Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Heredity (Edinb) ; 129(1): 64-74, 2022 07.
Article in English | MEDLINE | ID: mdl-35459932

ABSTRACT

Cancer is a disease of the genome, therefore, its development has a clear Mendelian component, demonstrated by well-studied genes such as BRCA1 and BRCA2 in breast cancer risk. However, it is known that a single genetic variant is not enough for cancer to develop leading to the theory of multistage carcinogenesis. In many cases, it is a sequence of events, acquired somatic mutations, or simply polygenic components with strong epigenetic effects, such as in the case of brain tumours. The expression of many genes is the product of the complex interplay between several factors, including the organism's genotype (in most cases Mendelian-inherited), genetic instability, epigenetic factors (non-Mendelian-inherited) as well as the immune response of the host, to name just a few. In recent years the importance of the immune system has been elevated, especially in the light of the immune checkpoint genes discovery and the subsequent development of their inhibitors. As the expression of these genes normally suppresses self-immunoreactivity, their expression by tumour cells prevents the elimination of the tumour by the immune system. These discoveries led to the rapid growth of the field of immuno-oncology that offers new possibilities of long-lasting and effective treatment options. Here we discuss the recent advances in the understanding of the key mechanisms controlling the expression of immune checkpoint genes in tumour cells.


Subject(s)
Breast Neoplasms , Immunological Synapses , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Genotype , Humans , Immunological Synapses/pathology , Mutation
2.
J Immunol ; 204(10): 2818-2828, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32269094

ABSTRACT

CTLs release cytotoxic proteins such as granzymes and perforin through fusion of cytotoxic granules (CG) at the target cell interface, the immune synapse, to kill virus-infected and tumorigenic target cells. A characteristic feature of these granules is their acidic pH inside the granule lumen, which is required to process precursors of granzymes and perforin to their mature form. However, the role of acidic pH in CG maturation, transport, and fusion is not understood. We demonstrate in primary murine CTLs that the a3-subunit of the vacuolar-type (H+)-adenosine triphosphatase is required for establishing a luminal pH of 6.1 inside CG using ClopHensorN(Q69M), a newly generated CG-specific pH indicator. Knockdown of the a3-subunit resulted in a significantly reduced killing of target cells and a >50% reduction in CG fusion in total internal reflection fluorescence microscopy, which was caused by a reduced number of CG at the immune synapse. Superresolution microscopy revealed a reduced interaction of CG with the microtubule network upon a3-subunit knockdown. Finally, we find by electron and structured illumination microscopy that knockdown of the a3-subunit altered the diameter and density of individual CG, whereas the number of CG per CTL was unaffected. We conclude that the a3-subunit of the vacuolar adenosine triphosphatase is not only responsible for the acidification of CG, but also contributes to the maturation and efficient transport of the CG to the immune synapse.


Subject(s)
Immunological Synapses/metabolism , Microtubules/metabolism , Secretory Vesicles/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Cells, Cultured , Cytotoxicity, Immunologic , Exocytosis , Hydrogen-Ion Concentration , Immunological Synapses/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , R-SNARE Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , Vacuolar Proton-Translocating ATPases/genetics
3.
J Immunol ; 202(1): 300-308, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30510065

ABSTRACT

Advances in imaging have led to the development of powerful multispectral, quantitative imaging techniques, like histo-cytometry. The utility of this approach is limited, however, by the need for time consuming manual image analysis. We therefore developed the software Chrysalis and a group of Imaris Xtensions to automate this process. The resulting automation allowed for high-throughput histo-cytometry analysis of three-dimensional confocal microscopy and two-photon time-lapse images of T cell-dendritic cell interactions in mouse spleens. It was also applied to epi-fluorescence images to quantify T cell localization within splenic tissue by using a "signal absorption" strategy that avoids computationally intensive distance measurements. In summary, this image processing and analysis software makes histo-cytometry more useful for immunology applications by automating image analysis.


Subject(s)
Dendritic Cells/pathology , Image Processing, Computer-Assisted/methods , Immunological Synapses/pathology , Software , T-Lymphocytes/pathology , Animals , Automation, Laboratory , Cells, Cultured , Female , High-Throughput Screening Assays , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Spleen/pathology , Time-Lapse Imaging
4.
Immunity ; 35(2): 208-22, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21820331

ABSTRACT

The immunological synapse (IS) serves a dual role for sustained T cell receptor (TCR) signaling and for TCR downregulation. TC21 (Rras2) is a RRas subfamily GTPase that constitutively associates with the TCR and is implicated in tonic TCR signaling by activating phosphatidylinositol 3-kinase. In this study, we demonstrate that TC21 both cotranslocates with the TCR to the IS and is necessary for TCR internalization from the IS through a mechanism dependent on RhoG, a small GTPase previously associated with phagocytosis. Indeed, we found that the TCR triggers T cells to phagocytose 1-6 µm beads through a TC21- and RhoG-dependent pathway. We further show that TC21 and RhoG are necessary for the TCR-promoted uptake of major histocompatibility complex (MHC) from antigen-presenting cells. Therefore, TC21 and RhoG dependence underlie the existence of a common phagocytic mechanism that drives TCR internalization from the IS together with its peptide-MHC ligand.


Subject(s)
Immunological Synapses/metabolism , Membrane Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Phagocytosis , Receptors, Antigen, T-Cell/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Antigen Presentation , Antigens/metabolism , Cell Communication , Histocompatibility Antigens Class II , Humans , Immunological Synapses/pathology , Jurkat Cells , Membrane Proteins/immunology , Mice , Mice, Knockout , Mice, Transgenic , Monomeric GTP-Binding Proteins/immunology , Peptide Fragments/immunology , Phagocytosis/immunology , Protein Transport , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , rho GTP-Binding Proteins/immunology
5.
Am J Hematol ; 95(12): 1495-1502, 2020 12.
Article in English | MEDLINE | ID: mdl-32815561

ABSTRACT

Nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) is a subtype of Hodgkin lymphoma with a preserved B-cell phenotype and follicular T helper (TFH ) cells rosetting around the tumor cells, the lymphocyte-predominant (LP) cells. As we recently described reactivity of the B-cell receptors of LP cells of some NLPHL cases with Moraxella spp. proteins, we hypothesized that LP cells could present peptides to rosetting T cells in a major histocompatibility complex class II (MHCII)-bound manner. Rosetting PD1+ T cells were present in the majority of NLPHL cases, both in typical (17/20) and variant patterns (16/19). In most cases, T-cell rosettes were CD69+ (typical NLPHL, 17/20; NLPHL variant, 14/19). Furthermore, both MHCII alpha and beta chains were expressed in the LP cells in 23/39 NLPHL. Proximity ligation assay and confocal laser imaging demonstrated interaction of the MHCII beta chain expressed by the LP cells and the T-cell receptor alpha chain expressed by rosetting T cells. We thus conclude that rosetting T cells in NLPHL express markers that are encountered after antigenic exposure, that MHCII is expressed by the LP cells, and that LP cells interact with rosetting T cells in an immunological synapse in a subset of cases. As they likely receive growth stimulatory signals in this way, blockade of this interaction, for example, by PD1-directed checkpoint inhibitors, could be a treatment option in a subset of cases in the future.


Subject(s)
Antigens, Differentiation/immunology , B-Lymphocytes , Hodgkin Disease , Immunological Synapses , Moraxella/immunology , Moraxellaceae Infections , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Female , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Humans , Immunological Synapses/immunology , Immunological Synapses/pathology , Male , Moraxellaceae Infections/immunology , Moraxellaceae Infections/pathology , Retrospective Studies , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology
6.
Int J Mol Sci ; 20(18)2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31547237

ABSTRACT

Upon tumor antigen recognition, cytotoxic T lymphocytes (CTLs) and target cells form specialized supramolecular structures, called cytotoxic immunological synapses, which are required for polarized delivery of cytotoxic granules. In previous reports, we described the accumulation of connexin 43 (Cx43)-formed gap junctions (GJs) at natural killer (NK) cell-tumor cell cytotoxic immunological synapse. In this report, we demonstrate the functional role of Cx43-GJs at the cytotoxic immunological synapse established between CTLs and melanoma cells during cytotoxicity. Using confocal microscopy, we evaluated Cx43 polarization to the contact site between CTLs isolated from pMEL-1 mice and B16F10 melanoma cells. We knocked down Cx43 expression in B16F10 cells and evaluated its role in the formation of functional GJs and the cytotoxic activity of CTLs, by calcein transfer and granzyme B activity assays, respectively. We found that Cx43 localizes at CTL/B16F10 intercellular contact sites via an antigen-dependent process. We also found that pMEL-1 CTLs but not wild-type naïve CD8+ T cells established functional GJs with B16F10 cells. Interestingly, we observed that Cx43-GJs were required for an efficient granzyme B activity in target B16F10 cells. Using an HLA-A2-restricted/MART-1-specific CD8+ T-cell clone, we confirmed these observations in human cells. Our results suggest that Cx43-channels are relevant components of cytotoxic immunological synapses and potentiate CTL-mediated tumor cell killing.


Subject(s)
Connexin 43/immunology , Gap Junctions/immunology , Immunological Synapses/immunology , Melanoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Gap Junctions/pathology , Humans , Immunological Synapses/pathology , Melanoma/pathology , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/pathology
7.
J Immunol ; 192(11): 5179-91, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24790152

ABSTRACT

B cell activation is regulated through the interplay of the BCR with the inhibitory coreceptor FcγRIIB and the activating coreceptor CD19. Recent studies suggest that Ag-driven BCR microclusters are efficiently converted to a signaling active state on colocalization with CD19 microclusters. Using total internal reflection fluorescence microscopy-based, high-resolution, high-speed live-cell and molecule imaging approaches, we show that when co-ligated to the BCR, the FcγRIIB can inhibit B cell activation by blocking the colocalization of BCR and CD19 microclusters within the B cell immunological synapse. Remarkably, this inhibitory function of FcγRIIB is dependent not on its well-characterized ITIM-containing cytoplasmic domain, but its transmembrane domain. Indeed, human primary B cells from systemic lupus erythematosus patients homozygous for gene encoding the loss-of-function transmembrane domain mutant FcγRIIB-I232T fail to block the synaptic colocalization of the BCR with CD19, leading to dysregulated recruitment of downstream signaling molecule p-PI3K to membrane proximal signalosome. This inhibitory function of FcγRIIB in impairing the spatial-temporal colocalization of BCR and CD19 microclusters in the B cell immunological synapse may help explain the hyper-reactive features of systemic lupus erythematosus patient B cells in reported studies. These observations may also provide new targets for therapies for systemic autoimmune disease.


Subject(s)
Antigens, CD19/immunology , B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Receptors, Antigen, B-Cell/immunology , Receptors, IgG/immunology , Amino Acid Substitution , Animals , Antigens, CD19/genetics , B-Lymphocytes/pathology , Humans , Immunological Synapses/genetics , Immunological Synapses/immunology , Immunological Synapses/pathology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Mice , Mutation, Missense , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Protein Structure, Tertiary , Receptors, Antigen, B-Cell/genetics , Receptors, IgG/genetics
8.
J Immunol ; 189(2): 689-700, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22689882

ABSTRACT

EWI motif-containing protein 2 (EWI-2) is a member of the Ig superfamily that links tetraspanin-enriched microdomains to the actin cytoskeleton. We found that EWI-2 colocalizes with CD3 and CD81 at the central supramolecular activation cluster of the T cell immune synapse. Silencing of the endogenous expression or overexpression of a cytoplasmic truncated mutant of EWI-2 in T cells increases IL-2 secretion upon Ag stimulation. Mass spectrometry experiments of pull-downs with the C-term intracellular domain of EWI-2 revealed the specific association of EWI-2 with the actin-binding protein α-actinin; this association was regulated by PIP2. α-Actinin regulates the immune synapse formation and is required for efficient T cell activation. We extended these observations to virological synapses induced by HIV and found that silencing of either EWI-2 or α-actinin-4 increased cell infectivity. Our data suggest that the EWI-2-α-actinin complex is involved in the regulation of the actin cytoskeleton at T cell immune and virological synapses, providing a link between membrane microdomains and the formation of polarized membrane structures involved in T cell recognition.


Subject(s)
Actinin/metabolism , Antigens, CD/metabolism , HIV Infections/immunology , HIV Infections/metabolism , Immunological Synapses/metabolism , Immunological Synapses/virology , Membrane Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , Actinin/physiology , Amino Acid Sequence , Antigen Presentation/immunology , Antigens, CD/physiology , Cell Line, Transformed , Cytoskeleton/immunology , Cytoskeleton/pathology , Cytoskeleton/virology , HIV Infections/pathology , HIV-1/immunology , Humans , Immunological Synapses/pathology , Jurkat Cells , Lymphocyte Activation/immunology , Membrane Microdomains/immunology , Membrane Microdomains/pathology , Membrane Microdomains/virology , Membrane Proteins/physiology , Molecular Sequence Data , T-Lymphocyte Subsets/pathology , Tumor Cells, Cultured
9.
J Immunol ; 187(3): 1081-9, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21772035

ABSTRACT

NKT cells that express the semi-invariant TCR are innate-like lymphocytes whose functions are regulated by self and foreign glycolipid ligands presented by the Ag-presenting, MHC class I-like molecule CD1d. Activation of NKT cells in vivo results in rapid release of copious amounts of effector cytokines and chemokines with which they regulate innate and adaptive immune responses to pathogens, certain types of cancers, and self-antigens. The nature of CD1d-restricted ligands, the manner in which they are recognized, and the unique effector functions of NKT cells suggest an immunoregulatory role for this T cell subset. Their ability to respond fast and our ability to steer NKT cell cytokine response to altered lipid ligands make them an important target for vaccine design and immunotherapies against autoimmune diseases. This review summarizes our current understanding of CD1d-restricted ligand recognition by NKT cells and how these innate-like lymphocytes regulate inflammation.


Subject(s)
Antigen Presentation/immunology , Immunological Synapses/pathology , Inflammation Mediators/metabolism , Lipid Metabolism/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Animals , Galactosylceramides/metabolism , Glycolipids/metabolism , Humans , Immunological Synapses/microbiology , Immunological Synapses/parasitology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/physiology , Ligands , Natural Killer T-Cells/pathology
10.
Clin Dev Immunol ; 2013: 785317, 2013.
Article in English | MEDLINE | ID: mdl-23861693

ABSTRACT

Recently, immune edition has been recognized as a new hallmark of cancer. In this respect, some clinical trials in breast cancer have reported imppressive outcomes related to laboratory immune findings, especially in the neoadjuvant and metastatic setting. Infiltration by tumor infiltrating lymphocytes (TIL) and their subtypes, tumor-associated macrophages (TAM) and myeloid-derived suppressive cells (MDSC) seem bona fide prognostic and even predictive biomarkers, that will eventually be incorporated into diagnostic and therapeutic algorithms of breast cancer. In addition, the complex interaction of costimulatory and coinhibitory molecules on the immune synapse and the different signals that they may exert represent another exciting field to explore. In this review we try to summarize and elucidate these new concepts and knowledge from a translational perspective focusing on breast cancer, paying special attention to those aspects that might have more significance in clinical practice and could be useful to design successful therapeutic strategies in the future.


Subject(s)
Breast Neoplasms/immunology , Carcinoma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Myeloid Cells/immunology , Tumor Microenvironment/immunology , Biomarkers/analysis , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Carcinoma/diagnosis , Carcinoma/pathology , Female , Humans , Immune Tolerance , Immunological Synapses/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/pathology , Myeloid Cells/pathology , Prognosis
11.
Anal Biochem ; 423(2): 261-8, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22274538

ABSTRACT

There is growing interest in the development of novel single-chain bispecific antibodies for retargeting of immune effector T cells to tumor cells. Until today, functional fusion constructs consisting of a single-chain bispecific antibody and a fluorescent protein were not reported. Such molecules could be useful for an in vivo visualization of this retargeting process. Recently, we established two novel single-chain bispecific antibodies. One is capable of retargeting T cells to CD33, and the other is capable of retargeting T cells to the prostate stem cell antigen (PSCA). CD33 is an attractive immunotarget on the surface of tumor cells from patients with acute myeloid leukemia (AML). The PSCA is a potential target on prostate cancer cells. Flanking the reading frame encoding the green fluorescent protein (GFP) with a recently described novel helical linker element allowed us to establish novel single-chain bispecific fusion antibodies. These fluorescent fusion antibodies were useful to efficiently retarget T cells to the respective tumor cells and visualize the formation of immune synapses between effector and target cells.


Subject(s)
Antibodies, Bispecific/metabolism , Green Fluorescent Proteins/metabolism , Immunological Synapses/pathology , Microscopy, Confocal , T-Lymphocytes/metabolism , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , CD3 Complex/immunology , Cell Line, Tumor , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Immunological Synapses/metabolism , Prostate-Specific Antigen/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Sialic Acid Binding Ig-like Lectin 3 , T-Lymphocytes/immunology
12.
Proc Natl Acad Sci U S A ; 106(15): 6250-5, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19332800

ABSTRACT

Preclinical animal models have largely ignored the immune-suppressive mechanisms that are important in human cancers. The identification and use of such models should allow better predictions of successful human responses to immunotherapy. As a model for changes induced in nonmalignant cells by cancer, we examined T-cell function in the chronic lymphocytic leukemia (CLL) Emu-TCL1 transgenic mouse model. With development of leukemia, Emu-TCL1 transgenic mice developed functional T-cell defects and alteration of gene and protein expression closely resembling changes seen in CLL human patients. Furthermore, infusion of CLL cells into young Emu-TCL1 mice induced defects comparable to those seen in mice with developed leukemia, demonstrating a causal relationship between leukemia and the T-cell defects. Altered pathways involved genes regulating actin remodeling, and T cells exhibited dysfunctional immunological synapse formation and T-cell signaling, which was reversed by the immunomodulatory drug lenalidomide. These results further demonstrate the utility of this animal model of CLL and define a versatile model to investigate both the molecular mechanisms of cancer-induced immune suppression and immunotherapeutic repair strategies.


Subject(s)
Disease Models, Animal , Immunotherapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Proto-Oncogene Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Animals , Disease Progression , Gene Expression Profiling , Humans , Immunological Synapses/drug effects , Immunological Synapses/immunology , Immunological Synapses/pathology , Lenalidomide , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , Thalidomide/analogs & derivatives , Thalidomide/pharmacology
13.
Blood ; 114(18): 3909-16, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19710498

ABSTRACT

Understanding how the immune system in patients with cancer interacts with malignant cells is critical for the development of successful immunotherapeutic strategies. We studied peripheral blood from newly diagnosed patients with acute myeloid leukemia (AML) to assess the impact of this disease on the patients' T cells. The absolute number of peripheral blood T cells is increased in AML compared with healthy controls. An increase in the absolute number of CD3+56+ cells was also noted. Gene expression profiling on T cells from AML patients compared with healthy donors demonstrated global differences in transcription suggesting aberrant T-cell activation patterns. These gene expression changes differ from those observed in chronic lymphocytic leukemia (CLL), indicating the heterogeneous means by which different tumors evade the host immune response. However, in common with CLL, differentially regulated genes involved in actin cytoskeletal formation were identified, and therefore the ability of T cells from AML patients to form immunologic synapses was assessed. Although AML T cells could form conjugates with autologous blasts, their ability to form immune synapses and recruit phosphotyrosine signaling molecules to the synapse was significantly impaired. These findings identify T-cell dysfunction in AML that may contribute to the failure of a host immune response against leukemic blasts.


Subject(s)
Blast Crisis/immunology , Immunological Synapses/immunology , Leukemia, Myeloid, Acute/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Blast Crisis/blood , Blast Crisis/diagnosis , Blast Crisis/genetics , Blast Crisis/pathology , CD3 Complex , CD36 Antigens , Gene Expression Profiling , Gene Expression Regulation, Leukemic/immunology , Genotype , Humans , Immunological Synapses/genetics , Immunological Synapses/metabolism , Immunological Synapses/pathology , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Lymphocyte Count , Male , Middle Aged , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
14.
Front Immunol ; 11: 600012, 2020.
Article in English | MEDLINE | ID: mdl-33343572

ABSTRACT

IL4I1 is an immunoregulatory enzyme that inhibits CD8 T-cell proliferation in vitro and in the tumoral context. Here, we dissected the effect of IL4I1 on CD8 T-cell priming by studying the differentiation of a transgenic CD8 T-cell clone and the endogenous repertoire in a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection. Unexpectedly, we show that IL4I1 accelerates the expansion of functional effector CD8 T cells during the first several days after infection and increases the average affinity of the elicited repertoire, supporting more efficient LCMV clearance in WT mice than IL4I1-deficient mice. Conversely, IL4I1 restrains the differentiation of CD8 T-cells into long-lived memory precursors and favors the memory response to the most immunodominant peptides. IL4I1 expression does not affect the phenotype or antigen-presenting functions of dendritic cells (DCs), but directly reduces the stability of T-DC immune synapses in vitro, thus dampening T-cell activation. Overall, our results support a model in which IL4I1 increases the threshold of T-cell activation, indirectly promoting the priming of high-affinity clones while limiting memory T-cell differentiation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Immunologic Memory , L-Amino Acid Oxidase/immunology , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Acute Disease , Animals , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Immunological Synapses/genetics , Immunological Synapses/immunology , Immunological Synapses/pathology , L-Amino Acid Oxidase/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Knockout
15.
Sci Rep ; 9(1): 8756, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217484

ABSTRACT

We examined the potential value of the natural killer (NK) cell line; NK-92, as immunotherapy tool for breast cancer (BC) treatment and searched for biomarker(s) of sensitivity to NK-92-mediated cytotoxicity. The cytotoxic activity of NK-92 cells towards one breast precancerous and nine BC cell lines was analyzed using calcein-AM and degranulation assays. The molecules associated with NK-92-responsiveness were determined by differential gene expression analysis using RNA-sequencing and validated by RT-PCR, immunostaining and flow cytometry. NK-target interactions and immunological synapse formation were assessed by fluorescence microscopy. Potential biomarker expression was determined by IHC in 99 patient-derived BC tissues and 10 normal mammary epithelial tissues. Most (8/9) BC cell lines were resistant while only one BC and the precancerous cell lines were effectively killed by NK-92 lymphocytes. NK-92-sensitive target cells specifically expressed CD56, which ectopic expression in CD56-negative BC cells induced their sensitivity to NK-92-mediated killing, suggesting that CD56 is not only a biomarker of responsiveness but actively regulates NK function. CD56 adhesion molecules which are also expressed on NK cells accumulate at the immunological synapse enhancing NK-target interactions, cytotoxic granzyme B transfer from NK-92 to CD56-expressing target cells and induction of caspase 3 activation in targets. Interestingly, CD56 expression was found to be reduced in breast tumor tissues (36%) with strong inter- and intratumoral heterogeneity in comparison to normal breast tissues (80%). CD56 is a potential predictive biomarker for BC responsiveness to NK-92-cell based immunotherapy and loss of CD56 expression might be a mechanism of escape from NK-immunity.


Subject(s)
Breast Neoplasms/immunology , CD56 Antigen/immunology , Immunity, Cellular , Immunological Synapses/metabolism , Killer Cells, Natural/immunology , Neoplasm Proteins/immunology , Breast Neoplasms/pathology , Female , Humans , Immunological Synapses/pathology , Killer Cells, Natural/pathology , MCF-7 Cells
16.
Mol Immunol ; 105: 137-149, 2019 01.
Article in English | MEDLINE | ID: mdl-30508726

ABSTRACT

Natural Killer (NK) cells suppress tumor initiation and metastasis. Most carcinomas are heterogeneous mixtures of epithelial, mesenchymal and hybrid tumor cells, but the relationships of these phenotypes to NK susceptibility are understood incompletely. Grainyhead-like-2 (GRHL2) is a master programmer of the epithelial phenotype, that is obligatorily down-regulated during experimentally induced Epithelial-Mesenchymal Transition (EMT). Here, we utilize GRHL2 re-expression to discover unifying molecular mechanisms that link the epithelial phenotype with NK-sensitivity. GRHL2 enhanced the expression of ICAM-1, augmenting NK-target cell synaptogenesis and NK killing of target cells. The expression of multiple interferon response genes, including ICAM1, anti-correlated with EMT. We identified two novel GRHL2-interacting proteins, the histone methyltransferases KMT2C and KMT2D. Mesenchymal-epithelial transition, NK-sensitization and ICAM-1 expression were promoted by GRHL2-KMT2C/D interactions and by GRHL2 inhibition of p300, revealing novel and potentially targetable epigenetic mechanisms connecting the epithelial phenotype with target cell susceptibility to NK killing.


Subject(s)
DNA-Binding Proteins/immunology , Epigenesis, Genetic/immunology , Epithelial-Mesenchymal Transition/immunology , Immunity, Cellular , Killer Cells, Natural/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Transcription Factors/immunology , Cell Line, Tumor , Humans , Immunological Synapses/immunology , Immunological Synapses/pathology , Intercellular Adhesion Molecule-1/immunology , Killer Cells, Natural/pathology , Neoplasms/pathology , p300-CBP Transcription Factors/immunology
17.
Adv Immunol ; 144: 23-63, 2019.
Article in English | MEDLINE | ID: mdl-31699219

ABSTRACT

B cells are essential to the adaptive immune system for providing the humoral immunity against cohorts of pathogens. The presentation of antigen to the B cell receptor (BCR) leads to the initiation of B cell activation, which is a process sensitive to the stiffness features of the substrates presenting the antigens. Mechanosensing of the B cells, potentiated through BCR signaling and the adhesion molecules, efficiently regulates B cell activation, proliferation and subsequent antibody responses. Defects in sensing of the antigen-presenting substrates can lead to the activation of autoreactive B cells in autoimmune diseases. The use of high-resolution, high-speed live-cell imaging along with the sophisticated biophysical materials, has uncovered the mechanisms underlying the initiation of B cell activation within seconds of its engagement with the antigen presenting substrates. In this chapter, we reviewed studies that have contributed to uncover the molecular mechanisms of B cell mechanosensing during the initiation of B cell activation.


Subject(s)
Antibody Formation , Antigen Presentation , B-Lymphocytes/immunology , Mechanotransduction, Cellular/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Animals , Autoimmune Diseases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/immunology , Humans , Immunological Synapses/chemistry , Immunological Synapses/genetics , Immunological Synapses/pathology , Integrins/immunology , Molecular Motor Proteins/immunology , Neoplasms/immunology , Neoplasms/metabolism , Receptors, Antigen, B-Cell/metabolism
18.
J Hematol Oncol ; 11(1): 31, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29482595

ABSTRACT

The advent of immunotherapy, especially checkpoint inhibitor-based immunotherapy, has provided novel and powerful weapons against cancer. Because only a subset of cancer patients exhibit durable responses, further exploration of the mechanisms underlying the resistance to immunotherapy in the bulk of cancer patients is merited. Such efforts may help to identify which patients could benefit from immune checkpoint blockade. Given the existence of a great number of pathways by which cancer can escape immune surveillance, and the complexity of tumor-immune system interaction, development of various combination therapies, including those that combine with conventional therapies, would be necessary. In this review, we summarize the current understanding of the mechanisms by which resistance to checkpoint blockade immunotherapy occurs, and outline how actionable combination strategies may be derived to improve clinical outcomes for patients.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Animals , Humans , Immunity , Immunological Synapses/immunology , Immunological Synapses/pathology , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment
19.
Methods Mol Biol ; 1584: 545-557, 2017.
Article in English | MEDLINE | ID: mdl-28255725

ABSTRACT

T cells are the main cellular targets of the human immunodeficiency virus 1 (HIV-1). HIV-1 infection induces pleiotropic effects on the infected T cell that modify the T cell capacity to respond to antigen and facilitates virus replication. HIV-1 infection subverts the formation and function of the immunological synapse altering both actin cytoskeleton remodeling and intracellular vesicle traffic. We describe here our methods to unveil how HIV-1 and in particular its protein Nef modify vesicle traffic to the immunological synapse, perturbing the synapse activation capacity.


Subject(s)
HIV Antigens/immunology , HIV Infections/immunology , HIV-1/physiology , Immunological Synapses/immunology , Virus Replication/immunology , nef Gene Products, Human Immunodeficiency Virus/immunology , HIV Infections/pathology , Humans , Immunological Synapses/pathology , Jurkat Cells
20.
Methods Mol Biol ; 1584: 533-544, 2017.
Article in English | MEDLINE | ID: mdl-28255724

ABSTRACT

Aberrant immune synapse formation between antigen-presenting and immune effector cells is a central mediator of immune dysfunction and can be observed across several haematologic malignancies. Here, we describe the cell preparation, conjugation and immune synapse quantification of B and T cells obtained from patients with leukaemia and the adaptions required when using cells from murine models of disease.


Subject(s)
Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , Immunological Synapses/immunology , Leukemia/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/pathology , B-Lymphocytes/pathology , Cell Line, Tumor , Humans , Immunological Synapses/pathology , Leukemia/pathology , Mice , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL