Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Mol Cancer ; 23(1): 68, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38561826

ABSTRACT

Cancer progression is continuously controlled by the immune system which can identify and destroy nascent tumor cells or inhibit metastatic spreading. However, the immune system and its deregulated activity in the tumor microenvironment can also promote tumor progression favoring the outgrowth of cancers capable of escaping immune control, in a process termed cancer immunoediting. This process, which has been classified into three phases, i.e. "elimination", "equilibrium" and "escape", is influenced by several cancer- and microenvironment-dependent factors. Senescence is a cellular program primed by cells in response to different pathophysiological stimuli, which is based on long-lasting cell cycle arrest and the secretion of numerous bioactive and inflammatory molecules. Because of this, cellular senescence is a potent immunomodulatory factor promptly recruiting immune cells and actively promoting tissue remodeling. In the context of cancer, these functions can lead to both cancer immunosurveillance and immunosuppression. In this review, the authors will discuss the role of senescence in cancer immunoediting, highlighting its context- and timing-dependent effects on the different three phases, describing how senescent cells promote immune cell recruitment for cancer cell elimination or sustain tumor microenvironment inflammation for immune escape. A potential contribution of senescent cells in cancer dormancy, as a mechanism of therapy resistance and cancer relapse, will be discussed with the final objective to unravel the immunotherapeutic implications of senescence modulation in cancer.


Subject(s)
Neoplasms , Humans , Neoplasms/pathology , Cellular Senescence , Immune System , Immunosuppression Therapy , Tumor Microenvironment
2.
Eur J Immunol ; 53(2): e2250198, 2023 02.
Article in English | MEDLINE | ID: mdl-36440686

ABSTRACT

Natural killer (NK) cell activation is regulated by activating and inhibitory receptors that facilitate diseased cell recognition. Among activating receptors, NKG2D and DNAM-1 play a pivotal role in anticancer immune responses since they bind ligands upregulated on transformed cells. During tumor progression, however, these receptors are frequently downmodulated and rendered functionally inactive. Of note, NKG2D internalization has been associated with the acquisition of a dysfunctional phenotype characterized by the cross-tolerization of unrelated activating receptors. However, our knowledge of the consequences of NKG2D engagement is still incomplete. Here, by cytotoxicity assays combined with confocal microscopy, we demonstrate that NKG2D engagement on human NK cells impairs DNAM-1-mediated killing through two different converging mechanisms: by the upregulation of the checkpoint inhibitory receptor TIGIT, that in turn suppresses DNAM-1-mediated cytotoxic function, and by direct inhibition of DNAM-1-promoted signaling. Our results highlight a novel interplay between NKG2D and DNAM-1/TIGIT receptors that may facilitate neoplastic cell evasion from NK cell-mediated clearance.


Subject(s)
Killer Cells, Natural , Neoplasms , Tumor Escape , Humans , Killer Cells, Natural/immunology , Neoplasms/genetics , Neoplasms/immunology , NK Cell Lectin-Like Receptor Subfamily K , Signal Transduction , Tumor Escape/genetics , Tumor Escape/immunology
3.
Int J Mol Sci ; 24(3)2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36768145

ABSTRACT

Nuclear factor-κB (NF-κB) transcription factors play a key role in the pathogenesis of multiple myeloma (MM). The survival, proliferation and chemoresistance of malignant plasma cells largely rely on the activation of canonical and noncanonical NF-κB pathways. They are triggered by cancer-associated mutations or by the autocrine and paracrine production of cytokines and growth factors as well as direct interaction with cellular and noncellular components of bone marrow microenvironment (BM). In this context, NF-κB also significantly affects the activity of noncancerous cells, including mesenchymal stromal cells (MSCs), which have a critical role in disease progression. Indeed, NF-κB transcription factors are involved in inflammatory signaling that alters the functional properties of these cells to support cancer evolution. Moreover, they act as regulators and/or effectors of pathways involved in the interplay between MSCs and MM cells. The aim of this review is to analyze the role of NF-κB in this hematologic cancer, focusing on NF-κB-dependent mechanisms in tumor cells, MSCs and myeloma-mesenchymal stromal cell crosstalk.


Subject(s)
Mesenchymal Stem Cells , Multiple Myeloma , Humans , NF-kappa B/metabolism , Multiple Myeloma/pathology , Signal Transduction , Mesenchymal Stem Cells/metabolism , Transcription Factors/metabolism , Stromal Cells/metabolism , Tumor Microenvironment
4.
Int J Mol Sci ; 24(11)2023 May 30.
Article in English | MEDLINE | ID: mdl-37298418

ABSTRACT

Natural Killer (NK) cells are innate cytotoxic lymphoid cells that play a crucial role in cancer immunosurveillance. NKG2D is an activating receptor that binds to MIC and ULBP molecules typically induced on damaged, transformed, or infected cells. The secretion of NKG2D ligands (NKG2DLs) through protease-mediated cleavage or in an extracellular vesicle (EV) is a mode to control their cell surface expression and a mechanism used by cancer cells to evade NKG2D-mediated immunosurveillance. EVs are emerging as important players in mediating cell-to-cell communication due to their ability to transfer biological material to acceptor cells. Herein, we investigated the spreading of NKG2DLs of both MIC and ULBP molecules through the EV-mediated cross-dressing on multiple myeloma (MM) cells. We focused our attention on two MICA allelic variants, namely MICA*008 and MICA*019, representing the prototype of short and long MICA alleles, respectively, and on ULBP-1, ULBP-2, and ULBP-3. Our findings demonstrate that both ULBP and MICA ligands can be acquired from tumor cells through EVs enhancing NK cell recognition and killing. Moreover, besides MICA, EVs expressing ULBP-1 but not ULBP-2 and 3 were detected in bone marrow aspirates derived from a cohort of MM patients. Our findings shed light on the role of EV-associated MICA allelic variants and ULBP molecules in the modulation of NKG2D-mediated NK cell immunosurveillance in the tumor microenvironment. Moreover, the EV-mediated transfer of NKG2DLs could suggest novel therapeutic approaches based on the usage of engineered nanoparticles aimed at increasing cancer cell immunogenicity.


Subject(s)
Extracellular Vesicles , Multiple Myeloma , Humans , Multiple Myeloma/metabolism , Ligands , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural , Extracellular Vesicles/metabolism , Cell Death , Bandages , Tumor Microenvironment
5.
Int J Mol Sci ; 24(7)2023 Apr 06.
Article in English | MEDLINE | ID: mdl-37047835

ABSTRACT

The success of senescence-based anticancer therapies relies on their anti-proliferative power and on their ability to trigger anti-tumor immune responses. Indeed, genotoxic drug-induced senescence increases the expression of NK cell-activating ligands on multiple myeloma (MM) cells, boosting NK cell recognition and effector functions. Senescent cells undergo morphological change and context-dependent functional diversification, acquiring the ability to secrete a vast pool of molecules termed the senescence-associated secretory phenotype (SASP), which affects neighboring cells. Recently, exosomes have been recognized as SASP factors, contributing to modulating a variety of cell functions. In particular, evidence suggests a key role for exosomal microRNAs in influencing many hallmarks of cancer. Herein, we demonstrate that doxorubicin treatment of MM cells leads to the enrichment of miR-433 into exosomes, which in turn induces bystander senescence. Our analysis reveals that the establishment of the senescent phenotype on neighboring MM cells is p53- and p21-independent and is related to CDK-6 down-regulation. Notably, miR-433-dependent senescence does not induce the up-regulation of activating ligands on MM cells. Altogether, our findings highlight the possibility of miR-433-enriched exosomes to reinforce doxorubicin-mediated cellular senescence.


Subject(s)
Antibiotics, Antineoplastic , Bystander Effect , Cellular Senescence , Doxorubicin , Exosomes , MicroRNAs , Multiple Myeloma , Topoisomerase II Inhibitors , Cellular Senescence/drug effects , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use , Humans , Cell Line, Tumor , Exosomes/drug effects , Exosomes/metabolism , DNA Damage , MicroRNAs/genetics , MicroRNAs/metabolism , Tumor Suppressor Protein p53/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism
6.
Eur J Immunol ; 51(11): 2607-2617, 2021 11.
Article in English | MEDLINE | ID: mdl-34392531

ABSTRACT

Rearrangement of the actin cytoskeleton is critical for cytotoxic and immunoregulatory functions as well as migration of natural killer (NK) cells. However, dynamic reorganization of actin is a complex process, which remains largely unknown. Here, we investigated the role of the protein Cereblon (CRBN), an E3 ubiquitin ligase complex co-receptor and the primary target of the immunomodulatory drugs, in NK cells. We observed that CRBN partially colocalizes with F-actin in chemokine-treated NK cells and is recruited to the immunological synapse, thus suggesting a role for this protein in cytoskeleton reorganization. Accordingly, silencing of CRBN in NK cells results in a reduced cytotoxicity that correlates with a defect in conjugate and lytic synapse formation. Moreover, CRBN depletion significantly impairs the ability of NK cells to migrate and reduces the enhancing effect of lenalidomide on NK cell migration. Finally, we provided evidence that CRBN is required for activation of the small GTPase Rac1, a critical mediator of cytoskeleton dynamics. Indeed, in CRBN-depleted NK cells, chemokine-mediated or target cell-mediated Rac1 activation is significantly reduced. Altogether our data identify a critical role for CRBN in regulating NK cell functions and suggest that this protein may mediate the stimulatory effect of lenalidomide on NK cells.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cell Movement/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Ubiquitin-Protein Ligases/immunology , rac1 GTP-Binding Protein/immunology , Cell Movement/drug effects , Cytotoxicity, Immunologic/drug effects , Humans , Immunomodulating Agents/pharmacology , Killer Cells, Natural/drug effects , Lenalidomide/pharmacology
7.
Int J Mol Sci ; 22(3)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33499314

ABSTRACT

The Ikaros zing-finger family transcription factors (IKZF TFs) are important regulators of lymphocyte development and differentiation and are also highly expressed in B cell malignancies, including Multiple Myeloma (MM), where they are required for cancer cell growth and survival. Moreover, IKZF TFs negatively control the functional properties of many immune cells. Thus, the targeting of these proteins has relevant therapeutic implications in cancer. Indeed, accumulating evidence demonstrated that downregulation of Ikaros and Aiolos, two members of the IKZF family, in malignant plasma cells as well as in adaptative and innate lymphocytes, is key for the anti-myeloma activity of Immunomodulatory drugs (IMiDs). This review is focused on IKZF TF-related pathways in MM. In particular, we will address how the depletion of IKZF TFs exerts cytotoxic effects on MM cells, by reducing their survival and proliferation, and concomitantly potentiates the antitumor immune response, thus contributing to therapeutic efficacy of IMiDs, a cornerstone in the treatment of this neoplasia.


Subject(s)
Ikaros Transcription Factor/physiology , Multiple Myeloma/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation , Cell Survival , Humans , Ikaros Transcription Factor/genetics , Immunity/drug effects , Immunomodulation/drug effects , Lenalidomide/pharmacology , Lymphocytes/cytology , Mice , Multiple Myeloma/immunology , Thalidomide/pharmacology
8.
Eur J Immunol ; 49(6): 873-883, 2019 06.
Article in English | MEDLINE | ID: mdl-30888046

ABSTRACT

Nectin2 is a member of immunoglobulin-like cell adhesion molecules and plays a prominent role in the establishment of adherens and tight junctions. It is also upregulated on the surface of tumor and virus-infected cells where it functions as a ligand for the activating receptor CD226, thus contributing to cytotoxic lymphocyte-mediated recognition and killing of damaged cells. Little is currently known about the regulation of Nectin2 expression and, in particular, whether posttranscriptional and posttranslational mechanisms are involved. Here, we analyzed Nectin2 expression on a panel of human tumor cell lines and primary cultures and we found that Nectin2 is mainly expressed in cytoplasmic pools. Moreover, we demonstrated that ubiquitination of Nectin2 promotes its degradation and is responsible for protein intracellular retention. Indeed, inhibition of the ubiquitin pathway results in increased Nectin2 surface expression and enhances tumor cell susceptibility to NK cell cytotoxicity. Our results demonstrate a previously unknown mechanism of Nectin2 regulation revealing that the ubiquitin pathway represents a potential target of intervention in order to increase susceptibility to NK cell-mediated lysis.


Subject(s)
Cytotoxicity, Immunologic/immunology , Gene Expression Regulation/immunology , Nectins/biosynthesis , Tumor Escape/immunology , Cells, Cultured , Humans , Killer Cells, Natural , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , Ubiquitination/immunology
9.
FASEB J ; 33(8): 9489-9504, 2019 08.
Article in English | MEDLINE | ID: mdl-31125275

ABSTRACT

NK cells have an important role in immunosurveillance of multiple myeloma (MM) progression, and their activity is enhanced by combination therapies able to regulate the expression of specific activating ligands. Liver X receptors (LXRs) are nuclear receptors and important regulators of intracellular cholesterol and lipid homeostasis. Moreover, they have regulatory roles in both cancer and immune response. Indeed, they can regulate inflammation and innate and acquired immunity. Furthermore, LXR activation directly acts in cancer cells (e.g., prostate, breast, melanoma, colon cancer, hepatocarcinoma, glioblastoma, and MM) that show an accumulation of cholesterol and alteration of LXR-mediated metabolic pathways. Here, we investigated the role of LXR and cholesterol on the expression of the NK cell-activating ligands major histocompatibility complex class I chain-related molecule A and B (MICA and MICB) in MM cells. The results shown in this work indicate that MM cells are responsive to LXR activation, which induces changes in the intracellular cholesterol content. These changes correlate with an enhanced expression of MICA and MICB in human MM cell lines and in primary malignant plasma cells, 2 ligands of the NK group 2D receptor (NKG2D)/CD314 activating receptor expressed in cytotoxic lymphocytes, rendering MM cells more sensitive to recognition, degranulation, and killing by NK cells. Mechanistically, we observed that LXR activation regulates MICA and MICB expression at different levels: MICA at the transcriptional level, enhancing mica promoter activity, and MICB by inhibiting its degradation in lysosomes. The present study provides evidence that activation of LXR, by enhancing NKG2D ligand expression, can promote NK cell-mediated cytotoxicity and suggests a novel immune-mediated mechanism involving modulation of intracellular cholesterol levels in cancer cells.-Bilotta, M. T., Abruzzese, M. P., Molfetta, R., Scarno, G., Fionda, C., Zingoni, A., Soriani, A., Garofalo, T., Petrucci, M. T., Ricciardi, M. R., Paolini, R., Santoni, A., Cippitelli, M. Activation of liver X receptor up-regulates the expression of the NKG2D ligands MICA and MICB in multiple myeloma through different molecular mechanisms.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Liver X Receptors/metabolism , Multiple Myeloma/metabolism , Adaptive Immunity/physiology , Apoptosis/genetics , Apoptosis/physiology , Cell Line , Cells, Cultured , Chromatography, Thin Layer , Flow Cytometry , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/genetics , Humans , Immunity, Innate/physiology , Inflammation/metabolism , Killer Cells, Natural/metabolism , Liver X Receptors/genetics , Microscopy, Confocal , Multiple Myeloma/genetics , Promoter Regions, Genetic/genetics
10.
Int J Mol Sci ; 21(3)2020 Jan 30.
Article in English | MEDLINE | ID: mdl-32019260

ABSTRACT

CD155 is an adhesion molecule belonging to the Nectin/Nectin-like family often overexpressed on tumor cells and involved in many different processes such as cell adhesion, migration and proliferation. In contrast to these pro-tumorigenic functions, CD155 is also a ligand for the activating receptor DNAM-1 expressed on cytotoxic lymphocytes including Natural Killer (NK) cells and involved in anti-tumor immune response. However, during tumor progression inhibitory receptors for CD155 are up-regulated on the surface of effector cells, contributing to an impairment of their cytotoxic capacity. In this review we will focus on the roles of CD155 as a ligand for the activating receptor DNAM-1 regulating immune surveillance against cancer and as pro-oncogenic molecule favoring tumor proliferation, invasion and immune evasion. A deeper understanding of the multiple roles played by CD155 in cancer development contributes to improving anti-tumor strategies aimed to potentiate immune response against cancer.


Subject(s)
Immunologic Surveillance/immunology , Neoplasms/immunology , Neoplasms/pathology , Receptors, Virus/metabolism , Disease Progression , Humans , Neoplasms/metabolism
11.
J Immunol ; 197(10): 4066-4078, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27733551

ABSTRACT

Elimination of virus-infected cells by cytotoxic lymphocytes is triggered by activating receptors, among which NKG2D and DNAM-1/CD226 play an important role. Their ligands, that is, MHC class I-related chain (MIC) A/B and UL16-binding proteins (ULBP)1-6 (NKG2D ligand), Nectin-2/CD112, and poliovirus receptor (PVR)/CD155 (DNAM-1 ligand), are often induced on virus-infected cells, although some viruses, including human CMV (HCMV), can block their expression. In this study, we report that infection of different cell types with laboratory or low-passage HCMV strains upregulated MICA, ULBP3, and PVR, with NKG2D and DNAM-1 playing a role in NK cell-mediated lysis of infected cells. Inhibition of viral DNA replication with phosphonoformic acid did not prevent ligand upregulation, thus indicating that early phases of HCMV infection are involved in ligand increase. Indeed, the major immediate early (IE) proteins IE1 and IE2 stimulated the expression of MICA and PVR, but not ULBP3. IE2 directly activated MICA promoter via its binding to an IE2-responsive element that we identified within the promoter and that is conserved among different alleles of MICA. Both IE proteins were instead required for PVR upregulation via a mechanism independent of IE DNA binding activity. Finally, inhibiting IE protein expression during HCMV infection confirmed their involvement in ligand increase. We also investigated the contribution of the DNA damage response, a pathway activated by HCMV and implicated in ligand regulation. However, silencing of ataxia telangiectasia mutated, ataxia telangiectasia and Rad3-related protein, and DNA-dependent protein kinase did not influence ligand expression. Overall, these data reveal that MICA and PVR are directly regulated by HCMV IE proteins, and this may be crucial for the onset of an early host antiviral response.


Subject(s)
Gene Expression Regulation , Histocompatibility Antigens Class I/genetics , Immediate-Early Proteins/metabolism , Receptors, Virus/genetics , Trans-Activators/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , Cell Line , Cytotoxicity, Immunologic , DNA Replication/drug effects , Fibroblasts/drug effects , Fibroblasts/virology , Foscarnet/pharmacology , GPI-Linked Proteins/genetics , HEK293 Cells , Host-Pathogen Interactions , Humans , Immediate-Early Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/genetics , Trans-Activators/pharmacology , Transcriptional Activation , Up-Regulation , Viral Proteins/genetics , Virus Replication/drug effects
12.
Crit Rev Immunol ; 36(6): 445-460, 2016.
Article in English | MEDLINE | ID: mdl-28845754

ABSTRACT

Natural killer (NK) cells are critical immune effector cells capable of mediating antitumor responses. These cytotoxic lymphocytes recognize transformed cells through a mechanism mainly dependent on the engagement of several activating receptors. However, many tumors have developed strategies to evade immunosurveillance and detection by NK cells. A relevant immune escape mechanism is the down regulation of NK cell activating ligands on the surface of tumor cells by proteolytic shedding mediated by different members of metalloproteinase families. Here, we consider two important NK activating receptors, namely NKG2D and NKp30, the ligands (i.e., MICA/B, ULBPs, and B7-H6) of which can be released by cancer cells through proteolytic cleavage. Modulation of ligand shedding in response to cancer therapy is also examined, and we discuss how metalloproteinases implicated in the ligand cleavage could be targeted in novel therapeutic schemes to counteract tumor escape from stress-elicited immune responses.

13.
J Immunol ; 195(2): 736-48, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26071561

ABSTRACT

Genotoxic stress can promote antitumor NK cell responses by upregulating the surface expression of activating ligands on cancer cells. Moreover, a number of studies suggested a role for soluble NK group 2D ligands in the impairment of NK cell tumor recognition and killing. We investigated whether genotoxic stress could promote the release of NK group 2D ligands (MHC class I-related chain [MIC]A and MICB), as well as the molecular mechanisms underlying this event in human multiple myeloma (MM) cells. Our results show that genotoxic agents used in the therapy of MM (i.e., doxorubicin and melphalan) selectively affect the shedding of MIC molecules that are sensitive to proteolytic cleavage, whereas the release of the short MICA*008 allele, which is frequent in the white population, is not perturbed. In addition, we found that a disintegrin and metalloproteinase 10 expression is upregulated upon chemotherapeutic treatment both in patient-derived CD138(+)/CD38(+) plasma cells and in several MM cell lines, and we demonstrate a crucial role for this sheddase in the proteolytic cleavage of MIC by means of silencing and pharmacological inhibition. Interestingly, the drug-induced upregulation of a disintegrin and metalloproteinase 10 on MM cells is associated with a senescent phenotype and requires generation of reactive oxygen species. Moreover, the combined use of chemotherapeutic drugs and metalloproteinase inhibitors enhances NK cell-mediated recognition of MM cells, preserving MIC molecules on the cell surface and suggesting that targeting of metalloproteinases in conjunction with chemotherapy could be exploited for NK cell-based immunotherapeutic approaches, thus contributing to avoid the escape of malignant cells from stress-elicited immune responses.


Subject(s)
ADAM Proteins/immunology , Amyloid Precursor Protein Secretases/immunology , Cytotoxins/pharmacology , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/drug effects , Membrane Proteins/immunology , Plasma Cells/drug effects , ADAM Proteins/genetics , ADAM10 Protein , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/immunology , Amyloid Precursor Protein Secretases/genetics , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Line, Tumor , Cellular Senescence , DNA Damage , Doxorubicin/pharmacology , Histocompatibility Antigens Class I/genetics , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Matrix Metalloproteinase Inhibitors/pharmacology , Melphalan/pharmacology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Proteins/genetics , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Plasma Cells/immunology , Plasma Cells/pathology , Primary Cell Culture , Proteolysis , Reactive Oxygen Species/immunology , Signal Transduction , Syndecan-1/genetics , Syndecan-1/immunology
14.
J Immunol ; 193(2): 950-60, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24913980

ABSTRACT

Increasing evidence indicates that cancer cell stress induced by chemotherapeutic agents promote antitumor immune responses and contribute to their full clinical efficacy. In this article, we identify the signaling events underlying chemotherapy-induced NKG2D and DNAM-1 ligand expression on multiple myeloma (MM) cells. Our findings indicate that sublethal doses of doxorubicin and melphalan initiate a DNA damage response (DDR) controlling ligand upregulation on MM cell lines and patient-derived malignant plasma cells in Chk1/2-dependent and p53-independent manner. Drug-induced MICA and PVR gene expression are transcriptionally regulated and involve DDR-dependent E2F1 transcription factor activity. We also describe the involvement of changes in the redox state in the control of DDR-dependent upregulation of ligand surface expression and gene transcriptional activity by using the antioxidant agent N-acetyl-L-cysteine. Finally, in accordance with much evidence indicating that DDR and oxidative stress are major determinants of cellular senescence, we found that redox-dependent DDR activation upon chemotherapeutic treatment is critical for MM cell entry in premature senescence and is required for the preferential ligand upregulation on senescent cells, which are preferentially killed by NK cells and trigger potent IFN-γ production. We propose immunogenic senescence as a mechanism that promotes the clearance of drug-treated tumor cells by innate effector lymphocytes, including NK cells.


Subject(s)
DNA Damage , E2F1 Transcription Factor/immunology , Killer Cells, Natural/immunology , Reactive Oxygen Species/immunology , Adult , Aged , Aged, 80 and over , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Doxorubicin/pharmacology , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Ligands , Lymphocyte Activation/immunology , Male , Melphalan/pharmacology , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , NK Cell Lectin-Like Receptor Subfamily K/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Reactive Oxygen Species/metabolism , Receptors, Virus/genetics , Receptors, Virus/immunology , Receptors, Virus/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , Up-Regulation/immunology
15.
BMC Cancer ; 15: 17, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25609078

ABSTRACT

BACKGROUND: DNAX accessory molecule-1 (DNAM-1) is an activating receptor constitutively expressed by macrophages/dendritic cells and by T lymphocytes and Natural Killer (NK) cells, having an important role in anticancer responses; in this regard, combination therapies able to enhance the expression of DNAM-1 ligands on tumor cells are of therapeutic interest. In this study, we investigated the effect of different nitric oxide (NO) donors on the expression of the DNAM-1 ligand Poliovirus Receptor/CD155 (PVR/CD155) in multiple myeloma (MM) cells. METHODS: Six MM cell lines, SKO-007(J3), U266, OPM-2, RPMI-8226, ARK and LP1 were used to investigate the activity of different nitric oxide donors [DETA-NO and the NO-releasing prodrugs NCX4040 (NO-aspirin) and JS-K] on the expression of PVR/CD155, using Flow Cytometry and Real-Time PCR. Western-blot and specific inhibitors were employed to investigate the role of soluble guanylyl cyclase/cGMP and activation of the DNA damage response (DDR). RESULTS: Our results indicate that increased levels of nitric oxide can upregulate PVR/CD155 cell surface and mRNA expression in MM cells; in addition, exposure to nitric oxide donors renders myeloma cells more efficient to activate NK cell degranulation and enhances their ability to trigger NK cell-mediated cytotoxicity. We found that activation of the soluble guanylyl cyclase and increased cGMP concentrations by nitric oxide is not involved in the up-regulation of ligand expression. On the contrary, treatment of MM cells with nitric oxide donors correlated with the activation of a DNA damage response pathway and inhibition of the ATM /ATR/Chk1/2 kinase activities by specific inhibitors significantly abrogates up-regulation. CONCLUSIONS: The present study provides evidence that regulation of the PVR/CD155 DNAM-1 ligand expression by nitric oxide may represent an additional immune-mediated mechanism and supports the anti-myeloma activity of nitric oxide donors.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/biosynthesis , Multiple Myeloma/metabolism , Nitric Oxide/metabolism , Receptors, Virus/biosynthesis , Antigens, Differentiation, T-Lymphocyte/genetics , Aspirin/administration & dosage , Aspirin/analogs & derivatives , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Nitro Compounds/administration & dosage , Receptors, Virus/genetics , Triazenes/administration & dosage
16.
J Immunol ; 190(12): 6662-72, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23686482

ABSTRACT

Engagement of NKG2D and DNAX accessory molecule-1 (DNAM-1) receptors on lymphocytes plays an important role for anticancer response and represents an interesting therapeutic target for pharmacological modulation. In this study, we investigated the effect of inhibitors targeting the glycogen synthase kinase-3 (GSK3) on the expression of NKG2D and DNAM-1 ligands in multiple myeloma (MM) cells. GSK3 is a pleiotropic serine-threonine kinase point of convergence of numerous cell-signaling pathways, able to regulate the proliferation and survival of cancer cells, including MM. We found that inhibition of GSK3 upregulates both MICA protein surface and mRNA expression in MM cells, with little or no effects on the basal expression of the MICB and DNAM-1 ligand poliovirus receptor/CD155. Moreover, exposure to GSK3 inhibitors renders myeloma cells more efficient to activate NK cell degranulation and to enhance the ability of myeloma cells to trigger NK cell-mediated cytotoxicity. We could exclude that increased expression of ß-catenin or activation of the heat shock factor-1 (transcription factors inhibited by active GSK3) is involved in the upregulation of MICA expression, by using RNA interference or viral transduction of constitutive active forms. On the contrary, inhibition of GSK3 correlated with a downregulation of STAT3 activation, a negative regulator of MICA transcription. Both Tyr(705) phosphorylation and binding of STAT3 on MICA promoter are reduced by GSK3 inhibitors; in addition, overexpression of a constitutively active form of STAT3 significantly inhibits MICA upregulation. Thus, we provide evidence that regulation of the NKG2D-ligand MICA expression may represent an additional immune-mediated mechanism supporting the antimyeloma activity of GSK3 inhibitors.


Subject(s)
Glycogen Synthase Kinase 3/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Multiple Myeloma/immunology , STAT3 Transcription Factor/immunology , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Cytotoxicity, Immunologic/immunology , Flow Cytometry , Fluorescent Antibody Technique , Glycogen Synthase Kinase 3/antagonists & inhibitors , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Multiple Myeloma/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Transduction, Genetic , Transfection
17.
J Infect Dis ; 210(7): 1083-9, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24737799

ABSTRACT

BACKGROUND: Natural killer (NK) cells provide a major defense against cytomegalovirus (CMV) infection through the interaction of their surface receptors, including the activating and inhibitory killer immunoglobulin-like receptors (KIRs), and human leukocyte antigens (HLA) class I molecules. This study assessed whether the KIR and HLA repertoire may influence the risk of developing symptomatic or asymptomatic disease after primary CMV infection in the immunocompetent host. METHODS: Sixty immunocompetent patients with primary symptomatic CMV infection were genotyped for KIR and their HLA ligands, along with 60 subjects with a previous asymptomatic infection as controls. RESULTS: The frequency of the homozygous A haplotype (only KIR2DS4 as activating KIR) was higher in symptomatic patients than controls (30% vs 12%, respectively; odds ratio [OR] = 3.24; P = .01). By logistic regression, the risk of developing symptomatic disease was associated with the homozygous A haplotype and the HLABw4(T) allele. Combining the 2 independent variables, we found that 37 out of 60 (62%) symptomatic patients but only 18 out of 60 (30%) of controls possessed the homozygous A haplotype or the HLABw4(T) allele with a highly significant OR (OR = 3.75, P < .0005). CONCLUSIONS: Immunocompetent subjects carrying the homozygous A haplotype or the HLABw4(T) allele are at higher risk of developing symptomatic disease after primary CMV infection.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus/immunology , Genetic Predisposition to Disease , Histocompatibility Antigens Class I/metabolism , Receptors, KIR/metabolism , Adolescent , Adult , Aged , Female , Gene Frequency , Genotype , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Male , Middle Aged , Receptors, KIR/genetics , Receptors, KIR/immunology , Young Adult
18.
iScience ; 27(5): 109814, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38746669

ABSTRACT

2'3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) is the endogenous agonist of STING; as such, cGAMP has powerful immunostimulatory activity, due to its capacity to stimulate type I interferon-mediated immunity. Recent evidence indicates that cancer cells, under certain conditions, can release cGAMP extracellularly, a phenomenon currently considered important for therapeutic responses and tumor rejection. Nonetheless, the mechanisms that regulate cGAMP activity in the extracellular environment are still largely unexplored. In this work, we collected evidence demonstrating that CD38 glycohydrolase can inhibit extracellular cGAMP activity through its direct binding. We firstly used different cell lines and clinical samples to demonstrate a link between CD38 and extracellular cGAMP activity; we then performed extensive in silico molecular modeling and cell-free biochemical assays to show a direct interaction between the catalytic pocket of CD38 and cGAMP. Altogether, our findings expand the current knowledge about the regulation of cGAMP activity.

19.
Eur J Immunol ; 42(5): 1129-39, 2012 May.
Article in English | MEDLINE | ID: mdl-22539288

ABSTRACT

CD127 is the IL-7 receptor α-chain and its expression is tightly regulated during T-cell differentiation. We previously showed that the bone marrow (BM) is a key organ for proliferation and maintenance of both antigen-specific and CD44(high) memory CD8(+) T cells. Interestingly, BM memory CD8(+) T cells express lower levels of membrane CD127 than do the corresponding spleen and lymph node cells. We investigated the requirements for CD127 downmodulation by CD44(high) memory-phenotype CD8(+) T cells in the BM of C57BL/6 mice. By comparing genetically modified (i.e. CD127tg, IL-7 KO, IL-15 KO, IL-15Rα KO) with wild-type (WT) mice, we found that the key molecule regulating CD127 downmodulation was IL-15 but not IL-7, and that the intact CD127 gene was required, including the promoter. Indeed, CD127 mRNA transcript levels were lower in CD44(high) CD8(+) T cells from the BM than in those from the spleen of WT mice, indicating organ-specific regulation. Although levels of the CD127 transactivator Foxo1 were low in BM CD44(high) CD8(+) T cells, Foxo1 was not involved in IL-15-induced CD127 downmodulation. Thus, recirculating CD44(high) CD8(+) T cells passing through the BM transiently downregulate CD127 in response to IL-15, with implications for human therapies acting on the IL-7/CD127 axis, for example cytokine treatments in cancer patients.


Subject(s)
Bone Marrow/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interleukin-15/immunology , Receptors, Interleukin-7/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes/metabolism , Down-Regulation/immunology , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/analysis , Hyaluronan Receptors/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Spleen/immunology
20.
Front Cell Dev Biol ; 11: 1213114, 2023.
Article in English | MEDLINE | ID: mdl-37313439

ABSTRACT

SUMOylation is a reversible modification that involves the covalent attachment of small ubiquitin-like modifier (SUMO) to target proteins, leading to changes in their localization, function, stability, and interactor profile. SUMOylation and additional related post-translational modifications have emerged as important modulators of various biological processes, including regulation of genomic stability and immune responses. Natural killer (NK) cells are innate immune cells that play a critical role in host defense against viral infections and tumors. NK cells can recognize and kill infected or transformed cells without prior sensitization, and their activity is tightly regulated by a balance of activating and inhibitory receptors. Expression of NK cell receptors as well as of their specific ligands on target cells is finely regulated during malignant transformation through the integration of different mechanisms including ubiquitin- and ubiquitin-like post-translational modifications. Our review summarizes the role of SUMOylation and other related pathways in the biology of NK cells with a special emphasis on the regulation of their response against cancer. The development of novel selective inhibitors as useful tools to potentiate NK-cell mediated killing of tumor cells is also briefly discussed.

SELECTION OF CITATIONS
SEARCH DETAIL