Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Immunol ; 19(7): 723-732, 2018 07.
Article in English | MEDLINE | ID: mdl-29915296

ABSTRACT

Checkpoint blockade enhances effector T cell function and has elicited long-term remission in a subset of patients with a broad spectrum of cancers. TIGIT is a checkpoint receptor thought to be involved in mediating T cell exhaustion in tumors; however, the relevance of TIGIT to the dysfunction of natural killer (NK) cells remains poorly understood. Here we found that TIGIT, but not the other checkpoint molecules CTLA-4 and PD-1, was associated with NK cell exhaustion in tumor-bearing mice and patients with colon cancer. Blockade of TIGIT prevented NK cell exhaustion and promoted NK cell-dependent tumor immunity in several tumor-bearing mouse models. Furthermore, blockade of TIGIT resulted in potent tumor-specific T cell immunity in an NK cell-dependent manner, enhanced therapy with antibody to the PD-1 ligand PD-L1 and sustained memory immunity in tumor re-challenge models. This work demonstrates that TIGIT constitutes a previously unappreciated checkpoint in NK cells and that targeting TIGIT alone or in combination with other checkpoint receptors is a promising anti-cancer therapeutic strategy.


Subject(s)
Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Receptors, Immunologic/metabolism , Adaptive Immunity , Animals , Cell Line , Colonic Neoplasms/immunology , Humans , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics
2.
Immunology ; 171(1): 60-76, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37753964

ABSTRACT

Tumorigenesis entails circumventing cell-intrinsic regulatory mechanisms while avoiding extrinsic immune surveillance and other host defence systems. Nevertheless, how tumour cells' ability to eliminate misfolded proteins affects immune surveillance remains poorly understood. In this study, we find that overexpression of murine tripartite motif-containing protein 30a (TRIM30a) sensitises tumour cells to natural killer (NK) cells-mediated cytolysis. TRIM30a has no effect on tumour cell proliferation or apoptosis in vitro. However, TRIM30a-overexpressing tumour cells grow substantially slower than control tumour cells in immune-competent mice but not in NK cell-depleted mice. [Correction added on 04 October 2023, after first online publication: 'NK-depleted' has been changed to 'NK cell-depleted' in the preceding sentence.] Mechanistically, TRIM30a overexpression impedes the clearance of misfolded proteins and increases the production of reactive oxygen species induced by proteotoxic stress, implying that TRIM30a impairs protein quality control (PQC) systems in tumour cells. Furthermore, TRIM30a reduces expression of genes encoding proteasome subunits and antioxidant proteins. Our study demonstrates that TRIM30a is a potential tumour suppressor and immune modulator that promotes tumour cytolysis by NK cells, and suggests that an enhanced PQC and antioxidant capacity is an integral part of the immune escape mechanism during tumorigenesis.


Subject(s)
Antioxidants , Neoplasms , Animals , Mice , Antioxidants/metabolism , Carcinogenesis/metabolism , Killer Cells, Natural , Reactive Oxygen Species/metabolism
3.
Biochem Biophys Res Commun ; 524(2): 418-423, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32007276

ABSTRACT

The antimalarial drug Artemisinin has been reported to possess direct anti-tumor effects on various types of tumor cells. However, its anti-tumor potential has not been fully revealed, and its effects on tumor susceptibility to immune surveillance by the host are still unknown. Natural killer (NK) cells are the first line in tumor surveillance by the host, and have been recognized as a promising target for tumor immunotherapy. Here, we reported that Artemisinin sensitized tumor cells to NK cell cytolysis. Both human K562 and Raji tumor cells, and mouse YAC-1 tumor cells were more susceptible to human or mouse NK cell cytolysis in vitro after Artemisinin pretreatment. Conjugation formation between tumor cells and NK cells was increased after pretreatment with Artemisinin. Such effects on tumor cells by Artemisinin might not be the results of tumor recognition by NK cells, since major ligands of NK cell surface receptors were not affected. Mechanistically, although Artemisinin didn't induce tumor cell apoptosis, Artemisinin enriched apoptosis-related gene sets in these tumor cells, which might predispose tumor cells to apoptosis upon NK cell cytolysis. Moreover, NK cell numbers, percentages, maturation and functions were preserved in the presence of Artemisinin in vitro, suggesting that Artemisinin displays detrimental effects only on tumor cells but not on immune cells. These data reveal a novel anti-tumor mechanism of Artemisinin and demonstrate that Artemisinin could be a promising drug candidate for cancer treatment.


Subject(s)
Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Artemisinins/pharmacology , Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/drug effects , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Immunologic Surveillance/drug effects , Killer Cells, Natural/immunology , Mice , Neoplasms/immunology
4.
Immunology ; 158(2): 63-69, 2019 10.
Article in English | MEDLINE | ID: mdl-31315144

ABSTRACT

Natural killer cells are potent cytotoxic lymphocytes specialized in recognizing and eliminating transformed cells, and in orchestrating adaptive anti-tumour immunity. However, NK cells are usually functionally exhausted in the tumour microenvironment. Strategies such as checkpoint blockades are under investigation to overcome NK cell exhaustion in order to boost anti-tumour immunity. The discovery and development of the CRISPR/Cas9 technology offer a flexible and efficient gene-editing capability in modulating various pathways that mediate NK cell exhaustion, and in arming NK cells with novel chimeric antigen receptors to specifically target tumour cells. Despite the high efficiency in its gene-editing capability, difficulty in the delivery of the CRISPR/Cas9 system remains a major bottleneck for its therapeutic applications, particularly for NK cells. The current review discusses feasible approaches to deliver the CRISPR/Cas9 systems, as well as potential strategies in gene-editing for NK cell immunotherapy for cancers.


Subject(s)
CRISPR-Cas Systems/immunology , Drug Delivery Systems/methods , Gene Transfer Techniques , Killer Cells, Natural/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/genetics , Adaptive Immunity , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/immunology , Cellular Reprogramming/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , Cytotoxicity, Immunologic , Gene Editing/methods , Humans , Immunotherapy/methods , Killer Cells, Natural/metabolism , Metal Nanoparticles/administration & dosage , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Plasmids/chemistry , Plasmids/immunology , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/immunology , Receptors, Chimeric Antigen/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
5.
J Immunol ; 198(8): 3336-3344, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28275135

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) play an important role in orchestrating type II immune responses. However, the cellular mechanisms of group 2 innate lymphoid cell regulation remain poorly understood. In this study, we found that activated NK cells inhibited the proliferation of, as well as IL-5 and IL-13 production by, ILC2s in vitro via IFN-γ. In addition, in a murine model of ILC2 expansion in the liver, polyinosinic-polycytidylic acid, an NK cell-activating agent, inhibited ILC2 proliferation, IL-5 and IL-13 production, and eosinophil recruitment. Such effects of polyinosinic-polycytidylic acid were abrogated in NK cell-depleted mice and in IFN-γ-deficient mice. Adoptively transferring wild-type NK cells into NK cell-depleted mice resulted in fewer ILC2s induced by IL-33 compared with the transfer of IFN-γ-deficient NK cells. Importantly, during the early stage of papain- or bleomycin-induced lung inflammation, depletion of NK cells resulted in increased ILC2 numbers and enhanced cytokine production by ILC2s, as well as aggravated eosinophilia and goblet cell hyperplasia. Collectively, these data show that NK cells negatively regulate ILC2s during the early stage of lung inflammation, which represents the novel cellular interaction between two family members of ILCs.


Subject(s)
Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Pneumonia/immunology , Adoptive Transfer , Animals , Cell Separation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunity, Innate/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Biochem Biophys Res Commun ; 483(1): 669-673, 2017 01 29.
Article in English | MEDLINE | ID: mdl-27988336

ABSTRACT

IgE is a key effector molecule in atopic diseases; however, the regulation mechanisms of IgE production in IgE B cells remain poorly understood. In the present study, we demonstrate that JSI-124 (cucurbitacin I), a selective STAT3 inhibitor, selectively inhibits production of IgE by a human IgE B cell line, CRL-8033 cells, while does not affect the IgG production by IgG B cell lines. In the aspect of molecular mechanism, we found that Igλ, but not Ighe, gene expression was suppressed by JSI-124. The above effects of JSI-124 were not mediated by affecting cellular proliferation or apoptosis. Furthermore, multiple B cell differentiation-related genes expression was not significantly affected by JSI-124. Taken together, we demonstrate a potential strategy of therapeutically suppressing IgE production without affecting IgG production in atopic patients.


Subject(s)
B-Lymphocytes/drug effects , Immunoglobulin E/biosynthesis , Triterpenes/pharmacology , Apoptosis/drug effects , B-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Line , Cell Proliferation , Gene Expression/drug effects , Genes, Immunoglobulin/drug effects , Humans , Immunoglobulin E/genetics , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , STAT3 Transcription Factor/antagonists & inhibitors
7.
Immunol Cell Biol ; 95(7): 593-600, 2017 08.
Article in English | MEDLINE | ID: mdl-28202908

ABSTRACT

Autoimmune Th1 and Th17 responses are critical for the development of central nervous system (CNS) pathology in experimental autoimmune encephalomyelitis (EAE), an animal model for human multiple sclerosis. Although macrophages play important roles in the development of Th1 and Th17 responses, whether modulating macrophage gene transcription can diminish the Th1- and Th17 cell-induced CNS pathology is unclear. In this study, we successfully silenced the expression of the transcription factor c-Rel in macrophages of mice with EAE (including those infiltrating the CNS) using chemically modified c-Rel-specific siRNAs delivered by nanoparticles. Knocking down c-Rel in macrophages in vitro inhibited expression of NF-κB targets, such as pro-inflammatory cytokines interleukin 1ß (IL-1ß) and p40 of interleukin 12 (IL-12)/interleukin 23 (IL-23), in macrophages, leading to reduced interferon γ (IFN-γ) and interleukin 17A (IL-17A) production by co-cultured MOG-specific T cells from EAE mice. Such effects correlated with diminished T-cell infiltration in the CNS, reduced clinical symptoms, as well as downregulated pathogenic Th1 and Th17 responses in EAE mice. Taken together, our findings indicate that targeting c-Rel in macrophages dampens CNS-specific Th1 and Th17 immune responses, and can be effective for treating autoimmune diseases of the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Silencing , Macrophages/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Central Nervous System/pathology , Cytokines/metabolism , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Knockdown Techniques , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , NF-kappa B/metabolism , Nanoparticles/chemistry , RAW 264.7 Cells , RNA, Small Interfering/metabolism , Up-Regulation
8.
Hepatology ; 60(4): 1389-98, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24912841

ABSTRACT

UNLABELLED: Overactivation of innate immunity, particularly natural killer (NK) cells, is harmful to liver regeneration; however, the molecular mechanisms that limit NK cell overactivation during liver regeneration are still elusive. Here we show that a coinhibitory receptor, T cell Ig and ITIM domain (TIGIT), was selectively up-regulated on NK cells, along with high expression of its ligand, poliovirus receptor (PVR/CD155), on hepatocytes during liver regeneration. The absence of TIGIT impaired liver regeneration in vivo, along with overactivation of NK cells and higher NK-derived interferon-gamma (IFN-γ) production. We also show that both depletion of NK cells and deficiency of IFN-γ, but not deficiency of RAG1, rescued impaired liver regeneration caused by the absence of TIGIT. Adoptive transfer of Tigit(-/-) NK cells into NK-deficient Nfil3(-/-) mice sufficiently led to impairment of liver regeneration. On the other hand, silencing PVR in hepatocytes rescued impaired liver regeneration caused by TIGIT deficiency in vivo, while blockade of TIGIT in NK-hepatocyte coculture increased IFN-γ production by NK cells in vitro. CONCLUSION: TIGIT is a safeguard molecule to improve liver regeneration through negatively regulating NK-hepatocyte crosstalk. This finding suggests a novel mechanism of NK cell self-tolerance towards regenerative hyperplasia of the host.


Subject(s)
Cell Communication/physiology , Hepatocytes/pathology , Killer Cells, Natural/pathology , Liver Regeneration/physiology , Receptors, Immunologic/physiology , Animals , Coculture Techniques , Homeodomain Proteins/metabolism , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Virus/metabolism , Up-Regulation
9.
Hepatology ; 59(5): 1715-25, 2014 May.
Article in English | MEDLINE | ID: mdl-24319005

ABSTRACT

UNLABELLED: Uncontrolled natural killer (NK) cell activation during the early response to acute viral infection can lead to severe immunopathology, and the mechanisms NK cells use to achieve self-tolerance in such contexts are currently unclear. Here, NK cells up-regulated a coinhibitory receptor, T-cell Ig and ITIM domain (TIGIT), during challenge with the viral double-stranded RNA (dsRNA) analog poly I:C. Blocking TIGIT by antibody treatment in vivo or a genetic deficiency in Tigit enhanced NK cell activation and aggravated liver injury in a poly I:C/D-GalN-induced model of acute fulminant hepatitis, suggesting that TIGIT is normally required for protecting against NK cell-mediated liver injury. Furthermore, adoptively transferring Tigit(-/-) NK cells into NK cell-deficient Nfil3(-/-) mice also resulted in elevated liver injury. Reconstituting Kupffer cell-depleted mice with poliovirus receptor (PVR/CD155, a TIGIT ligand)-silenced Kupffer cells led to aggravated liver injury in a TIGIT-dependent manner. Blocking TIGIT in an NK-Kupffer cell coculture in vitro enhanced NK cell activation and interferon-gamma (IFN-γ) production in a PVR-dependent manner. We also found that TIGIT was up-regulated selectively on NK cells and protected against liver injury in an acute adenovirus infection model in both an NK cell- and Kupffer cell-dependent manner. Knocking down PVR in Kupffer cells resulted in aggravated liver injury in response to adenovirus infection in a TIGIT-dependent manner. CONCLUSION: TIGIT negatively regulates NK-Kupffer cell crosstalk and alleviates liver injury in response to poly I:C/D-GalN challenge or acute adenovirus infection, suggesting a novel mechanism of NK cell self-tolerance in liver homeostasis during acute viral infection.


Subject(s)
Hepatitis, Viral, Animal/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Receptors, Immunologic/physiology , Acute Disease , Animals , Antigens, CD/analysis , Antigens, CD/physiology , Antigens, Differentiation, T-Lymphocyte/analysis , Antigens, Differentiation, T-Lymphocyte/physiology , Interferon-gamma/blood , Kupffer Cells/physiology , Mice , Mice, Inbred C57BL , Poly I-C/pharmacology , Receptors, Virus/physiology
10.
J Immunother Cancer ; 11(2)2023 02.
Article in English | MEDLINE | ID: mdl-36725083

ABSTRACT

BACKGROUND: To enhance the efficacy of adoptive NK cell therapy against solid tumors, NK cells must be modified to resist exhaustion in the tumor microenvironment (TME). However, the molecular checkpoint underlying NK cell exhaustion in the TME remains elusive. METHODS: We analyzed the correlation between TIPE2 expression and NK cell functional exhaustion in the TME both in humans and mice by single-cell transcriptomic analysis and by using gene reporter mice. We investigated the effects of TIPE2 deletion on adoptively transferred NK cell therapy against cancers by using NK cells from NK-specific Tipe2-deficient mice or peripheral blood-derived or induced pluripotent stem cell (iPSC)-derived human NK cells with TIPE2 deletion by CRISPR/Cas9. We also investigated the potential synergy of double deletion of TIPE2 and another checkpoint molecule, CISH. RESULTS: By single-cell transcriptomic analysis and by using gene reporter mice, we found that TIPE2 expression correlated with NK cell exhaustion in the TME both in humans and mice and that the TIPE2 high NK cell subset correlated with poorer survival of tumor patients. TIPE2 deletion promoted the antitumor activity of adoptively transferred mouse NK cells and adoptively transferred human NK cells, either derived from peripheral blood or differentiated from iPSCs. TIPE2 deletion rendered NK cells with elevated capacities for tumor infiltration and effector functions. TIPE2 deletion also synergized with CISH deletion to further improve antitumor activity in vivo. CONCLUSIONS: This study highlighted TIPE2 targeting as a promising approach for enhancing adoptive NK cell therapy against solid tumors.


Subject(s)
Immunotherapy, Adoptive , Intracellular Signaling Peptides and Proteins , Killer Cells, Natural , Neoplasms , Animals , Humans , Mice , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/metabolism , Neoplasms/metabolism , Tumor Microenvironment
11.
Adv Sci (Weinh) ; 10(12): e2207499, 2023 04.
Article in English | MEDLINE | ID: mdl-36807566

ABSTRACT

Natural killer (NK) cells not only are innate effector lymphocytes that directly participate in tumor surveillance but are also essential helpers in the antitumor CD8+ T-cell response. However, the molecular mechanisms and potential checkpoints regulating NK cell helper functions remain elusive. Here, it is shown that the T-bet/Eomes-IFN-γ axis in NK cells is essential for CD8+ T cell-dependent tumor control, whereas T-bet-dependent NK cell effector functions are required for an optimal response to anti-PD-L1 immunotherapy. Importantly, NK cell-expressed TIPE2 (tumor necrosis factor-alpha-induced protein-8 like-2) represents a checkpoint molecule for NK cell helper function, since Tipe2 deletion in NK cells not only enhances NK-intrinsic antitumor activity but also indirectly improves the antitumor CD8+ T cell response by promoting T-bet/Eomes-dependent NK cell effector functions. These studies thus reveal TIPE2 as a checkpoint for NK cell helper function, whose targeting might boost the antitumor T cell response in addition to T cell-based immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Killer Cells, Natural , Neoplasms/therapy , Neoplasms/pathology , Proteins , Immunotherapy
12.
Biomark Res ; 11(1): 63, 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37287049

ABSTRACT

The ATP-adenosine pathway has emerged as a promising target for cancer therapy, but challenges remain in achieving effective tumor control. Early research focused on blocking the adenosine generating enzyme CD73 and the adenosine receptors A2AR or A2BR in cancer. However, recent studies have shown that targeting CD39, the rate-limiting ecto-enzyme of the ATP-adenosine pathway, can provide more profound anti-tumor efficacy by reducing immune-suppressive adenosine accumulation and increasing pro-inflammatory ATP levels. In addition, combining CD39 blocking antibody with PD-1 immune checkpoint therapy may have synergistic anti-tumor effects and improve patient survival. This review will discuss the immune components that respond to CD39 targeting in the tumor microenvironment. Targeting CD39 in cancer has been shown to not only decrease adenosine levels in the tumor microenvironment (TME), but also increase ATP levels. Additionally, targeting CD39 can limit the function of Treg cells, which are known to express high levels of CD39. With phase I clinical trials of CD39 targeting currently underway, further understanding and rational design of this approach for cancer therapy are expected.

13.
Front Immunol ; 13: 1084109, 2022.
Article in English | MEDLINE | ID: mdl-36591230

ABSTRACT

Effective and long-term treatment is required for controlling chronic Hepatitis B Virus (HBV) infection. Natural killer (NK) cells are antiviral innate lymphocytes and represent an essential arm of current immunotherapy. In chronic HBV (CHB), NK cells display altered changes in phenotypes and functions, but preserve antiviral activity, especially for cytolytic activity. On the other hand, NK cells might also cause liver injury in the disease. NK -based immunotherapy, including adoptive NK cell therapy and NK -based checkpoint inhibition, could potentially exploit the antiviral aspect of NK cells for controlling CHB infection while preventing liver tissue damage. Here, we review recent progress in NK cell biology under the context of CHB infection, and discuss potential NK -based immunotherapy strategies for the disease.


Subject(s)
Hepatitis B, Chronic , Humans , Hepatitis B virus/genetics , Antiviral Agents/therapeutic use , Immunotherapy
14.
Cell Mol Immunol ; 19(2): 192-209, 2022 02.
Article in English | MEDLINE | ID: mdl-35043005

ABSTRACT

Cytokines exert powerful immunomodulatory effects that are critical to physiology and pathology in humans. The application of natural cytokines in clinical studies has not been clearly established, and there are often problems associated with toxicity or lack of efficacy. The key reasons can be attributed to the pleiotropy of cytokine receptors and undesired activation of off-target cells. With a deeper understanding of the structural principles and functional signals of cytokine-receptor interactions, artificial modification of cytokine signaling through protein engineering and synthetic immunology has become an increasingly feasible and powerful approach. Engineered cytokines are designed to selectively target cells. Herein, the theoretical and experimental evidence of cytokine engineering is reviewed, and the "supercytokines" resulting from structural enhancement and the "immunocytokines" generated by antibody fusion are described. Finally, the "engager cytokines" formed by the crosslinking of cytokines and bispecific immune engagers and other synthetic cytokines formed by nonnatural analogs are also discussed.


Subject(s)
Cytokines , Immunotherapy , Animals , Cytokines/metabolism , Disease Models, Animal , Extracellular Space , Humans , Immunologic Factors , Immunotherapy/methods
15.
Front Immunol ; 12: 761920, 2021.
Article in English | MEDLINE | ID: mdl-34675939

ABSTRACT

Natural killer (NK) cells are cytotoxic innate lymphocytes that play an important role in immune surveillance. The development, maturation and effector functions of NK cells are orchestrated by the T-box transcription factor T-bet, whose expression is induced by cytokines such as IFN-γ, IL-12, IL-15 and IL-21 through the respective cytokine receptors and downstream JAK/STATs or PI3K-AKT-mTORC1 signaling pathways. In this review, we aim to discuss the expression and regulation of T-bet in NK cells, the role of T-bet in mouse NK cell development, maturation, and function, as well as the role of T-bet in acute, chronic infection, inflammation, autoimmune diseases and tumors.


Subject(s)
Killer Cells, Natural/immunology , T-Box Domain Proteins/immunology , Animals , Autoimmune Diseases/immunology , Humans , Infections/immunology , Inflammation/immunology , Neoplasms/immunology
16.
Int Immunopharmacol ; 96: 107658, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33887610

ABSTRACT

Natural Killer (NK) cells are components of innate immune surveillance against transformed cells. NK cell immunotherapy has attracted attention as a promising strategy for cancer treatment, whose antitumor effects, however, require further improvement. The use of small molecules with immunomodulatory potentials and selective tumor-killing possesses the potential to complement immunotherapy. This study demonstrated that Piperlongumine (PL), a natural alkaloid obtained from long pepper fruit, alone has antitumor and anti-proliferative potential on all the tested tumors in vitro. PL pretreatment of tumor cells also potentiates their susceptibility to NK cell cytolysis at the doses where NK cell functions were preserved. Importantly, PL suppresses both NK -sensitive MHC-I -deficient and MHC-I -sufficient tumor growth in vivo. Mechanistically, PL induces misfolded proteins, impedes autophagy, increases ROS and tumor conjugation with NK cells. Furthermore, PL enhances the expression of NK cell-activating receptors on NK cells and its ligands on tumor cells, possibly leading to increased susceptibility to NK cell killing. Our findings showed the antitumor and immunomodulatory potential of PL, which could be explored to complement NK cell immunotherapy for cancer treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biological Products/pharmacology , Dioxolanes/pharmacology , Killer Cells, Natural/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Animals , Antineoplastic Agents, Phytogenic/immunology , Apoptosis/drug effects , Autophagy/drug effects , Biological Products/immunology , Cell Line, Tumor , Cell Survival/drug effects , Cytotoxicity, Immunologic/drug effects , Dioxolanes/immunology , Humans , Killer Cells, Natural/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Receptors, Natural Killer Cell/drug effects , Receptors, Natural Killer Cell/metabolism , Xenograft Model Antitumor Assays
17.
Int Immunopharmacol ; 95: 107541, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33756232

ABSTRACT

Although exogenous oxidative stress has been suggested to promote the pathogenesis of airway inflammation, previous trials using conventional antioxidant therapy in asthma have been largely ineffective, suggesting the complex roles of oxidative stress in the regulation of airway inflammation and of its critical mediating lymphocyte populations. Group 2 innate lymphoid cells (ILC2s) proliferate and induce eosinophilia in response to tissue alarminsin the early phase of airway inflammation. We previously showed that IL-33 -induced endogenous reactive oxygen species is required for optimal metabolic activation of ILC2 functions, however, the role of exogenous oxidative stress in regulating ILC2 functions has not been investigated. Here, we found that exogenous oxidative stress induced by injection of ROS -generating reagent alleviated IL-33 -triggered ILC2 response and inflammation both in the airway and in the liver. Exogenous oxidative stress in ILC2s also compromised IL-33 -mediated accumulation of these cells, as well as subsequent recruitment of eosinophils, after adoptive transferring into ILC2 deficient hosts. Mechanistically, exogenous oxidative stress in ILC2s compromised the proliferation program, while preserving the expression levels of effector molecules in ILC2s. Impaired proliferation under exogenous oxidative stress led to reduced numbers of ILC2s, and an overall decrease in ILC2 response to IL-33 stimulation. Collectively, these data indicate that exogenous oxidative stress suppresses the proliferation program in ILC2s and alleviates IL-33 -triggered inflammation, suggesting that therapeutic induction of oxidative stress might alleviate ILC2 -mediated inflammation in the airway, and possibly also in other organs.


Subject(s)
Interleukin-33/immunology , Lymphocytes/immunology , Oxidative Stress , Animals , Cell Proliferation , Cells, Cultured , Humans , Liver/immunology , Lung/immunology , Mice, Inbred C57BL , Mice, Transgenic , Transcriptome
18.
Sci Adv ; 7(38): eabi6515, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34524845

ABSTRACT

The maturation process of NK cells determines their functionality during which IL-15 plays a critical role. However, very few checkpoints specifically targeting this process have been discovered. Here, we report that TIPE2 expression gradually increased during NK cell ontogenesis correlating to their maturation stages in both mice and humans. NK-specific TIPE2 deficiency increased mature NK cells in mice, and these TIPE2-deficient NK cells exhibited enhanced activation, cytotoxicity, and IFN-γ production upon stimulation and enhanced response to IL-15 for maturation. Moreover, TIPE2 suppressed IL-15­triggered mTOR activity in both human and murine NK cells. Consequently, blocking mTOR constrained the effect of TIPE2 deficiency on NK cell maturation in response to IL-15. Last, NK-specific TIPE2-deficient mice were resistant to tumor growth in vivo. Our results uncover a potent checkpoint in NK cell maturation and antitumor immunity in both mice and humans, suggesting a promising approach of targeting TIPE2 for NK cell­based immunotherapies.


Subject(s)
Interleukin-15 , Killer Cells, Natural , Animals , Cell Differentiation , Humans , Immunotherapy/methods , Interleukin-15/metabolism , Interleukin-15/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , TOR Serine-Threonine Kinases/metabolism
19.
Front Immunol ; 12: 701671, 2021.
Article in English | MEDLINE | ID: mdl-34531855

ABSTRACT

Histone deacetylases (HDAC) are frequently overexpressed in tumors, and their inhibition has shown promising anti-tumor effects. However, the synergistic effects of HDAC inhibition with immune cell therapy have not been fully explored. Natural killer (NK) cells are cytotoxic lymphocytes for anti-tumor immune surveillance, with immunotherapy potential. We showed that a pan-HDAC inhibitor, panobinostat, alone demonstrated anti-tumor and anti-proliferative activities on all tested tumors in vitro. Additionally, panobinostat co-treatment or pretreatment synergized with NK cells to mediate tumor cell cytolysis. Mechanistically, panobinostat treatment increased the expression of cell adhesion and tight junction-related genes, promoted conjugation formation between NK and tumor cells, and modulates NK cell-activating receptors and ligands on tumor cells, contributing to the increased tumor cytolysis. Finally, panobinostat therapy led to better tumor control and synergized with anti-PD-L1 therapy. Our data highlights the anti-tumor potential of HDAC inhibition through tumor-intrinsic toxicity and enhancement of NK -based immunotherapy.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Killer Cells, Natural/drug effects , Panobinostat/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Line , Cell Line, Tumor , Drug Synergism , HeLa Cells , Hep G2 Cells , Humans , Immunologic Surveillance/drug effects , Immunotherapy/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Tight Junctions/drug effects
20.
Front Immunol ; 12: 712936, 2021.
Article in English | MEDLINE | ID: mdl-34489962

ABSTRACT

The engineered "obligate" anaerobic Salmonella typhimurium strain YB1 shows a prominent ability to repress tumor growth and metastasis, which has great potential as a novel cancer immunotherapy. However, the antitumor mechanism of YB1 remains unelucidated. To resolve the proteome dynamics induced by the engineered bacteria, we applied tumor temporal proteome profiling on murine bladder tumors after intravenous injection of either YB1 or PBS as a negative control. Our data suggests that during the two weeks treatment of YB1 injections, the cured tumors experienced three distinct phases of the immune response. Two days after injection, the innate immune response was activated, particularly the complement and blood coagulation pathways. In the meantime, the phagocytosis was initiated. The professional phagocytes such as macrophages and neutrophils were recruited, especially the infiltration of iNOS+ and CD68+ cells was enhanced. Seven days after injection, substantial amount of T cells was observed at the invasion margin of the tumor. As a result, the tumor shrunk significantly. Overall, the temporal proteome profiling can systematically reveal the YB1 induced immune responses in tumor, showing great promise for elucidating the mechanism of bacteria-mediated cancer immunotherapy.


Subject(s)
Biological Therapy/methods , Neoplasms/etiology , Neoplasms/metabolism , Proteome , Proteomics , Salmonella typhimurium , Animals , Blood Coagulation , Cell Line, Tumor , Chromatography, Liquid , Complement System Proteins/immunology , Computational Biology/methods , Disease Management , Disease Models, Animal , Disease Susceptibility , Female , Genetic Engineering , Humans , Lymphocyte Activation , Neoplasms/pathology , Neoplasms/therapy , Phagocytosis , Proteomics/methods , Salmonella typhimurium/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tandem Mass Spectrometry , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL