1
|
Qin J, Zhang Z, Cui H, Yang J, Liu A. Biological characteristics and immune responses of NK Cells in commonly used experimental mouse models. Front Immunol 2024; 15:1478323. [PMID: 39628473 PMCID: PMC11611892 DOI: 10.3389/fimmu.2024.1478323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
The biology of natural killer (NK) cells in commonly used mouse models is discussed in this review, along with their crucial function in a variety of immunological responses. It has been demonstrated that the formation, maturation, subtype variety, and immunological recognition mechanisms of NK cells from various mice strains exhibit notable differences. These variations shed light on the intricacy of NK cell function and offer crucial information regarding their possible uses in treating human illnesses. The application of flow cytometry in mouse NK cell research is also covered in the article. Improved knowledge of the biology of NK cells across species may facilitate the development of new NK cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Jingwen Qin
- Department of Gastroenterology and Respiratory Internal Medicine & Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhaokai Zhang
- Department of General Surgery II, Cenxi People’s Hospital, Wuzhou, Guangxi, China
| | - Haopeng Cui
- Department of Gastroenterology and Respiratory Internal Medicine & Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jinhua Yang
- Department of Gastroenterology and Respiratory Internal Medicine & Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Aiqun Liu
- Department of Gastroenterology and Respiratory Internal Medicine & Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
2
|
Ustiuzhanina MO, Streltsova MA, Timofeev ND, Kryukov MA, Chudakov DM, Kovalenko EI. Autologous T-Cell-Free Antigen Presentation System Unveils hCMV-Specific NK Cell Response. Cells 2024; 13:530. [PMID: 38534374 PMCID: PMC10969127 DOI: 10.3390/cells13060530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
NK cells play a decisive role in controlling hCMV infection by combining innate and adaptive-like immune reactions. The hCMV-derived VMAPRTLFL (LFL) peptide is a potent activator of NKG2C+ NK cells. Proposed here is an autologous system of LFL stimulation without T lymphocytes and exogenous cytokines that allows us to evaluate NK-cell hCMV-specific responses in more native settings. In this model, we evaluated LFL-induced IFNγ production, focusing on signaling pathways and the degranulation and proliferation of NK cells orchestrated by microenvironment cytokine production and analyzed the transcriptome of expanded NK cells. NK cells of individuals having high anti-hCMV-IgG levels, in contrast to NK cells of hCMV-seronegative and low-positive donors, displayed increased IFNγ production and degranulation and activation levels and enhanced proliferation upon LFL stimulation. Cytokine profiles of these LFL-stimulated cultures demonstrated a proinflammatory shift. LFL-induced NK-cell IFNγ production was dependent on the PI3K and Ras/Raf/Mek signaling pathways, independently of cytokines. In hCMV-seropositive individuals, this model allowed obtaining NK-cell antigen-specific populations proliferating in response to LFL. The transcriptomic profile of these expanded NK cells showed increased adaptive gene expression and metabolic activation. The results complement the existing knowledge about hCMV-specific NK-cell response. This model may be further exploited for the identification and characterization of antigen-specific NK cells.
Collapse
Affiliation(s)
- Maria O. Ustiuzhanina
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (M.O.U.); (M.A.S.); (N.D.T.); (D.M.C.)
| | - Maria A. Streltsova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (M.O.U.); (M.A.S.); (N.D.T.); (D.M.C.)
| | - Nikita D. Timofeev
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (M.O.U.); (M.A.S.); (N.D.T.); (D.M.C.)
| | - Maxim A. Kryukov
- Ecole Polytechnique Federale de Lausanne, 1015 Lausanne, Switzerland;
| | - Dmitriy M. Chudakov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (M.O.U.); (M.A.S.); (N.D.T.); (D.M.C.)
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Central European Institute of Technology, Masaryk University, 60200 Brno, Czech Republic
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
| | - Elena I. Kovalenko
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (M.O.U.); (M.A.S.); (N.D.T.); (D.M.C.)
| |
Collapse
|
3
|
Muriuki BM, Forconi CS, Kirwa EK, Maina TK, Ariera BO, Bailey JA, Ghansah A, Moormann AM, Ong’echa JM. Evaluation of KIR3DL1/KIR3DS1 allelic polymorphisms in Kenyan children with endemic Burkitt lymphoma. PLoS One 2023; 18:e0275046. [PMID: 37647275 PMCID: PMC10468049 DOI: 10.1371/journal.pone.0275046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 08/15/2023] [Indexed: 09/01/2023] Open
Abstract
Endemic Burkitt lymphoma (eBL) is a fast-growing germinal center B cell lymphoma, affecting 5-10 per 100,000 children annually, in the equatorial belt of Africa. We hypothesize that co-infections with Plasmodium falciparum (Pf) malaria and Epstein-Barr virus (EBV) impair host natural killer (NK) and T cell responses to tumor cells, and thus increase the risk of eBL pathogenesis. NK cell education is partially controlled by killer immunoglobulin-like receptors and variable expression of KIR3DL1 has been associated with other malignancies. Here, we investigated whether KIR3D-mediated mechanisms contribute to eBL, by testing for an association of KIR3DL1/KIR3DS1 genotypes with the disease in 108 eBL patients and 99 healthy Kenyan children. KIR3DL1 allelic typing and EBV loads were assessed by PCR. We inferred previously observed phenotypes from the genotypes. The frequencies of KIR3DL1/KIR3DL1 and KIR3DL1/KIR3DS1 did not differ significantly between cases and controls. Additionally, none of the study participants was homozygous for KIR3DS1 alleles. EBV loads did not differ by the KIR3DL1 genotypes nor were they different between eBL survivors and non-survivors. Our results suggest that eBL pathogenesis may not simply involve variations in KIR3DL1 and KIR3DS1 genotypes. However, considering the complexity of the KIR3DL1 locus, this study could not exclude a role for copy number variation in eBL pathogenesis.
Collapse
Affiliation(s)
- Beatrice M. Muriuki
- West African Center for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Catherine S. Forconi
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States of America
| | - Erastus K. Kirwa
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Titus K. Maina
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Bonface O. Ariera
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Jeffrey A. Bailey
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States of America
| | - Anita Ghansah
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Ann M. Moormann
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States of America
| | - John M. Ong’echa
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| |
Collapse
|
4
|
Mosher BS, Kowalik TF, Yurochko AD. Overview of how HCMV manipulation of host cell intracellular trafficking networks can promote productive infection. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2022.1026452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human cytomegalovirus (HCMV) is a significant cause of morbidity and mortality in the immunocompromised and developing fetuses. Infection has also been linked to chronic inflammatory diseases, cardiovascular disease, and the development of certain cancers. The wide range of pathologies associated with HCMV infection is attributable to the broad cellular tropism of the virus where infection affects every organ system. Like other viruses, HCMV must tailor host cells to support productive infection. In particular, HCMV dedicates many resources and various strategies to manipulate host intracellular trafficking networks to facilitate various aspects of infection across all infected cell types. The dysregulation of host intracellular trafficking networks allows the virus to translocate to the host cell nucleus for genome replication, facilitate nuclear import/export of viral proteins and immature virions, subvert the host immune response, form new organelles for progeny virion assembly, maturation and egress, and promote cellular migration and viral spread. However, due to their complex nature, many aspects of these processes are not well-studied. New research and omics-based technologies have recently begun to elucidate the extent to which HCMV dysregulates host cell trafficking machinery. Here we review the variety of strategies HCMV utilizes to dysregulate intracellular trafficking networks to promote productive infection.
Collapse
|
5
|
Jones AB, Rocco A, Lamb LS, Friedman GK, Hjelmeland AB. Regulation of NKG2D Stress Ligands and Its Relevance in Cancer Progression. Cancers (Basel) 2022; 14:2339. [PMID: 35565467 PMCID: PMC9105350 DOI: 10.3390/cancers14092339] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
Under cellular distress, multiple facets of normal homeostatic signaling are altered or disrupted. In the context of the immune landscape, external and internal stressors normally promote the expression of natural killer group 2 member D (NKG2D) ligands that allow for the targeted recognition and killing of cells by NKG2D receptor-bearing effector populations. The presence or absence of NKG2D ligands can heavily influence disease progression and impact the accessibility of immunotherapy options. In cancer, tumor cells are known to have distinct regulatory mechanisms for NKG2D ligands that are directly associated with tumor progression and maintenance. Therefore, understanding the regulation of NKG2D ligands in cancer will allow for targeted therapeutic endeavors aimed at exploiting the stress response pathway. In this review, we summarize the current understanding of regulatory mechanisms controlling the induction and repression of NKG2D ligands in cancer. Additionally, we highlight current therapeutic endeavors targeting NKG2D ligand expression and offer our perspective on considerations to further enhance the field of NKG2D ligand biology.
Collapse
Affiliation(s)
- Amber B. Jones
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Abbey Rocco
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.R.); (G.K.F.)
| | | | - Gregory K. Friedman
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.R.); (G.K.F.)
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| |
Collapse
|
6
|
Nie Y, Liu D, Yang W, Li Y, Zhang L, Cheng X, Chen R, Yuan B, Zhang G, Wang H. Increased expression of TIGIT and KLRG1 correlates with impaired CD56 bright NK cell immunity in HPV16-related cervical intraepithelial neoplasia. Virol J 2022; 19:68. [PMID: 35413989 PMCID: PMC9003970 DOI: 10.1186/s12985-022-01776-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/03/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The onset and progression of cervical intraepithelial neoplasia (CIN) are closely associated with the persistent infection of high-risk HPV (especially type16), which is mainly caused by immune escape. Natural killer (NK) cells play an important role against virally infected cells and tumor cells through a fine balance of signals from multiple surface receptors. Overexpression of non-MHC-I specific inhibitory receptors TIGIT, KLRG1, Siglec-7, LAIR-1, and CD300a on NK cells correlates with cellular exhaustion and immune evasion, but these receptors have not been investigated in CIN. The aim of the present study was to examine the potential role of NK cell non-MHC-I specific inhibitory receptors expression in immune escape from HPV16(+)CIN patients. METHODS The subset distribution, IFN-γ and TNF-α expression levels and immunophenotype of TIGIT, KLRG1, Siglec-7, LAIR-1, and CD300a of NK cells were investigated in peripheral blood mononuclear cell samples by flow cytometry from 82 women who were HPV16(+) with CIN grades 0, I, II-III or HPV(-) CIN 0. Immunohistochemistry was applied to detect the expression of ligands for NK receptors in the cervical tissues. HPV types were identified by PCR assays. RESULTS The HPV16(+) subjects with high-grade lesions had an increased number of circulating peripheral blood CD56bright NK cells with reduced functionality and IFN-γ secretion. The expression levels of the inhibitory molecules TIGIT and KLRG1 on CD56bright NK cells increased in parallel with increasing CIN grade. In addition, TIGIT and KLRG1 related ligands, Poliovirus receptor (PVR), N-Cadherin and E-Cadherin expression level was also elevated with increasing CIN grade. CONCLUSIONS Our results suggest that up-regulation of the inhibitory TIGIT, KLRG1 and their ligands may negatively regulate cervical CD56bright NK-mediated immunity to HPV16 and contribute to the progression of CIN. These results may facilitate the development of early-warning immune predictors and therapeutic strategies for HPV16(+) CIN based on the TIGIT and KLRG1 inhibitory pathways of NK cells.
Collapse
Affiliation(s)
- You Nie
- Department of Pathology, Fourth Medical Centre of Chinese PLA (People's Liberation Army) General Hospital, 51 Fucheng Road, Beijing, China.,Basic Medicine College, Zhengzhou University, 100 Science Avenue, Henan, China
| | - Dandan Liu
- Department of Pathology, Fourth Medical Centre of Chinese PLA (People's Liberation Army) General Hospital, 51 Fucheng Road, Beijing, China
| | - Wen Yang
- Department of Gynaecology and Obstetrics, Chinese PLA (People's Liberation Army) General Hospital, Beijing, China
| | - Yazhuo Li
- Department of Pathology, Fourth Medical Centre of Chinese PLA (People's Liberation Army) General Hospital, 51 Fucheng Road, Beijing, China
| | - Lihua Zhang
- Department of Pathology, Fourth Medical Centre of Chinese PLA (People's Liberation Army) General Hospital, 51 Fucheng Road, Beijing, China
| | - Xia Cheng
- Department of Pathology, Fourth Medical Centre of Chinese PLA (People's Liberation Army) General Hospital, 51 Fucheng Road, Beijing, China
| | - Ruyu Chen
- Department of Pathology, Fourth Medical Centre of Chinese PLA (People's Liberation Army) General Hospital, 51 Fucheng Road, Beijing, China
| | - Bingbing Yuan
- Basic Medicine College, Zhengzhou University, 100 Science Avenue, Henan, China
| | - Guangzheng Zhang
- Basic Medicine College, Zhengzhou University, 100 Science Avenue, Henan, China.
| | - Hongwei Wang
- Department of Pathology, Fourth Medical Centre of Chinese PLA (People's Liberation Army) General Hospital, 51 Fucheng Road, Beijing, China.
| |
Collapse
|
7
|
Kheshtchin N, Bakhshi P, Arab S, Nourizadeh M. Immunoediting in SARS-CoV-2: Mutual relationship between the virus and the host. Int Immunopharmacol 2022; 105:108531. [PMID: 35074569 PMCID: PMC8743495 DOI: 10.1016/j.intimp.2022.108531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 11/05/2022]
Abstract
Immunoediting is a well-known concept that occurs in cancer through three steps of elimination, equilibrium, and escape (3Es), where the immune system first suppresses the growth of tumor cells and then promotes them towards the malignancy. This phenomenon has been conceptualized in some chronic viral infections such as HTLV-1 and HIV by obtaining the resistance to elimination and making a persistent form of infected cells especially in untreated patients. Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a heterogeneous disease characterizing from mild/asymptomatic to severe/critical courses with some behavioral aspects in an immunoediting setting. In this context, a coordinated effort between innate and adaptive immune system leads to detection and destruction of early infection followed by equilibrium between virus-specific responses and infected cells, which eventually ends up with an uncontrolled inflammatory response in severe/critical patients. Although the SARS-CoV-2 applies several escape strategies such as mutations in viral epitopes, modulating the interferon response and inhibiting the MHC I molecules similar to the cancer cells, the 3Es hallmark may not occur in all clinical conditions. Here, we discuss how the lesson learnt from cancer immunoediting and accurate understanding of these pathophysiological mechanisms helps to develop more effective therapeutic strategies for COVID-19.
Collapse
|
8
|
Christodoulou I, Rahnama R, Ravich JW, Seo J, Zolov SN, Marple AN, Markovitz DM, Bonifant CL. Glycoprotein Targeted CAR-NK Cells for the Treatment of SARS-CoV-2 Infection. Front Immunol 2022; 12:763460. [PMID: 35003077 PMCID: PMC8732772 DOI: 10.3389/fimmu.2021.763460] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022] Open
Abstract
H84T-Banana Lectin (BanLec) CAR-NK cells bind high mannose glycosites that decorate the SARS-CoV-2 envelope, thereby decreasing cellular infection in a model of SARS-CoV-2. H84T-BanLec CAR-NK cells are innate effector cells, activated by virus. This novel cellular agent is a promising therapeutic, capable of clearing circulating SARS-CoV-2 virus and infected cells. Banana Lectin (BanLec) binds high mannose glycans on viral envelopes, exerting an anti-viral effect. A point mutation (H84T) divorces BanLec mitogenicity from antiviral activity. SARS-CoV-2 contains high mannose glycosites in proximity to the receptor binding domain of the envelope Spike (S) protein. We designed a chimeric antigen receptor (CAR) that incorporates H84T-BanLec as the extracellular moiety. Our H84T-BanLec CAR was devised to specifically direct NK cell binding of SARS-CoV-2 envelope glycosites to promote viral clearance. The H84T-BanLec CAR was stably expressed at high density on primary human NK cells during two weeks of ex vivo expansion. H84T-BanLec CAR-NK cells reduced S-protein pseudotyped lentiviral infection of 293T cells expressing ACE2, the receptor for SARS-CoV-2. NK cells were activated to secrete inflammatory cytokines when in culture with virally infected cells. H84T-BanLec CAR-NK cells are a promising cell therapy for further testing against wild-type SARS-CoV-2 virus in models of SARS-CoV-2 infection. They may represent a viable off-the-shelf immunotherapy for patients suffering from COVID-19.
Collapse
Affiliation(s)
- Ilias Christodoulou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ruyan Rahnama
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonas W Ravich
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jaesung Seo
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sergey N Zolov
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Andrew N Marple
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David M Markovitz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.,Division of Infectious Diseases, Department of Internal Medicine, and the Programs in Immunology, Cellular and Molecular Biology, and Cancer Biology, University of Michigan, Ann Arbor, MI, United States
| | - Challice L Bonifant
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Khan AA, Liu ZK, Xu X. Recent advances in immunotherapy for hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2021; 20:511-520. [PMID: 34344612 DOI: 10.1016/j.hbpd.2021.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Treatment of hepatocellular carcinoma (HCC) is challenging as most patients are diagnosed at advanced stage with underlying chronic liver conditions. Conventional systemic chemotherapy has failed in HCC, and the clinical efficacy of FDA-approved molecular targeted agents such as sorafenib and lenvatinib remains unsatisfactory. DATA SOURCES Literature search was conducted in PubMed for relevant articles published before January 2021. The search aimed to identify recent developments in immune-based treatment approaches for HCC. Information of clinical trials was obtained from https://clinicaltrials.gov/. RESULTS Two immune checkpoint inhibitors (ICIs), nivolumab and pembrolizumab were approved as monotherapies, which has revolutionized HCC treatment. Besides, combination ICIs have also got accelerated FDA approval recently. Immune-based therapies have challenged targeted drugs owing to their safety, tolerability, and survival benefits. In addition to the significant success in ICIs, other immunotherapeutic strategies such as cancer vaccine, chimeric antigen receptor T-cells, natural killer cells, cytokines, and combination therapy, have also shown promising outcomes in clinical trials. Various diagnostic and prognostic biomarkers have been identified which can help in clinical decision making when starting treatment with ICIs. CONCLUSIONS Immunotherapy has emerged as one of the mainstream treatment modalities for advanced HCC in recent years. However, challenges such as low response rate and acquired resistance in previously respondent patients still exist. Further research is needed to understand the unique resistance mechanism to immunotherapy and to discover more predictive biomarkers to guide clinical decision making.
Collapse
Affiliation(s)
- Abid Ali Khan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Lab of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Public Health, Hangzhou 310003, China
| | - Zhi-Kun Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Lab of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Public Health, Hangzhou 310003, China
| | - Xiao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Lab of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Public Health, Hangzhou 310003, China; Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
10
|
Clinical trial to analyze the effects of oral intake of Phellinus linteus (sanghuang) extract on immune function: a study protocol for a randomized, double-blind controlled trial. Trials 2021; 22:849. [PMID: 34838112 PMCID: PMC8627044 DOI: 10.1186/s13063-021-05740-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022] Open
Abstract
Background As the population of Korea ages, interest in healthcare has increased. In particular, there is an increasing demand for immune-function improvement to prevent infectious diseases. Phellinus linteus (PL) has previously been shown to exert immune-enhancing and anticancer effects. We aim to evaluate whether PL mycelium extract, cultured from the PL KCTC0399BP strain, can increase immune function, as measured using blood-test indicators. This clinical trial protocol is designed as the main trial and is based on the results of a pilot study. Methods This clinical trial is a randomized, double-blinded, placebo-controlled trial. Ninety-eight participants are enrolled and randomly divided into two groups: the experimental group (PL 1000 mg) and the control group (placebo). Participants are administered with experimental food or placebo for eight weeks. Blood tests are performed before trial initiation and 8 weeks later, at trial completion. Laboratory evaluation items are as follows: natural killer cell activity, tumor necrosis factor-α, interferon-γ, interleukin (IL)-1β, IL-2, IL-6, IL-12, immunoglobulin (Ig)G1, IgG2, and IgM. We will mainly use the full analysis dataset to statistically analyze the effectiveness of the treatment. Discussion This study evaluates the effects of PL extract on immune function and will contribute to knowledge on the value of PL as an immune-function–boosting functional food. Trial registration Clinical Research Information Service (CRIS) of Korea CRIS-KCT0005460. Registered on 12 October 2020
Collapse
|
11
|
Mortimer NT, Fischer ML, Waring AL, Kr P, Kacsoh BZ, Brantley SE, Keebaugh ES, Hill J, Lark C, Martin J, Bains P, Lee J, Vrailas-Mortimer AD, Schlenke TA. Extracellular matrix protein N-glycosylation mediates immune self-tolerance in Drosophila melanogaster. Proc Natl Acad Sci U S A 2021; 118:e2017460118. [PMID: 34544850 PMCID: PMC8488588 DOI: 10.1073/pnas.2017460118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2021] [Indexed: 12/26/2022] Open
Abstract
In order to respond to infection, hosts must distinguish pathogens from their own tissues. This allows for the precise targeting of immune responses against pathogens and also ensures self-tolerance, the ability of the host to protect self tissues from immune damage. One way to maintain self-tolerance is to evolve a self signal and suppress any immune response directed at tissues that carry this signal. Here, we characterize the Drosophila tuSz1 mutant strain, which mounts an aberrant immune response against its own fat body. We demonstrate that this autoimmunity is the result of two mutations: 1) a mutation in the GCS1 gene that disrupts N-glycosylation of extracellular matrix proteins covering the fat body, and 2) a mutation in the Drosophila Janus Kinase ortholog that causes precocious activation of hemocytes. Our data indicate that N-glycans attached to extracellular matrix proteins serve as a self signal and that activated hemocytes attack tissues lacking this signal. The simplicity of this invertebrate self-recognition system and the ubiquity of its constituent parts suggests it may have functional homologs across animals.
Collapse
Affiliation(s)
- Nathan T Mortimer
- School of Biological Sciences, Illinois State University, Normal, IL 61790;
| | - Mary L Fischer
- School of Biological Sciences, Illinois State University, Normal, IL 61790
| | - Ashley L Waring
- School of Biological Sciences, Illinois State University, Normal, IL 61790
| | - Pooja Kr
- School of Biological Sciences, Illinois State University, Normal, IL 61790
| | - Balint Z Kacsoh
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - Susanna E Brantley
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| | | | - Joshua Hill
- School of Biological Sciences, Illinois State University, Normal, IL 61790
| | - Chris Lark
- School of Biological Sciences, Illinois State University, Normal, IL 61790
| | - Julia Martin
- School of Biological Sciences, Illinois State University, Normal, IL 61790
| | - Pravleen Bains
- School of Biological Sciences, Illinois State University, Normal, IL 61790
| | - Jonathan Lee
- School of Biological Sciences, Illinois State University, Normal, IL 61790
| | | | - Todd A Schlenke
- Department of Entomology, University of Arizona, Tucson, AZ 85719
| |
Collapse
|
12
|
Maucourant C, Nonato Queiroz GA, Corneau A, Leandro Gois L, Meghraoui-Kheddar A, Tarantino N, Bandeira AC, Samri A, Blanc C, Yssel H, Rios Grassi MF, Vieillard V. NK Cell Responses in Zika Virus Infection Are Biased towards Cytokine-Mediated Effector Functions. THE JOURNAL OF IMMUNOLOGY 2021; 207:1333-1343. [PMID: 34408012 DOI: 10.4049/jimmunol.2001180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 06/23/2021] [Indexed: 12/30/2022]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that has emerged as a global concern because of its impact on human health. ZIKV infection during pregnancy can cause microcephaly and other severe brain defects in the developing fetus and there have been reports of the occurrence of Guillain-Barré syndrome in areas affected by ZIKV. NK cells are activated during acute viral infections and their activity contributes to a first line of defense because of their ability to rapidly recognize and kill virus-infected cells. To provide insight into NK cell function during ZIKV infection, we have profiled, using mass cytometry, the NK cell receptor-ligand repertoire in a cohort of acute ZIKV-infected female patients. Freshly isolated NK cells from these patients contained distinct, activated, and terminally differentiated, subsets expressing higher levels of CD57, NKG2C, and KIR3DL1 as compared with those from healthy donors. Moreover, KIR3DL1+ NK cells from these patients produced high levels of IFN-γ and TNF-α, in the absence of direct cytotoxicity, in response to in vitro stimulation with autologous, ZIKV-infected, monocyte-derived dendritic cells. In ZIKV-infected patients, overproduction of IFN-γ correlated with STAT-5 activation (r = 0.6643; p = 0.0085) and was mediated following the recognition of MHC class 1-related chain A and chain B molecules expressed by ZIKV-infected monocyte-derived dendritic cells, in synergy with IL-12 production by the latter cells. Together, these findings suggest that NK cells contribute to the generation of an efficacious adaptive anti-ZIKV immune response that could potentially affect the outcome of the disease and/or the development of persistent symptoms.
Collapse
Affiliation(s)
- Christopher Maucourant
- Sorbonne Université, UPMC, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | | | - Aurelien Corneau
- UPMC Univ Paris 06, Plateforme de Cytométrie, UMS30-LUMIC, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris, France; and
| | - Luana Leandro Gois
- FIOCRUZ, Salvador, Brazil.,Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Aida Meghraoui-Kheddar
- Sorbonne Université, UPMC, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Nadine Tarantino
- Sorbonne Université, UPMC, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | | | - Assia Samri
- Sorbonne Université, UPMC, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Catherine Blanc
- UPMC Univ Paris 06, Plateforme de Cytométrie, UMS30-LUMIC, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris, France; and
| | - Hans Yssel
- Sorbonne Université, UPMC, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | | | - Vincent Vieillard
- Sorbonne Université, UPMC, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France;
| |
Collapse
|
13
|
Gardner G, Fraker CA. Natural Killer Cells as Key Mediators in Type I Diabetes Immunopathology. Front Immunol 2021; 12:722979. [PMID: 34489972 PMCID: PMC8417893 DOI: 10.3389/fimmu.2021.722979] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/05/2021] [Indexed: 01/03/2023] Open
Abstract
The immunopathology of type I diabetes (T1D) presents a complicated case in part because of the multifactorial origin of this disease. Typically, T1D is thought to occur as a result of autoimmunity toward islets of Langerhans, resulting in the destruction of insulin-producing cells (β cells) and thus lifelong reliance on exogenous insulin. However, that explanation obscures much of the underlying mechanism, and the actual precipitating events along with the associated actors (latent viral infection, diverse immune cell types and their roles) are not completely understood. Notably, there is a malfunctioning in the regulation of cytotoxic CD8+ T cells that target endocrine cells through antigen-mediated attack. Further examination has revealed the likelihood of an imbalance in distinct subpopulations of tolerogenic and cytotoxic natural killer (NK) cells that may be the catalyst of adaptive immune system malfunction. The contributions of components outside the immune system, including environmental factors such as chronic viral infection also need more consideration, and much of the recent literature investigating the origins of this disease have focused on these factors. In this review, the details of the immunopathology of T1D regarding NK cell disfunction is discussed, along with how those mechanisms stand within the context of general autoimmune disorders. Finally, the rarer cases of latent autoimmune, COVID-19 (viral), and immune checkpoint inhibitor (ICI) induced diabetes are discussed as their exceptional pathology offers insight into the evolution of the disease as a whole.
Collapse
Affiliation(s)
| | - Christopher A. Fraker
- Tissue and Biomedical Engineering Laboratory, Leonard M. Miller School of Medicine, Diabetes Research Institute, University of Miami, Miami, FL, United States
| |
Collapse
|
14
|
Mahgoub S, Abosalem H, Emara M, Kotb N, Maged A, Soror S. Restoring NK cells functionality via cytokine activation enhances cetuximab-mediated NK-cell ADCC: A promising therapeutic tool for HCC patients. Mol Immunol 2021; 137:221-227. [PMID: 34284214 DOI: 10.1016/j.molimm.2021.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/08/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022]
Abstract
Natural Killer (NK) cells are considered the first line of defense against viral infections and tumors. Several factors affect NK cytotoxic activity rendering it dysfunctional and thereby impeding the ability to scavenge abnormal cells as a part of immune escaping mechanisms induced by different types of cancers. NK cells play a crucial role augmenting the activity of various types of anticancer mAb since dysfunctional NK cells are the main reason for the low response to these therapies. To this light, we examined the phenotypic characters of the circulating NK cells isolated from HCC patients compared to healthy controls. Then, dysfunctional NK cells, from HCC patients, were reactivated with cytokines cocktail and their cytotoxic activity with the anti-EGFR mAb "cetuximab" was investigated. This showed a downregulation of patients NK cells activating receptors (NKP30, NKP46, NKG2D and CD16) as well as CD56 and up-regulation of NKG2A inhibitory receptor. We also reported an increase in aberrant CD56- NK cells subset in peripheral blood of HCC patients compared to healthy controls. Thus, confirming the dysfunctionality of peripheral NK cells isolated from HCC patients. Cytokines re-activation of those NK cells lead to upregulation of NK activating receptors and downregulation of inhibitory receptor. Moreover, the percentage of aberrant CD56- NK cells subset was reduced. Here, we proved that advanced HCC patients have an increased percentage of more immature and noncytotoxic NK cell subsets in their peripheral blood, which might account for the low cytotoxicity noticed in those patients. A significant improvement in the cytotoxicity against HCC was noticed upon using reactivated NK cells combined with cetuximab. Therefore, this study highlights the potential recruitment of NK immune cells along with cetuximab to enhance cytotoxicity against HCC.
Collapse
Affiliation(s)
- Shahenda Mahgoub
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Ein-Helwan, Cairo, 11795, Egypt.
| | - Hadeer Abosalem
- Deputy of Technical Manager, Biotechnology Unit, Egyptian Drug Authority (EDA), Giza, 12654, Egypt.
| | - Mohamed Emara
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ein-Helwan, Cairo, 11795, Egypt.
| | - Nahla Kotb
- Manager of Blood Derivative Unit, Egyptian Drug Authority (EDA), Giza, 12654, Egypt.
| | - A Maged
- National Hepatology and Tropical Medicine Research Institute (NHTMRI), 11441, Cairo, Egypt.
| | - Sameh Soror
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Ein-Helwan, Cairo, 11795, Egypt.
| |
Collapse
|
15
|
Santaclara V, Torres-Moreno D, Bernal-Mañas CM, Isaac MA, Ortiz-Reina S, Conesa-Zamora P. Relationship between polymorphisms in the FAS/FASL death receptor system and progression of low-grade precursor lesions infected with high-risk human papilloma virus. Hum Immunol 2021; 82:621-624. [PMID: 34127318 DOI: 10.1016/j.humimm.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 11/19/2022]
Abstract
Squamous intraepithelial lesions (SIL) and cervical cancer are primary due to suboptimal immune response against human papillomavirus (HPV). The FASL/FAS system is a trigger of extrinsic pathway apoptosis. The distribution of polymorphisms rs1800682 (-670 A > G) FAS and rs763110 (-844C > T) FASL was studied in cervical smears from 372 females (182 with stable or regressed low-grade SIL (LSIL) (groupI) and a group of 190 high-grade SIL (HSIL) (groupII). No significant differences were observed for rs1800682 in FAS between the study groups. In contrast, rs763110 CC genotype of FASL was found in 35.7% of group I females, and in 50.5% of group II (p = 0.0027; OR = 1.83 (95% CI = 1.21-2.79)). When only females infected with high-risk HPV were analysed, these differences were even higher (p = 0.0024; OR = 2.21 (95% CI = 1.30-3.75)). CC genotype in FASL seems to be associated with increased risk of LSIL to HSIL progression suggesting a role in HPV tolerance, persistent infection, and HSIL development.
Collapse
Affiliation(s)
| | | | | | - María Alejandra Isaac
- Pathology Department, Santa Lucía University Hospital (HGUSL), Spain; Facultad de Ciencias de la Salud, Catholic University of Murcia (UCAM), Murcia, Spain
| | | | - Pablo Conesa-Zamora
- Facultad de Ciencias de la Salud, Catholic University of Murcia (UCAM), Murcia, Spain; Clinical Analysis Department, HGUSL, Cartagena, Spain; Molecular Pathology and Pharmacogenetic Group. Institute for Biohealth Research from Murcia (IMIB), HGUSL, Cartagena, Spain.
| |
Collapse
|
16
|
Muriuki BM, Forconi CS, Oluoch PO, Bailey JA, Ghansah A, Moormann AM, Ong'echa JM. Association of killer cell immunoglobulin-like receptors with endemic Burkitt lymphoma in Kenyan children. Sci Rep 2021; 11:11343. [PMID: 34059753 PMCID: PMC8166913 DOI: 10.1038/s41598-021-90596-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/09/2021] [Indexed: 02/04/2023] Open
Abstract
Endemic Burkitt lymphoma (eBL) is an aggressive pediatric B cell lymphoma, common in Equatorial Africa. Co-infections with Epstein-Barr virus (EBV) and Plasmodium falciparum, coupled with c-myc translocation are involved in eBL etiology. Infection-induced immune evasion mechanisms to avoid T cell cytotoxicity may increase the role of Natural killer (NK) cells in anti-tumor immunosurveillance. Killer immunoglobulin-like receptor (KIR) genes on NK cells exhibit genotypic and allelic variations and are associated with susceptibility to diseases and malignancies. However, their role in eBL pathogenesis remains undefined. This retrospective study genotyped sixteen KIR genes and compared their frequencies in eBL patients (n = 104) and healthy geographically-matched children (n = 104) using sequence-specific primers polymerase chain reaction (SSP-PCR) technique. The relationship between KIR polymorphisms with EBV loads and eBL pathogenesis was investigated. Possession of ≥ 4 activating KIRs predisposed individuals to eBL (OR = 3.340; 95% CI 1.530-7.825; p = 0.004). High EBV levels were observed in Bx haplogroup (p = 0.016) and AB genotypes (p = 0.042) relative to AA haplogroup and AA genotype respectively, in eBL patients but not in healthy controls. Our results suggest that KIR-mediated NK cell stimulation could mute EBV control, contributing to eBL pathogenesis.
Collapse
Affiliation(s)
- Beatrice M Muriuki
- West African Center for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Catherine S Forconi
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Peter O Oluoch
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jeffrey A Bailey
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Anita Ghansah
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Ann M Moormann
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - John M Ong'echa
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.
| |
Collapse
|
17
|
Grudzien M, Pawlak A, Kutkowska J, Ziolo E, Wysokińska E, Hildebrand W, Obmińska-Mrukowicz B, Strzadala L, Rapak A. A newly established canine NK-type cell line and its cytotoxic properties. Vet Comp Oncol 2021; 19:567-577. [PMID: 33774906 DOI: 10.1111/vco.12695] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/30/2022]
Abstract
We established a canine natural killer (NK)-type cell line called CNK-89 derived from a dog with NK cell neoplasia. Immunophenotyping analysis showed positive staining for CD5, CD8, CD45, CD56, CD79a and NKp46, while negative for CD3, CD4, CD14, CD20, CD21, CD34, Thy1, IgG, IgM and MHCII. Polymerase chain reaction analysis revealed the presence of CD56, NKG2D, NKp30, NKp44, NKp46 and perforin, but the absence of CD16, Ly49 and granzyme B mRNA. Treating CNK-89 cells with IL-2 did not change the expression of activating receptors, TNFα and IFNγ secretion and cytotoxic activity, however, treatment with IL-12 alone or in combinations with IL-15, IL-18 and IL-21 caused an increase in granzyme B and CD16 mRNA, IFNγ secretion and cytotoxic properties of the CNK-89 cell line.
Collapse
Affiliation(s)
- Malgorzata Grudzien
- Laboratory of Tumor Molecular Immunobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Aleksandra Pawlak
- Department of Biochemistry, Pharmacology and Toxicology, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Justyna Kutkowska
- Laboratory of Tumor Molecular Immunobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Ewa Ziolo
- Laboratory of Tumor Molecular Immunobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Edyta Wysokińska
- Laboratory of Tumor Molecular Immunobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | | | - Bożena Obmińska-Mrukowicz
- Department of Biochemistry, Pharmacology and Toxicology, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Leon Strzadala
- Laboratory of Tumor Molecular Immunobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Andrzej Rapak
- Laboratory of Tumor Molecular Immunobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| |
Collapse
|
18
|
Malouli D, Hansen SG, Hancock MH, Hughes CM, Ford JC, Gilbride RM, Ventura AB, Morrow D, Randall KT, Taher H, Uebelhoer LS, McArdle MR, Papen CR, Espinosa Trethewy R, Oswald K, Shoemaker R, Berkemeier B, Bosche WJ, Hull M, Greene JM, Axthelm MK, Shao J, Edlefsen PT, Grey F, Nelson JA, Lifson JD, Streblow D, Sacha JB, Früh K, Picker LJ. Cytomegaloviral determinants of CD8 + T cell programming and RhCMV/SIV vaccine efficacy. Sci Immunol 2021; 6:eabg5413. [PMID: 33766849 PMCID: PMC8244349 DOI: 10.1126/sciimmunol.abg5413] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/04/2021] [Indexed: 12/15/2022]
Abstract
Simian immunodeficiency virus (SIV) insert-expressing, 68-1 rhesus cytomegalovirus (RhCMV/SIV) vectors elicit major histocompatibility complex E (MHC-E)- and MHC-II-restricted, SIV-specific CD8+ T cell responses, but the basis of these unconventional responses and their contribution to demonstrated vaccine efficacy against SIV challenge in the rhesus monkeys (RMs) have not been characterized. We show that these unconventional responses resulted from a chance genetic rearrangement in 68-1 RhCMV that abrogated the function of eight distinct immunomodulatory gene products encoded in two RhCMV genomic regions (Rh157.5/Rh157.4 and Rh158-161), revealing three patterns of unconventional response inhibition. Differential repair of these genes with either RhCMV-derived or orthologous human CMV (HCMV)-derived sequences (UL128/UL130; UL146/UL147) leads to either of two distinct CD8+ T cell response types-MHC-Ia-restricted only or a mix of MHC-II- and MHC-Ia-restricted CD8+ T cells. Response magnitude and functional differentiation are similar to RhCMV 68-1, but neither alternative response type mediated protection against SIV challenge. These findings implicate MHC-E-restricted CD8+ T cell responses as mediators of anti-SIV efficacy and indicate that translation of RhCMV/SIV vector efficacy to humans will likely require deletion of all genes that inhibit these responses from the HCMV/HIV vector.
Collapse
Affiliation(s)
- Daniel Malouli
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Meaghan H Hancock
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Colette M Hughes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Julia C Ford
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Abigail B Ventura
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - David Morrow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Kurt T Randall
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Husam Taher
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Luke S Uebelhoer
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Matthew R McArdle
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Courtney R Papen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Renee Espinosa Trethewy
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Kelli Oswald
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Rebecca Shoemaker
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Brian Berkemeier
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - William J Bosche
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Michael Hull
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Justin M Greene
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Jason Shao
- Population Sciences and Computational Biology Programs, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Paul T Edlefsen
- Population Sciences and Computational Biology Programs, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Finn Grey
- Division of Infection and Immunity, Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Jay A Nelson
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Daniel Streblow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA.
| | - Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
19
|
Roles of the Fc Receptor γ-Chain in Inducing Protective Immune Responses after Heterologous Vaccination against Respiratory Syncytial Virus Infection. Vaccines (Basel) 2021; 9:vaccines9030232. [PMID: 33800349 PMCID: PMC7998258 DOI: 10.3390/vaccines9030232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 11/17/2022] Open
Abstract
The roles of the Fc receptor (FcR) in protection or inflammatory disease after respiratory syncytial virus (RSV) vaccination and infection remain unknown. Virus-like particles containing RSV fusion proteins (RSV F-VLPs) induce T-helper type 1 antibody responses and protection against RSV. Heterologous RSV F-VLP prime and formalin-inactivated RSV (FI-RSV) boost vaccination has been reported to be effective in providing protection without inflammatory disease. Here, we investigated whether the FcRγ-chain is important for immune protection by the heterologous F-VLP and FI-RSV vaccination using FcRγ-chain knockout (-/-) mice. RSV F-VLP-primed and FI-RSV-boosted FcRγ -/- mice displayed less protective efficacy, as shown by higher lung viral titers upon RSV challenge, compared to RSV F-VLP-primed and FI-RSV-boosted immunized wild-type mice. RSV F-VLP and FI-RSV immunization induced lower levels of neutralizing activity and interferon-γ-producing CD8 T-cells in the bronchoalveolar lavage cells of FcRγ -/- mice than in those of wild-type mice. In addition, FcRγ -/- mice displayed a trend of enhancing lung histopathology after RSV vaccination and infection. This study suggests that the FcRγ-chain plays an important role in inducing antiviral protection and CD8 T-cell responses in RSV F-VLP prime and FI-RSV boost vaccination after RSV infections.
Collapse
|
20
|
The NKG2D ligand ULBP4 is not expressed by human monocytes. PLoS One 2021; 16:e0246726. [PMID: 33556116 PMCID: PMC7870063 DOI: 10.1371/journal.pone.0246726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 01/26/2021] [Indexed: 11/21/2022] Open
Abstract
The C-type lectin-like receptor NKG2D contributes to the immunosurveillance of virally infected and malignant cells by cytotoxic lymphocytes. A peculiar and puzzling feature of the NKG2D-based immunorecognition system is the high number of ligands for this single immunoreceptor. In humans, there are a total of eight NKG2D ligands (NKG2DL) comprising two members of the MIC (MICA, MICB) and six members of the ULBP family of glycoproteins (ULBP1 to ULBP6). While MICA has been extensively studied with regard to its biochemistry, cellular expression and function, very little is known about the NKG2DL ULBP4. This is, at least in part, due to its rather restricted expression by very few cell lines and tissues. Recently, constitutive ULBP4 expression by human monocytes was reported, questioning the view of tissue-restricted ULBP4 expression. Here, we scrutinized ULBP4 expression by human peripheral blood mononuclear cells and monocytes by analyzing ULBP4 transcripts and ULBP4 surface expression. In contrast to MICA, there was no ULBP4 expression detectable, neither by freshly isolated monocytes nor by PAMP-activated monocytes. However, a commercial antibody erroneously indicated surface ULBP4 on monocytes due to a non-ULBP4-specific binding activity, emphasizing the critical importance of validated reagents for life sciences. Collectively, our data show that ULBP4 is not expressed by monocytes, and likely also not by other peripheral blood immune cells, and therefore exhibits an expression pattern rather distinct from other human NKG2DL.
Collapse
|
21
|
Baugh R, Khalique H, Seymour LW. Convergent Evolution by Cancer and Viruses in Evading the NKG2D Immune Response. Cancers (Basel) 2020; 12:E3827. [PMID: 33352921 PMCID: PMC7766243 DOI: 10.3390/cancers12123827] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
The natural killer group 2 member D (NKG2D) receptor and its family of NKG2D ligands (NKG2DLs) are key components in the innate immune system, triggering NK, γδ and CD8+ T cell-mediated immune responses. While surface NKG2DL are rarely found on healthy cells, expression is significantly increased in response to various types of cellular stress, viral infection, and tumour cell transformation. In order to evade immune-mediated cytotoxicity, both pathogenic viruses and cancer cells have evolved various mechanisms of subverting immune defences and preventing NKG2DL expression. Comparisons of the mechanisms employed following virus infection or malignant transformation reveal a pattern of converging evolution at many of the key regulatory steps involved in NKG2DL expression and subsequent immune responses. Exploring ways to target these shared steps in virus- and cancer-mediated immune evasion may provide new mechanistic insights and therapeutic opportunities, for example, using oncolytic virotherapy to re-engage the innate immune system towards cancer cells.
Collapse
Affiliation(s)
| | | | - Leonard W. Seymour
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (R.B.); (H.K.)
| |
Collapse
|
22
|
Lyu YR, Jung SJ, Lee SW, Yang WK, Kim SH, Jung IC, Kim KH, Kim HY, Yang YJ, Lee Y, Yoon SR, Park YC. Efficacy and safety of CAEC (Canavalia gladiata arctium lappa extract complex) on immune function enhancement: An 8 week, randomised, double-blind, placebo-controlled clinical trial. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
23
|
Hosseini A, Hashemi V, Shomali N, Asghari F, Gharibi T, Akbari M, Gholizadeh S, Jafari A. Innate and adaptive immune responses against coronavirus. Biomed Pharmacother 2020; 132:110859. [PMID: 33120236 PMCID: PMC7580677 DOI: 10.1016/j.biopha.2020.110859] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 01/08/2023] Open
Abstract
Coronaviruses (CoVs) are a member of the Coronaviridae family with positive-sense single- stranded RNA. In recent years, the CoVs have become a global problem to public health. The immune responses (innate and adaptive immunity) are essential for elimination and clearance of CoVs infections, however, uncontrolled immune responses can result in aggravating acute lung injury and significant immunopathology. Gaining profound understanding about the interaction between CoVs and the innate and adaptive immune systems could be a critical step in the field of treatment. In this review, we present an update on the host innate and adaptive immune responses against SARS-CoV, MERS-CoV and newly appeared SARS-CoV-2.
Collapse
Affiliation(s)
- Arezoo Hosseini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vida Hashemi
- Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Asghari
- Department of Immunology, School of Medicine, Tarbiat Modares University of Medical Sciences, Tehran, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saber Gholizadeh
- Department of Medical Entomology and Vector Control, School of Public Health, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Jafari
- Department of Toxicology and Cellular and Molecular Research Center, School of Public Health, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
24
|
Soleimanian S, Yaghobi R. Harnessing Memory NK Cell to Protect Against COVID-19. Front Pharmacol 2020; 11:1309. [PMID: 32973527 PMCID: PMC7468462 DOI: 10.3389/fphar.2020.01309] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023] Open
Abstract
The worldwide struggle against the coronavirus disease 2019 (COVID-19) as a public health crisis continues to sweep across the globe. Up to now, effective antiviral treatment against COVID-19 is not available. Therefore, throughout virus infections, a thorough clarification of the virus-host immune system interactions will be most probably helpful to encounter these challenges. Emerging evidence suggests that just like SARS and MERS, COVID-19 primarily suppresses the innate immune system, enabling its stable propagation during the early stage of infection. Consequently, proinflammatory cytokines and chemokines have been increasing during infection progression associated with severe lung pathology. It is imperative to consider hyper inflammation in vaccine designing, as vaccine-induced immune responses must have a protective role against infection without leading to immunopathology. Among the front-line responders to viral infections, Natural Killer (NK) cells have immense therapeutic potential, forming a bridge between innate and adaptive responses. A subset of NK cells exhibits putatively increased effector functions against viruses following pathogen-specific and immunization. Memory NK cells have higher cytotoxicity and effector activity, compared with the conventional NK cells. As a pioneering strategy, prompt accumulation and long-term maintenance of these memory NK cells could be an efficacious viral treatment. According to the high prevalence of human cytomegalovirus (HCMV) infection in the world, it remains to be determined whether HCMV adaptive NK cells could play a protective role against this new emerging virus. In addition, the new adaptive-like KIR+NKG2C+ NK cell subset (the adaptive-like lung tissue residue [tr]NK cell) in the context of the respiratory infection at this site could specifically exhibit the expansion upon COVID-19. Another aspect of NK cells we should note, utilizing modified NK cells such as allogeneic off-the-shelf CAR-NK cells as a state-of-the-art strategy for the treatment of COVID-19. In this line, we speculate introducing NKG2C into chimeric antigen receptors in NK cells might be a potential approach in future viral immunotherapy for emerging viruses. In this contribution, we will briefly discuss the current status and future perspective of NK cells, which provide to successfully exploit NK cell-mediated antiviral activity that may offer important new tools in COVID-19 treatment.
Collapse
Affiliation(s)
| | - Ramin Yaghobi
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
25
|
Hashemi E, Malarkannan S. Tissue-Resident NK Cells: Development, Maturation, and Clinical Relevance. Cancers (Basel) 2020; 12:cancers12061553. [PMID: 32545516 PMCID: PMC7352973 DOI: 10.3390/cancers12061553] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells belong to type 1 innate lymphoid cells (ILC1) and are essential in killing infected or transformed cells. NK cells mediate their effector functions using non-clonotypic germ-line-encoded activation receptors. The utilization of non-polymorphic and conserved activating receptors promoted the conceptual dogma that NK cells are homogeneous with limited but focused immune functions. However, emerging studies reveal that NK cells are highly heterogeneous with divergent immune functions. A distinct combination of several activation and inhibitory receptors form a diverse array of NK cell subsets in both humans and mice. Importantly, one of the central factors that determine NK cell heterogeneity and their divergent functions is their tissue residency. Decades of studies provided strong support that NK cells develop in the bone marrow. However, evolving evidence supports the notion that NK cells also develop and differentiate in tissues. Here, we summarize the molecular basis, phenotypic signatures, and functions of tissue-resident NK cells and compare them with conventional NK cells.
Collapse
Affiliation(s)
- Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA;
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA;
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence:
| |
Collapse
|
26
|
Müller-Durovic B, Grählert J, Devine OP, Akbar AN, Hess C. CD56-negative NK cells with impaired effector function expand in CMV and EBV co-infected healthy donors with age. Aging (Albany NY) 2020; 11:724-740. [PMID: 30686790 PMCID: PMC6366961 DOI: 10.18632/aging.101774] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/14/2019] [Indexed: 12/14/2022]
Abstract
Natural killer cells lacking expression of CD56 (CD56neg NK cells) have been described in chronic HIV and hepatitis C virus infection. Features and functions of CD56neg NK cells in the context of latent infection with CMV and / or EBV with age are not known. In a cohort of healthy donors >60 years of age, we found that co-infection with CMV and EBV drives expansion of CD56neg NK cells. Functionally, CD56neg NK cells displayed reduced cytotoxic capacity and IFN-γ production, a feature that was enhanced with CMV / EBV co-infection. Further, the frequency of CD56neg NK cells correlated with accumulation of end-stage-differentiated T cells and a reduced CD4 / CD8 T cell ratio, reflecting an immune risk profile. CD56neg NK cells had a mature phenotype characterized by low CD57 and KIR expression and lacked characteristics of cell senescence. No changes in their activating NK cell receptor expression, and no upregulation of the negative co-stimulation receptors PD-1 or TIM-3 were observed. In all, our data identify expansion of dysfunctional CD56neg NK cells in CMV+EBV+ elderly individuals suggesting that these cells may function as shape-shifters of cellular immunity and argue for a previously unrecognized role of EBV in mediating immune risk in the elderly.
Collapse
Affiliation(s)
| | - Jasmin Grählert
- University Hospital Basel, Department of Biomedicine, Basel, Switzerland
| | - Oliver P Devine
- Division of Infection and Immunity, University College London, London, UK
| | - Arne N Akbar
- Division of Infection and Immunity, University College London, London, UK
| | - Christoph Hess
- University Hospital Basel, Department of Biomedicine, Basel, Switzerland.,Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
27
|
Wyatt RC, Lanzoni G, Russell MA, Gerling I, Richardson SJ. What the HLA-I!-Classical and Non-classical HLA Class I and Their Potential Roles in Type 1 Diabetes. Curr Diab Rep 2019; 19:159. [PMID: 31820163 PMCID: PMC6901423 DOI: 10.1007/s11892-019-1245-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Hyperexpression of classical HLA class I (HLA-I) molecules in insulin-containing islets has become a widely accepted hallmark of type 1 diabetes pathology. In comparison, relatively little is known about the expression, function and role of non-classical subtypes of HLA-I. This review focuses on the current understanding of the non-classical HLA-I subtypes: HLA-E, HLA-F and HLA-G, within and outside the field of type 1 diabetes, and considers the possible impacts of these molecules on disease etiology. RECENT FINDINGS Evidence is growing to suggest that non-classical HLA-I proteins are upregulated, both at the RNA and protein levels in the pancreas of individuals with recent-onset type 1 diabetes. Moreover, associations between non-classical HLA-I genotypes and age at onset of type 1 diabetes have been reported in some studies. As with classical HLA-I, it is likely that hyperexpression of non-classical HLA-I is driven by the release of diffusible interferons by stressed β cells (potentially driven by viral infection) and exacerbated by release of cytokines from infiltrating immune cells. Non-classical HLA-I proteins predominantly (but not exclusively) transduce negative signals to immune cells infiltrating at the site of injury/inflammation. We propose a model in which the islet endocrine cells, through expression of non-classical HLA-I are fighting back against the infiltrating immune cells. By inhibiting the activity and function on NK, B and select T cells, the non-classical HLA-I, proteins will reduce the non-specific bystander effects of inflammation, while at the same time still allowing the targeted destruction of β cells by specific islet-reactive CD8+ T cells.
Collapse
Affiliation(s)
- Rebecca C. Wyatt
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW UK
| | - Giacomo Lanzoni
- Diabetes Research Institute, University of Miami – Miller School of Medicine, 1450 NW 10th Avenue, Miami, FL 33136 USA
- Department of Biochemistry and Molecular Biology, University of Miami – Miller School of Medicine, 1011 NW 15th Street, Miami, FL 33136 USA
| | - Mark A. Russell
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW UK
| | - Ivan Gerling
- Department of Medicine University of Tennessee Health Science Center and VA Medical Center Research Service, 1030 Jefferson Avenue, Memphis, TN 38128 USA
| | - Sarah J. Richardson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW UK
| |
Collapse
|
28
|
Immunization with a murine cytomegalovirus based vector encoding retrovirus envelope confers strong protection from Friend retrovirus challenge infection. PLoS Pathog 2019; 15:e1008043. [PMID: 31568492 PMCID: PMC6786657 DOI: 10.1371/journal.ppat.1008043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 10/10/2019] [Accepted: 08/25/2019] [Indexed: 02/04/2023] Open
Abstract
Immunization vectors based on cytomegalovirus (CMV) have attracted a lot of interest in recent years because of their high efficacy in the simian immunodeficiency virus (SIV) macaque model, which has been attributed to their ability to induce strong, unusually broad, and unconventionally restricted CD8+ T cell responses. To evaluate the ability of CMV-based vectors to mediate protection by other immune mechanisms, we evaluated a mouse CMV (MCMV)-based vector encoding Friend virus (FV) envelope (Env), which lacks any known CD8+ T cell epitopes, for its protective efficacy in the FV mouse model. When we immunized highly FV-susceptible mice with the Env-encoding MCMV vector (MCMV.env), we could detect high frequencies of Env-specific CD4+ T cells after a single immunization. While the control of an early FV challenge infection was highly variable, an FV infection applied later after immunization was tightly controlled by almost all immunized mice. Protection of mice correlated with their ability to mount a robust anamnestic neutralizing antibody response upon FV infection, but Env-specific CD4+ T cells also produced appreciable levels of interferon γ. Depletion and transfer experiments underlined the important role of antibodies for control of FV infection but also showed that while no Env-specific CD8+ T cells were induced by the MCMV.env vaccine, the presence of CD8+ T cells at the time of FV challenge was required. The immunity induced by MCMV.env immunization was long-lasting, but was restricted to MCMV naïve animals. Taken together, our results demonstrate a novel mode of action of a CMV-based vaccine for anti-retrovirus immunization that confers strong protection from retrovirus challenge, which is conferred by CD4+ T cells and antibodies. CMV-based vectors have attracted a lot of attention in the vaccine development field, since they were shown to induce unconventionally restricted CD8+ T cell responses and strong protection in the SIV rhesus macaque model. In a mouse retrovirus model, we show now that immunization with a mouse CMV-based vector encoding retrovirus envelope conferred very strong protection, even though it was not designed to induce any CD8+ T cell responses. In this MCMV.env immunization, protection relied on the induction of CD4+ T cells and the ability to mount a strong anamnestic neutralizing antibody response upon retrovirus infection, but it was restricted to MCMV pre-naïve mice. In our model system, the MCMV based vector shows very high efficacy that is comparable to an attenuated retrovirus-based vaccine, and encourages the pursuit of this vaccination strategy.
Collapse
|
29
|
Intracellular Pathogens: Host Immunity and Microbial Persistence Strategies. J Immunol Res 2019; 2019:1356540. [PMID: 31111075 PMCID: PMC6487120 DOI: 10.1155/2019/1356540] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 01/18/2023] Open
Abstract
Infectious diseases caused by pathogens including viruses, bacteria, fungi, and parasites are ranked as the second leading cause of death worldwide by the World Health Organization. Despite tremendous improvements in global public health since 1950, a number of challenges remain to either prevent or eradicate infectious diseases. Many pathogens can cause acute infections that are effectively cleared by the host immunity, but a subcategory of these pathogens called "intracellular pathogens" can establish persistent and sometimes lifelong infections. Several of these intracellular pathogens manage to evade the host immune monitoring and cause disease by replicating inside the host cells. These pathogens have evolved diverse immune escape strategies and overcome immune responses by residing and multiplying inside host immune cells, primarily macrophages. While these intracellular pathogens that cause persistent infections are phylogenetically diverse and engage in diverse immune evasion and persistence strategies, they share common pathogen type-specific mechanisms during host-pathogen interaction inside host cells. Likewise, the host immune system is also equipped with a diverse range of effector functions to fight against the establishment of pathogen persistence and subsequent host damage. This article provides an overview of the immune effector functions used by the host to counter pathogens and various persistence strategies used by intracellular pathogens to counter host immunity, which enables their extended period of colonization in the host. The improved understanding of persistent intracellular pathogen-derived infections will contribute to develop improved disease diagnostics, therapeutics, and prophylactics.
Collapse
|
30
|
Lu Y, Liu X, Huang Y, Liao Y, Xi T, Zhang Y, Shu S, Fang F. The Effects of IL10 and NK Cells on the Susceptibility to Mouse Cytomegalovirus in BALB/c Mice despite the Compensation of IFNγ. Intervirology 2018; 61:111-122. [PMID: 30336455 DOI: 10.1159/000493316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/27/2018] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The aim of this study was to determine which factors lead to the susceptibility to mouse cytomegalovirus (MCMV) in the spleens of BALB/c mice. METHODS BALB/c and C57BL/6 mice were randomly divided into a control group and an infection group and sacrificed on day 0, 1, 3, 7, 14, and 28 postinfection. The cytotoxicity of NK cells was determined by evaluating lactate dehydrogenase contents. Flow cytometry was used to analyze activated NK cells, IFNγ+ NK cells, and total NK cells in the spleen. The pathological changes of spleens in each group were analyzed by HE staining. The expression of IL10, IL18, IFNγ, Thpok, and IFNβ of spleens was determined by quantitative reverse transcriptase PCR. The viral loads of MCMV in spleens and salivary glands were also detected. RESULTS We found that spleen NK cells and IL10 in C57BL/6 mice possessed more powerful immunity to MCMV than BALB/c mice. In BALB/c mice, combined effects of the cytotoxicity of NK cells and IFNγ in spleens still ended up with deficient control of infection. CONCLUSION The functional shortage of NK cells and inappropriate expression of IL10 result in the susceptibility to MCMV in BALB/c mice.
Collapse
|
31
|
Lai HC, Chang CJ, Lin CS, Wu TR, Hsu YJ, Wu TS, Lu JJ, Martel J, Ojcius DM, Ku CL, Young JD, Lu CC. NK Cell-Derived IFN-γ Protects against Nontuberculous Mycobacterial Lung Infection. THE JOURNAL OF IMMUNOLOGY 2018; 201:1478-1490. [PMID: 30061197 DOI: 10.4049/jimmunol.1800123] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 07/05/2018] [Indexed: 01/09/2023]
Abstract
In developed countries, pulmonary nontuberculous mycobacteria (NTM) infections are more prevalent than Mycobacterium tuberculosis infections. Given the differences in the pathogenesis of NTM and M. tuberculosis infections, separate studies are needed to investigate the pathological effects of NTM pathogens. Our previous study showed that anti-IFN-γ autoantibodies are detected in NTM-infected patients. However, the role of NK cells and especially NK cell-derived IFN-γ in this context has not been studied in detail. In the current study, we show that NK1.1 cell depletion increases bacterial load and mortality in a mouse model of pulmonary NTM infection. NK1.1 cell depletion exacerbates NTM-induced pathogenesis by reducing macrophage phagocytosis, dendritic cell development, cytokine production, and lung granuloma formation. Similar pathological phenomena are observed in IFN-γ-deficient (IFN-γ-/-) mice following NTM infection, and adoptive transfer of wild-type NK cells into IFN-γ-/- mice considerably reduces NTM pathogenesis. Injection of rIFN-γ also prevents NTM-induced pathogenesis in IFN-γ-/- mice. We observed that NK cells represent the main producers of IFN-γ in the lungs and production starts as soon as 1 d postinfection. Accordingly, injection of rIFN-γ into IFN-γ-/- mice 1 d (but not 2 wk) postinfection significantly improves immunity against NTM infection. NK cells also stimulate mycobacterial killing and IL-12 production by macrophages. Our results therefore indicate that IFN-γ production by NK cells plays an important role in activating and enhancing innate and adaptive immune responses at early stages of pulmonary NTM infection.
Collapse
Affiliation(s)
- Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taoyuan 33303, Taiwan.,Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taoyuan 33303, Taiwan
| | - Chih-Jung Chang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Chuan-Sheng Lin
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Tsung-Ru Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Ya-Jing Hsu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Ting-Shu Wu
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan
| | - Jang-Jih Lu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan
| | - Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Department of Biomedical Sciences, Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103
| | - Cheng-Lung Ku
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Laboratory of Cellular Physiology and Immunology, Rockefeller University, New York, NY 10021; and
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, Xinzhuang, New Taipei City 24205, Taiwan
| |
Collapse
|
32
|
Hangai S, Kimura Y, Taniguchi T, Yanai H. Innate Immune Receptors in the Regulation of Tumor Immunity. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
33
|
Subramanian N, Wu Z, Reister F, Sampaio KL, Frascaroli G, Cicin-Sain L, Mertens T. Naïve T cells are activated by autologous HCMV-infected endothelial cells through NKG2D and can control HCMV transmission in vitro. J Gen Virol 2017; 98:3068-3085. [PMID: 29165229 DOI: 10.1099/jgv.0.000976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Apart from classical antigen-presenting cells (APCs) like dendritic cells and macrophages, there are semiprofessional APCs such as endothelial cells (ECs) and Langerhans' cells. Human cytomegalovirus (HCMV) infects a wide range of cell types including the ECs which are involved in the trafficking and homing of T cells. By investigating the interaction of naïve T cells obtained from HCMV-seronegative umbilical cord blood with autologous HCMV-infected human umbilical vein ECs (HUVECs), we could show that the activation of naïve T cells occurred after 1 day of peripheral blood mononuclear cell (PBMC) exposure to HCMV-infected HUVECs. The percentage of activated T cells increased over time and the activation of naïve T cells was not induced by either autologous uninfected HUVECs or by autologous HCMV-infected fibroblasts. The activation of T cells occurred also when purified T cells were co-cultured with HCMV-infected HUVECs. In addition, in most of the donors only CD8+ T cells were activated, when the purified T cells were exposed to HCMV-infected HUVECs. The activation of naïve T cells was inhibited when the NKG2D receptor was blocked on the surface of T cells and among the different NKG2D ligands, we identified two ligands (ULBP4 and MICA) on HCMV-infected HUVECs which might be the interaction partners of the NKG2D receptor. Using a functional cell culture assay, we could show that these activated naïve T cells specifically inhibited HCMV transmission. Altogether, we identified a novel specific activation mechanism of naïve T cells from the umbilical cord by HCMV-infected autologous HUVECs through interaction with NKG2D.
Collapse
Affiliation(s)
| | - Zeguang Wu
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| | - Frank Reister
- Gynecology and Obstetrics Clinics, Ulm University Hospital, Ulm, Germany
| | | | - Giada Frascaroli
- Institute of Virology, Ulm University Medical Center, Ulm, Germany.,Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Virology, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Location Hannover-Braunschweig, Germany
| | - Thomas Mertens
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
34
|
Hölzemer A, Garcia-Beltran WF, Altfeld M. Natural Killer Cell Interactions with Classical and Non-Classical Human Leukocyte Antigen Class I in HIV-1 Infection. Front Immunol 2017; 8:1496. [PMID: 29184550 PMCID: PMC5694438 DOI: 10.3389/fimmu.2017.01496] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/24/2017] [Indexed: 11/23/2022] Open
Abstract
Natural killer (NK) cells are effector lymphocytes of the innate immune system that are able to mount a multifaceted antiviral response within hours following infection. This is achieved through an array of cell surface receptors surveilling host cells for alterations in human leukocyte antigen class I (HLA-I) expression and other ligands as signs of viral infection, malignant transformation, and cellular stress. This interaction between HLA-I ligands and NK-cell receptor is not only important for recognition of diseased cells but also mediates tuning of NK-cell-effector functions. HIV-1 alters the expression of HLA-I ligands on infected cells, rendering them susceptible to NK cell-mediated killing. However, over the past years, various HIV-1 evasion strategies have been discovered to target NK-cell-receptor ligands and allow the virus to escape from NK cell-mediated immunity. While studies have been mainly focusing on the role of polymorphic HLA-A, -B, and -C molecules, less is known about how HIV-1 affects the more conserved, non-classical HLA-I molecules HLA-E, -G, and -F. In this review, we will focus on the recent progress in understanding the role of non-classical HLA-I ligands in NK cell-mediated recognition of HIV-1-infected cells.
Collapse
Affiliation(s)
- Angelique Hölzemer
- First Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | | | - Marcus Altfeld
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
35
|
Dose of Retroviral Infection Determines Induction of Antiviral NK Cell Responses. J Virol 2017; 91:JVI.01122-17. [PMID: 28904191 PMCID: PMC5660477 DOI: 10.1128/jvi.01122-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/01/2017] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells are part of the innate immune system and recognize virus-infected cells as well as tumor cells. Conflicting data about the beneficial or even detrimental role of NK cells in different infectious diseases have been described previously. While the type of pathogen strongly influences NK cell functionality, less is known about how the infection dose influences the quality of a NK cell response against retroviruses. In this study, we used the well-established Friend retrovirus (FV) mouse model to investigate the impact of virus dose on the induction of antiviral NK cell functions. High-dose virus inoculation increased initial virus replication compared to that with medium- or low-dose viral challenge and significantly improved NK cell activation. Antiviral NK cell activity, including in vivo cytotoxicity toward infected target cells, was also enhanced by high-dose virus infection. NK cell activation following high-dose viral challenge was likely mediated by activated dendritic cells (DCs) and macrophages and the NK cell-stimulating cytokines interleukin 15 (IL-15) and IL-18. Neutralization of these cytokines decreased NK cell functions and increased viral loads, whereas IL-15 and IL-18 therapy improved NK cell activity. Here we demonstrate that virus dose positively correlates with antiviral NK cell activity and function, which are at least partly driven by IL-15 and IL-18. Our results suggest that NK cell activity may be therapeutically enhanced by administering IL-15 and IL-18 in virus infections that inadequately activate NK cells. IMPORTANCE In infections with retroviruses, like HIV and FV infection of mice, NK cells clearly mediate antiviral activities, but they are usually not sufficient to prevent severe pathology. Here we show that the initial infection dose impacts the induction of an antiviral NK cell response during an acute retroviral infection, which had not investigated before. High-dose infection resulted in a strong NK cell functionality, whereas no antiviral activities were detected after low- or medium-dose infection. Interestingly, DCs and macrophages were highly activated after high-dose FV challenge, which corresponded with increased levels of NK cell-stimulating cytokines IL-15 and IL-18. IL-15 and IL-18 neutralization decreased NK cell functions, whereas IL-15 and IL-18 therapy improved NK cell activity. Here we show the importance of cytokines for NK cell activation in retroviral infections; our findings suggest that immunotherapy combining the well-tolerated cytokines IL-15 and IL-18 might be an interesting approach for antiretroviral treatment.
Collapse
|
36
|
Sharma P, Kumar P, Sharma R. Natural Killer Cells - Their Role in Tumour Immunosurveillance. J Clin Diagn Res 2017; 11:BE01-BE05. [PMID: 28969116 DOI: 10.7860/jcdr/2017/26748.10469] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/01/2017] [Indexed: 01/09/2023]
Abstract
An important component of the innate immune system, the natural killer cells that originate from the lymphoid cell lineage, hold tremendous potential as an effective therapeutic tool to combat a variety of cancers. Their vast capability to kill altered cells such as opsonized cells (antibody coated), tumour cells, genotoxically changed cells without affecting the healthy cells of the body, make them an effective therapeutic agent for various types of cancers. Besides, through interplay and molecular crosstalk via several cytokines, they also augment the adaptive immune response by, promoting the differentiation, activation and recruitment of component cells of the system. With the current advance knowledge of Natural Killer (NK) cells, their receptor-ligand interactions involved in functional regulation, various mechanistic approaches involving the role of cytokines led to desired modulation of NK cell activity in a tailor-made manner, for triggering clinically relevant responces. Several strategies have been adopted by researchers, to augment the efficacy of NK cells. Still many challenges exist for increasing the therapeutic relevance of these cells.
Collapse
Affiliation(s)
- Preeti Sharma
- Associate Professor, Department of Biochemistry, Santosh Medical University, Ghaziabad, Uttar Pradesh, India
| | - Pradeep Kumar
- Professor, Department of Biochemistry, Santosh Medical University, Ghaziabad, Uttar Pradesh, India
| | - Rachna Sharma
- Lecturer, Department of Biochemistry, TSM Medical College and Hospital, Lucknow, Uttar Pradesh, India
| |
Collapse
|
37
|
Avian influenza virus directly infects human natural killer cells and inhibits cell activity. Virol Sin 2017; 32:122-129. [PMID: 28255852 DOI: 10.1007/s12250-016-3918-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/30/2016] [Indexed: 12/18/2022] Open
Abstract
Natural killer (NK) cell is a key component of innate immunity and plays an important role in host defense against virus infection by directly destroying infected cells. Influenza is a respiratory disease transmitted in the early phase of virus infection. Evasion of host innate immunity including NK cells is critical for the virus to expand and establish a successful acute infection. Previously, we showed that human influenza H1N1 virus infects NK cells and induces cell apoptosis, as well as inhibits NK cell activity. In this study, we further demonstrated that avian influenza virus also directly targeted NK cells as an immunoevasion strategy. The avian virus infected human NK cells and induced cell apoptosis. In addition, avian influenza virion and HA protein inhibited NK cell cytotoxicity. This novel strategy has obvious advantages for avian influenza virus, allowing the virus sufficient time to expand and subsequent spread before the onset of the specific immune response. Our findings provide an important clue for the immunopathogenesis of avian influenza, and also suggest that direct targeting NK cells may be a common strategy used by both human and avian influenza viruses to evade NK cell immunity.
Collapse
|
38
|
Granato M, Rizzello C, Gilardini Montani MS, Cuomo L, Vitillo M, Santarelli R, Gonnella R, D'Orazi G, Faggioni A, Cirone M. Quercetin induces apoptosis and autophagy in primary effusion lymphoma cells by inhibiting PI3K/AKT/mTOR and STAT3 signaling pathways. J Nutr Biochem 2017; 41:124-136. [PMID: 28092744 DOI: 10.1016/j.jnutbio.2016.12.011] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/23/2016] [Accepted: 12/28/2016] [Indexed: 12/15/2022]
Abstract
Quercetin, a bioflavonoid contained in several vegetables daily consumed, has been studied for long time for its antiinflammatory and anticancer properties. Quercetin interacts with multiple cancer-related pathways such as PI3K/AKT, Wnt/β-catenin and STAT3. These pathways are hyperactivated in primary effusion lymphoma (PEL), an aggressive B cell lymphoma whose pathogenesis is strictly linked to the oncogenic virus Kaposis' Sarcoma-associated Herpesvirus (KSHV). In this study, we found that quercetin inhibited PI3K/AKT/mTOR and STAT3 pathways in PEL cells, and as a consequence, it down-regulated the expression of the prosurvival cellular proteins such as c-FLIP, cyclin D1 and cMyc. It also reduced the release of IL-6 and IL-10 cytokines, leading to PEL cell death. Moreover, quercetin induced a prosurvival autophagy in these cells and increased the cytotoxic effect of bortezomib, a proteasomal inhibitor, against them. Interestingly, quercetin decreased also the expression of latent and lytic KSHV proteins involved in PEL tumorigenesis and up-regulated the surface expression of HLA-DR and calreticulin, rendering the dying cells more likely detectable by the immune system. The results obtained in this study indicate that quercetin, which does not exert any cytotoxicity against normal B cells, may represent a good candidate for the treatment of this aggressive B cell lymphoma, especially in combination with autophagy inhibitors or with bortezomib.
Collapse
Affiliation(s)
- Marisa Granato
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Celeste Rizzello
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | | | - Laura Cuomo
- U. O.C. Patologia Clinica, Ospedale San Filippo Neri, 00135 Rome, Italy
| | - Marina Vitillo
- U. O.C. Patologia Clinica, Ospedale San Filippo Neri, 00135 Rome, Italy
| | - Roberta Santarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Roberta Gonnella
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Gabriella D'Orazi
- Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy; Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", 66013, Chieti, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy.
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy.
| |
Collapse
|
39
|
Bahador M, Gras Navarro A, Rahman M, Dominguez-Valentin M, Sarowar S, Ulvestad E, Njølstad G, Lie S, Kristoffersen E, Bratland E, Chekenya M. Increased infiltration and tolerised antigen-specific CD8 + T EM cells in tumor but not peripheral blood have no impact on survival of HCMV + glioblastoma patients. Oncoimmunology 2017; 6:e1336272. [PMID: 28919997 PMCID: PMC5593710 DOI: 10.1080/2162402x.2017.1336272] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/07/2017] [Accepted: 05/24/2017] [Indexed: 12/27/2022] Open
Abstract
Human cytomegalovirus (HCMV) antigens in glioblastoma (GBM) present opportunities for personalised immunotherapy. However, their presence in GBM tissue is still under debate, and evidence of their impact on functional immune responses and prognosis is sparse. Here, we investigated the presence of pp65 (UL83) and immediate early 1 (IE-1) HCMV antigens in a cohort of Norwegian GBM patients (n = 177), using qPCR, immunohistochemistry, and serology. HCMV status was then used to investigate whether viral antigens influenced immune cell phenotype, infiltration, activation and patient survival. Pp65 and IE-1 were detected by qPCR in 23% and 43% of GBM patients, respectively. Furthermore, there was increased seropositivity in GBM patients relative to donors (79% vs. 48%, respectively; Logistic regression, OR = 4.05, 95%CI [1.807-9.114], P = 0.001, also when adjusted for age (OR = 2.84, 95%CI [1.110-7.275], P = 0.029). Tissue IE-1-positivity correlated with increased CD3+CD8+ T-cell infiltration (P < 0.0001), where CD8+ effector memory T (TEM) cells accounted for the majority of CD8+T cells compared with peripheral blood of HCMV+ patients (P < 0.0001), and HCMV+ (P < 0.001) and HCMV- (P < 0.001) donors. HLA-A2/B8-restricted HCMV-specific CD8+ T cells were more frequent in blood and tumor of HCMV+ GBM patients compared with seronegative patients, and donors irrespective of their serostatus. In biopsies, the HCMV-specific CD8+ TEM cells highly expressed CTLA-4 and PD-1 immune checkpoint protein markers compared with populations in peripheral blood (P < 0.001 and P < 0.0001), which expressed 3-fold greater levels of CD28 (P < 0.001 and P < 0.0001). These peripheral blood T cells correspondingly secreted higher levels of IFNγ in response to pp65 and IE-1 peptide stimulation (P < 0.001). Thus, despite apparent increased immunogenicity of HCMV compared with tumor antigens, the T cells were tolerised, and HCMV status did not impact patient survival (Log Rank3.53 HR = 0.85 95%CI [0.564-1.290], P = 0.45). Enhancing immune functionality in the tumor microenvironment thus may improve patient outcome.
Collapse
Affiliation(s)
- M. Bahador
- University of Bergen, Department of Biomedicine, Bergen, Norway
| | - A. Gras Navarro
- University of Bergen, Department of Biomedicine, Bergen, Norway
| | - M.A. Rahman
- University of Bergen, Department of Biomedicine, Bergen, Norway
| | | | - S. Sarowar
- University of Bergen, Department of Biomedicine, Bergen, Norway
| | - E. Ulvestad
- University of Bergen, Department of Clinical Science, Bergen, Norway
- Haukeland University Hospital, Department of Microbiology, Bergen, Norway
| | - G. Njølstad
- Haukeland University Hospital, Department of Microbiology, Bergen, Norway
| | - S.A. Lie
- University of Bergen, Department of Clinical Dentistry, Bergen, Norway
| | - E.K. Kristoffersen
- University of Bergen, Department of Clinical Science, Bergen, Norway
- Haukeland University Hospital, Department of Immunology and Transfusion Medicine, Bergen, Norway
| | - E. Bratland
- University of Bergen, Department of Clinical Science, Bergen, Norway
- Eirik Bratland, PhD University of Bergen, Department of clinical science, Jonas Lies vei 91, 5020, Bergen
| | - M. Chekenya
- University of Bergen, Department of Biomedicine, Bergen, Norway
- CONTACT Professor Martha Chekenya, PhD, Dr Philos University of Bergen, Department of Biomedicine, Jonas Lies vei 91, 5020, Bergen
| |
Collapse
|
40
|
Bayer AL, Fraker CA. The Folate Cycle As a Cause of Natural Killer Cell Dysfunction and Viral Etiology in Type 1 Diabetes. Front Endocrinol (Lausanne) 2017; 8:315. [PMID: 29218028 PMCID: PMC5703744 DOI: 10.3389/fendo.2017.00315] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022] Open
Abstract
The folate pathway is critical to proper cellular function and metabolism. It is responsible for multiple functions, including energy (ATP) production, methylation reactions for DNA and protein synthesis and the production of immunomodulatory molecules, inosine and adenosine. These play an important role in immune signaling and cytotoxicity. Herein, we hypothesize that defects in the folate pathway in genetically susceptible individuals could lead to immune dysfunction, permissive environments for chronic cyclical latent/lytic viral infection, and, ultimately, the development of unchecked autoimmune responses to infected tissue, in this case islet beta cells. In the context of type 1 diabetes (T1D), there has been a recent increase in newly diagnosed cases of T1D in the past 20 years that has exceeded previous epidemiological predictions with yet unidentified factor(s). This speaks to a potential environmental trigger that adversely affects immune responses. Most research into the immune dysfunction of T1D has focused on downstream adaptive responses of T and B cells neglecting the role of the upstream innate players such as natural killer (NK) cells. Constantly, surveilling the blood and tissues for pathogens, NK cells remove threats through direct cytotoxic responses and recruitment of adaptive responses using cytokines, such as IL-1β and IFN-γ. One long-standing hypothesis suggests viral infection as a potential trigger for the autoimmune response in T1D. Recent data suggest multiple viruses as potential causal agents. Intertwined with this is an observed reduced NK cell enumeration, cytotoxicity, and cytokine signaling in T1D patients. Many of the viruses implicated in T1D are chronic latent/lysogenic infections with demonstrated capacity to reduce NK cell response and number through mechanisms that resemble those of pregnancy tolerance. Defects in the folate pathway in T1D patients could result in decreased immune response to viral infection or viral reactivation. Dampened NK responses to infections result in improper signaling, improper antigen presentation, and amplified CD8+ lymphocyte proliferation and cytotoxicity, a hallmark of beta cell infiltrates in patients with T1D onset. This would suggest a critical role for NK cells in T1D development linked to viral infection and the importance of the folate pathway in maintaining proper NK response.
Collapse
Affiliation(s)
- Allison L. Bayer
- Immunobiology Laboratory, Leonard M. Miller School of Medicine, Diabetes Research Institute, University of Miami, Miami, FL, United States
| | - Christopher A. Fraker
- Tissue and Biomedical Engineering Laboratory, Leonard M. Miller School of Medicine, Diabetes Research Institute, University of Miami, Miami, FL, United States
- *Correspondence: Christopher A. Fraker,
| |
Collapse
|
41
|
Béziat V, Hilton HG, Norman PJ, Traherne JA. Deciphering the killer-cell immunoglobulin-like receptor system at super-resolution for natural killer and T-cell biology. Immunology 2016; 150:248-264. [PMID: 27779741 PMCID: PMC5290243 DOI: 10.1111/imm.12684] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/12/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022] Open
Abstract
Killer-cell immunoglobulin-like receptors (KIRs) are components of two fundamental biological systems essential for human health and survival. First, they contribute to host immune responses, both innate and adaptive, through their expression by natural killer cells and T cells. Second, KIR play a key role in regulating placentation, and hence reproductive success. Analogous to the diversity of their human leucocyte antigen class I ligands, KIR are extremely polymorphic. In this review, we describe recent developments, fuelled by methodological advances, that are helping to decipher the KIR system in terms of haplotypes, polymorphisms, expression patterns and their ligand interactions. These developments are delivering deeper insight into the relevance of KIR in immune system function, evolution and disease.
Collapse
Affiliation(s)
- Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.,Imagine Institute, Paris Descartes University, Paris, France
| | - Hugo G Hilton
- Departments of Structural Biology and Microbiology & Immunology, Stanford University, Stanford, CA, USA
| | - Paul J Norman
- Departments of Structural Biology and Microbiology & Immunology, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
42
|
Abstract
Treatments for autoimmune diseases including type 1 diabetes (T1D) are aimed at resetting the immune system, especially its adaptive arm. The innate immune system is often ignored in the design of novel immune-based therapies. There is increasing evidence for multiple natural killer (NK) subpopulations, but their role is poorly understood in autoimmunity and likely is contributing to the controversial role reported for NKs. In this review, we will summarize NK subsets and their roles in tolerance, autoimmune diabetes, and immunotherapy.
Collapse
Affiliation(s)
- Chris Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Allison L Bayer
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
43
|
Goncalves A, Makalo P, Joof H, Burr S, Ramadhani A, Massae P, Malisa A, Mtuy T, Derrick T, Last AR, Nabicassa M, Cassama E, Houghton J, Palmer CD, Pickering H, Burton MJ, Mabey DCW, Bailey RL, Goodier MR, Holland MJ, Roberts CH. Differential frequency of NKG2C/KLRC2 deletion in distinct African populations and susceptibility to Trachoma: a new method for imputation of KLRC2 genotypes from SNP genotyping data. Hum Genet 2016; 135:939-51. [PMID: 27312142 PMCID: PMC4947484 DOI: 10.1007/s00439-016-1694-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/02/2016] [Indexed: 11/26/2022]
Abstract
NKG2C is an activating receptor that is preferentially expressed on natural killer (NK) cells. The gene encoding NKG2C (killer cell lectin-like receptor C2, KLRC2) is present at different copy numbers in the genomes of different individuals. Deletion at the NKG2C locus was investigated in a case-control study of 1522 individuals indigenous to East- and West-Africa and the association with the ocular Chlamydia trachomatis infection and its sequelae was explored. The frequency of homozygous KLRC2 deletion was 13.7 % in Gambians and 4.7 % in Tanzanians. A significantly higher frequency of the deletion allele was found in West-Africans from the Gambia and Guinea-Bissau (36.2 % p = 2.105 × 10(-8), 26.8 % p = 0.050; respectively) in comparison to East-African Tanzanians where the frequency of the deletion is comparable to other human populations (20.9 %). We found no evidence for an association between the numbers of KLRC2 gene copies and the clinical manifestations of trachoma (follicular trachoma or conjunctival scarring). A new method for imputation of KLRC2 genotypes from single nucleotide polymorphism (SNP) data in 2621 individuals from the Gambia further confirmed these results. Our data suggest that NKG2C does not play a major role in trachomatous disease. We found that the deletion allele is present at different frequencies in different populations but the reason behind these differences is currently not understood. The new method offers the potential to use SNP arrays from genome wide association studies to study the frequency of KLRC2 deletion in other populations and its association with other diseases.
Collapse
Affiliation(s)
- Adriana Goncalves
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Pateh Makalo
- Disease Control and Elimination Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Hassan Joof
- Disease Control and Elimination Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Sarah Burr
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Disease Control and Elimination Theme, Medical Research Council Unit, Fajara, The Gambia
| | | | | | - Aiweda Malisa
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Tara Mtuy
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- International Centre for Eye Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Tamsyn Derrick
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Anna R Last
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Meno Nabicassa
- Programa Nacional de Saude de Visao, Ministerio de Saude Publica, Bissau, Guinea-Bissau
| | - Eunice Cassama
- Programa Nacional de Saude de Visao, Ministerio de Saude Publica, Bissau, Guinea-Bissau
| | - Joanna Houghton
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Christine D Palmer
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Harry Pickering
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Matthew J Burton
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- International Centre for Eye Health, London School of Hygiene and Tropical Medicine, London, UK
| | - David C W Mabey
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Robin L Bailey
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Martin R Goodier
- Department of Immunology and Infectious Disease, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Martin J Holland
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Disease Control and Elimination Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Chrissy H Roberts
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
44
|
Burga RA, Nguyen T, Zulovich J, Madonna S, Ylisastigui L, Fernandes R, Yvon E. Improving efficacy of cancer immunotherapy by genetic modification of natural killer cells. Cytotherapy 2016; 18:1410-1421. [PMID: 27421740 DOI: 10.1016/j.jcyt.2016.05.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 11/17/2022]
Abstract
Natural killer (NK) cells are members of the innate immune system that recognize target cells via activating and inhibitory signals received through cell receptors. Derived from the lymphoid lineage, NK cells are able to produce cytokines and exert a cytotoxic effect on viral infected and malignant cells. It is their unique ability to lyse target cells rapidly and without prior education that renders NK cells a promising effector cell for adoptive cell therapy. However, both viruses and tumors employ evasion strategies to avoid attack by NK cells, which represent biological challenges that need to be harnessed to fully exploit the cytolytic potential of NK cells. Using genetic modification, the function of NK cells can be enhanced to improve their homing, cytolytic activity, in vivo persistence and safety. Examples include gene modification to express chemokine, high-affinity Fc receptor and chimeric antigen receptors, suicide genes and the forced expression of cytokines such as interleukin (IL)-2 and IL-15. Preclinical studies have clearly demonstrated that such approaches are effective in improving NK-cell function, homing and safety. In this review, we summarize the recent advances in the genetic manipulations of NK cells and their application for cellular immunotherapeutic strategies.
Collapse
Affiliation(s)
- Rachel A Burga
- Institute for Biomedical Sciences, The George Washington University, Washington, DC, USA; Children's National Health System, Washington, DC, USA
| | - Tuongvan Nguyen
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jane Zulovich
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sarah Madonna
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Loyda Ylisastigui
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Rohan Fernandes
- Institute for Biomedical Sciences, The George Washington University, Washington, DC, USA; Children's National Health System, Washington, DC, USA
| | - Eric Yvon
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
45
|
Bang BR, Elmasry S, Saito T. Organ system view of the hepatic innate immunity in HCV infection. J Med Virol 2016; 88:2025-2037. [PMID: 27153233 DOI: 10.1002/jmv.24569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2016] [Indexed: 12/12/2022]
Abstract
An orchestration of innate and adaptive immunity determines the infection outcome and whether the host achieves clearance or allows the pathogen to establish persistent infection. The robust activation of the innate immune response plays the most critical role in both limiting viral replication and halting the spread of the pathogen immediately after infection. The magnitude of innate immune activation is coupled with the efficient mounting of the adaptive immunity. Although immunity against HCV infection is known to be inadequate as most cases transitions to chronicity, approximately 25% of acute infection cases result in spontaneous clearance. The exact immune mechanisms that govern the infection outcome remain largely unknown; recent discoveries suggest that the innate immune system facilitates this event. Both infected hepatocytes and local innate immune cells trigger the front line defense program of the liver as well as the recruitment of diverse adaptive immune cells to the site of infection. Although hepatocyte is the target of HCV infection, nearly all cell types that exist in the liver are involved in the innate defense and contribute to the pathophysiology of hepatic inflammation. The main focus of this comprehensive review is to discuss the current knowledge on how each hepatic cell type contributes to the organ system level innate immunity against HCV infection as well as interplays with the viral evasion program. Furthermore, this review article also aims to synchronize the observations from both molecular biological studies and clinical studies with the ultimate goal of improving our understanding of HCV mediated hepatitis. J. Med. Virol. 88:2025-2037, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bo-Ram Bang
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, USC Research Center for Liver Diseases, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Sandra Elmasry
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, USC Research Center for Liver Diseases, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Takeshi Saito
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, USC Research Center for Liver Diseases, University of Southern California, Keck School of Medicine, Los Angeles, California. .,Department of Molecular Microbiology and Immunology, University of Southern California, Keck School of Medicine, Los Angeles, California. .,Department of Pathology, University of Southern California, Keck School of Medicine, Los Angeles, California.
| |
Collapse
|
46
|
Natural killer cells enhance the immune surveillance of cancer. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2016. [DOI: 10.1016/j.ejmhg.2015.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
47
|
Hansen SG, Wu HL, Burwitz BJ, Hughes CM, Hammond KB, Ventura AB, Reed JS, Gilbride RM, Ainslie E, Morrow DW, Ford JC, Selseth AN, Pathak R, Malouli D, Legasse AW, Axthelm MK, Nelson JA, Gillespie GM, Walters LC, Brackenridge S, Sharpe HR, López CA, Früh K, Korber BT, McMichael AJ, Gnanakaran S, Sacha JB, Picker LJ. Broadly targeted CD8⁺ T cell responses restricted by major histocompatibility complex E. Science 2016; 351:714-20. [PMID: 26797147 PMCID: PMC4769032 DOI: 10.1126/science.aac9475] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 01/06/2016] [Indexed: 12/22/2022]
Abstract
Major histocompatibility complex E (MHC-E) is a highly conserved, ubiquitously expressed, nonclassical MHC class Ib molecule with limited polymorphism that is primarily involved in the regulation of natural killer (NK) cells. We found that vaccinating rhesus macaques with rhesus cytomegalovirus vectors in which genes Rh157.5 and Rh157.4 are deleted results in MHC-E-restricted presentation of highly varied peptide epitopes to CD8αβ(+) T cells, at ~4 distinct epitopes per 100 amino acids in all tested antigens. Computational structural analysis revealed that MHC-E provides heterogeneous chemical environments for diverse side-chain interactions within a stable, open binding groove. Because MHC-E is up-regulated to evade NK cell activity in cells infected with HIV, simian immunodeficiency virus, and other persistent viruses, MHC-E-restricted CD8(+) T cell responses have the potential to exploit pathogen immune-evasion adaptations, a capability that might endow these unconventional responses with superior efficacy.
Collapse
Affiliation(s)
- Scott G. Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Helen L. Wu
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Benjamin J. Burwitz
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Colette M. Hughes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Katherine B. Hammond
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Abigail B. Ventura
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Jason S. Reed
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Roxanne M. Gilbride
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Emily Ainslie
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - David W. Morrow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Julia C. Ford
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Andrea N. Selseth
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Reesab Pathak
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Alfred W. Legasse
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Jay A. Nelson
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | | | - Lucy C. Walters
- Nuffield Department of Medicine, University of Oxford, OX37FZ, United Kingdom
| | - Simon Brackenridge
- Nuffield Department of Medicine, University of Oxford, OX37FZ, United Kingdom
| | - Hannah R. Sharpe
- Nuffield Department of Medicine, University of Oxford, OX37FZ, United Kingdom
| | - César A. López
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Bette T. Korber
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory
- The New Mexico Consortium, Los Alamos, NM 87545
| | - Andrew J. McMichael
- Nuffield Department of Medicine, University of Oxford, OX37FZ, United Kingdom
| | - S. Gnanakaran
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory
| | - Jonah B. Sacha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| |
Collapse
|
48
|
Zhou K, Wang J, Li A, Zhao W, Wang D, Zhang W, Yan J, Gao GF, Liu W, Fang M. Swift and Strong NK Cell Responses Protect 129 Mice against High-Dose Influenza Virus Infection. THE JOURNAL OF IMMUNOLOGY 2016; 196:1842-54. [DOI: 10.4049/jimmunol.1501486] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 12/15/2015] [Indexed: 11/19/2022]
|
49
|
Wagner S, Wittekindt C, Reuschenbach M, Hennig B, Thevarajah M, Würdemann N, Prigge ES, von Knebel Doeberitz M, Dreyer T, Gattenlöhner S, Peter Klussmann J. CD56-positive lymphocyte infiltration in relation to human papillomavirus association and prognostic significance in oropharyngeal squamous cell carcinoma. Int J Cancer 2016; 138:2263-73. [DOI: 10.1002/ijc.29962] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 11/25/2015] [Accepted: 12/03/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Steffen Wagner
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Giessen; Giessen Germany
| | - Claus Wittekindt
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Giessen; Giessen Germany
| | - Miriam Reuschenbach
- Department of Applied Tumor Biology; University of Heidelberg and Clinical Cooperation Unit, German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Ben Hennig
- Institute of Pathology, University of Giessen; Giessen Germany
| | - Mauran Thevarajah
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Giessen; Giessen Germany
| | - Nora Würdemann
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Giessen; Giessen Germany
| | - Elena-Sophie Prigge
- Department of Applied Tumor Biology; University of Heidelberg and Clinical Cooperation Unit, German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumor Biology; University of Heidelberg and Clinical Cooperation Unit, German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Thomas Dreyer
- Institute of Pathology, University of Giessen; Giessen Germany
| | | | - Jens Peter Klussmann
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Giessen; Giessen Germany
| |
Collapse
|
50
|
Raudenska M, Gumulec J, Fribley AM, Masarik M. HNSCC Biomarkers Derived from Key Processes of Cancerogenesis. TARGETING ORAL CANCER 2016:115-160. [DOI: 10.1007/978-3-319-27647-2_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|