Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.130
1.
J Transl Med ; 22(1): 274, 2024 Mar 13.
Article En | MEDLINE | ID: mdl-38475814

BACKGROUND: Chimeric antigen receptor natural killer (CAR-NK) cells represent a promising advancement in CAR cell therapy, addressing limitations observed in CAR-T cell therapy. However, our prior study revealed challenges in CAR-NK cells targeting CD19 antigens, as they failed to eliminate CD19+ Raji cells in NSG tumor-bearing mice, noting down-regulation or loss of CD19 antigen expression in some Raji cells. In response, this study aims to enhance CD19 CAR-NK cell efficacy and mitigate the risk of tumor recurrence due to target antigen escape by developing CD19 and CD20 (CD19/CD20) dual-targeted CAR-NK cells. METHODS: Initially, mRNA encoding anti-CD19 CARs (FMC63 scFv-CD8α-4-1BB-CD3ζ) and anti-CD20 CARs (LEU16 scFv-CD8α-4-1BB-CD3ζ) was constructed via in vitro transcription. Subsequently, CD19/CD20 dual-targeted CAR-NK cells were generated through simultaneous electrotransfection of CD19/CD20 CAR mRNA into umbilical cord blood-derived NK cells (UCB-NK). RESULTS: Following co-electroporation, the percentage of dual-CAR expression on NK cells was 86.4% ± 1.83%, as determined by flow cytometry. CAR expression was detectable at 8 h post-electric transfer, peaked at 24 h, and remained detectable at 96 h. CD19/CD20 dual-targeted CAR-NK cells exhibited increased specific cytotoxicity against acute lymphoblastic leukemia (ALL) cell lines (BALL-1: CD19+CD20+, REH: CD19+CD20-, Jurkat: CD19-CD20-) compared to UCB-NK, CD19 CAR-NK, and CD20 CAR-NK cells. Moreover, CD19/CD20 dual-targeted CAR-NK cells released elevated levels of perforin, IFN-γ, and IL-15. Multiple activation markers such as CD69 and cytotoxic substances were highly expressed. CONCLUSIONS: The creation of CD19/CD20 dual-targeted CAR-NK cells addressed the risk of tumor escape due to antigen heterogeneity in ALL, offering efficient and safe 'off-the-shelf' cell products. These cells demonstrate efficacy in targeting CD20 and/or CD19 antigens in ALL, laying an experimental foundation for their application in ALL treatment.


Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Mice , Animals , Receptors, Chimeric Antigen/metabolism , Antigens, CD19/genetics , Antigens, CD19/metabolism , Cytotoxicity, Immunologic/genetics , Cell Line, Tumor , Killer Cells, Natural , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/metabolism
2.
Immunity ; 56(12): 2816-2835.e13, 2023 Dec 12.
Article En | MEDLINE | ID: mdl-38091953

Cancer cells can evade natural killer (NK) cell activity, thereby limiting anti-tumor immunity. To reveal genetic determinants of susceptibility to NK cell activity, we examined interacting NK cells and blood cancer cells using single-cell and genome-scale functional genomics screens. Interaction of NK and cancer cells induced distinct activation and type I interferon (IFN) states in both cell types depending on the cancer cell lineage and molecular phenotype, ranging from more sensitive myeloid to less sensitive B-lymphoid cancers. CRISPR screens in cancer cells uncovered genes regulating sensitivity and resistance to NK cell-mediated killing, including adhesion-related glycoproteins, protein fucosylation genes, and transcriptional regulators, in addition to confirming the importance of antigen presentation and death receptor signaling pathways. CRISPR screens with a single-cell transcriptomic readout provided insight into underlying mechanisms, including regulation of IFN-γ signaling in cancer cells and NK cell activation states. Our findings highlight the diversity of mechanisms influencing NK cell susceptibility across different cancers and provide a resource for NK cell-based therapies.


Hematologic Neoplasms , Neoplasms , Humans , Killer Cells, Natural , Neoplasms/genetics , Antigen Presentation , Genomics , Cytotoxicity, Immunologic/genetics , Cell Line, Tumor
3.
Int J Mol Sci ; 23(19)2022 Sep 27.
Article En | MEDLINE | ID: mdl-36232695

Bone Marrow Stromal Cell Antigen 2 (BST2) is a type II transmembrane protein expressed on various cell types that tethers the release of viruses. Natural killer (NK) cells express low levels of BST2 under normal conditions but exhibit increased expression of BST2 upon activation. In this study, we show for the first time that murine BST2 can control the cytotoxicity of NK cells. The cytoplasmic tail of murine BST2 contains an immunoreceptor tyrosine-based inhibitory motif (ITIM). The absence of BST2 on NK cells can enhance their cytotoxicity against tumor cells compared to wild type NK cells. NK cells isolated from NZW mice, which express ITIM-deficient BST2, also showed higher cytotoxicity than wild type NK cells. In addition, we found that galectin-8 and galectin-9 were ligands of BST2, since blocking galectin-8 or -9 with monoclonal antibodies enhanced the cytotoxicity of NK cells. These results suggested that BST2 might be a novel NK cell inhibitory receptor as it was involved in regulating NK cell cytotoxicity through its interaction with galectins.


Bone Marrow Stromal Antigen 2 , Cytotoxicity, Immunologic , Killer Cells, Natural , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Bone Marrow Stromal Antigen 2/genetics , Bone Marrow Stromal Antigen 2/immunology , Carrier Proteins/immunology , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Galectins/immunology , Killer Cells, Natural/immunology , Ligands , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Tyrosine/metabolism
4.
Front Immunol ; 13: 941880, 2022.
Article En | MEDLINE | ID: mdl-36072599

Eliminating major xenoantigens in pig cells has drastically reduced human antibody-mediated hyperacute xenograft rejection (HXR). Despite these advancements, acute xenograft rejection (AXR) remains one of the major obstacles to clinical xenotransplantation, mediated by innate immune cells, including macrophages, neutrophils, and natural killer (NK) cells. NK cells play an 'effector' role by releasing cytotoxicity granules against xenogeneic cells and an 'affecter' role on other immune cells through cytokine secretion. We highlight the key receptor-ligand interactions that determine the NK cell response to target cells, focusing on the regulation of NK cell activating receptor (NKG2D, DNAM1) and inhibitory receptor (KIR2DL1-4, NKG2A, and LIR-1) signaling pathways. Inhibition of NK cell activity may protect xenografts from cytotoxicity. Recent successful approaches to reducing NK cell-mediated HXR and AXR are reviewed, including genetic modifications of porcine xenografts aimed at improving pig-to-human compatibility. Future directions to promote xenograft acceptance are discussed, including NK cell tolerance in pregnancy and NK cell evasion in viral infection.


Cytotoxicity, Immunologic , Killer Cells, Natural , Animals , Cytotoxicity, Immunologic/genetics , Humans , Immune Tolerance , Receptors, Natural Killer Cell/metabolism , Swine , Transplantation, Heterologous
5.
Proc Natl Acad Sci U S A ; 119(33): e2208522119, 2022 08 16.
Article En | MEDLINE | ID: mdl-35939714

Apoptosis is a genetically regulated program of cell death that plays a key role in immune disease processes. We identified EBF4, a little-studied member of the early B cell factor (EBF) family of transcription factors, in a whole-genome CRISPR screen for regulators of Fas/APO-1/CD95-mediated T cell death. Loss of EBF4 increases the half-life of the c-FLIP protein, and its presence in the Fas signaling complex impairs caspase-8 cleavage and apoptosis. Transcriptome analysis revealed that EBF4 regulates molecules such as TBX21, EOMES, granzyme, and perforin that are important for human natural killer (NK) and CD8+ T cell functions. Proximity-dependent biotin identification (Bio-ID) mass spectrometry analyses showed EBF4 binding to STAT3, STAT5, and MAP kinase 3 and a strong pathway relationship to interleukin-2 regulated genes, which are known to govern cytotoxicity pathways. Chromatin immunoprecipitation and DNA sequencing analysis defined a canonical EBF4 binding motif, 5'-CCCNNGG/AG-3', closely related to the EBF1 binding site; using a luciferase-based reporter, we found a dose-dependent transcriptional response of this motif to EBF4. We also conducted assay for transposase-accessible chromatin sequencing in EBF4-overexpressing cells and found increased chromatin accessibility upstream of granzyme and perforin and in topologically associated domains in human lymphocytes. Finally, we discovered that the EBF4 has basal expression in human but not mouse NK cells and CD8+ T cells and vanishes following activating stimulation. Together, our data reveal key features of a previously unknown transcriptional regulator of human cytotoxic immune function.


Apoptosis , CD8-Positive T-Lymphocytes , Cytotoxicity, Immunologic , Fas Ligand Protein , T-Lymphocytes, Cytotoxic , Transcription Factors , Animals , Apoptosis/physiology , Chromatin/metabolism , Cytotoxicity, Immunologic/genetics , Fas Ligand Protein/metabolism , Granzymes/genetics , Humans , Mice , Perforin/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
ACS Nano ; 16(8): 12695-12710, 2022 08 23.
Article En | MEDLINE | ID: mdl-35939651

Fas ligand (FasL), expressed on the surface of activated cytotoxic T lymphocytes (CTLs), is the physiological ligand for the cell surface death receptor, Fas. The Fas-FasL engagement initiates diverse signaling pathways, including the extrinsic cell death signaling pathway, which is one of the effector mechanisms that CTLs use to kill tumor cells. Emerging clinical and experimental data indicate that Fas is essential for the efficacy of CAR-T cell immunotherapy. Furthermore, loss of Fas expression is a hallmark of human melanoma. We hypothesize that restoring Fas expression in tumor cells reverses human melanoma resistance to T cell cytotoxicity. DNA hypermethylation, at the FAS promoter, down-regulates FAS expression and confers melanoma cell resistance to FasL-induced cell death. Forced expression of Fas in tumor cells overcomes melanoma resistance to FasL-induced cell death in vitro. Lipid nanoparticle-encapsulated mouse Fas-encoding plasmid therapy eliminates Fas+ tumor cells and suppresses established melanoma growth in immune-competent syngeneic mice. Similarly, lipid nanoparticle-encapsulated human FAS-encoding plasmid (hCOFAS01) therapy significantly increases Fas protein levels on tumor cells of human melanoma patient-derived xenograft (PDX) and suppresses the established human melanoma PDX growth in humanized NSG mice. In human melanoma patients, FasL is expressed in activated and exhausted T cells, Fas mRNA level positively correlates with melanoma patient survival, and nivolumab immunotherapy increases FAS expression in tumor cells. Our data demonstrate that hCOFAS01 is an effective immunotherapeutic agent for human melanoma therapy with dual efficacy in increasing tumor cell FAS expression and in enhancing CTL tumor infiltration.


Melanoma , fas Receptor , Humans , Mice , Animals , fas Receptor/genetics , fas Receptor/metabolism , Cytotoxicity, Immunologic/genetics , Tumor Cells, Cultured , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , T-Lymphocytes, Cytotoxic , Melanoma/pathology , Plasmids/genetics , Apoptosis
7.
Diabetes ; 71(3): 566-577, 2022 03 01.
Article En | MEDLINE | ID: mdl-35007320

Type 1 diabetes in children is heralded by a preclinical phase defined by circulating autoantibodies to pancreatic islet antigens. How islet autoimmunity is initiated and then progresses to clinical diabetes remains poorly understood. Only one study has reported gene expression in specific immune cells of children at risk associated with progression to islet autoimmunity. We analyzed gene expression with RNA sequencing in CD4+ and CD8+ T cells, natural killer (NK) cells, and B cells, and chromatin accessibility by assay for transposase-accessible chromatin sequencing (ATAC-seq) in CD4+ T cells, in five genetically at risk children with islet autoantibodies who progressed to diabetes over a median of 3 years ("progressors") compared with five children matched for sex, age, and HLA-DR who had not progressed ("nonprogressors"). In progressors, differentially expressed genes (DEGs) were largely confined to CD4+ T cells and enriched for cytotoxicity-related genes/pathways. Several top-ranked DEGs were validated in a semi-independent cohort of 13 progressors and 11 nonprogressors. Flow cytometry confirmed that progression was associated with expansion of CD4+ cells with a cytotoxic phenotype. By ATAC-seq, progression was associated with reconfiguration of regulatory chromatin regions in CD4+ cells, some linked to differentially expressed cytotoxicity-related genes. Our findings suggest that cytotoxic CD4+ T cells play a role in promoting progression to type 1 diabetes.


CD4-Positive T-Lymphocytes/metabolism , Chromatin/chemistry , Cytotoxicity, Immunologic/genetics , Diabetes Mellitus, Type 1/immunology , Disease Progression , Gene Expression Regulation , Adolescent , Autoimmunity/genetics , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , CD8-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Humans , Islets of Langerhans/immunology , Killer Cells, Natural/metabolism , Sequence Analysis, RNA
8.
Cancer Immunol Immunother ; 71(3): 613-625, 2022 Mar.
Article En | MEDLINE | ID: mdl-34282497

BACKGROUND: Natural killer (NK) cell-based immunotherapy is a promising treatment approach for multiple myeloma (MM), but obtaining a sufficient number of activated NK cells remains challenging. Here, we report an improved method to generate ex vivo expanded NK (eNK) cells from MM patients based on genetic engineering of K562 cells to express OX40 ligand and membrane-bound (mb) IL-18 and IL-21. METHODS: K562-OX40L-mbIL-18/-21 cells were generated by transducing K562-OX40L cells with a lentiviral vector encoding mbIL-18 and mbIL-21, and these were used as feeder cells to expand NK cells from peripheral blood mononuclear cells of healthy donors (HDs) and MM patients in the presence of IL-2/IL-15. Purity, expansion rate, receptor expression, and functions of eNK cells were determined over four weeks of culture. RESULTS: NK cell expansion was enhanced by short exposure of soluble IL-18 and IL-21 with K562-OX40L cells. Co-culture of NK cells with K562-OX40L-mbIL-18/-21 cells resulted in remarkable expansion of NK cells from HDs (9,860-fold) and MM patients (4,929-fold) over the 28-day culture period. Moreover, eNK cells showed increased expression of major activation markers and enhanced cytotoxicity towards target K562, U266, and RPMI8226 cells. CONCLUSIONS: Our data suggest that genetically engineered K562 cells expressing OX40L, mbIL-18, and mbIL-21 improve the expansion of NK cells, increase activation signals, and enhance their cytolytic activity towards MM cells.


Cytotoxicity, Immunologic , Interleukin-18/metabolism , Interleukins/metabolism , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Multiple Myeloma/immunology , OX40 Ligand/metabolism , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic/genetics , Gene Expression , Humans , Immunophenotyping , Interleukin-18/genetics , Interleukins/genetics , K562 Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , OX40 Ligand/genetics , Transduction, Genetic , Transgenes
9.
Front Immunol ; 12: 792609, 2021.
Article En | MEDLINE | ID: mdl-34880879

Background: We previously reported algorithms based on clinical parameters and plasma cell characteristics to identify patients with smoldering multiple myeloma (SMM) with higher risk of progressing who could benefit from early treatment. In this work, we analyzed differences in the immune bone marrow (BM) microenvironment in SMM to better understand the role of immune surveillance in disease progression and to identify immune biomarkers associated to higher risk of progression. Methods: Gene expression analysis of BM cells from 28 patients with SMM, 22 patients with monoclonal gammopathy of undetermined significance (MGUS) and 22 patients with symptomatic MM was performed by using Nanostring Technology. Results: BM cells in SMM compared to both MGUS and symptomatic MM showed upregulation of genes encoding for key molecules in cytotoxicity. However, some of these cytotoxic molecules positively correlated with inhibitory immune checkpoints, which may impair the effector function of BM cytotoxic cells. Analysis of 28 patients with SMM revealed 4 distinct clusters based on immune composition and activation markers. Patients in cluster 2 showed a significant increase in expression of cytotoxic molecules but also inhibitory immune checkpoints compared to cluster 3, suggesting the presence of cytotoxic cells with an exhausted phenotype. Accordingly, patients in cluster 3 had a significantly longer progression free survival. Finally, individual gene expression analysis showed that higher expression of TNF superfamily members (TNF, TNFAIP3, TNFRSF14) was associated with shorter progression free survival. Conclusions: Our results suggest that exhausted cytotoxic cells are associated to high-risk patients with SMM. Biomarkers overexpressed in patients with this immune gene profile in combination with clinical parameters and PC characterization may be useful to identify SMM patients with higher risk of progression.


Bone Marrow/physiology , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/immunology , Smoldering Multiple Myeloma/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinogenesis , Cellular Microenvironment , Cohort Studies , Cytotoxicity, Immunologic/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Immune Checkpoint Proteins/genetics , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/mortality , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Phenotype , Smoldering Multiple Myeloma/genetics , Smoldering Multiple Myeloma/mortality , Survival Analysis , Tumor Necrosis Factor-alpha/genetics
10.
Science ; 374(6565): eabe9977, 2021 Oct 15.
Article En | MEDLINE | ID: mdl-34648346

T cell receptor activation of naïve CD8+ T lymphocytes initiates their maturation into effector cytotoxic T lymphocytes (CTLs), which can kill cancer and virally infected cells. Although CTLs show an increased reliance on glycolysis upon acquisition of effector function, we found an essential requirement for mitochondria in target cell­killing. Acute mitochondrial depletion in USP30 (ubiquitin carboxyl-terminal hydrolase 30)­deficient CTLs markedly diminished killing capacity, although motility, signaling, and secretion were all intact. Unexpectedly, the mitochondrial requirement was linked to mitochondrial translation, inhibition of which impaired CTL killing. Impaired mitochondrial translation triggered attenuated cytosolic translation, precluded replenishment of secreted killing effectors, and reduced the capacity of CTLs to carry out sustained killing. Thus, mitochondria emerge as a previously unappreciated homeostatic regulator of protein translation required for serial CTL killing.


Cytotoxicity, Immunologic/immunology , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Thiolester Hydrolases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Movement/genetics , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondrial Proteins/genetics , Protein Biosynthesis , T-Lymphocytes, Cytotoxic/enzymology , Thiolester Hydrolases/genetics
11.
Nat Commun ; 12(1): 5405, 2021 09 13.
Article En | MEDLINE | ID: mdl-34518538

Tumor cells evade T cell-mediated immunosurveillance via the interaction between programmed death-1 (PD-1) ligand 1 (PD-L1) on tumor cells and PD-1 on T cells. Strategies disrupting PD-1/PD-L1 have shown clinical benefits in various cancers. However, the limited response rate prompts us to investigate the molecular regulation of PD-L1. Here, we identify trafficking protein particle complex subunit 4 (TRAPPC4), a major player in vesicular trafficking, as a crucial PD-L1 regulator. TRAPPC4 interacts with PD-L1 in recycling endosomes, acting as a scaffold between PD-L1 and RAB11, and promoting RAB11-mediated recycling of PD-L1, thus replenishing its distribution on the tumor cell surface. TRAPPC4 depletion leads to a significant reduction of PD-L1 expression in vivo and in vitro. This reduction in PD-L1 facilitates T cell-mediated cytotoxicity. Overexpression of Trappc4 sensitizes tumor cells to checkpoint therapy in murine tumor models, suggesting TRAPPC4 as a therapeutic target to enhance anti-tumor immunity.


B7-H1 Antigen/immunology , Colorectal Neoplasms/immunology , Gene Expression Regulation, Neoplastic/immunology , Nerve Tissue Proteins/immunology , Vesicular Transport Proteins/immunology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Endosomes/immunology , Endosomes/metabolism , HCT116 Cells , Humans , Intracellular Space/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Protein Transport , RNA Interference , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
12.
Immunohorizons ; 5(8): 687-702, 2021 08 25.
Article En | MEDLINE | ID: mdl-34433624

Tyrosine kinase inhibitor (TKI)-treated chronic myeloid leukemia (CML) patients with increased NK cell number have a better prognosis, and thus, NK cells may suppress CML. However, the efficacy of TKIs varies for reasons yet to be fully elucidated. As NK cell activity is modulated by interactions between their killer cell Ig-like receptors (KIRs) and HLAs of target cells, the combination of their polymorphisms may have functional significance. We previously showed that allelic polymorphisms of KIR3DL1 and HLAs were associated with the prognosis of TKI-treated CML patients. In this study, we focus on differential NK cell activity modulation through KIR3DL1 allotypes. KIR3DL1 expression levels varied according to their alleles. The combination of KIR3DL1 expression level and HLA-Bw4 motifs defined NK cell activity in response to the CML-derived K562 cell line, and Ab-mediated KIR3DL1 blocking reversed this activity. The TKI dasatinib enhanced NK cell activation and cytotoxicity in a KIR3DL1 allotype-dependent manner but did not significantly decrease effector regulatory T cells, suggesting that it directly activated NK cells. Dasatinib also enhanced NK cell cytotoxicity against K562 bearing the BCR-ABL1 T315I TKI resistance-conferring mutation, depending on KIR3DL1/HLA-Bw4 allotypes. Transduction of KIR3DL1*01502 into the NK cell line NK-92 resulted in KIR3DL1 expression and suppression of NK-92 activity by HLA-B ligation, which was reversed by anti-KIR3DL1 Ab. Finally, KIR3DL1 expression levels also defined activation patterns in CML patient-derived NK cells. Our findings raise the possibility of a novel strategy to enhance antitumor NK cell immunity against CML in a KIR3DL1 allotype-dependent manner.


Gene Expression Regulation, Leukemic/immunology , Killer Cells, Natural/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Receptors, KIR3DL1/immunology , Alleles , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Dasatinib/pharmacology , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/immunology , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , HLA-B Antigens/genetics , HLA-B Antigens/immunology , HLA-B Antigens/metabolism , Humans , K562 Cells , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mutation , Protein Kinase Inhibitors/pharmacology , Receptors, KIR3DL1/genetics , Receptors, KIR3DL1/metabolism
13.
Int J Mol Sci ; 22(16)2021 Aug 10.
Article En | MEDLINE | ID: mdl-34445292

The genes involved in implantation and placentation are tightly regulated to ensure a healthy pregnancy. The endoplasmic reticulum aminopeptidase 2 (ERAP2) gene is associated with preeclampsia (PE). Our studies have determined that an isoform of ERAP2-arginine (N), expressed in trophoblast cells (TC), significantly activates immune cells, and ERAP2N-expressing TCs are preferentially killed by both cytotoxic T lymphocytes (CTLs) and Natural Killer cells (NKCs). To understand the cause of this phenomenon, we surveyed differentially expressed genes (DEGs) between ERAP2N expressing and non-expressing TCs. Our RNAseq data revealed 581 total DEGs between the two groups. 289 genes were up-regulated, and 292 genes were down-regulated. Interestingly, most of the down-regulated genes of significance were pro-survival genes that play a crucial role in cell survival (LDHA, EGLN1, HLA-C, ITGB5, WNT7A, FN1). However, the down-regulation of these genes in ERAP2N-expressing TCs translates into a propensity for cell death. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that 64 DEGs were significantly enriched in nine pathways, including "Protein processing in endoplasmic reticulum" and "Antigen processing and presentation", suggesting that the genes may be associated with peptide processes involved in immune recognition during the reproductive cycle.


Aminopeptidases/genetics , Cell Death/genetics , Trophoblasts/metabolism , Amino Acid Substitution/genetics , Aminopeptidases/metabolism , Arginine/genetics , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Trophoblasts/pathology , Trophoblasts/physiology , Up-Regulation/genetics
14.
Nat Genet ; 53(8): 1196-1206, 2021 08.
Article En | MEDLINE | ID: mdl-34253920

To systematically define molecular features in human tumor cells that determine their degree of sensitivity to human allogeneic natural killer (NK) cells, we quantified the NK cell responsiveness of hundreds of molecularly annotated 'DNA-barcoded' solid tumor cell lines in multiplexed format and applied genome-scale CRISPR-based gene-editing screens in several solid tumor cell lines, to functionally interrogate which genes in tumor cells regulate the response to NK cells. In these orthogonal studies, NK cell-sensitive tumor cells tend to exhibit 'mesenchymal-like' transcriptional programs; high transcriptional signature for chromatin remodeling complexes; high levels of B7-H6 (NCR3LG1); and low levels of HLA-E/antigen presentation genes. Importantly, transcriptional signatures of NK cell-sensitive tumor cells correlate with immune checkpoint inhibitor (ICI) resistance in clinical samples. This study provides a comprehensive map of mechanisms regulating tumor cell responses to NK cells, with implications for future biomarker-driven applications of NK cell immunotherapies.


Cytotoxicity, Immunologic/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Immune Checkpoint Inhibitors/pharmacology , Killer Cells, Natural/physiology , Allogeneic Cells/physiology , Animals , B7 Antigens/genetics , Cell Line, Tumor , Chromatin Assembly and Disassembly/physiology , Cytotoxicity Tests, Immunologic/methods , Cytotoxicity, Immunologic/physiology , Drug Resistance, Neoplasm/drug effects , Female , Genome, Human , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Mice, Inbred NOD , Xenograft Model Antitumor Assays , HLA-E Antigens
15.
Eur J Immunol ; 51(10): 2513-2521, 2021 10.
Article En | MEDLINE | ID: mdl-34323289

Recent studies have indicated the antitumor activity and reduced allogeneic response of universal chimeric antigen receptor-modified T (UCAR T) cells lacking endogenous T cell receptors and beta-2 microglobulin (B2M) generated using gene-editing technologies. However, these cells are vulnerable to lysis by allogeneic natural killer (NK) cells due to their lack of human leukocyte antigen (HLA) class I molecule expression. Here, constitutive expression of mutant B2M-HLA-E (mBE) and B2M-HLA-G (mBG) fusion proteins in anti-CD19 UCAR T (UCAR T-19) cells was conducted to protect against allogeneic NK cell-mediated lysis. The ability of cells expressing mBE or mBG to resist NK cell-mediated lysis was observed in gene-edited Jurkat CAR19 cells. UCAR T-19 cells constitutively expressing the mBE and mBG fusion proteins were manufactured and showed effective and specific anti-tumor activity. Constitutive expression of the mBE and mBG fusion proteins in UCAR T-19 cells prevented allogeneic NK cell-mediated lysis. In addition, these cells were not recognizable by allogeneic T cells. Additional experiments, including those in animal models and clinical trials, are required to evaluate the safety and efficacy of UCAR T-19 cells that constitutively express mBE and mBG.


Cytotoxicity, Immunologic/genetics , HLA-G Antigens/genetics , Histocompatibility Antigens Class I/genetics , Mutation , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , beta 2-Microglobulin/genetics , Antigens, CD19/immunology , Gene Knockout Techniques , HLA-G Antigens/immunology , Histocompatibility Antigens Class I/immunology , Humans , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , beta 2-Microglobulin/immunology , HLA-E Antigens
16.
Cancer Res ; 81(19): 5074-5088, 2021 10 01.
Article En | MEDLINE | ID: mdl-34321242

Epstein-Barr virus (EBV) infection is an established cause of nasopharyngeal carcinoma (NPC) and is involved in a variety of malignant phenotypes, including tumor immune escape. EBV can encode a variety of circular RNAs (circRNA), however, little is known regarding the biological functions of these circRNAs in NPC. In this study, EBV-encoded circBART2.2 was found to be highly expressed in NPC where it upregulated PD-L1 expression and inhibited T-cell function in vitro and in vivo. circBART2.2 promoted transcription of PD-L1 by binding the helicase domain of RIG-I and activating transcription factors IRF3 and NF-κB, resulting in tumor immune escape. These results elucidate the biological function of circBART2.2, explain a novel mechanism of immune escape caused by EBV infection, and provide a new immunotherapy target for treating NPC. SIGNIFICANCE: This work demonstrates that circBART2.2 binding to RIG-I is essential for the regulation of PD-L1 and subsequent immune escape in nasopharyngeal carcinoma.


B7-H1 Antigen/genetics , Herpesvirus 4, Human/genetics , Nasopharyngeal Carcinoma/etiology , RNA, Circular/genetics , RNA, Viral/genetics , Tumor Escape/genetics , Animals , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Disease Models, Animal , Disease Susceptibility , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Neoplastic , Humans , Mice , NF-kappa B/metabolism , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Protein Binding , RNA Interference , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
17.
Cell Rep ; 36(3): 109401, 2021 07 20.
Article En | MEDLINE | ID: mdl-34289371

Respiratory syncytial virus (RSV) is a major cause of serious acute lower respiratory tract infection in infants and the elderly. The lack of a licensed RSV vaccine calls for the development of vaccines with other targets and vaccination strategies. Here, we construct a recombinant protein, designated P-KFD1, comprising RSV phosphoprotein (P) and the E.-coli-K12-strain-derived flagellin variant KFD1. Intranasal immunization with P-KFD1 inhibits RSV replication in the upper and lower respiratory tract and protects mice against lung disease without vaccine-enhanced disease (VED). The P-specific CD4+ T cells provoked by P-KFD1 intranasal (i.n.) immunization either reside in or migrate to the respiratory tract and mediate protection against RSV infection. Single-cell RNA sequencing (scRNA-seq) and carboxyfluorescein succinimidyl ester (CFSE)-labeled cell transfer further characterize the Th1 and Th17 responses induced by P-KFD1. Finally, we find that anti-viral protection depends on either interferon-γ (IFN-γ) or interleukin-17A (IL-17A). Collectively, P-KFD1 is a promising safe and effective mucosal vaccine candidate for the prevention of RSV infection.


Flagellin/genetics , Immunity, Mucosal/immunology , Mutation/genetics , Phosphoproteins/metabolism , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Clone Cells , Cytotoxicity, Immunologic/genetics , Female , Humans , Immunity , Immunization , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lung Diseases/pathology , Lung Diseases/virology , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Phosphorylation , Recombinant Proteins/immunology , Single-Cell Analysis , Th1 Cells/immunology , Th17 Cells/immunology
18.
Cell Oncol (Dordr) ; 44(5): 1051-1063, 2021 Oct.
Article En | MEDLINE | ID: mdl-34189679

PURPOSE: Glioblastoma, the most aggressive type of brain cancer, is composed of heterogeneous populations of differentiated cells, cancer stem cells and immune cells. Cystatin F, an endogenous inhibitor of lysosomal cysteine peptidases, regulates the function of cytotoxic immune cells. The aim of this study was to determine which type of cells expresses cystatin F in glioblastoma and to determine the role of cystatin F during disease progression. METHODS: RT-qPCR and immunohistochemistry were used to determine cystatin F mRNA and protein levels in glioblastoma tissue samples. The internalization of cystatin F was analyzed by Western blotting. Enzyme kinetics, real time invasion and calcein release cytotoxicity assays were used to assess the role of internalized cystatin F. RESULTS: We found that cystatin F was not expressed in non-cancer brain tissues, but that its expression increased with glioma progression. In tumor tissues, extensive staining was observed in cancer stem-like cells and microglia/monocytes, which secrete cystatin F into their microenvironment. In trans activity of cystatin F was confirmed using an in vitro glioblastoma cell model. Internalized cystatin F affected cathepsin L activity in glioblastoma cells and decreased their invasiveness. In addition, we found that cystatin F decreased the susceptibility of glioblastoma cells to the cytotoxic activity of natural killer (NK) cells. CONCLUSIONS: Our data implicate cystatin F as a mediator of immune suppression in glioblastoma. Increased cystatin F mRNA and protein levels in immune, glioblastoma and glioblastoma stem-like cells or trans internalized cystatin F may have an impact on decreased susceptibility of glioblastoma cells to NK cytotoxicity.


Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Cystatins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Killer Cells, Natural/metabolism , Neoplastic Stem Cells/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Line, Tumor , Cells, Cultured , Cystatins/metabolism , Cytotoxicity, Immunologic/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunohistochemistry , Microglia/metabolism , Monocytes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
19.
J Exp Med ; 218(8)2021 08 02.
Article En | MEDLINE | ID: mdl-34115115

Naturally occurring cases of monogenic type 1 diabetes (T1D) help establish direct mechanisms driving this complex autoimmune disease. A recently identified de novo germline gain-of-function (GOF) mutation in the transcriptional regulator STAT3 was found to cause neonatal T1D. We engineered a novel knock-in mouse incorporating this highly diabetogenic human STAT3 mutation (K392R) and found that these mice recapitulated the human autoimmune diabetes phenotype. Paired single-cell TCR and RNA sequencing revealed that STAT3-GOF drives proliferation and clonal expansion of effector CD8+ cells that resist terminal exhaustion. Single-cell ATAC-seq showed that these effector T cells are epigenetically distinct and have differential chromatin architecture induced by STAT3-GOF. Analysis of islet TCR clonotypes revealed a CD8+ cell reacting against known antigen IGRP, and STAT3-GOF in an IGRP-reactive TCR transgenic model demonstrated that STAT3-GOF intrinsic to CD8+ cells is sufficient to accelerate diabetes onset. Altogether, these findings reveal a diabetogenic CD8+ T cell response that is restrained in the presence of normal STAT3 activity and drives diabetes pathogenesis.


CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Immune Tolerance/genetics , Mutation/genetics , STAT3 Transcription Factor/genetics , Animals , Autoimmunity , Cell Proliferation , Chemotaxis/genetics , Cross-Priming/immunology , Cytotoxicity, Immunologic/genetics , Disease Models, Animal , Epigenesis, Genetic , Gain of Function Mutation , Heterozygote , Humans , Mice , Phenotype , Up-Regulation
20.
J Immunother Cancer ; 9(5)2021 05.
Article En | MEDLINE | ID: mdl-34006631

BACKGROUND: Autologous T cells engineered to express a chimeric antigen receptor (CAR) specific for CD19 molecule have transformed the therapeutic landscape in patients with highly refractory leukemia and lymphoma, and the use of donor-generated allogeneic CAR T is paving the way for further breakthroughs in the treatment of cancer. However, it remains unknown how the intrinsic heterogeneities of these engineered cells mediate therapeutic efficacy and whether allogeneic products match the effectiveness of autologous therapies. METHODS: Using single-cell mRNA sequencing in conjunction with CITE-seq, we performed multiomics characterization of CAR T cells generated from healthy donor and patients with acute lymphoblastic leukemia. CAR T cells used in this study were manufactured at the University of Pennsylvania through lentiviral transduction with a CD19-4-1BB-CD3ζ construct. Besides the baseline condition, we engineered NIH-3T3 cells with human CD19 or mesothelin expression to conduct ex vivo antigen-specific or non-antigen stimulation of CAR T cells through 6-hour coculture at a 1:1 ratio. RESULTS: We delineated the global cellular and molecular CAR T landscape and identified that transcriptional CAR tonic signaling was regulated by a mixture of early activation, exhaustion signatures, and cytotoxic activities. On CD19 stimulation, we illuminated the disparities of CAR T cells derived from different origins and found that donor CAR T had more pronounced activation level in correlation with the upregulation of major histocompatibility complex class II genes compared with patient CAR T cells. This finding was independently validated in additional datasets from literature. Furthermore, GM-CSF(CSF2) expression was found to be associated with functional gene productions, but it induced little impact on the CAR T activation. CONCLUSIONS: Through integrated multiomics profiling and unbiased canonical pathway analyses, our results unveil heterogeneities in the transcriptional, phenotypic, functional, and metabolic profiles of donor and patient CAR T cells, providing mechanistic basis for ameliorating clinical outcomes and developing next-generation 'off- the-shelf' allogeneic products.


Antigens, CD19/genetics , Gene Expression Profiling , Immunotherapy, Adoptive , Lymphocyte Activation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/genetics , Single-Cell Analysis , T-Lymphocytes/transplantation , Transcriptome , Animals , Antigens, CD19/immunology , Antigens, CD19/metabolism , Case-Control Studies , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic/genetics , Humans , Mice , NIH 3T3 Cells , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA-Seq , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
...