodimeric heavy chain antibodies, somatic hypermutation of their T-cell receptor genes and unusually low diversity of their major histocompatibility complex genes, camels represent an important model for immunogenetic studies. Here, we analyzed genes encoding selected natural killer cell receptors with a special focus on genes encoding receptors for major histocompatibility complex (MHC) class I ligands in the two domestic camel species, Camelus dromedarius and Camelus bactrianus. Based on the dromedary genome assembly CamDro2, we characterized the genetic contents, organization, and variability of two complex genomic regions, the leukocyte receptor complex and the natural killer complex, along with the natural cytotoxicity receptor genes NCR1, NCR2, and NCR3. The genomic organization of the natural killer complex region of camels differs from cattle, the phylogenetically most closely related species. With its minimal set of KLR genes, it resembles this complex in the domestic pig. Similarly, the leukocyte receptor complex of camels is strikingly different from its cattle counterpart. With KIR pseudogenes and few LILR genes, it seems to be simpler than in the pig. The syntenies and protein sequences of the NCR1, NCR2, and NCR3 genes in the dromedary suggest that they could be human orthologues. However, only NCR1 and NCR2 have a structure of functional genes, while NCR3 appears to be a pseudogene. High sequence similarities between the two camel species as well as with the alpaca Vicugna pacos were observed. The polymorphism in all genes analyzed seems to be generally low, similar to the rest of the camel genomes. This first report on natural killer cell receptor genes in camelids adds new data to our understanding of specificities of the camel immune system and its functions, extends our genetic knowledge of the innate immune variation in dromedaries and Bactrian camels, and contributes to studies of natural killer cell receptors evolution in mammals.
Collapse
Affiliation(s)
- Jan Futas
- Department of Animal Genetics, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
- RG Animal Immunogenomics, CEITEC-VFU, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| | - Jan Oppelt
- RG Animal Immunogenomics, CEITEC-VFU, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
- National Centre for Biomolecular research, CEITEC-MU, Faculty of Science, Masaryk University, Brno, Czechia
| | - April Jelinek
- Department of Animal Genetics, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| | - Jean P. Elbers
- Research Institute for Wildlife Ecology, Department of Integrative Biology and Evolution, Vetmeduni Vienna, Vienna, Austria
| | - Jan Wijacki
- Department of Animal Morphology, Physiology and Genetics, Faculty of Agronomy, Mendel University in Brno, Brno, Czechia
- RG Animal Immunogenomics, CEITEC-MENDELU, Mendel University in Brno, Brno, Czechia
| | - Ales Knoll
- Department of Animal Morphology, Physiology and Genetics, Faculty of Agronomy, Mendel University in Brno, Brno, Czechia
- RG Animal Immunogenomics, CEITEC-MENDELU, Mendel University in Brno, Brno, Czechia
| | - Pamela A. Burger
- Research Institute for Wildlife Ecology, Department of Integrative Biology and Evolution, Vetmeduni Vienna, Vienna, Austria
| | - Petr Horin
- Department of Animal Genetics, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
- RG Animal Immunogenomics, CEITEC-VFU, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| |
Collapse
|
298
|
Zhang L, Li Y, Meng W, Ni Y, Gao Y. Dynamic urinary proteomic analysis in a Walker 256 intracerebral tumor model. Cancer Med 2019; 8:3553-3565. [PMID: 31090175 PMCID: PMC6601583 DOI: 10.1002/cam4.2240] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/17/2019] [Accepted: 04/19/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Patients with primary and metastatic brain cancer have an extremely poor prognosis, mostly due to the late diagnosis of disease. Urine, which lacks homeostatic mechanisms, is an ideal biomarker source that accumulates early and highly sensitive changes to provide information about the early stage of disease. METHODS A rat model mimicking the local tumor growth process in the brain was established with intracerebral Walker 256 (W256) cell injection. Urine samples were collected on days 3, 5, and 8 after injection, and then analyzed by liquid chromatography coupled with tandem mass spectrometry. RESULTS In the intracerebral W256 model, no obvious clinical manifestations or abnormal magnetic resonance imaging (MRI) signals were found on days 3 or 5; at these time points, 9 proteins were changed significantly in the urine of all eight tumor rats. On day 8, when tumors were detected by MRI, 25 differential proteins were identified, including 10 that have been reported to be closely related to brain metastasis or primary tumors. The differential urinary proteome was compared with those from the subcutaneous W256 model and the intracerebral C6 model. Few differential proteins overlapped, and specific differential protein patterns were observed among the three models. CONCLUSIONS These findings demonstrate that early changes in the urine proteome can be detected in the intracerebral W256 model. The urinary proteome can reflect the difference when tumor cells with different growth characteristics are inoculated into the brain and when identical tumor cells are inoculated into different areas, specifically, the subcutis and the brain.
Collapse
Affiliation(s)
- Linpei Zhang
- Department of Biochemistry and Molecular BiologyBeijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key LaboratoryBeijingChina
- BiobankThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yuqiu Li
- Department of Biochemistry and Molecular BiologyBeijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key LaboratoryBeijingChina
| | - Wenshu Meng
- Department of Biochemistry and Molecular BiologyBeijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key LaboratoryBeijingChina
| | - Yanying Ni
- Department of PathologyAviation General Hospital of China Medical UniversityBeijingChina
| | - Youhe Gao
- Department of Biochemistry and Molecular BiologyBeijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key LaboratoryBeijingChina
| |
Collapse
|
299
|
Juengpanich S, Shi L, Iranmanesh Y, Chen J, Cheng Z, Khoo AKJ, Pan L, Wang Y, Cai X. The role of natural killer cells in hepatocellular carcinoma development and treatment: A narrative review. Transl Oncol 2019; 12:1092-1107. [PMID: 31176993 PMCID: PMC6558093 DOI: 10.1016/j.tranon.2019.04.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 12/12/2022] Open
Abstract
A major obstacle for treatment of HCC is the inadequate efficacy and limitation of the available therapeutic options. Despite the recent advances in developing novel treatment options, HCC still remains one of the major causes of cancer morbidity and mortality around the world. Achieving effective treatment and eradication of HCC is a challenging task, however recent studies have shown that targeting Natural Killer cells, as major regulators of immune system, can help with the complete treatment of HCC, restoration of normal liver function and subsequently higher survival rate of HCC patients. Studies have shown that decrease in the frequency of NK cells, their dysfunction due to several factors such as dysregulation of receptors and their ligands, and imbalance of different types of inhibitory and stimulating microRNA expression is associated with higher rate of HCC progression and development, and poor survival outcome. Here in our review, we mainly focused on the importance of NK cells in HCC development and treatment.
Collapse
Affiliation(s)
- Sarun Juengpanich
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China; School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Liang Shi
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China.
| | | | - Jiang Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China; Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Zhenzhe Cheng
- School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Aaron Kah-Jin Khoo
- Faculty of Medicine, The University of Queensland, St Lucia, QLD, 4027, Australia.
| | - Long Pan
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China; School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Yifan Wang
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China.
| | - Xiujun Cai
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China.
| |
Collapse
|
300
|
Peixoto A, Relvas-Santos M, Azevedo R, Santos LL, Ferreira JA. Protein Glycosylation and Tumor Microenvironment Alterations Driving Cancer Hallmarks. Front Oncol 2019; 9:380. [PMID: 31157165 PMCID: PMC6530332 DOI: 10.3389/fonc.2019.00380] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Decades of research have disclosed a plethora of alterations in protein glycosylation that decisively impact in all stages of disease and ultimately contribute to more aggressive cell phenotypes. The biosynthesis of cancer-associated glycans and its reflection in the glycoproteome is driven by microenvironmental cues and these events act synergistically toward disease evolution. Such intricate crosstalk provides the molecular foundations for the activation of relevant oncogenic pathways and leads to functional alterations driving invasion and disease dissemination. However, it also provides an important source of relevant glyco(neo)epitopes holding tremendous potential for clinical intervention. Therefore, we highlight the transversal nature of glycans throughout the currently accepted cancer hallmarks, with emphasis on the crosstalk between glycans and the tumor microenvironment stromal components. Focus is also set on the pressing need to include glycans and glycoconjugates in comprehensive panomics models envisaging molecular-based precision medicine capable of improving patient care. We foresee that this may provide the necessary rationale for more comprehensive studies and molecular-based intervention.
Collapse
Affiliation(s)
- Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal.,Tumour and Microenvironment Interactions Group, INEB-Institute for Biomedical Engineering, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Marta Relvas-Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal
| | - Rita Azevedo
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Department of Surgical Oncology, Portuguese Institute of Oncology, Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal.,Porto Comprehensive Cancer Center, Porto, Portugal
| |
Collapse
|