Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (109)

Search Parameters:
Keywords = NKG2D receptor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 2588 KiB  
Article
Immunophenotyping and Functional Characterization of NK Cells in SARS-CoV-2 Infection
by Steliyan Petrov, Martina Bozhkova, Mariya Ivanovska, Teodora Kalfova, Alexandra Baldzhieva, Angel Todev, Dilyana Kirova, Yoana Kicheva, Stoyno Stoynov, Marianna Murdjeva and Hristo Taskov
Immuno 2025, 5(3), 35; https://doi.org/10.3390/immuno5030035 - 15 Aug 2025
Abstract
The immune response to SARS-CoV-2 infection involves significant alterations in the phenotype and function of natural killer (NK) cells. This study aimed to investigate the dynamic changes in NK cell subsets during COVID-19 by analyzing their activation and inhibitory markers [CD3, CD14, CD16, [...] Read more.
The immune response to SARS-CoV-2 infection involves significant alterations in the phenotype and function of natural killer (NK) cells. This study aimed to investigate the dynamic changes in NK cell subsets during COVID-19 by analyzing their activation and inhibitory markers [CD3, CD14, CD16, CD19, CD25, CD45, CD56, CD57, CD69, CD159a (NKG2A), CD159c (NKG2C), CD314 (NKG2D), CD335 (NKp46)], cytotoxic potential (perforin, interferon-gamma, granzyme B), and direct cytotoxicity against a newly genetically modified K562 cell line. Peripheral blood samples were collected from COVID-19 patients on days 3–5 and day 30 post-symptom onset and were compared to healthy controls. 16-color flow cytometry analysis revealed distinct shifts in NK cell subpopulations, characterized by increased expression of the inhibitory receptor NKG2A and the activating receptors NKG2D and NKG2C, particularly in the CD56+CD16 subset. Elevated IFN-γ production on day 30 suggested a recovery-phase immune response, while the persistent upregulation of NKG2A indicated an ongoing regulatory mechanism. The CD16+CD56 subpopulation exhibited increased expression of the markers CD69 and CD25 over time; however, its cytotoxic potential, assessed through granzyme B levels and direct cytotoxicity assays, remained lower than that of healthy controls. Significant correlations were observed between CD57 and CD69 expression, as well as NKp46 and IFN-γ production, highlighting a coordinated balance between activation and regulatory mechanisms. These findings suggest that NK cells undergo functional adaptation during COVID-19, displaying signs of partial exhaustion while retaining antiviral potential. Understanding the interplay between NK cell activation and suppression may provide valuable insights into immune dysregulation in COVID-19 and inform potential therapeutic interventions. Full article
(This article belongs to the Section Innate Immunity and Inflammation)
29 pages, 3651 KiB  
Article
Donor Variability and Seeding Density Shape NK-Cell Proliferation and Surface Receptor Expression: Insights from an Integrated Phenotypic and Genetic Analysis
by Neele Kusch, Jonathan Storm, Antonia Macioszek, Cornelius Knabbe, Barbara Kaltschmidt and Christian Kaltschmidt
Cells 2025, 14(16), 1252; https://doi.org/10.3390/cells14161252 - 14 Aug 2025
Viewed by 26
Abstract
Natural killer (NK) cells are promising candidates for adoptive immunotherapy, but their clinical application requires standardized expansion protocols that yield functional cells in sufficient numbers. This study examined how initial seeding density and donor-intrinsic variability affect NK cell proliferation and receptor phenotype during [...] Read more.
Natural killer (NK) cells are promising candidates for adoptive immunotherapy, but their clinical application requires standardized expansion protocols that yield functional cells in sufficient numbers. This study examined how initial seeding density and donor-intrinsic variability affect NK cell proliferation and receptor phenotype during in vitro expansion in a G-Rex® 24-well plate under IL-2 stimulation. NK cells from healthy donors were analyzed longitudinally by flow cytometry, and targeted SNP sequencing of selected receptor genes (IL2RA, IL2RB, FCGR3A, NCR1, KLRK1, and ICAM-1) was performed to assess potential genetic contributions. A seeding density of 2.0 × 106 cells/cm2 promoted high expansion rates and favorable expression of activating receptors including CD16a, NKp46, and NKG2D. Nonetheless, marked inter-donor differences were observed. Some donors exhibited impaired proliferation and aberrant receptor expression, possibly associated with high-priority SNPs and distinct haplotype structures. Others showed robust proliferation despite the absence of identifiable genetic drivers, suggesting the involvement of variants in other genes or non-genetic mechanisms such as epigenetic priming or adaptive NK-cell differentiation. These results highlight the influence of both culture conditions and donor-intrinsic factors on NK-cell expansion outcomes. Integrating phenotypic and genetic analyses may improve the reproducibility and personalization of NK-cell-based manufacturing protocols for therapeutic use. Full article
(This article belongs to the Special Issue Natural Killer (NK) Cells in Immunity: Limitations and Potential)
Show Figures

Figure 1

28 pages, 939 KiB  
Review
Targets for CAR Therapy in Multiple Myeloma
by Olga A. Bezborodova, Galina V. Trunova, Elena R. Nemtsova, Varvara A. Khokhlova, Julia B. Venediktova, Natalia B. Morozova, Maria S. Vorontsova, Anna D. Plyutinskaya, Elena P. Zharova, Peter V. Shegai and Andrey D. Kaprin
Int. J. Mol. Sci. 2025, 26(13), 6051; https://doi.org/10.3390/ijms26136051 - 24 Jun 2025
Viewed by 824
Abstract
Multiple myeloma (MM or plasma cell myeloma) is a heterogenous B-cell malignant tumor that typically exhibits a high recurrence rate, resistance to drugs, and molecular diversity of tumor subclones. Given the limited efficacy of standard therapy options, cellular immunotherapy featuring a chimeric antigen [...] Read more.
Multiple myeloma (MM or plasma cell myeloma) is a heterogenous B-cell malignant tumor that typically exhibits a high recurrence rate, resistance to drugs, and molecular diversity of tumor subclones. Given the limited efficacy of standard therapy options, cellular immunotherapy featuring a chimeric antigen receptor (CAR) has proven tangible potential in treatment for relapsed and refractory forms of MM. The rational choice of a tumor target which shows high selectivity, stable expression, and biological significance is key to the successful implementation of CAR therapy. This review has summarized and analyzed data from the literature on biological properties, the features of expression, and the clinical development stages of CAR cell products for MM treatment which target BCMA, GPRC5D, FcRH5, SLAMF7, CD38, CD138, TACI, APRIL, CD19, TNFR2, CD44v6, CD70, NKG2D ligands, etc. Special focus is on strategic approaches to overcoming antigenic escape, such as multi-specific CAR constructs, logical activation sequences, and controlled safety systems. The analysis underscores the need for integrating the molecular selection of targets with cutting-edge bioengineering solutions as a key trend for raising the efficacy, stability, and safety of cellular therapy in the case of MM. Full article
(This article belongs to the Special Issue Advancements in Cancer Biomarkers)
Show Figures

Graphical abstract

20 pages, 2917 KiB  
Review
HIF-1α: A Key Factor Mediating Tumor Cells from Digestive System to Evade NK Cell Killing via Activating Metalloproteinases to Hydrolyze MICA/B
by Quan Zhu, Shuyi Tang, Ting Huang, Chunjing Chen, Biyuan Liu, Chuyu Xiao, Liugu Chen, Wang Wang and Fangguo Lu
Biomolecules 2025, 15(6), 899; https://doi.org/10.3390/biom15060899 - 19 Jun 2025
Viewed by 713
Abstract
Malignant tumors of the digestive system are widespread and pose a serious threat to humans. Immune escape is an important factor promoting the deterioration of malignant tumors in the digestive system. Natural killer cells (NK cells) are key members of the anti-tumor and [...] Read more.
Malignant tumors of the digestive system are widespread and pose a serious threat to humans. Immune escape is an important factor promoting the deterioration of malignant tumors in the digestive system. Natural killer cells (NK cells) are key members of the anti-tumor and immune surveillance system, mainly exerting cytotoxic effects by binding to the activating receptor natural killer cell group 2D (NKG2D) on their cell surface with the corresponding ligands (major histocompatibility complex class I chain-related protein A/B, MICA/B) on the surface of tumor cells. Malignant tumors of epithelial origin usually highly express NKG2D ligands such as MICA, which can attract NK cells to kill tumor cells and also serve as an important basis for NK cell-based immunotherapy. Tumor cells highly express hypoxia-inducible factor-1α (HIF-1α), which promotes the expression of matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinases (ADAMs). These metalloproteinases hydrolyze MICA and other ligands on the surface of tumor cells to generate soluble molecules. These soluble ligands, when binding to NKG2D, cannot activate NK cells and also block the binding of NKG2D to MICA on the surface of tumor cells, enabling tumor cells to evade the killing effect of NK cells. Almost all organs in the digestive system originate from epithelial tissue, so the soluble ligands generated by the HIF-1α/MMPs or HIF-1α/ADAMs signaling pathways play a crucial role in evading NK cell killing. A comprehensive understanding of this immune escape process is helpful for a deeper understanding of the molecular mechanism of NK cell anti-tumor activity. This article reviews the molecular mechanisms of common digestive system malignancies evading NK cell killing, providing new insights into the mechanism of tumor immune escape. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

24 pages, 18981 KiB  
Article
Ectopic ULBP2 Is Associated with Decreased NKG2D Expression in CD8+ T Cells Under T Cell-Modulatory Conditions in a Murine Tumor Model
by Yasuhiko Teruya, Kosuke Yamaguchi, Kohei Yamane, Naomi Miyake, Yuji Nakayama, Takafumi Nonaka, Hiroki Chikumi and Akira Yamasaki
Cells 2025, 14(12), 893; https://doi.org/10.3390/cells14120893 - 13 Jun 2025
Viewed by 651
Abstract
UL16-binding protein 2 (ULBP2), a ligand for the activating receptor NKG2D, plays a dual role in tumor immunity, promoting immune activation or suppression, depending on the context. To investigate its impact on CD4+CD25+ T cell-targeted immunotherapies, we used a syngeneic [...] Read more.
UL16-binding protein 2 (ULBP2), a ligand for the activating receptor NKG2D, plays a dual role in tumor immunity, promoting immune activation or suppression, depending on the context. To investigate its impact on CD4+CD25+ T cell-targeted immunotherapies, we used a syngeneic CT26 colon cancer model engineered to express ULBP2 and compared tumor growth and tumor-infiltrating lymphocyte (TIL) profiles in control and ULBP2-expressing tumors treated with anti-CD4, anti-CD25, or anti-CTLA-4 antibodies. Tumor growth was uniformly assessed on day 21 post-transplantation, and TIL analysis was performed in groups with evaluable residual tumors. Anti-CD4 antibody significantly suppressed tumor growth in mock-transfected tumors, while no significant suppression was observed in ULBP2-expressing tumors. Anti-CD25 antibody had limited efficacy in mock tumors and tended to promote tumor growth in ULBP2-expressing tumors. Following these treatments, ULBP2 expression was associated with reduced NKG2D expression in CD8+ effector memory T cells, particularly PD-1high subsets. In contrast, anti-CTLA-4 antibody treatment induced marked tumor regression irrespective of ULBP2 expression. These findings suggest that ULBP2–NKG2D signaling may contribute to altered CD8+ T cell phenotypes under T cell-modulatory conditions, potentially impacting the outcome of CD4+CD25+ T cell-targeted therapies and providing insights for optimizing immunotherapeutic strategies. Full article
(This article belongs to the Section Cellular Immunology)
Show Figures

Figure 1

16 pages, 4733 KiB  
Article
Inhibition of MCP1 (CCL2) Enhances Antitumor Activity of NK Cells Against HCC Cells Under Hypoxia
by Hwan Hee Lee, Juhui Kim, Eunbi Park, Hyojeung Kang and Hyosun Cho
Int. J. Mol. Sci. 2025, 26(10), 4900; https://doi.org/10.3390/ijms26104900 - 20 May 2025
Cited by 1 | Viewed by 656
Abstract
Hypoxia, a low-oxygen state, is a common feature of solid tumors. MCP1 (CCL2) is a small cytokine that is closely related to hypoxia and has a positive effect on tumor development. Hypoxia causes resistance to various treatments for solid tumors and the evasion [...] Read more.
Hypoxia, a low-oxygen state, is a common feature of solid tumors. MCP1 (CCL2) is a small cytokine that is closely related to hypoxia and has a positive effect on tumor development. Hypoxia causes resistance to various treatments for solid tumors and the evasion of cancer immune surveillance by lymphocytes. Natural killer (NK) cells are innate lymphocytes that play an important role in cancer development, particularly in the liver. First, it was found that the incubation of HCC in hypoxia (2–5% O2) significantly increased the production of several inflammatory cytokines, including MCP1, compared to that of normal oxygen (20% O2). Subsequently, blocking MCP1 with an anti-MCP1 antibody in HCC cultures inhibited the growth and migration of HCC cells in vitro and in vivo. This was associated with a decrease in the expression of HIF-1α/STAT3 in HCC under hypoxia. Furthermore, blocking MCP1 in HCC cell cultures under hypoxia significantly increased the chemotaxis and activation of NK-92 cells against HCC cells. MCP1 blockade in HCC cell cultures under hypoxia induced a shift in NK cells to the CD56+dim population and an increase in the expression of the activation receptors NKG2D and NKp44. In conclusion, modulation of MCP1 could enhance NK activity against hypoxic HCC cells. Full article
(This article belongs to the Special Issue Molecular Biology of Hypoxia)
Show Figures

Figure 1

24 pages, 1536 KiB  
Review
A Concise Review of the Role of the NKG2D Receptor and Its Ligands in Cancer
by Elitsa Boneva, Velizar Shivarov and Milena Ivanova
Immuno 2025, 5(1), 9; https://doi.org/10.3390/immuno5010009 - 2 Mar 2025
Cited by 3 | Viewed by 2899
Abstract
The immune system’s ability to detect and eliminate transformed cells is a critical factor in suppressing cancer development. However, immune surveillance in tumors is often disrupted by various immune escape mechanisms, many of which remain poorly understood. The Natural Killer Group 2D (NKG2D) [...] Read more.
The immune system’s ability to detect and eliminate transformed cells is a critical factor in suppressing cancer development. However, immune surveillance in tumors is often disrupted by various immune escape mechanisms, many of which remain poorly understood. The Natural Killer Group 2D (NKG2D) receptor is an activating receptor expressed on natural killer (NK) cells and cytotoxic T lymphocytes. It can recognize and bind with varying affinities to a wide range of structurally diverse ligands, including MHC class I chain-related proteins A and B (MICA and MICB) and members of the ULBP family (ULBP1-6). The expression of these ligands plays a crucial role in immune antitumor responses and cancer immunoevasion mechanisms. Some evidence suggests that functional polymorphisms in the NKG2D receptor and the genes encoding its ligands significantly influence HLA-independent cancer immunosurveillance. Consequently, the NKG2D-NKG2D ligands (NKG2DLs) axis represents a promising target for developing novel therapeutic strategies. This review aims to provide a general overview of the role of NKG2D and its ligands in various malignancies and explore their potential in advancing personalized cancer treatment protocols. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

19 pages, 7047 KiB  
Article
Overexpression of NKG2D and IL24 in NK Cell-Derived Exosomes for Cancer Therapy
by Chujun Huang, Qian Hu, Peiyun Wang, Mi Xie, Ying Zhang, Zhixing Li, Shuqing Tang, Yuxuan Zhang, Zhixin Tian, Xionghao Liu, Zhiqing Hu and Desheng Liang
Int. J. Mol. Sci. 2025, 26(5), 2098; https://doi.org/10.3390/ijms26052098 - 27 Feb 2025
Cited by 1 | Viewed by 944
Abstract
Natural killer (NK) cell-derived exosomes (NK-Exos) are emerging as a promising avenue in cancer immunotherapy due to their inherent tumor-targeting properties and their capacity to deliver therapeutic agents directly to malignant cells. This research delves into the boosted anti-tumor potency of NK-Exos that [...] Read more.
Natural killer (NK) cell-derived exosomes (NK-Exos) are emerging as a promising avenue in cancer immunotherapy due to their inherent tumor-targeting properties and their capacity to deliver therapeutic agents directly to malignant cells. This research delves into the boosted anti-tumor potency of NK-Exos that has been genetically enhanced to overexpress NKG2D, a vital activating receptor, along with interleukin-24 (IL24), a cytokine renowned for its selective suppressive impact on tumor cells. NKG2D facilitates the recognition of tumor cells by binding to stress-induced ligands, while IL24 induces apoptosis and modulates immune responses to enhance tumor destruction. The NK-Exos engineered to express both NKG2D and IL24 significantly enhanced tumor targeting and increased the apoptosis rate of tumor cells by 30% in A549 and by 20% in HELA at 48 h compared with non-modified NK-Exos, respectively. Furthermore, this enhancement also impacted cell proliferation, with inhibition rates increasing by 30%, 15%, and 15% in A549, HELA, and MCF-7 cells, respectively, and it reduced A549 cell migration by 10%. The integration of NKG2D and IL24 within NK-Exos confers a dual therapeutic mechanism, synergistically amplifying their efficacy in cancer treatment. The utility of NK-Exos co-expressing NKG2D and IL24 offers a novel approach to overcome the limitations of current therapies, providing prolonged tumor suppression and precise targeting of malignant cells and holding great promise for clinical application. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

18 pages, 1392 KiB  
Review
Enhancing CAR T-Cell Function with Domains of Innate Immunity Sensors
by Tjaša Mlakar, Mojca Skrbinek, Tina Fink and Duško Lainšček
Int. J. Mol. Sci. 2025, 26(3), 1339; https://doi.org/10.3390/ijms26031339 - 5 Feb 2025
Viewed by 2098
Abstract
The innate immune system plays an important role in protecting the organism via recognizing the danger signals and pathogens through pattern recognition receptors. By sensing the danger signal and conveying the signaling towards the elimination of the threat, several families of these receptors, [...] Read more.
The innate immune system plays an important role in protecting the organism via recognizing the danger signals and pathogens through pattern recognition receptors. By sensing the danger signal and conveying the signaling towards the elimination of the threat, several families of these receptors, expressed on different myeloid and innate lymphoid cells, serve as the first defense line in the innate immunity. Toll-like receptors, C-type lectin receptors, and many other receptors therefore illustrate the importance of the protective role of the immune system. This was additionally confirmed by CAR T-cell-based cancer immunotherapy, where the patient’s own immune system is being used for successful tumor elimination. CAR T-cells have proven themselves to be a potent therapeutic option, yet in some cases their efficiency could be enhanced. Innate immune sensors that include strong activation and signaling domains, for instance, part of the Toll-like receptors, MyD88 (Myeloid Differentiation Primary Response gene), NKG2D (Natural killer group 2-member D), and many other domains, could be used as a CAR building module to increase the functionality and potency of the CAR T-cells. Full article
(This article belongs to the Special Issue Advanced Research on CAR-T Cell Therapy)
Show Figures

Figure 1

19 pages, 3097 KiB  
Article
Natural Killer Cells in Graves’ Disease: Increased Frequency but Impaired Degranulation Ability Compared to Healthy Controls
by Daniela Gallo, Eliana Piantanida, Raffaella Bombelli, Silvia Lepanto, Antonino Bruno, Matteo Gallazzi, Giorgia Bilato, Marina Borgese, Denisa Baci, Lorenzo Mortara and Maria Laura Tanda
Int. J. Mol. Sci. 2025, 26(3), 977; https://doi.org/10.3390/ijms26030977 - 24 Jan 2025
Viewed by 1386
Abstract
Graves’ disease (GD) is an autoimmune disorder, driven by the appearance of circulating autoantibodies (Ab) against the thyroid stimulating hormone (TSH) receptor, thus causing hyperthyroidism. While antithyroid drugs, the only available treatment for GD, carry a significant risk of relapse, advances in immunology [...] Read more.
Graves’ disease (GD) is an autoimmune disorder, driven by the appearance of circulating autoantibodies (Ab) against the thyroid stimulating hormone (TSH) receptor, thus causing hyperthyroidism. While antithyroid drugs, the only available treatment for GD, carry a significant risk of relapse, advances in immunology could pave the way for more effective therapies. Natural killer (NK) cells, divided into cytotoxic CD56dim and cytokine-secreting CD56bright subsets, regulate immune responses through cytokine production and cell lysis and may play a role in the pathogenesis of GD. To investigate their involvement, we conducted flow cytometry on peripheral blood samples from 131 GD patients at various stages (disease onset, on antithyroid drugs, and in remission) and 97 age- and sex-matched healthy controls (HC). We analyzed NK cell subsets, activating (CD16, CD69, NKG2D, NKp30) and inhibitory receptors (CD161, NKG2A), degranulation (CD107a), and intracellular cytokines expression (interferon γ, tumor necrosis factor α). Statistical comparisons were made between GD patients and HC and across disease stages. GD patients had a higher frequency of total NK cells (p < 0.028) and CD56bright NK cells (p < 0.01) but a lower frequency of CD56dim NK cells (p = 0.005) compared to HC. NK cells in GD patients expressed activating receptors more frequently, except for NKG2D, but had decreased cytokine expression and degranulation ability. At GD onset, patients had higher frequencies of total NK cells, CD56bright NK cells, and NK cells expressing activating receptors compared to patients receiving ATD treatment and those in remission. CD161+ NK cells were lower at GD onset and returned to levels of HC following treatment. Correlation analysis revealed that free thyroxine (FT4) levels were inversely correlated with CD107a+ NK cells (p < 0.05) and positively correlated with CD69+ NK cells (p < 0.01). These findings suggest that hyperthyroidism impairs NK cell degranulation, with the increased frequency of NK cells potentially compensating for their reduced function. This dysfunction may contribute to the unregulated immune response in GD, highlighting NK cells as a potential target for novel therapeutic strategies. Full article
(This article belongs to the Collection Feature Papers in Molecular Immunology)
Show Figures

Figure 1

24 pages, 2895 KiB  
Article
A New Antibody–Cytokine Construct Targeting Natural Killer Cells: An Immunotherapeutic Approach to Chronic Lymphocytic Leukemia
by Michela Flego, Mauro Andreotti, Francesca Romana Mauro, Maria Beatrice Arasi, Silvia Zamboni, Zuleika Michelini, Sara Pepe, Clementina Maria Galluzzo, Roberta Amici, Diego Moricoli, Chiara Mazzei, Alessandro Ascione and Alessandra Mallano
Biomolecules 2025, 15(1), 117; https://doi.org/10.3390/biom15010117 - 13 Jan 2025
Cited by 1 | Viewed by 1109
Abstract
In chronic lymphocytic leukemia (CLL), natural killer (NK) cells show a dysfunctional phenotype that correlates with disease progression. Our aim was to restore NK cell functionality in CLL through a specifically targeted IL15-stimulating activity; IL15 targeting could, in fact, potentiate the activity of [...] Read more.
In chronic lymphocytic leukemia (CLL), natural killer (NK) cells show a dysfunctional phenotype that correlates with disease progression. Our aim was to restore NK cell functionality in CLL through a specifically targeted IL15-stimulating activity; IL15 targeting could, in fact, potentiate the activity of NK cells and reduce off-target effects. We designed and developed a cis-acting immunocytokine composed of an anti-CD56 single-chain Fragment variable (scFv) and IL15, labeled scFvB1IL15. scFvB1IL15 was tested in vitro on peripheral blood mononuclear cells (PBMCs) obtained from both different healthy donors (HDs) and CLL patients in order to evaluate its ability to target NK cells and enhance their activation and NK-mediated directed cytotoxicity. scFvB1IL15 specifically induced strong degranulation and cytokine and chemokine production in NK cells in both HD- and CLL patient-derived PBMC samples. Furthermore, compared to IL15 alone, it was able to induce higher levels of NKG2D- and NKp30-activating receptors and restore NK-mediated direct killing in the CLL patient-derived samples. The preliminary data presented in this work suggest that IL15’s targeting of NK cells via scFvB1 potentiates the effects of IL15 and that scFvB1IL15 can be a useful agent for overcoming NK functional gaps and contribute to NK-cell-based immunotherapies. Full article
(This article belongs to the Special Issue Cytokine Signaling in Immunity and Immune Cell-Mediated Diseases)
Show Figures

Graphical abstract

16 pages, 8657 KiB  
Article
Characterization of the Bone Marrow Lymphoid Microenvironment and Discovery of Prognostic Immune-Related Factors in Acute Myeloid Leukemia
by Yoon-Ju Kim, Daehun Kwag, Bo-Reum Kim, Hyunsong Son, Silvia Park, Hee-Je Kim and Byung-Sik Cho
Int. J. Mol. Sci. 2024, 25(23), 13039; https://doi.org/10.3390/ijms252313039 - 4 Dec 2024
Cited by 1 | Viewed by 1344
Abstract
Given the limited comprehensive data on the bone marrow (BM) immune environment in acute myeloid leukemia (AML), we analyzed the distribution and phenotype of T cell subsets, including γδ T cells, and their immune checkpoint (IC) ligands on blasts. We performed multiparametric flow [...] Read more.
Given the limited comprehensive data on the bone marrow (BM) immune environment in acute myeloid leukemia (AML), we analyzed the distribution and phenotype of T cell subsets, including γδ T cells, and their immune checkpoint (IC) ligands on blasts. We performed multiparametric flow cytometry with BM samples taken from 89 AML patients at the time of diagnosis, remission, and relapse/refractory status after chemotherapy and 13 healthy controls (HCs) to identify immune-related risk factors. Compared to the HCs, the T cells of the AML patients exhibited exhausted features including higher TIGIT levels and similar levels of PD-1 and TIM-3. The γδ T cells were exhausted by the upregulation of TIGIT and/or TIM-3 and downregulation of NKG2D and NKp30, with different patterns in the Vδ1 and Vδ2 subtypes. A successful chemotherapeutic response partially restored the exhausted phenotypes of the T cell subsets. The simultaneous analysis of IC receptors on the T cell subsets and their ligands on blasts showed the prognostic value of a specific IC receptor–ligand pair and the feasibility of risk stratification based on their diverse patterns. Our findings clarified the BM T cell landscape in AML, unveiling the prognostic value of γδ T cells in both diagnosis and remission predictions. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Graphical abstract

21 pages, 2792 KiB  
Article
Three-Dimensional Model Analysis Revealed Differential Cytotoxic Effects of the NK-92 Cell Line and Primary NK Cells on Breast and Ovarian Carcinoma Cell Lines Mediated by Variations in Receptor–Ligand Interactions and Soluble Factor Profiles
by Nadezhda A. Alekseeva, Anna A. Boyko, Marina A. Shevchenko, Maria V. Grechikhina, Maria A. Streltsova, Ludmila G. Alekseeva, Alexander M. Sapozhnikov, Sergey M. Deyev and Elena I. Kovalenko
Biomedicines 2024, 12(10), 2398; https://doi.org/10.3390/biomedicines12102398 - 20 Oct 2024
Cited by 2 | Viewed by 2082
Abstract
Background/objectives: The functional activity of a certain tumor determines the effectiveness of primary NK cells and NK-92 cell line-based cancer therapy; their therapeutic effectiveness against different tumors can vary. This work provides a direct simultaneous comparison of the cytotoxic effects of in vitro-activated [...] Read more.
Background/objectives: The functional activity of a certain tumor determines the effectiveness of primary NK cells and NK-92 cell line-based cancer therapy; their therapeutic effectiveness against different tumors can vary. This work provides a direct simultaneous comparison of the cytotoxic effects of in vitro-activated peripheral NK (pNK) cells and NK-92 cells in spheroid models of BT-474, MCF7 and SKOV-3 carcinomas and uncovers the reasons for the differential effectiveness of NK cells against tumors. Methods: Tumor spheroids of similar size and shape, obtained from agarose molds, were incubated with NK-92 or pNK cells for 24 h. Tumor cell death was detected using flow cytometry or confocal microscopy. Cytokine production, granzyme B levels and NK cell degranulation analyses were performed, along with pNK and target-cell phenotypic characterization. Results: While NK-92 and pNK cells lysed BT-474 spheroids with comparably low efficiency, pNK cells were more capable of eliminating MCF7 and SKOV-3 spheroids than NK-92 cells were. The results of the functional and phenotypic analyses strongly support the participation of the NKG2D-NKG2DL pathway in pNK cell activation induced by the most sensitive cytotoxic attack on SKOV-3 spheroids, whereas the CX3CR1-CX3CL1 axis appears to be involved in the pNK reaction against MCF-7 spheroids. Conclusions: We provide a new approach for the preliminary identification of the most promising NK cell receptors that can alter the effectiveness of cancer therapy depending on the specific tumor type. Using this approach, NK-92 cells or pNK subsets can be selected for further accumulation and/or genetic modification to improve specificity and reactivity. Full article
(This article belongs to the Special Issue The Role of NK Cells in Health and Diseases)
Show Figures

Figure 1

24 pages, 4655 KiB  
Article
Dihydrotestosterone Enhances MICA-Mediated Immune Responses to Epstein–Barr Virus-Associated Gastric Carcinoma
by Donghyun Seo, Hyeji Byun, Miyeon Cho, Sun Hee Lee, Sohyun Youn, Junho Lee, Inuk Jung, Hyosun Cho and Hyojeung Kang
Cancers 2024, 16(18), 3219; https://doi.org/10.3390/cancers16183219 - 21 Sep 2024
Cited by 3 | Viewed by 1461
Abstract
Background: Epstein–Barr virus-associated gastric carcinoma (EBVaGC) is a subset of gastric cancers linked to EBV infection. While the role of male hormones in cancers such as prostate, breast, and ovarian cancers is well-studied, their impact on EBVaGC remains less understood. This study aims [...] Read more.
Background: Epstein–Barr virus-associated gastric carcinoma (EBVaGC) is a subset of gastric cancers linked to EBV infection. While the role of male hormones in cancers such as prostate, breast, and ovarian cancers is well-studied, their impact on EBVaGC remains less understood. This study aims to examine the effect of dihydrotestosterone (DHT) on EBVaGC, particularly focusing on its influence on the immune response. Methods: The study utilized the SNU719 EBVaGC cell line. Cells were treated with DHT to assess androgen receptor (AR) expression and the activation of signaling pathways, including NF-κB. The expression of MHC class I polypeptide-related sequence A (MICA) and its interaction with the NKG2D receptor on NK and T cells was evaluated. Cytotoxicity assays were conducted to determine DHT’s effect on NK and T cell-mediated cytotoxicity, and proinflammatory cytokine gene expression was analyzed. Results: DHT significantly increased AR expression in EBVaGC cells and activated the NF-κB pathway, which led to increased transcription of target genes such as MICA and EBNA1. These changes enhanced the interaction with receptors on NK and T cells, thereby boosting their cytotoxicity against EBVaGC cells. Importantly, DHT did not upregulate proinflammatory cytokine genes. Conclusion: DHT enhances the immune response against EBVaGC by upregulating MICA and activating NK and T cells. These findings suggest potential therapeutic strategies targeting androgen signaling to improve anti-tumor immunity in EBVaGC. Full article
(This article belongs to the Special Issue Epstein–Barr Virus (EBV) Associated Cancers)
Show Figures

Figure 1

15 pages, 2671 KiB  
Article
MAIT Cells in the Bone Marrow of Patients with Aplastic Anemia
by Lam Quang Vu, J. Luis Espinoza, Hoang Thao Giang Nguyen, Shohei Mizuno and Akiyoshi Takami
Int. J. Mol. Sci. 2024, 25(18), 10160; https://doi.org/10.3390/ijms251810160 - 21 Sep 2024
Viewed by 1746
Abstract
Mucosal-associated invariant T cells (MAIT cells) are a subset of T cells with innate, effector-like properties that play an essential role in the immune response to microbial infections. In humans, MAIT cells are detectable in the blood, liver, and lungs, but little is [...] Read more.
Mucosal-associated invariant T cells (MAIT cells) are a subset of T cells with innate, effector-like properties that play an essential role in the immune response to microbial infections. In humans, MAIT cells are detectable in the blood, liver, and lungs, but little is known about the frequency of these cells in the bone marrow. Also, the pathogenic role, if any, of MAIT cells in the development of aplastic anemia, a disease with an exquisite origin in the bone marrow, is currently unknown. We investigated the frequency and clinical relevance of bone marrow MAIT cells in a cohort of 14 patients (60.6 ± 23 and 57% women) with aplastic anemia. MAIT cells in the bone marrow samples obtained at diagnosis were evaluated by flow cytometry, and their association with various blood cell parameters and the patients’ clinical features was analyzed. MAIT cells were detectable in the bone marrow of all patients, with considerable variations among them. Bone marrow MAIT cells expressing the activator receptor natural killer group 2D - NKG2D (NKG2D+ MAIT cells) were significantly more abundant in the specimens of the aplastic anemia patients than in patients with bone marrow failure distinct from aplastic anemia. In addition, the NKG2D+ MAIT cells positively correlated with whole blood cell counts (WBC), platelet counts, and neutrophil counts, as well as with various inflammatory markers, including neutrophil-to-lymphocyte rate (NLR), platelet-to-lymphocyte rate (PLR), and systemic inflammatory index (SII). In functional studies, bone marrow CD34+ hematopoietic cells exposed to phytohemagglutinin or bacterial-derived lipopolysaccharide and acetyl-6-formylpterin upregulated MR1 (major histocompatibility complex, class I-related, known to interact with MAIT cells) and MICA/B (MHC class I chain-related gene A, a ligand of NKG2D) proteins on their cell surface, suggesting that under stress conditions, CD34+ hematopoietic cells are more likely to interact with NKG2D+ MAIT cells. In addition, NKG2D+ MAIT cells upregulated perforin and granzyme B in response to their interaction with recombinant MICA protein in vitro. This study reports for the first time the frequency of MAIT cells in the bone marrow of patients with aplastic anemia and assesses the potential implications of these cells in the pathogenesis or progression of aplastic anemia. Full article
(This article belongs to the Special Issue Molecular Immunology in Hematological Disorders 2.0)
Show Figures

Figure 1

Back to TopTop