1
|
Mousavi S, Khazaee-Nasirabadi MH, Seyedmehdi MS, Bazi A, Mirzaee Khalilabadi R. Natural killer cells: a new promising source for developing chimeric antigen receptor anti-cancer cells in hematological malignancies. Leuk Lymphoma 2025; 66:594-616. [PMID: 39656564 DOI: 10.1080/10428194.2024.2438802] [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: 06/10/2024] [Revised: 11/18/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
In recent times, the application of CAR-T cell treatment has significantly progressed, showing auspicious treatment outcomes in hematologic malignancies. However, along with these advances, certain limitations and challenges hurdle the widespread utilization of this technology. Recently, CAR-NK cells have gained attention in cancer treatment, as this approach has an important advantage over CART therapy (i.e. no need for HLA matching) for targeting foreign cells. This review aims to explore the benefits of CAR NK cell therapy, and generation strategies, as well as the challenges and limitations hindering the application of CAR NK cells in experimental studies and trials on hematologic malignancies.
Collapse
Affiliation(s)
- Shahrzad Mousavi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Maryam Sadat Seyedmehdi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Islamic Republic of Iran
| | - Ali Bazi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Roohollah Mirzaee Khalilabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
2
|
Hooda V, Sharma A. Interactions of NK Cells and Macrophages: From Infections to Cancer Therapeutics. Immunology 2025; 174:287-295. [PMID: 39739619 DOI: 10.1111/imm.13886] [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: 08/25/2024] [Revised: 11/13/2024] [Accepted: 12/07/2024] [Indexed: 01/02/2025] Open
Abstract
The interaction between immune cells brings a consequence either on their role and functioning or the functioning of the other immune cells, modulating the whole mechanistic pathway. The interaction between natural killer (NK) cells and macrophages is one such interaction which is relatively less explored amongst diseased conditions. Their significance comes from their innate nature and secretion of large proportions of cytokines and chemokines which results in influencing adaptive immune responses. Their interplay can lead to several functional outcomes such as NK cell activation/inhibition, increased cytotoxicity and IFNγ release by NK cells, inhibition of macrophage function, etc. This paper delves into the interaction amongst NK cells and macrophages via different receptor-ligands and cytokines, particularly emphasising microbial infections and tumours. The review has the potential to uncover new insights and approaches that could lead to the development of innovative therapeutic tools and targets.
Collapse
Affiliation(s)
- Vishakha Hooda
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
3
|
Tao JH, Zhang J, Li HS, Zhou Y, Guan CX. Nature killer cell for solid tumors: Current obstacles and prospective remedies in NK cell therapy and beyond. Crit Rev Oncol Hematol 2025; 205:104553. [PMID: 39515404 DOI: 10.1016/j.critrevonc.2024.104553] [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: 11/25/2023] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, cell therapy has emerged as an innovative treatment method for the management of clinical tumors following immunotherapy. Among them, Natural killer (NK) cell therapy has achieved a significant breakthrough in the treatment of hematological tumors. However, the therapeutic effectiveness of NK cells in the treatment of solid tumors remains challenging. With the progress of gene editing and culture techniques and their application to NK cell engineering, it is expected that NK cell therapy will revolutionize the treatment of solid tumors. In this review, we explore the discovery and biological properties of NK cells, their role in the tumor microenvironment, and the therapeutic strategies, clinical trials, challenges, and prospects of NK cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Jia-Hao Tao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Jun Zhang
- Ascle Therapeutics, Suzhou, Jiangsu 215000, China
| | - Hua-Shun Li
- Ascle Therapeutics, Suzhou, Jiangsu 215000, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
4
|
Appiah C, Chen S, Pori AI, Retyunskiy V, Tzeng C, Zhao Y. Study of alloferon, a novel immunomodulatory antimicrobial peptide (AMP), and its analogues. Front Pharmacol 2024; 15:1359261. [PMID: 38434708 PMCID: PMC10904621 DOI: 10.3389/fphar.2024.1359261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Antimicrobial peptides (AMPs) are widely distributed throughout the biosphere and represent a class of conserved peptide molecules with intrinsic antimicrobial properties. Their broad-spectrum antimicrobial activity and low risk to induce resistance have led to increased interest in AMPs as potential alternatives to traditional antibiotics. Among the AMPs, alloferon has been addressed due to its immunomodulatory properties that augment both innate and adaptive immune responses against various pathogens. Alloferon and its analogues have demonstrated pharmaceutical potential through their ability to enhance Natural Killer (NK) cell cytotoxicity and stimulate interferon (IFN) synthesis in both mouse and human models. Additionally, they have shown promise in augmenting antiviral and antitumor activities in mice. In this article, we provide a comprehensive review of the biological effects of alloferon and its analogues, incorporating our own research findings as well. These insights may contribute to a deeper understanding of the therapeutic potential of these novel AMPs.
Collapse
Affiliation(s)
- Clara Appiah
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| | - Shitian Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| | - Afia Ibnat Pori
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| | | | - Chimeng Tzeng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| |
Collapse
|
5
|
Lee S, Kim TD. Breakthroughs in Cancer Immunotherapy: An Overview of T Cell, NK Cell, Mφ, and DC-Based Treatments. Int J Mol Sci 2023; 24:17634. [PMID: 38139461 PMCID: PMC10744055 DOI: 10.3390/ijms242417634] [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: 11/14/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
Efforts to treat cancer using chimeric antigen receptor (CAR)-T therapy have made astonishing progress and clinical trials against hematopoietic malignancies have demonstrated their use. However, there are still disadvantages which need to be addressed: high costs, and side effects such as Graft-versus-Host Disease (GvHD) and Cytokine Release Syndrome (CRS). Therefore, recent efforts have been made to harness the properties of certain immune cells to treat cancer-not just T cells, but also natural killer (NK) cells, macrophages (Mφ), dendritic cells (DC), etc. In this paper, we will introduce immune cell-based cellular therapies that use various immune cells and describe their characteristics and their clinical situation. The development of immune cell-based cancer therapy fully utilizing the unique advantages of each and every immune cell is expected to enhance the survival of tumor patients owing to their high efficiency and fewer side effects.
Collapse
Affiliation(s)
- Sunyoung Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea;
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Tae-Don Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea;
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
6
|
D’Silva SZ, Singh M, Pinto AS. NK cell defects: implication in acute myeloid leukemia. Front Immunol 2023; 14:1112059. [PMID: 37228595 PMCID: PMC10203541 DOI: 10.3389/fimmu.2023.1112059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is a complex disease with rapid progression and poor/unsatisfactory outcomes. In the past few years, the focus has been on developing newer therapies for AML; however, relapse remains a significant problem. Natural Killer cells have strong anti-tumor potential against AML. This NK-mediated cytotoxicity is often restricted by cellular defects caused by disease-associated mechanisms, which can lead to disease progression. A stark feature of AML is the low/no expression of the cognate HLA ligands for the activating KIR receptors, due to which these tumor cells evade NK-mediated lysis. Recently, different Natural Killer cell therapies have been implicated in treating AML, such as the adoptive NK cell transfer, Chimeric antigen receptor-modified NK (CAR-NK) cell therapy, antibodies, cytokine, and drug treatment. However, the data available is scarce, and the outcomes vary between different transplant settings and different types of leukemia. Moreover, remission achieved by some of these therapies is only for a short time. In this mini-review, we will discuss the role of NK cell defects in AML progression, particularly the expression of different cell surface markers, the available NK cell therapies, and the results from various preclinical and clinical trials.
Collapse
Affiliation(s)
- Selma Z. D’Silva
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Meenakshi Singh
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Andrea S. Pinto
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| |
Collapse
|
7
|
Current Progress of CAR-NK Therapy in Cancer Treatment. Cancers (Basel) 2022; 14:cancers14174318. [PMID: 36077853 PMCID: PMC9454439 DOI: 10.3390/cancers14174318] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/23/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Chimeric antigen receptor (CAR)-T and -natural killer (NK) therapies are promising in cancer treatment. CAR-NK therapy gains great attention due to the lack of adverse effects observed in CAR-T therapies and to the NK cells’ unique mechanisms of recognizing target cells. Off-the-shelf products are in urgent need, not only for good yields, but also for lower cost and shorter preparation time. The current progress of CAR-NK therapy is discussed. Abstract CD8+ T cells and natural killer (NK) cells eliminate target cells through the release of lytic granules and Fas ligand (FasL)-induced target cell apoptosis. The introduction of chimeric antigen receptor (CAR) makes these two types of cells selective and effective in killing cancer cells. The success of CAR-T therapy in the treatment of acute lymphoblastic leukemia (ALL) and other types of blood cancers proved that the immunotherapy is an effective approach in fighting against cancers, yet adverse effects, such as graft versus host disease (GvHD) and cytokine release syndrome (CRS), cannot be ignored for the CAR-T therapy. CAR-NK therapy, then, has its advantage in lacking these adverse effects and works as effective as CAR-T in terms of killing. Despite these, NK cells are known to be hard to transduce, expand in vitro, and sustain shorter in vivo comparing to infiltrated T cells. Moreover, CAR-NK therapy faces challenges as CAR-T therapy does, e.g., the time, the cost, and the potential biohazard due to the use of animal-derived products. Thus, enormous efforts are needed to develop safe, effective, and large-scalable protocols for obtaining CAR-NK cells. Here, we reviewed current progress of CAR-NK therapy, including its biological properties, CAR compositions, preparation of CAR-NK cells, and clinical progresses. We also discussed safety issues raised from genetic engineering. We hope this review is instructive to the research community and a broad range of readers.
Collapse
|
8
|
Chu J, Gao F, Yan M, Zhao S, Yan Z, Shi B, Liu Y. Natural killer cells: a promising immunotherapy for cancer. J Transl Med 2022; 20:240. [PMID: 35606854 PMCID: PMC9125849 DOI: 10.1186/s12967-022-03437-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022] Open
Abstract
As a promising alternative platform for cellular immunotherapy, natural killer cells (NK) have recently gained attention as an important type of innate immune regulatory cell. NK cells can rapidly kill multiple adjacent cancer cells through non-MHC-restrictive effects. Although tumors may develop multiple resistance mechanisms to endogenous NK cell attack, in vitro activation, expansion, and genetic modification of NK cells can greatly enhance their anti-tumor activity and give them the ability to overcome drug resistance. Some of these approaches have been translated into clinical applications, and clinical trials of NK cell infusion in patients with hematological malignancies and solid tumors have thus far yielded many encouraging clinical results. CAR-T cells have exhibited great success in treating hematological malignancies, but their drawbacks include high manufacturing costs and potentially fatal toxicity, such as cytokine release syndrome. To overcome these issues, CAR-NK cells were generated through genetic engineering and demonstrated significant clinical responses and lower adverse effects compared with CAR-T cell therapy. In this review, we summarize recent advances in NK cell immunotherapy, focusing on NK cell biology and function, the types of NK cell therapy, and clinical trials and future perspectives on NK cell therapy.
Collapse
Affiliation(s)
- Junfeng Chu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Fengcai Gao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Meimei Yan
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Shuang Zhao
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Zheng Yan
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Bian Shi
- Department of Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China.
| | - Yanyan Liu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
9
|
Hossian AKMN, Hackett CS, Brentjens RJ, Rafiq S. Multipurposing CARs: Same engine, different vehicles. Mol Ther 2022; 30:1381-1395. [PMID: 35151842 PMCID: PMC9077369 DOI: 10.1016/j.ymthe.2022.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
T cells genetically engineered to recognize and eliminate tumor cells through synthetic chimeric antigen receptors (CARs) have demonstrated remarkable clinical efficacy against B cell leukemia over the past decade. This therapy is a form of highly personalized medicine that involves genetically modifying a patient's T cells to recognize and kill cancer cells. With the FDA approval of 5 CAR T cell products, this approach has been validated as a powerful new drug in the therapeutic armamentarium against cancer. Researchers are now studying how to expand this technology beyond its use in conventional polyclonal αβ T cells to address limitations to the current therapy in cancer and applications beyond it. Considering the specific characteristics of immune cell from diverse lineages, several preclinical and clinical studies are under way to assess the advantages of CAR-redirected function in these cells and apply the lessons learned from CAR T cell therapy in cancer to other diseases.
Collapse
Affiliation(s)
- A K M Nawshad Hossian
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
| | - Christopher S Hackett
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Renier J Brentjens
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA.
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
10
|
Biederstädt A, Rezvani K. Engineering the next generation of CAR-NK immunotherapies. Int J Hematol 2021; 114:554-571. [PMID: 34453686 PMCID: PMC8397867 DOI: 10.1007/s12185-021-03209-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022]
Abstract
Over the past few years, cellular immunotherapy has emerged as a novel treatment option for certain forms of hematologic malignancies with multiple CAR-T therapies now routinely administered in the clinic. The limitations of generating an autologous cell product and the challenges of toxicity with CAR-T cells underscore the need to develop novel cell therapy products that are universal, safe, and potent. Natural killer (NK) cells are part of the innate immune system with unique advantages, including the potential for off-the-shelf therapy. A recent first-in-human trial of CD19-CAR-NK infusion in patients with relapsed/refractory lymphoid malignancies proved safe with promising clinical activity. Building on these encouraging clinical responses, research is now actively exploring ways to further enhance CAR-NK cell potency by prolonging in vivo persistence and overcoming mechanisms of functional exhaustion. Besides these strategies to modulate CAR-NK cell intrinsic properties, there are increasing efforts to translate the successes seen in hematologic malignancies to the solid tumor space. This review will provide an overview on current trends and evolving concepts to genetically engineer the next generation of CAR-NK therapies. Emphasis will be placed on innovative multiplexed engineering approaches including CRISPR/Cas9 to overcome CAR-NK functional exhaustion and reprogram immune cell metabolism for enhanced potency.
Collapse
Affiliation(s)
- Alexander Biederstädt
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 423, Houston, TX, USA
- Department of Medicine III, Hematology/Oncology, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 423, Houston, TX, USA.
| |
Collapse
|
11
|
Wu X, Sharma A, Oldenburg J, Weiher H, Essler M, Skowasch D, Schmidt-Wolf IGH. NKG2D Engagement Alone Is Sufficient to Activate Cytokine-Induced Killer Cells While 2B4 Only Provides Limited Coactivation. Front Immunol 2021; 12:731767. [PMID: 34691037 PMCID: PMC8529192 DOI: 10.3389/fimmu.2021.731767] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/08/2021] [Indexed: 12/29/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are an ex vivo expanded heterogeneous cell population with an enriched NK-T phenotype (CD3+CD56+). Due to the convenient and relatively inexpensive expansion capability, together with low incidence of graft versus host disease (GVHD) in allogeneic cancer patients, CIK cells are a promising candidate for immunotherapy. It is well known that natural killer group 2D (NKG2D) plays an important role in CIK cell-mediated antitumor activity; however, it remains unclear whether its engagement alone is sufficient or if it requires additional co-stimulatory signals to activate the CIK cells. Likewise, the role of 2B4 has not yet been identified in CIK cells. Herein, we investigated the individual and cumulative contribution of NKG2D and 2B4 in the activation of CIK cells. Our analysis suggests that (a) NKG2D (not 2B4) is implicated in CIK cell (especially CD3+CD56+ subset)-mediated cytotoxicity, IFN-γ secretion, E/T conjugate formation, and degranulation; (b) NKG2D alone is adequate enough to induce degranulation, IFN-γ secretion, and LFA-1 activation in CIK cells, while 2B4 only provides limited synergy with NKG2D (e.g., in LFA-1 activation); and (c) NKG2D was unable to costimulate CD3. Collectively, we conclude that NKG2D engagement alone suffices to activate CIK cells, thereby strengthening the idea that targeting the NKG2D axis is a promising approach to improve CIK cell therapy for cancer patients. Furthermore, CIK cells exhibit similarities to classical invariant natural killer (iNKT) cells with deficiencies in 2B4 stimulation and in the costimulation of CD3 with NKG2D. In addition, based on the current data, the divergence in receptor function between CIK cells and NK (or T) cells can be assumed, pointing to the possibility that molecular modifications (e.g., using chimeric antigen receptor technology) on CIK cells may need to be customized and optimized to maximize their functional potential.
Collapse
Affiliation(s)
- Xiaolong Wu
- Department of Integrated Oncology, Center of Integrated Oncology (CIO) Bonn, University Hospital Bonn, Bonn, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center of Integrated Oncology (CIO) Bonn, University Hospital Bonn, Bonn, Germany.,Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Hans Weiher
- Department of Applied Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Markus Essler
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Dirk Skowasch
- Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center of Integrated Oncology (CIO) Bonn, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
12
|
Marofi F, Saleh MM, Rahman HS, Suksatan W, Al-Gazally ME, Abdelbasset WK, Thangavelu L, Yumashev AV, Hassanzadeh A, Yazdanifar M, Motavalli R, Pathak Y, Naimi A, Baradaran B, Nikoo M, Khiavi FM. CAR-engineered NK cells; a promising therapeutic option for treatment of hematological malignancies. Stem Cell Res Ther 2021; 12:374. [PMID: 34215336 PMCID: PMC8252313 DOI: 10.1186/s13287-021-02462-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022] Open
Abstract
Adoptive cell therapy has received a great deal of interest in the treatment of advanced cancers that are resistant to traditional therapy. The tremendous success of chimeric antigen receptor (CAR)-engineered T (CAR-T) cells in the treatment of cancer, especially hematological cancers, has exposed CAR's potential. However, the toxicity and significant limitations of CAR-T cell immunotherapy prompted research into other immune cells as potential candidates for CAR engineering. NK cells are a major component of the innate immune system, especially for tumor immunosurveillance. They have a higher propensity for immunotherapy in hematologic malignancies because they can detect and eliminate cancerous cells more effectively. In comparison to CAR-T cells, CAR-NK cells can be prepared from allogeneic donors and are safer with a lower chance of cytokine release syndrome and graft-versus-host disease, as well as being a more efficient antitumor activity with high efficiency for off-the-shelf production. Moreover, CAR-NK cells may be modified to target various antigens while also increasing their expansion and survival in vivo. Extensive preclinical research has shown that NK cells can be effectively engineered to express CARs with substantial cytotoxic activity against both hematological and solid tumors, establishing evidence for potential clinical trials of CAR-NK cells. In this review, we discuss recent advances in CAR-NK cell engineering in a variety of hematological malignancies, as well as the main challenges that influence the outcomes of CAR-NK cell-based tumor immunotherapies.
Collapse
Affiliation(s)
- Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marwan Mahmood Saleh
- Department of Biophysics, College of Applied Science, University of Anbar, Ramadi, Iraq
| | - Heshu Sulaiman Rahman
- College of Medicine, University of Sulaimani, Sulaymaniyah, Iraq
- Department of Medical Laboratory Sciences, Komar University of Science and Technology, Chaq-Chaq Qularaise, Sulaimaniyah, Iraq
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210 Thailand
| | | | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | | | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA USA
| | - Roza Motavalli
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yashwant Pathak
- Professor and Associate Dean for Faculty Affairs, Taneja College of Pharmacy, University of South Florida, Tampa, FL USA
- Faculty of Pharmacy, Airlangga University, Surabaya, Indonesia
| | - Adel Naimi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Behzad Baradaran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | |
Collapse
|
13
|
Sun L, Gang X, Li Z, Zhao X, Zhou T, Zhang S, Wang G. Advances in Understanding the Roles of CD244 (SLAMF4) in Immune Regulation and Associated Diseases. Front Immunol 2021; 12:648182. [PMID: 33841431 PMCID: PMC8024546 DOI: 10.3389/fimmu.2021.648182] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
Proteins in the signaling lymphocytic activating molecule (SLAM) family play crucial roles in regulating the immune system. CD244 (SLAMF4) is a protein in this family, and is also a member of the CD2 subset of the immunoglobulin (Ig) superfamily. CD244 is a cell surface protein expressed by NK cells, T cells, monocytes, eosinophils, myeloid-derived suppressor cells, and dendritic cells. CD244 binds to the ligand CD48 on adjacent cells and transmits stimulatory or inhibitory signals that regulate immune function. In-depth studies reported that CD244 functions in many immune-related diseases, such as autoimmune diseases, infectious diseases, and cancers, and its action is essential for the onset and progression of these diseases. The discovery of these essential roles of CD244 suggests it has potential as a prognostic indicator or therapeutic target. This review describes the molecular structure and function of CD244 and its roles in various immune cells and immune-related diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Daher M, Rezvani K. Outlook for New CAR-Based Therapies with a Focus on CAR NK Cells: What Lies Beyond CAR-Engineered T Cells in the Race against Cancer. Cancer Discov 2021; 11:45-58. [PMID: 33277313 PMCID: PMC8137521 DOI: 10.1158/2159-8290.cd-20-0556] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/15/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022]
Abstract
Chimeric antigen receptor (CAR) engineering of T cells has revolutionized the field of cellular therapy for the treatment of cancer. Despite this success, autologous CAR-T cells have recognized limitations that have led to the investigation of other immune effector cells as candidates for CAR modification. Recently, natural killer (NK) cells have emerged as safe and effective platforms for CAR engineering. In this article, we review the advantages, challenges, and preclinical and clinical research advances in CAR NK cell engineering for cancer immunotherapy. We also briefly consider the feasibility and potential benefits of applying other immune effector cells as vehicles for CAR expression. SIGNIFICANCE: CAR engineering can redirect the specificity of immune effector cells, converting them to a much more potent weapon to combat cancer cells. Expanding this strategy to immune effectors beyond conventional T lymphocytes could overcome some of the limitations of CAR T cells, paving the way for safer and more effective off-the-shelf cellular therapy products.
Collapse
Affiliation(s)
- May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
15
|
Subversion of natural killer cell responses by a cytomegalovirus-encoded soluble CD48 decoy receptor. PLoS Pathog 2019; 15:e1007658. [PMID: 30947296 PMCID: PMC6448830 DOI: 10.1371/journal.ppat.1007658] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
Throughout evolution, cytomegaloviruses (CMVs) have been capturing genes from their hosts, employing the derived proteins to evade host immune defenses. We have recently reported the presence of a number of CD48 homologs (vCD48s) encoded by different pathogenic viruses, including several CMVs. However, their properties and biological relevance remain as yet unexplored. CD48, a cosignaling molecule expressed on the surface of most hematopoietic cells, modulates the function of natural killer (NK) and other cytotoxic cells by binding to its natural ligand 2B4 (CD244). Here, we have characterized A43, the vCD48 exhibiting the highest amino acid sequence identity with host CD48. A43, which is encoded by owl monkey CMV, is a soluble molecule released from the cell after being proteolytically processed through its membrane proximal region. A43 is expressed with immediate-early kinetics, yielding a protein that is rapidly detected in the supernatant of infected cells. Remarkably, surface plasmon resonance assays revealed that this viral protein binds to host 2B4 with high affinity and slow dissociation rates. We demonstrate that soluble A43 is capable to abrogate host CD48:2B4 interactions. Moreover, A43 strongly binds to human 2B4 and prevents 2B4-mediated NK-cell adhesion to target cells, therefore reducing the formation of conjugates and the establishment of immunological synapses between human NK cells and CD48-expressing target cells. Furthermore, in the presence of this viral protein, 2B4-mediated cytotoxicity and IFN-γ production by NK cells are severely impaired. In summary, we propose that A43 may serve as a functional soluble CD48 decoy receptor by binding and masking 2B4, thereby impeding effective NK cell immune control during viral infections. Thus, our findings provide a novel example of the immune evasion strategies developed by viruses. In order to evade detection and destruction by cytotoxic lymphocytes and successfully persist within their hosts, cytomegalovirus (CMVs) have evolved a number of genes dedicated to block immune recognition. Certain CMVs and other large DNA viruses encode homologs of the cell-surface molecule CD48, a ligand of the 2B4 receptor involved in regulating the function of cytotoxic lymphocytes. Here, we have investigated for the first time the immunomodulatory potential of one of these viral molecules. We show that A43, a CD48 homolog encoded by owl monkey CMV, is a soluble molecule that exhibits exceptional binding kinetics for 2B4, and is furthermore capable of blocking the interaction with its counter-receptor CD48. Moreover, we reveal how this viral protein interferes with human NK cell-mediated cytotoxicity by inhibiting the immune synapse between human NK cells and target cells. Thus, these findings not only underscore the importance of 2B4-mediated immune responses in controlling CMV infections, but also unveil the shedding of a virally-encoded soluble variant of CD48 as a new viral counteract mechanism for subverting immune surveillance.
Collapse
|
16
|
Cabinian A, Sinsimer D, Tang M, Jang Y, Choi B, Laouar Y, Laouar A. Gut symbiotic microbes imprint intestinal immune cells with the innate receptor SLAMF4 which contributes to gut immune protection against enteric pathogens. Gut 2018; 67:847-859. [PMID: 28341747 PMCID: PMC5890651 DOI: 10.1136/gutjnl-2016-313214] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 02/27/2017] [Accepted: 03/04/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Interactions between host immune cells and gut microbiota are crucial for the integrity and function of the intestine. How these interactions regulate immune cell responses in the intestine remains a major gap in the field. AIM We have identified the signalling lymphocyte activation molecule family member 4 (SLAMF4) as an immunomodulator of the intestinal immunity. The aim is to determine how SLAMF4 is acquired in the gut and what its contribution to intestinal immunity is. METHODS Expression of SLAMF4 was assessed in mice and humans. The mechanism of induction was studied using GFPtg bone marrow chimaera mice, lymphotoxin α and TNLG8A-deficient mice, as well as gnotobiotic mice. Role in immune protection was revealed using oral infection with Listeria monocytogenes and Cytobacter rodentium. RESULTS SLAMF4 is a selective marker of intestinal immune cells of mice and humans. SLAMF4 induction occurs directly in the intestinal mucosa without the involvement of the gut-associated lymphoid tissue. Gut bacterial products, particularly those of gut anaerobes, and gut-resident antigen-presenting cell (APC) TNLG8A are key contributors of SLAMF4 induction in the intestine. Importantly, lack of SLAMF4 expression leads the increased susceptibility of mice to infection by oral pathogens culminating in their premature death. CONCLUSIONS SLAMF4 is a marker of intestinal immune cells which contributes to the protection against enteric pathogens and whose expression is dependent on the presence of the gut microbiota. This discovery provides a possible mechanism for answering the long-standing question of how the intertwining of the host and gut microbial biology regulates immune cell responses in the gut.
Collapse
Affiliation(s)
- Allison Cabinian
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Daniel Sinsimer
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - May Tang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Youngsoon Jang
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Bongkum Choi
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Yasmina Laouar
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Amale Laouar
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| |
Collapse
|
17
|
Arjunaraja S, Angelus P, Su HC, Snow AL. Impaired Control of Epstein-Barr Virus Infection in B-Cell Expansion with NF-κB and T-Cell Anergy Disease. Front Immunol 2018; 9:198. [PMID: 29472930 PMCID: PMC5809398 DOI: 10.3389/fimmu.2018.00198] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 01/23/2018] [Indexed: 11/13/2022] Open
Abstract
B-cell expansion with NF-κB and T-cell anergy (BENTA) disease is a B-cell-specific lymphoproliferative disorder caused by germline gain-of-function mutations in CARD11. These mutations force the CARD11 scaffold into an open conformation capable of stimulating constitutive NF-κB activation in lymphocytes, without requiring antigen receptor engagement. Many BENTA patients also suffer from recurrent infections, with 7 out of 16 patients exhibiting chronic, low-grade Epstein–Barr virus (EBV) viremia. In this mini-review, we discuss EBV infection in the pathogenesis and clinical management of BENTA disease, and speculate on mechanisms that could explain inadequate control of viral infection in BENTA patients.
Collapse
Affiliation(s)
- Swadhinya Arjunaraja
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Pamela Angelus
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., National Cancer Institute at Frederick, Frederick, MD, United States
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
18
|
Hanaoka N, Jabri B, Dai Z, Ciszewski C, Stevens AM, Yee C, Nakakuma H, Spies T, Groh V. NKG2D initiates caspase-mediated CD3zeta degradation and lymphocyte receptor impairments associated with human cancer and autoimmune disease. THE JOURNAL OF IMMUNOLOGY 2010; 185:5732-42. [PMID: 20926796 DOI: 10.4049/jimmunol.1002092] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Deficiencies of the T cell and NK cell CD3ζ signaling adapter protein in patients with cancer and autoimmune diseases are well documented, but mechanistic explanations are fragmentary. The stimulatory NKG2D receptor on T and NK cells mediates tumor immunity but can also promote local and systemic immune suppression in conditions of persistent NKG2D ligand induction that include cancer and certain autoimmune diseases. In this paper, we provide evidence that establishes a causative link between CD3ζ impairment and chronic NKG2D stimulation due to pathological ligand expression. We describe a mechanism whereby NKG2D signaling in human T and NK cells initiates Fas ligand/Fas-mediated caspase-3/-7 activation and resultant CD3ζ degradation. As a consequence, the functional capacities of the TCR, the low-affinity Fc receptor for IgG, and the NKp30 and NKp46 natural cytotoxicity receptors, which all signal through CD3ζ, are impaired. These findings are extended to ex vivo phenotypes of T and NK cells among tumor-infiltrating lymphocytes and in peripheral blood from patients with juvenile-onset lupus. Collectively, these results indicate that pathological NKG2D ligand expression leads to simultaneous impairment of multiple CD3ζ-dependent receptor functions, thus offering an explanation that may be applicable to CD3ζ deficiencies associated with diverse disease conditions.
Collapse
Affiliation(s)
- Nobuyoshi Hanaoka
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Betser-Cohen G, Mizrahi S, Elboim M, Alsheich-Bartok O, Mandelboim O. The association of MHC class I proteins with the 2B4 receptor inhibits self-killing of human NK cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:2761-8. [PMID: 20164429 DOI: 10.4049/jimmunol.0901572] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The killing activity of NK cells is carried out by several activating NK receptors, which includes NKp46, NKp44, NKp30, NKp80, NKG2D, and 2B4. The ligands of these receptors are either self-derived, pathogen-derived, stress-induced ligands or tumor ligands. Importantly, none of these killer ligands are expressed on NK cells and thus self-killing of NK cells is prevented. A notable exception with this regard, is the ligand of the 2B4 receptor. This unusual receptor can exert both activating and inhibiting signals; however, in human NK cells, it serves mainly as an activating receptor. The ligand of 2B4 is CD48 and in contrast to the ligands of all the other NK activating receptors, CD48 is also present on NK cells. Thus, NK cells might be at risk for self-killing that is mediated via the 2B4-CD48 interaction. In this study, we identify a novel mechanism that prevents this self-killing as we show that the association of the MHC class I proteins with the 2B4 receptor, both present on NK cells, results in the attenuation of the 2B4-mediated self-killing of NK cells.
Collapse
Affiliation(s)
- Gili Betser-Cohen
- The Lautenberg Center for General and Tumor Immunology, The Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
20
|
The natural killer cell: a further innate mediator of gouty inflammation? Immunol Cell Biol 2009; 88:24-31. [DOI: 10.1038/icb.2009.91] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
21
|
Functional SNPs in CD244 increase the risk of rheumatoid arthritis in a Japanese population. Nat Genet 2008; 40:1224-9. [DOI: 10.1038/ng.205] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Accepted: 06/23/2008] [Indexed: 12/25/2022]
|
22
|
Calpe S, Wang N, Romero X, Berger SB, Lanyi A, Engel P, Terhorst C. The SLAM and SAP gene families control innate and adaptive immune responses. Adv Immunol 2008; 97:177-250. [PMID: 18501771 DOI: 10.1016/s0065-2776(08)00004-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The nine SLAM-family genes, SLAMF1-9, a subfamily of the immunoglobulin superfamily, encode differentially expressed cell-surface receptors of hematopoietic cells. Engagement with their ligands, which are predominantly homotypic, leads to distinct signal transduction events, for instance those that occur in the T or NK cell immune synapse. Upon phosphorylation of one or more copies of a unique tyrosine-based signaling motif in their cytoplasmic tails, six of the SLAM receptors recruit the highly specific single SH2-domain adapters SLAM-associated protein (SAP), EAT-2A, and/or EAT-2B. These adapters in turn bind to the tyrosine kinase Fyn and/or other protein tyrosine kinases connecting the receptors to signal transduction networks. Individuals deficient in the SAP gene, SH2D1A, develop an immunodeficiency syndrome: X-linked lympho-proliferative disease. In addition to operating in the immune synapse, SLAM receptors initiate or partake in multiple effector functions of hematopoietic cells, for example, neutrophil and macrophage killing and platelet aggregation. Here we discuss the current understanding of the structure and function of these recently discovered receptors and adapter molecules in the regulation of adaptive and innate immune responses.
Collapse
Affiliation(s)
- Silvia Calpe
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Roda-Navarro P, Reyburn HT. Intercellular protein transfer at the NK cell immune synapse: mechanisms and physiological significance. FASEB J 2007; 21:1636-46. [PMID: 17314139 DOI: 10.1096/fj.06-7488rev] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Immune synapses (IS) are supramolecular clusters providing intercellular communication among cells of the immune system. While the physiological role and consequences of IS formation are beginning to be understood, these studies have given rise to a new research topic in the biology of lymphocyte interactions: synaptic transfer of proteins between lymphocytes. During natural killer (NK) cell immunosurveillance, clustering and transfer of receptor and ligand molecules have been observed at both the inhibitory and cytotoxic NK cell immune synapse (NK-IS). The transfer of activating receptors seems to be associated with receptor distribution to thin membrane connective structures (MCS)/nanotubes that communicate effector and susceptible target cells. Strikingly, bidirectional transfer of the activating receptor NKG2D and its cellular ligand MICB correlates with a reduction in NK cell cytotoxic function. In this regard, synaptic uptake of MICB may represent a novel strategy of tumor immune evasion. Finally, synaptic acquisition of receptors by NK cells may also favor the spread of pathogens. In this review we discuss possible mechanisms of synaptic protein transfer and propose different testable hypotheses about the physiological and pathological significance of this process for NK cell function.
Collapse
|
24
|
Wendt K, Wilk E, Buyny S, Buer J, Schmidt RE, Jacobs R. Gene and protein characteristics reflect functional diversity of CD56dim and CD56bright NK cells. J Leukoc Biol 2006; 80:1529-41. [PMID: 16966385 DOI: 10.1189/jlb.0306191] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Recent findings underline the role of NK cell subsets in regulating adaptive immunity. To define characteristics of NK cell subpopulations, purified CD56(dim) and CD56(bright) NK cells were analyzed by using gene chip arrays covering more than 39,000 transcripts. Gene profiling revealed resting NK cells to differ in respect to 473 transcripts with 176 exclusively expressed in CD56(dim) and 130 solely in CD56(bright) NK cells. Results were compared with array analyses using mRNA obtained from activated CD56(dim) and CD56(bright) NK cells. In this approach, NK cell receptors, cytolytic molecules, adhesion structures, and chemokine ligands showed differential expression patterns in the two subpopulations. These data were validated using FACS, RT-qPCR, or cytokine bead array (CBA) techniques. Cytokines produced by CD56(dim) and CD56(bright) NK cells were determined using a protein array covering 79 different bioactive mediators. GDNF, IGFBP-1, EGF, and TIMP-2 were detected in both subsets. In contrast, IGFBP-3 and IGF-1 were mainly produced by CD56(dim), while GM-CSF, TARC, and TGFbeta3 were expressed by CD56(bright) NK cells. In summary, we report new characteristic features of CD56(dim) and CD56(bright) NK cells, further underscoring that they represent independent populations with functionally diverse capabilities. The information on NK cells generated in this study will help to define corresponding NK cell populations in other species that lack CD56 expression on NK cells, such as mice. This will subsequently lead to the establishment of suitable animal models for detailed analysis of NK cell populations in vivo.
Collapse
Affiliation(s)
- Katy Wendt
- Department of Clinical Immunology, OE 6830, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover D-30625, Germany
| | | | | | | | | | | |
Collapse
|
25
|
Messmer B, Eissmann P, Stark S, Watzl C. CD48 Stimulation by 2B4 (CD244)-Expressing Targets Activates Human NK Cells. THE JOURNAL OF IMMUNOLOGY 2006; 176:4646-50. [PMID: 16585556 DOI: 10.4049/jimmunol.176.8.4646] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human NK cells can be activated by a variety of different cell surface receptors. Members of the SLAM-related receptors (SRR) are important modulators of NK cell activity. One interesting feature of the SRR is their homophilic interaction, combining receptor and ligand in the same molecule. Therefore, SRR cannot only function as activating NK cell receptors, but also as activating NK cell ligands. 2B4 (CD244) is the only SRR that does not show homophilic interaction. Instead, 2B4 is activated by binding to CD48, a GPI-anchored surface molecule that is widely expressed in the hemopoietic system. In this study, we show that 2B4 also can function as an activating NK cell ligand. 2B4-expressing target cells can efficiently stimulate NK cell cytotoxicity and IFN-gamma production. Using soluble receptor fusion proteins and SRR-transfected cells, we show that 2B4 does not bind to any other SRR expressed on NK cells, but only interacts with CD48. Lysis of 2B4-expressing target cells can be blocked by anti-CD48 Abs and triggering of CD48 in a redirected lysis assay can stimulate NK cell cytotoxicity. This demonstrates that 2B4 can stimulate NK cell cytotoxicity and cytokine production by interacting with NK cell expressed CD48 and adds CD48 to the growing number of activating NK cell receptors.
Collapse
Affiliation(s)
- Birgitta Messmer
- Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | | | | | | |
Collapse
|
26
|
Saborit-Villarroya I, Del Valle JM, Romero X, Esplugues E, Lauzurica P, Engel P, Martín M. The adaptor protein 3BP2 binds human CD244 and links this receptor to Vav signaling, ERK activation, and NK cell killing. THE JOURNAL OF IMMUNOLOGY 2005; 175:4226-35. [PMID: 16177062 DOI: 10.4049/jimmunol.175.7.4226] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adaptor proteins, molecules that mediate intermolecular interactions, are crucial for cellular activation. The adaptor 3BP2 has been shown to positively regulate NK cell-mediated cytotoxicity. In this study we present evidence for a physical interaction between 3BP2 and the CD244 receptor. CD244, a member of the CD150 family, is a cell surface protein expressed on NK, CD8+ T, and myeloid cells. CD244 interacts via its Src homology 2 domain with the X-linked lymphoproliferative disease gene product signaling lymphocytic activation molecule-associated protein (SAP)/SH2 domain protein 1A. 3BP2 interacts with human but not murine CD244. CD244-3BP2 interaction was direct and regulated by phosphorylation, as shown by a three-hybrid analysis in yeast and NK cells. Tyr337 on CD244, part of a consensus motif for SAP/SH2 domain protein 1A binding, was critical for the 3BP2 interaction. Although mutation of Tyr337 to phenylalanine abrogated human 3BP2 binding, we still observed SAP association, indicating that this motif is not essential for SAP recruitment. CD244 ligation induced 3BP2 phosphorylation and Vav-1 recruitment. Overexpression of 3BP2 led to an increase in the magnitude and duration of ERK activation, after CD244 triggering. This enhancement was concomitant with an increase in cytotoxicity due to CD244 ligation. However, no differences in IFN-gamma secretion were found when normal and 3BP2-transfected cells were compared. These results indicate that CD244-3BP2 association regulates cytolytic function but not IFN-gamma release, reinforcing the hypothesis that, in humans, CD244-mediated cytotoxicity and IFN-gamma release involve distinct NK pathways.
Collapse
Affiliation(s)
- Ifigènia Saborit-Villarroya
- Unitat d'Immunología, Departament de Biologia Cellular i Anatomia Patològica, Facultat de Medicina, Universitat de Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
27
|
Zieker D, Fehrenbach E, Dietzsch J, Fliegner J, Waidmann M, Nieselt K, Gebicke-Haerter P, Spanagel R, Simon P, Niess AM, Northoff H. cDNA microarray analysis reveals novel candidate genes expressed in human peripheral blood following exhaustive exercise. Physiol Genomics 2005; 23:287-94. [PMID: 16118270 DOI: 10.1152/physiolgenomics.00096.2005] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
It is generally accepted that exhausting endurance exercise exhibits strong effects on the immune system. Such effects have been attributed to changes in the cellular composition of peripheral blood as well as to changes in the expression of plausible candidate genes. The list of candidate genes is far from being complete, since this issue has not yet been investigated in a systematic way. In this study, we used a custom-made cDNA microarray focused on inflammation as a screening approach to study gene expression in eight one-half marathon runners before, immediately after, and 24 h after exercise. Significant differential gene expression was verified by quantitative real-time PCR. Linear regression analysis showed that microarray expression analysis of cell type-specific surface molecules reflects the observed individual cellular shifts in peripheral blood cells with high statistical significance. In line with the results of former studies, we observed an upregulation of mitogen-activated protein kinase-activated protein kinase-2 (MAPKAP-K2), l-selectin, and IL-1 receptor antagonist (IL-1ra) after exhaustive exercise. The main results of this study report, for the first time, the downregulation of CD81; the upregulation of thioredoxin, which may play an important part in anti-oxidative defense; and, surprisingly, the downregulation of the anti-carcinogenic gene glutathione- S-transferase-3 (GSTM3) in peripheral blood. The study shows cDNA microarray expression analysis as a reliable systematic instrument to complete the list of candidate genes that may play a role in exhaustive exercise-induced modulation of the immune response.
Collapse
Affiliation(s)
- Derek Zieker
- Department of Transfusions Medicine, University of Tuebingen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ostrowski SR, Ullum H, Pedersen BK, Gerstoft J, Katzenstein TL. 2B4 expression on natural killer cells increases in HIV-1 infected patients followed prospectively during highly active antiretroviral therapy. Clin Exp Immunol 2005; 141:526-33. [PMID: 16045743 PMCID: PMC1809452 DOI: 10.1111/j.1365-2249.2005.02869.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2005] [Indexed: 11/28/2022] Open
Abstract
Human immunodeficiency virus (HIV)-1 infection influences natural killer (NK) cell expression of inhibitory NK receptors and activating natural cytotoxicity receptors. It is unknown whether expression of the co-stimulatory NK cell receptor 2B4 (CD244) on NK cells and CD3+ CD8+ cells are affected by highly active antiretroviral therapy (HAART), low-level viraemia, proviral-DNA or immune activation in HIV-1 infected patients. A total of 101 HAART-treated HIV-1 infected patients with < or = 200 HIV-RNA copies/ml were followed prospectively for 24 months. HIV-RNA was investigated 3-monthly and 2B4 expression on CD3- CD16+ NK cells and CD3+ CD8+ cells, proviral-DNA and plasma soluble tumour necrosis factor receptor (sTNFr)-II were investigated 6-monthly. For comparison, 2B4 expression was investigated in 20 healthy individuals. The concentration of 2B4+ NK cells was initially reduced in HIV-1 infected patients (P < 0.001) but increased to a normal level during the 24 months' follow-up. The concentration of CD3+ CD8+ 2B4+ cells in HIV-1 infected patients was normal and did not change during follow-up. The relative fluorescence intensity (RFI) of 2B4 increased on both NK cells and CD3+ CD8+ cells during follow-up (both P < 0.001). Higher levels of proviral-DNA carrying cells and plasma sTNFrII were associated with reductions in the concentration of 2B4+ NK cells (all P < 0.05). HIV-RNA had no effect on 2B4 expression on NK cells or CD3+ CD8+ cells. These findings demonstrate that the concentration of 2B4+ NK cells normalizes during long-term HAART in HIV-1 infected patients. The finding that proviral-DNA and sTNFrII were associated negatively with the concentration of 2B4+ NK cells suggests that immune activation in HIV-1 infected patients receiving HAART influences the target cell recognition by NK cells.
Collapse
Affiliation(s)
- S R Ostrowski
- Department of Infectious Diseases, Rishospitalet, Copenhagen, Denmark.
| | | | | | | | | |
Collapse
|
29
|
Aoukaty A, Tan R. Role for glycogen synthase kinase-3 in NK cell cytotoxicity and X-linked lymphoproliferative disease. THE JOURNAL OF IMMUNOLOGY 2005; 174:4551-8. [PMID: 15814676 DOI: 10.4049/jimmunol.174.8.4551] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
NK cells from individuals with X-linked lymphoproliferative (XLP) disease exhibit functional defects when stimulated through the NK receptor, 2B4 (CD244). These defects are likely a consequence of aberrant intracellular signaling initiated by mutations of the adaptor molecule SLAM-associated protein. In this report, we show that NK cells from individuals with XLP but not healthy individuals fail to phosphorylate and thereby inactivate glycogen synthase kinase-3 (GSK-3) following 2B4 stimulation. Lack of GSK-3 phosphorylation prevented the accumulation of the transcriptional coactivator beta-catenin in the cytoplasm and its subsequent translocation to the nucleus. Potential signaling pathways leading from 2B4 stimulation to GSK-3 phosphorylation were also investigated. Ligation of 2B4 resulted in the phosphorylation of the guanine nucleotide exchange factor, Vav-1, and subsequent activation of the GTP-binding protein Rac-1 (but not Ras) and the serine-threonine kinase Raf-1 in healthy but not XLP-derived NK cells. In addition, the activity of MEK-2 (but not MEK-1) was up-regulated, and Erk1/2 was phosphorylated in normal NK cells but not those from an individual with XLP suggesting that these proteins relay SLAM-associated protein-dependent signals from 2B4. Finally, inactivation of GSK-3 using a specific inhibitor of GSK-3beta increased the cytotoxicity and cytokine secretion of both healthy and XLP NK cells. These data indicate that the signaling of 2B4 in NK cells is mediated by GSK-3 and beta-catenin, possibly through a signal transduction pathway that involves Vav-1, Rac-1, Raf-1, MEK-2, and Erk1/2 and that this pathway is aberrant in individuals with XLP.
Collapse
Affiliation(s)
- Ala Aoukaty
- Department of Pathology and Laboratory Medicine, British Columbia's Children's Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
30
|
Assarsson E, Kambayashi T, Persson CM, Ljunggren HG, Chambers BJ. 2B4 co-stimulation: NK cells and their control of adaptive immune responses. Mol Immunol 2005; 42:419-23. [PMID: 15607793 DOI: 10.1016/j.molimm.2004.07.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
NK cells have primarily been defined by their ability to kill infected cells, tumor cells and some normal cells expressing low levels of MHC class I molecules. NK cells have also been shown to affect adaptive immune responses by their production of both pro- and anti-inflammatory cytokines. Recently it has been shown that adaptive immune responses can be enhanced or maintained also through direct lymphocyte-lymphocyte interactions. One of these interactions was identified to occur between 2B4 and CD48, where 2B4 acted as a co-stimulatory ligand for both NK cells and T cells. In the current article, we discuss the role of 2B4 in the development of adaptive immune responses and the role of NK-T cell interactions in these responses.
Collapse
Affiliation(s)
- Erika Assarsson
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, F59, Karolinska University Hospital Huddinge, S-14186 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
31
|
Bigger CB, Guerra B, Brasky KM, Hubbard G, Beard MR, Luxon BA, Lemon SM, Lanford RE. Intrahepatic gene expression during chronic hepatitis C virus infection in chimpanzees. J Virol 2004; 78:13779-92. [PMID: 15564486 PMCID: PMC533929 DOI: 10.1128/jvi.78.24.13779-13792.2004] [Citation(s) in RCA: 220] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Accepted: 07/30/2004] [Indexed: 12/24/2022] Open
Abstract
Hepatitis C virus (HCV) infections represent a global health problem and are a major contributor to end-stage liver disease including cirrhosis and hepatocellular carcinoma. An improved understanding of the parameters involved in disease progression is needed to develop better therapies and diagnostic markers of disease manifestation. To better understand the dynamics of host gene expression resulting from persistent virus infection, DNA microarray analyses were conducted on livers from 10 chimpanzees persistently infected with HCV. A total of 162 genes were differentially regulated in chronically infected animals compared to uninfected controls. Many genes exhibited a remarkable consistency in changes in expression in the 10 chronically infected animals. A second method of analysis identified 971 genes altered in expression during chronic infection at a 99% confidence level. As with acute-resolving HCV infections, many interferon (IFN)-stimulated genes (ISGs) were transcriptionally elevated, suggesting an ongoing response to IFN and/or double-stranded RNA which is amplified in downstream ISG expression. Thus, persistent infection with HCV results in a complex and partially predictable pattern of gene expression, although the underlying mechanisms regulating the different pathways are not well defined. A single genotype 3-infected animal was available for analysis, and this animal exhibited reduced levels of ISG expression compared to levels of expression with genotype 1 infections and increased expression of a number of genes potentially involved in steatosis. Gene expression data in concert with other observations from HCV infections permit speculation on the regulation of specific aspects of HCV infection.
Collapse
Affiliation(s)
- Catherine B Bigger
- Department of Virology and Immunology, Southwest National Primate Research Center, Southwest Foundation for Biomedical Research, 7620 NW Loop 410, San Antonio, TX 78227, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Assarsson E, Kambayashi T, Schatzle JD, Cramer SO, von Bonin A, Jensen PE, Ljunggren HG, Chambers BJ. NK Cells Stimulate Proliferation of T and NK Cells through 2B4/CD48 Interactions. THE JOURNAL OF IMMUNOLOGY 2004; 173:174-80. [PMID: 15210772 DOI: 10.4049/jimmunol.173.1.174] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Few studies have addressed the consequences of physical interactions between NK and T cells, as well as physical interactions among NK cells themselves. We show in this study that NK cells can enhance T cell activation and proliferation in response to CD3 cross-linking and specific Ag through interactions between 2B4 (CD244) on NK cells and CD48 on T cells. Furthermore, 2B4/CD48 interactions between NK cells also enhanced proliferation of NK cells in response to IL-2. Overall, these results suggest that NK cells augment the proliferation of neighboring T and NK cells through direct cell-cell contact. These results provide new insights into NK cell-mediated control of innate and adaptive immunity and demonstrate that receptor/ligand-specific cross talk between lymphocytes may occur in settings other than T-B cell or T-T cell interactions.
Collapse
Affiliation(s)
- Erika Assarsson
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Huddinge University Hospital, SE-141 86 Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Grau R, Lang KS, Wernet D, Lang P, Niethammer D, Pusch CM, Handgretinger R. Cytotoxic activity of natural killer cells lacking killer-inhibitory receptors for self-HLA class I molecules against autologous hematopoietic stem cells in healthy individuals. Exp Mol Pathol 2004; 76:90-8. [PMID: 15010286 DOI: 10.1016/j.yexmp.2003.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2003] [Indexed: 11/22/2022]
Abstract
Killer-inhibitory receptors (KIR) are receptors for self-HLA class I molecules, which are expressed on natural killer (NK) cells and small subsets of T-lymphocytes. KIR receptors that do not bind to self-HLA class I have been implicated in the pathogenesis of pure red-cell aplasia and other autoimmune diseases. However, NK cells whose inhibitory receptors lack any apparent self-ligand can also be found in healthy individuals. We therefore tested whether these NK cells are capable of exerting cytotoxic activity against autologous CD34(+) hematopoietic precursors. We detected NK cells whose sole inhibitory receptors were CD94/NKG2-A and that had no affinity for autologous HLA-C molecules. In vitro, such cells were able to kill autologous CD34(+) stem cells that expressed MHC class I antigen at a high level in about 50% of the cases of HLA-C group 2 donors. Two individual clones derived from this NK subpopulation were stimulated by autologous HLA-Cw5/6-positive stem cells, but not by allogeneic HLA-Cw7-positive stem cells. Our findings demonstrate the presence of potentially autoreactive natural killer cells in otherwise healthy individuals.
Collapse
Affiliation(s)
- Roger Grau
- Department of Hematology, Children's University Hospital, Tuebingen, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Prominent blood and tissue eosinophilia is manifested in a number of inflammatory states, particularly in allergic diseases. Eosinophils are a source of numerous cytokines and growth factors, thus in principle they can display both pro-inflammatory and anti-inflammatory activities as well as immunoregulatory ones. In this review, we will discuss the cross-talk between eosinophils and other cell types that they come in contact with in the inflammatory milieu, such as mast cells, fibroblasts and endothelial cells. 'New' roles for eosinophils in cancer and novel activatory signals will also be described.
Collapse
Affiliation(s)
- A Munitz
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | |
Collapse
|
35
|
Engel P, Eck MJ, Terhorst C. The SAP and SLAM families in immune responses and X-linked lymphoproliferative disease. Nat Rev Immunol 2003; 3:813-21. [PMID: 14523387 DOI: 10.1038/nri1202] [Citation(s) in RCA: 232] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SAP (signalling lymphocytic activation molecule (SLAM)-associated protein) is a T- and natural killer (NK)-cell-specific protein containing a single SH2 domain encoded by a gene that is defective or absent in patients with X-linked lymphoproliferative syndrome (XLP). The SH2 domain of SAP binds with high affinity to the cytoplasmic tail of the haematopoietic cell-surface glycoprotein SLAM and five related receptors. SAP regulates signal transduction of the SLAM-family receptors by recruiting SRC kinases. Similarly, the SAP-related proteins EAT2A and EAT2B are thought to control signal transduction that is initiated by SLAM-related receptors in professional antigen-presenting cells. In this review, we discuss recent findings on the structure and function of proteins of the SAP and SLAM families.
Collapse
Affiliation(s)
- Pablo Engel
- Immunology Unit, Department of Cellular Biology and Pathology, Medical School, University of Barcelona, Barcelona 08036, Spain.
| | | | | |
Collapse
|
36
|
Chuang SS, Lee JK, Mathew PA. Protein kinase C is involved in 2B4 (CD244)-mediated cytotoxicity and AP-1 activation in natural killer cells. Immunology 2003; 109:432-9. [PMID: 12807490 PMCID: PMC1782976 DOI: 10.1046/j.1365-2567.2003.01662.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
2B4 (CD244) is a member of the CD2 subset of the immunoglobulin superfamily and functions as a triggering molecule on natural killer (NK) cells. Previously, we have found that 2B4-mediated activation of NK cells involves complex interactions involving LAT, Ras, Raf, ERK and p38 and that cytolytic function and cytokine production may be regulated by distinct pathways. Here we assessed the role of protein kinase C (PKC) in 2B4-mediated cytotoxicity of YT cells, a human NK cell line. Our data indicate that PKC-delta is activated upon stimulation with monoclonal antibody against 2B4. Treatment with the PKC inhibitor, bisindolylmaleimide I (Gö6850), of YT cells or YT cells depleted of Ca2+-dependent isoforms of PKC prior to 2B4 stimulation, resulted in inhibition of natural cytotoxicity and redirected antibody-dependent cellular cytotoxicity. However, inhibition of PKC failed to block 2B4 stimulation of interferon-gamma secretion as opposed to pretreatment with LY294002, a phosphoinositide 3-kinase inhibitor. We also examined the effect of phorbol 12-myristate 13-acetate (PMA) induction on 2B4 gene transcription. PMA induction resulted in a more than two-fold increase of 2B4 transcription. However, when we introduced a three-base substitution mutation to disrupt the activator protein-1 binding site at (-106 to -100) in the 2B4 promoter, we found complete loss of transcriptional activity, including the two-fold increase due to PMA induction of PKC. The present study indicated that PKC may play an important role in 2B4 signalling and activator protein-1 activation.
Collapse
Affiliation(s)
- Samuel S Chuang
- Department of Molecular Biology and Immunology and Institute for Cancer Research, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX 76107-2699, USA
| | | | | |
Collapse
|
37
|
Abstract
The 'signalling lymphocyte activation molecule' (SLAM) family is a newly appreciated group of immune-cell specific receptors that has the ability to regulate the function of several immune cell types. Recent studies show that the SLAM-related receptors mediate intracellular protein tyrosine phosphorylation signals. This property is dependent on the aptitude of SLAM-family receptors to bind with high affinity to SAP and/or EAT-2, two small adaptor molecules composed almost exclusively of a Src homology 2 domain. SAP is mutated in X-linked lymphoproliferative disease, a human immune dysfunction characterised by an inappropriate response to Epstein-Barr virus infection, thereby suggesting that the SLAM-related receptors may be critical for a normal immune response. The existence of the SLAM family broadens the spectrum of receptors known to be involved in immunomodulation.
Collapse
Affiliation(s)
- André Veillette
- Laboratory of Molecular Oncology, Clinical Research Institute of Montreal, Montréal, Québec, Canada.
| | | |
Collapse
|
38
|
Parolini O, Weinhäusel A, Kagerbauer B, Sassmann J, Holter W, Gadner H, Haas OA, Knapp W. Differential methylation pattern of the X-linked lymphoproliferative (XLP) disease gene SH2D1A correlates with the cell lineage-specific transcription. Immunogenetics 2003; 55:116-21. [PMID: 12709835 DOI: 10.1007/s00251-003-0557-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2003] [Revised: 02/06/2003] [Indexed: 10/25/2022]
Abstract
SH2D1A, the X-linked lymphoproliferative disease (XLP) gene, encodes a cytoplasmic protein that plays an essential role in controlling Epstein-Barr virus infection. It is expressed in T and NK cells, but not in B cells or in granulocytes. The promoter, the regulatory regions, as well as the mechanisms controlling its tissue-specific expression, are still unknown. We tested the hypothesis that DNA methylation might contribute to tissue-specific SH2D1A gene expression and analyzed the methylation status of 2,300 bp upstream of the ATG starting codon, the coding region and part of intron 1. By bisulfite sequencing and methylation-sensitive restriction enzyme digestion, we show that a differential methylation pattern of CpG-rich regions in the 5' region and the adjacent exon 1 of the SH2D1A gene indeed correlates with the tissue-specific gene transcription.
Collapse
Affiliation(s)
- Ornella Parolini
- Institute of Immunology, University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Chiorean EG, Dylla SJ, Olsen K, Lenvik T, Soignier Y, Miller JS. BCR/ABL alters the function of NK cells and the acquisition of killer immunoglobulin-like receptors (KIRs). Blood 2003; 101:3527-33. [PMID: 12511422 DOI: 10.1182/blood-2002-04-1172] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Natural killer (NK) cells decrease in function during chronic myelogenous leukemia (CML) progression from chronic phase to blast crisis, and they can become BCR/ABL(+) late in the disease course. To study this altered function, NK92 cells were transduced with the BCR/ABL oncogene. In contrast to the parental cells, which died when deprived of interleukin 2 (IL-2), p210(+) NK92 cells proliferated and survived indefinitely in the absence of IL-2. BCR/ABL also decreased the natural cytotoxicity of NK92 cells against K562 targets, without affecting IL-2, interferon gamma (IFN-gamma), or tumor necrosis factor alpha (TNF-alpha) production. Although the ABL-specific tyrosine kinase inhibitor imatinib mesylate (STI-571) had no effect on parental NK92 cells, it markedly decreased the growth and survival of IL-2-independent p210(+) NK92 cells. In contrast to the parental cell line, serial analysis of p210(+) NK92 cells detected small populations that clonally expressed one or more killer immunoglobulin-like receptors (KIRs). Unlike the decreased natural cytotoxicity, the function of the activating CD158j receptor remained intact. Southern blotting and hybridization with an enhanced green fluorescence protein (eGFP) probe showed that KIR(-) and KIR(+) NK92 cells were all derived from the same clone, suggesting that KIR acquisition remains dynamic at the maturational stage represented by the NK92 cell line. When tested in primary CD56(+bright) NK cells, p210 induced partial IL-2-independent growth and increased KIR expression similar to findings in NK92 cells. This is the first study to show that BCR/ABL, well known for its effects on the myeloid lineage, can alter the function of lymphoid cells, which may be associated with the defect in innate immunity associated with CML progression.
Collapse
MESH Headings
- Benzamides
- CD56 Antigen/analysis
- Cell Lineage
- Cells, Cultured/immunology
- Clone Cells/immunology
- Clone Cells/metabolism
- Cytotoxicity, Immunologic
- Enzyme Inhibitors/pharmacology
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/physiology
- Genes, Reporter
- Humans
- Imatinib Mesylate
- Interferon-gamma/biosynthesis
- Interleukin-2/biosynthesis
- Interleukin-2/pharmacology
- K562 Cells
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocytes/metabolism
- Piperazines/pharmacology
- Pyrimidines/pharmacology
- Receptors, Immunologic/biosynthesis
- Receptors, KIR
- Recombinant Fusion Proteins/physiology
- Transduction, Genetic
- Tumor Necrosis Factor-alpha/biosynthesis
Collapse
Affiliation(s)
- Elena G Chiorean
- Division of Hematology-Oncology and Transplantation, University of Minnesota Cancer Center, Minneapolis, 55455, USA
| | | | | | | | | | | |
Collapse
|
40
|
Watzl C, Long EO. Natural killer cell inhibitory receptors block actin cytoskeleton-dependent recruitment of 2B4 (CD244) to lipid rafts. J Exp Med 2003; 197:77-85. [PMID: 12515815 PMCID: PMC2193803 DOI: 10.1084/jem.20020427] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
A dynamic balance of positive and negative signals regulates target cell lysis by natural killer (NK) cells upon engagement of a variety of different activation receptors and of inhibitory receptors that recruit the tyrosine phosphatase SHP-1. However, the step at which activation signals are blocked by SHP-1 is not known. We have been using activation receptor 2B4 (CD244) to study the influence of inhibitory receptors on NK cell activation. Engagement of inhibitory receptors by HLA class I on target cells blocks phosphorylation of 2B4, placing the inhibitory step at the level, or upstream of 2B4 phosphorylation. Here we show that phosphorylated 2B4, after engagement with either antibodies or target cells that express the 2B4 ligand, is found exclusively in a detergent-resistant membrane fraction that contains lipid rafts. Integrity of lipid rafts was essential for phosphorylation and activating function of 2B4. Coengagement of inhibitory receptors blocked 2B4 phosphorylation and 2B4 association with detergent-resistant membranes, indicating that inhibitory receptors function upstream of raft-dependent signals. Recruitment of 2B4 into detergent-resistant membrane fractions and 2B4 phosphorylation were dependent on actin polymerization. Blocking actin cytoskeleton-dependent raft recruitment of different receptors may be a general mechanism by which inhibitory receptors control NK cell activation.
Collapse
Affiliation(s)
- Carsten Watzl
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | | |
Collapse
|
41
|
Abstract
The CD150 subfamily within the CD2 family is a growing group of dual-function receptors that have within their cytoplasmic tails a characteristic signaling motif. The ITSM (immunoreceptor tyrosine-based switch motif) enables these receptors to bind to and be regulated by small SH2 domain adaptor proteins, including SH2D1A (SH2-containing adaptor protein SH2 domain protein 1A) and EAT-2 (EWS-activated transcript 2). A major signaling pathway through the prototypic receptor in this subfamily, CD150, leads to the activation of interferon-gamma, a key cytokine for viral immunity. As a result, many viruses have designed strategies to usurp or alter CD150 functions. Measles virus uses CD150 as a receptor and Molluscum contagiosum virus encodes proteins that are homologous to CD150. Thus, viruses use CD150 subfamily receptors to create a favorable environment to elude detection and destruction. Understanding the CD150 subfamily may lead to new strategies for vaccine development and antiviral therapies.
Collapse
Affiliation(s)
- Svetlana P Sidorenko
- Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology NAS Ukraine, 45 Vasylkivska str., Kiev 03022, Ukraine
| | | |
Collapse
|
42
|
Falk CS, Mach M, Schendel DJ, Weiss EH, Hilgert I, Hahn G. NK cell activity during human cytomegalovirus infection is dominated by US2-11-mediated HLA class I down-regulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:3257-66. [PMID: 12218145 DOI: 10.4049/jimmunol.169.6.3257] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A highly attractive approach to investigate the influence and hierarchical organization of viral proteins on cellular immune responses is to employ mutant viruses carrying deletions of various virus-encoded, immune-modulating genes. Here, we introduce a novel set of deletion mutants of the human CMV (HCMV) lacking the UL40 region either alone or on the background of a deletion mutant devoid of the entire US2-11 region. Deletion of UL40 had no significant effect on lysis of infected cells by NK cells, indicating that the expected enhancement of HLA-E expression by specific peptides derived from HCMV-encoded gpUL40 leader sequences was insufficient to confer target cell protection. Moreover, the kinetics of MHC class I down-regulation by US2-11 genes observed at early and late phases postinfection with wild-type virus correlated with increased susceptibility to NK lysis. Thus, the influence of HCMV genes on NK reactivity follows a hierarchy dominated by the US2-11 region, which encodes all viral genes capable of down-modulating expression of classical and non-classical MHC class I molecules. The insights gained from studies of such virus mutants may impact on future therapeutic strategies and vaccine development and incorporate NK cells in the line of defense mechanisms against HCMV infection.
Collapse
Affiliation(s)
- Christine S Falk
- Institute of Molecular Immunology, GSF National Research Center for the Environment and Health, Munich, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Aoukaty A, Tan R. Association of the X-linked lymphoproliferative disease gene product SAP/SH2D1A with 2B4, a natural killer cell-activating molecule, is dependent on phosphoinositide 3-kinase. J Biol Chem 2002; 277:13331-7. [PMID: 11815622 DOI: 10.1074/jbc.m112029200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Natural killer (NK) cells express an activating receptor, 2B4, that enhances cellular cytotoxicity. Upon NK cell activation by ligation of 2B4, the intracellular domain of 2B4 associates with the X-linked lymphoproliferative disease (XLP) gene product, signaling lymphocytic activation molecule-associated protein/SH2D1A (SAP/SH2D1A). Defective intracellular association of 2B4 with mutated SAP/SH2D1A is likely to underlie the defects in cytotoxicity observed in NK cells from patients with XLP. We report here a role for phosphoinositide 3-kinase (PI3K) in the recruitment and association of SAP/SH2D1A to 2B4 in human NK cells. The activation of normal NK cells by ligation of 2B4 leads to the phosphorylation of 2B4, recruitment of SAP/SH2D1A, and association of the p85 regulatory subunit of PI3K. The inhibition of PI3K enzymatic activity with either wortmannin or LY294002 prior to 2B4 ligation does not alter the association of 2B4 with the p85 subunit but prevents the recruitment of SAP/SH2D1A to 2B4. In addition, PI3K inhibitors significantly diminish the cytotoxic function of primary NK cells. This observed inhibition of cytotoxicity, present in normal NK cells, was less apparent or absent in NK cells derived from a patient with XLP. These data indicate that the cytotoxicity of activated NK cells is mediated by the association of 2B4 and SAP/SH2D1A, and that this association is dependent upon the activity of PI3K.
Collapse
Affiliation(s)
- Ala Aoukaty
- Department of Pathology and Laboratory Medicine, British Columbia's Children's Hospital, University of British Columbia, 4480 Oak Street, Vancouver, BC V6H 3V4, Canada
| | | |
Collapse
|
44
|
Murphy JJ, Hobby P, Vilarino-Varela J, Bishop B, Iordanidou P, Sutton BJ, Norton JD. A novel immunoglobulin superfamily receptor (19A) related to CD2 is expressed on activated lymphocytes and promotes homotypic B-cell adhesion. Biochem J 2002; 361:431-6. [PMID: 11802771 PMCID: PMC1222324 DOI: 10.1042/0264-6021:3610431] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A novel lymphocyte-specific immunoglobulin superfamily protein (19A) has been cloned. The predicted 335-amino-acid sequence of 19A represents a Type 1 membrane protein with homology with the CD2 family of receptors. A molecular model of the two predicted extracellular immunoglobulin-like domains of 19A has been generated using the crystal structure of CD2 as a template. In isolated lymphocytes, expression of 19A is induced by various activation stimuli, and enforced expression of the 19A gene promotes homotypic cell adhesion in a B-cell-line model. Collectively these data imply that the 19A protein plays a role in regulation of lymphocyte adhesion.
Collapse
Affiliation(s)
- John J Murphy
- Infection and Immunity Research Group, Division of Life Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NN, UK.
| | | | | | | | | | | | | |
Collapse
|
45
|
Borrego F, Kabat J, Kim DK, Lieto L, Maasho K, Peña J, Solana R, Coligan JE. Structure and function of major histocompatibility complex (MHC) class I specific receptors expressed on human natural killer (NK) cells. Mol Immunol 2002; 38:637-60. [PMID: 11858820 DOI: 10.1016/s0161-5890(01)00107-9] [Citation(s) in RCA: 193] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Natural killer (NK) cells express receptors that are specific for MHC class I molecules. These receptors play a crucial role in regulating the lytic and cytokine expression capabilities of NK cells. In humans, three distinct families of genes have been defined that encode for receptors of HLA class I molecules. The first family identified consists of type I transmembrane molecules belonging to the immunoglobulin (Ig) superfamily and are called killer cell Ig-like receptors (KIR). A second group of receptors belonging to the Ig superfamily, named ILT (for immunoglobulin like transcripts), has more recently been described. ILTs are expressed mainly on B, T and myeloid cells, but some members of this group are also expressed on NK cells. They are also referred to as LIRs (for leukocyte Ig-like receptor) and MIRs (for macrophage Ig-like receptor). The ligands for the KIR and some of the ILT receptors include classical (class Ia) HLA class I molecules, as well as the nonclassical (class Ib) HLA-G molecule. The third family of HLA class I receptors are C-type lectin family members and are composed of heterodimers of CD94 covalently associated with a member of the NKG2 family of molecules. The ligand for most members is the nonclassical class I molecule HLA-E. NKG2D, a member of the NKG2 family, is expressed as a homodimer, along with the adaptor molecule DAP10. The ligands of NKG2D include the human class I like molecules MICA and MICB, and the recently described ULBPs. Each of these three families of receptors has individual members that can recognize identical or similar ligands yet signal for activation or inhibition of cellular functions. This dichotomy correlates with particular structural features present in the transmembrane and intracytoplasmic portions of these molecules. In this review we will discuss the molecular structure, specificity, cellular expression patterns, and function of these HLA class I receptors, as well as the chromosomal location and genetic organization.
Collapse
Affiliation(s)
- Francisco Borrego
- Receptor Cell Biology Section, Laboratory of Allergic Diseases, NIAID, NIH, Twinbrook II, Room 205, 12441 Parklawn Dr., Rockville, MD 20852, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Falk CS, Schendel DJ. Allogeneic MHC class I ligands and their role in positive and negative regulation of human cytotoxic effector cells. Hum Immunol 2002; 63:8-19. [PMID: 11916173 DOI: 10.1016/s0198-8859(01)00360-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The allogeneic mixed lymphocyte culture (MLC) has served as an important experimental system for elucidating the cellular and molecular basis of human lymphocyte responses. Complex mixtures of lymphocytes are stimulated by disparate alloantigens, inducing cellular activation and generating a cytokine milieu that is an excellent breeding ground for the proliferation and differentiation of many distinct lymphocyte subsets. Cloning of individual lymphocytes following alloactivation has allowed various cytotoxic lymphocytes to be isolated and characterized with respect to phenotype and specificity. These analyses have revealed that all types of cytotoxic effector cells are regulated by interactions with MHC-peptide ligands, however, the consequences of these interactions can result in opposite functional outcomes. In this review we summarize how allogeneic MHC class I-peptide ligands positively or negatively regulate the activities of four distinct groups of cytotoxic lymphocytes and how this information might be transferred into clinical use.
Collapse
Affiliation(s)
- Christine S Falk
- Institute of Molecular Immunology, GSF National Research Center for the Environment and Health, Munich, Germany.
| | | |
Collapse
|
47
|
Chuang SS, Kumaresan PR, Mathew PA. 2B4 (CD244)-mediated activation of cytotoxicity and IFN-gamma release in human NK cells involves distinct pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6210-6. [PMID: 11714782 DOI: 10.4049/jimmunol.167.11.6210] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
2B4 (CD244), a member of the CD2 subset of the Ig superfamily receptors, is expressed on all human NK cells, a subpopulation of T cells, basophils and monocytes. 2B4 activates NK cell mediated cytotoxicity, induces secretion of IFN-gamma and matrix metalloproteinases, and NK cell invasiveness. Although there have been several molecules shown to interact with 2B4, the signaling mechanism of 2B4-mediated activation of NK cells is still unknown. In this study, we found cross-linking of 2B4 on YT cells, a human NK cell line, results in the increased DNA binding activity of activator protein-1 (AP-1), an important regulator of nuclear gene expression in leukocytes. We investigated the possible role of various signaling molecules that may be involved in the activation of lytic function of YT cells via 2B4. Treatment of YT cells with various specific inhibitors indicate that 2B4-stimulation of YT cells in spontaneous and Ab-dependent cytotoxicity is Ras/Raf dependent and involves multiple MAPK signaling pathways (ERK1/2 and p38). However, only inhibitors of transcription and p38 inhibited 2B4-mediated IFN-gamma release indicating distinct pathways are involved in cytotoxicity and cytokine release. In this study we also show that 2B4 constitutively associates with the linker for activation of T cells (LAT) and that 2B4 may mediate NK cell activation via a LAT-dependent signaling pathway. These results indicate that 2B4-mediated activation of NK cells involves complex interactions involving LAT, Ras, Raf, ERK and p38 and that cytolytic function and cytokine production may be regulated by distinct pathways.
Collapse
Affiliation(s)
- S S Chuang
- Department of Molecular Biology and Immunology and Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | | | | |
Collapse
|
48
|
Forte P, Pazmany L, Matter-Reissmann UB, Stussi G, Schneider MK, Seebach JD. HLA-G inhibits rolling adhesion of activated human NK cells on porcine endothelial cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6002-8. [PMID: 11698480 DOI: 10.4049/jimmunol.167.10.6002] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human NK cells adhere to and lyse porcine endothelial cells (pEC) and therefore may contribute to the cell-mediated rejection of vascularized pig-to-human xenografts. Since MHC class I molecules inhibit the cytotoxic activity of NK cells, the expression of HLA genes in pEC has been proposed as a potential solution to overcome NK cell-mediated xenogeneic cytotoxicity. HLA-G, a minimally polymorphic HLA class I molecule that can inhibit a wide range of NK cells, is an especially attractive candidate for this purpose. In this study we tested whether the expression of HLA-G on pEC inhibits the molecular mechanisms that lead to adhesion of human NK cells to pEC and subsequent xenogeneic NK cytotoxicity. To this end two immortalized pEC lines (2A2 and PED) were stably transfected with HLA-G1. Rolling adhesion of activated human NK cells to pEC monolayers and xenogeneic cytotoxicity against pEC mediated by polyclonal human NK lines as well as NK clones were inhibited by the expression of HLA-G. The adhesion was partially reversed by masking HLA-G on pEC with anti-HLA mAbs or by masking the HLA-G-specific inhibitory receptor ILT-2 on NK cells with the mAb HP-F1. The inhibition of NK cytotoxicity by HLA-G was only partially mediated by ILT-2, indicating a role for other unknown NK receptors. In conclusion, transgenic expression of HLA-G may be useful to prevent human NK cell responses to porcine xenografts, but is probably not sufficient on its own. Moreover, the blocking of rolling adhesion by HLA-G provides evidence for a novel biological function of HLA molecules.
Collapse
Affiliation(s)
- P Forte
- Department of Internal Medicine, Laboratory for Transplantation Immunology, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
49
|
Morra M, Lu J, Poy F, Martin M, Sayos J, Calpe S, Gullo C, Howie D, Rietdijk S, Thompson A, Coyle AJ, Denny C, Yaffe MB, Engel P, Eck MJ, Terhorst C. Structural basis for the interaction of the free SH2 domain EAT-2 with SLAM receptors in hematopoietic cells. EMBO J 2001; 20:5840-52. [PMID: 11689425 PMCID: PMC125701 DOI: 10.1093/emboj/20.21.5840] [Citation(s) in RCA: 115] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The T and natural killer (NK) cell-specific gene SAP (SH2D1A) encodes a 'free SH2 domain' that binds a specific tyrosine motif in the cytoplasmic tail of SLAM (CD150) and related cell surface proteins. Mutations in SH2D1A cause the X-linked lymphoproliferative disease, a primary immunodeficiency. Here we report that a second gene encoding a free SH2 domain, EAT-2, is expressed in macrophages and B lympho cytes. The EAT-2 structure in complex with a phosphotyrosine peptide containing a sequence motif with Tyr281 of the cytoplasmic tail of CD150 is very similar to the structure of SH2D1A complexed with the same peptide. This explains the high affinity of EAT-2 for the pTyr motif in the cytoplasmic tail of CD150 but, unlike SH2D1A, EAT-2 does not bind to non-phosphorylated CD150. EAT-2 binds to the phosphorylated receptors CD84, CD150, CD229 and CD244, and acts as a natural inhibitor, which interferes with the recruitment of the tyrosine phosphatase SHP-2. We conclude that EAT-2 plays a role in controlling signal transduction through at least four receptors expressed on the surface of professional antigen-presenting cells.
Collapse
Affiliation(s)
- Massimo Morra
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | - Jun Lu
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | - Florence Poy
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | - Margarita Martin
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | | | | | | | | | | | - Andrew Thompson
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | - Anthony J. Coyle
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | - Christopher Denny
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | - Michael B. Yaffe
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | - Pablo Engel
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | - Michael J. Eck
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School and
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, Molecular Biology Institute, University of California, Los Angeles, CA 90095, Millennium Pharmaceuticals Inc., Inflammation Division, Cambridge, MA 02139, Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Cellular Biology and Pathology, Faculty of Medicine, University of Barcelona, Spain Corresponding authors e-mail: or
| |
Collapse
|
50
|
Abstract
OBJECTIVE NK cells are important cells of the immune system. They are ultimately derived from pluripotent hematopoietic stem cells. NK cell cytotoxicity and other functions are tightly regulated by numerous activating and inhibitory receptors including newly discovered receptors that selectively recognize major histocompatibility complex class I alleles. Based on their defining function of spontaneous cytotoxicity without prior immunization, NK cells have been thought to play a critical role in immune surveillance and cancer therapy. However, new insights into NK cell biology have suggested major roles for NK cells in infection control and uterine function. The purpose of this review is to provide an update on NK cell function, ontogeny, and biology in order to better understand the role of NK cells in health and disease. DATA SOURCES In the Medline database, the major subject heading "Natural Killer Cells" was introduced in 1983, identifying 16,848 citations as of December 31, 2000. Since 1986, there have been approximately 1000 citations per year under this subject heading. In this database, 68% of manuscripts are limited to human NK cells; 40% of citations cross with the major sub-heading of cytotoxicity, 40% with cytokines, 36% with neoplasm, 5% with antibody-dependent cellular cytotoxicity, 2.8% with pregnancy, and 1.3% with infection. Of references from the year 2000-2001, 46 were selected to combine with contributions from earlier literature. CONCLUSIONS NK cells should no longer be thought of as direct cytotoxic killers alone as they clearly serve a critical role in cytokine production which may be important to control cancer, infection, and fetal implantation. Understanding mechanisms of NK cell functions may lead to novel therapeutic strategies for the treatment of human disease.
Collapse
Affiliation(s)
- J S Miller
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota Cancer Center, Minneapolis, Minn. 55455, USA.
| |
Collapse
|