Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(2): e0245917, 2021.
Article in English | MEDLINE | ID: mdl-33596227

ABSTRACT

Pre-clinical murine models are critical for translating drug candidates from the bench to the bedside. There is interest in better understanding how anti-human CD3 therapy works based on recent longitudinal studies of short-term administration. Although several models have been created in this pursuit, each have their own advantages and disadvantages in Type-1 diabetes. In this study, we report a murine genetic knock-in model which expresses both a murine and a humanized-CD3ε-exon, rendering it sensitive to manipulation with anti-human CD3. These huCD3εHET mice are viable and display no gross abnormalities. Specifically, thymocyte development and T cell peripheral homeostasis is unaffected. We tested immune functionality of these mice by immunizing them with T cell-dependent antigens and no differences in antibody titers compared to wild type mice were recorded. Finally, we performed a graft-vs-host disease model that is driven by effector T cell responses and observed a wasting disease upon transfer of huCD3εHET T cells. Our results show a viable humanized CD3 murine model that develops normally, is functionally engaged by anti-human CD3 and can instruct on pre-clinical tests of anti-human CD3 antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , CD3 Complex/genetics , CD3 Complex/immunology , Gene Knock-In Techniques , Animals , Humans , Mice , Mice, Inbred C57BL , Phenotype , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymocytes/cytology , Thymocytes/immunology
2.
Nature ; 585(7824): 277-282, 2020 09.
Article in English | MEDLINE | ID: mdl-32879489

ABSTRACT

Abnormal epigenetic patterns correlate with effector T cell malfunction in tumours1-4, but the cause of this link is unknown. Here we show that tumour cells disrupt methionine metabolism in CD8+ T cells, thereby lowering intracellular levels of methionine and the methyl donor S-adenosylmethionine (SAM) and resulting in loss of dimethylation at lysine 79 of histone H3 (H3K79me2). Loss of H3K79me2 led to low expression of STAT5 and impaired T cell immunity. Mechanistically, tumour cells avidly consumed methionine and outcompeted T cells for methionine by expressing high levels of the methionine transporter SLC43A2. Genetic and biochemical inhibition of tumour SLC43A2 restored H3K79me2 in T cells, thereby boosting spontaneous and checkpoint-induced tumour immunity. Moreover, methionine supplementation improved the expression of H3K79me2 and STAT5 in T cells, and this was accompanied by increased T cell immunity in tumour-bearing mice and patients with colon cancer. Clinically, tumour SLC43A2 correlated negatively with T cell histone methylation and functional gene signatures. Our results identify a mechanistic connection between methionine metabolism, histone patterns, and T cell immunity in the tumour microenvironment. Thus, cancer methionine consumption is an immune evasion mechanism, and targeting cancer methionine signalling may provide an immunotherapeutic approach.


Subject(s)
Amino Acid Transport System L/metabolism , CD8-Positive T-Lymphocytes/metabolism , Histones/metabolism , Methionine/metabolism , Methylation , Neoplasms/metabolism , Amino Acid Transport System L/deficiency , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Epigenesis, Genetic , Female , Histones/chemistry , Humans , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Receptors, Antigen, T-Cell/metabolism , STAT5 Transcription Factor/metabolism
3.
PLoS Pathog ; 15(1): e1007560, 2019 01.
Article in English | MEDLINE | ID: mdl-30682165

ABSTRACT

Bacterial lung infections, particularly with methicillin-resistant Staphylococcus aureus (MRSA), increase mortality following influenza infection, but the mechanisms remain unclear. Here we show that expression of TLR9, a microbial DNA sensor, is increased in murine lung macrophages, dendritic cells, CD8+ T cells and epithelial cells post-influenza infection. TLR9-/- mice did not show differences in handling influenza nor MRSA infection alone. However, TLR9-/- mice have improved survival and bacterial clearance in the lung post-influenza and MRSA dual infection, with no difference in viral load during dual infection. We demonstrate that TLR9 is upregulated on macrophages even when they are not themselves infected, suggesting that TLR9 upregulation is related to soluble mediators. We rule out a role for elevations in interferon-γ (IFNγ) in mediating the beneficial MRSA clearance in TLR9-/- mice. While macrophages from WT and TLR9-/- mice show similar phagocytosis and bacterial killing to MRSA alone, following influenza infection, there is a marked upregulation of scavenger receptor A and MRSA phagocytosis as well as inducible nitric oxide synthase (Inos) and improved bacterial killing that is specific to TLR9-deficient cells. Bone marrow transplant chimera experiments and in vitro experiments using TLR9 antagonists suggest TLR9 expression on non-hematopoietic cells, rather than the macrophages themselves, is important for regulating myeloid cell function. Interestingly, improved bacterial clearance post-dual infection was restricted to MRSA, as there was no difference in the clearance of Streptococcus pneumoniae. Taken together these data show a surprising inhibitory role for TLR9 signaling in mediating clearance of MRSA that manifests following influenza infection.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/metabolism , Toll-Like Receptor 9/metabolism , Animals , Humans , Influenza, Human/immunology , Lung/immunology , Macrophages , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Knockout , Orthomyxoviridae Infections/immunology , Phagocytosis , Staphylococcal Infections/microbiology , Staphylococcus aureus/immunology , Toll-Like Receptor 9/genetics
4.
J Immunol ; 201(2): 814-820, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29802127

ABSTRACT

Naive T cells are thought to be functionally quiescent. In this study, we studied and compared the phenotype, cytokine profile, and potential function of human naive CD4+ T cells in umbilical cord and peripheral blood. We found that naive CD4+ T cells, but not memory T cells, expressed high levels of chemokine CXCL8. CXCL8+ naive T cells were preferentially enriched CD31+ T cells and did not express T cell activation markers or typical Th effector cytokines, including IFN-γ, IL-4, IL-17, and IL-22. In addition, upon activation, naive T cells retained high levels of CXCL8 expression. Furthermore, we showed that naive T cell-derived CXCL8 mediated neutrophil migration in the in vitro migration assay, supported tumor sphere formation, and promoted tumor growth in an in vivo human xenograft model. Thus, human naive T cells are phenotypically and functionally heterogeneous and can carry out active functions in immune responses.


Subject(s)
Blood Cells/physiology , Interleukin-8/metabolism , Neoplasms, Experimental/immunology , Neutrophils/physiology , T-Lymphocytes/physiology , Umbilical Cord/pathology , Animals , Carcinogenesis , Cell Line, Tumor , Cell Movement , Cytokines/metabolism , Gene Expression Regulation , Humans , Interleukin-8/genetics , Lymphocyte Activation , Mice , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Xenograft Model Antitumor Assays
5.
Oncoimmunology ; 6(12): e1362529, 2017.
Article in English | MEDLINE | ID: mdl-29209568

ABSTRACT

CD28H is a newly discovered co-receptor of the human B7 family. CD28H interacts with its ligand B7-H5 and regulates T cell response. Here we showed that CD28H was not expressed on granulocytes, monocytes, myeloid dendritic cells (MDCs), and B cells, but constitutively expressed with moderate levels on memory T cells and with high levels on naïve T cells, innate lymphoid cells (ILCs), natural killer (NK) cells, and plasmacytoid dendritic cells (PDCs) in human peripheral blood. Similar CD28H+ cell profile existed in secondary lymphoid organs and pathological tissues including multiple types of cancers. Further analysis demonstrated that CD28H+ naïve and CD28H+ memory T cells were characterized with increased naïve feature and less effector functional phenotype, respectively. High levels of constitutive CD28H expression on naïve T cells and innate immune cells suggest a potential role of CD28H in innate and adaptive immunity.

6.
Sci Immunol ; 2(17)2017 11 17.
Article in English | MEDLINE | ID: mdl-29150439

ABSTRACT

Naïve T cells are poorly studied in cancer patients. We report that naïve T cells are prone to undergo apoptosis due to a selective loss of FAK family-interacting protein of 200 kDa (FIP200) in ovarian cancer patients and tumor-bearing mice. This results in poor antitumor immunity via autophagy deficiency, mitochondria overactivation, and high reactive oxygen species production in T cells. Mechanistically, loss of FIP200 disables the balance between proapoptotic and antiapoptotic Bcl-2 family members via enhanced argonaute 2 (Ago2) degradation, reduced Ago2 and microRNA1198-5p complex formation, less microRNA1198-5p maturation, and consequently abolished microRNA1198-5p-mediated repression on apoptotic gene Bak1 Bcl-2 overexpression and mitochondria complex I inhibition rescue T cell apoptosis and promoted tumor immunity. Tumor-derived lactate translationally inhibits FIP200 expression by down-regulating the nicotinamide adenine dinucleotide level while potentially up-regulating the inhibitory effect of adenylate-uridylate-rich elements within the 3' untranslated region of Fip200 mRNA. Thus, tumors metabolically target naïve T cells to evade immunity.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Lactic Acid/pharmacology , Ovarian Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/drug effects , Animals , Apoptosis/genetics , Autophagy/genetics , Autophagy-Related Proteins , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lactic Acid/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein-Tyrosine Kinases/genetics , T-Lymphocytes/metabolism
7.
Nat Immunol ; 18(12): 1332-1341, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29083399

ABSTRACT

Live regulatory T cells (Treg cells) suppress antitumor immunity, but how Treg cells behave in the metabolically abnormal tumor microenvironment remains unknown. Here we show that tumor Treg cells undergo apoptosis, and such apoptotic Treg cells abolish spontaneous and PD-L1-blockade-mediated antitumor T cell immunity. Biochemical and functional analyses show that adenosine, but not typical suppressive factors such as PD-L1, CTLA-4, TGF-ß, IL-35, and IL-10, contributes to apoptotic Treg-cell-mediated immunosuppression. Mechanistically, apoptotic Treg cells release and convert a large amount of ATP to adenosine via CD39 and CD73, and mediate immunosuppression via the adenosine and A2A pathways. Apoptosis in Treg cells is attributed to their weak NRF2-associated antioxidant system and high vulnerability to free oxygen species in the tumor microenvironment. Thus, the data support a model wherein tumor Treg cells sustain and amplify their suppressor capacity through inadvertent death via oxidative stress. This work highlights the oxidative pathway as a metabolic checkpoint that controls Treg cell behavior and affects the efficacy of therapeutics targeting cancer checkpoints.


Subject(s)
Apoptosis/immunology , B7-H1 Antigen/metabolism , Immune Tolerance/immunology , Ovarian Neoplasms/immunology , Oxidative Stress/physiology , T-Lymphocytes, Regulatory/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , CTLA-4 Antigen/metabolism , Female , GPI-Linked Proteins/genetics , Humans , Interleukin-10/metabolism , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxygen/metabolism , Receptor, Adenosine A2A/metabolism , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/immunology
8.
Nat Immunol ; 17(1): 95-103, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26523864

ABSTRACT

Aerobic glycolysis regulates T cell function. However, whether and how primary cancer alters T cell glycolytic metabolism and affects tumor immunity in cancer patients remains a question. Here we found that ovarian cancers imposed glucose restriction on T cells and dampened their function via maintaining high expression of microRNAs miR-101 and miR-26a, which constrained expression of the methyltransferase EZH2. EZH2 activated the Notch pathway by suppressing Notch repressors Numb and Fbxw7 via trimethylation of histone H3 at Lys27 and, consequently, stimulated T cell polyfunctional cytokine expression and promoted their survival via Bcl-2 signaling. Moreover, small hairpin RNA-mediated knockdown of human EZH2 in T cells elicited poor antitumor immunity. EZH2(+)CD8(+) T cells were associated with improved survival in patients. Together, these data unveil a metabolic target and mechanism of cancer immune evasion.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , MicroRNAs , Neoplasms/immunology , Polycomb Repressive Complex 2/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Animals , Cell Separation , Chromatin Immunoprecipitation , Enhancer of Zeste Homolog 2 Protein , Female , Flow Cytometry , Fluorescent Antibody Technique , Glycolysis , Humans , Immunoblotting , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Ovarian Neoplasms/immunology , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , Transfection
9.
Oncoimmunology ; 2(8): e25752, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-24083082

ABSTRACT

The ability of Listeria monocytogenes-based anticancer vaccines to induce tumor regression depends on the responsiveness of malignant cells to interferon γ (IFNγ). Inhibition of IFNγ limits the recruitment of T cells to the tumors of vaccinated mice. We hypothesized that vaccination with immunotherapeutic L. monocytogenes induces the IFNγ-dependent production of chemokines that regulate the migration of tumor-infiltrating T cells. To gain further insights into this issue, we examined the chemokine responses of a transplantable, human papillomavirus (HPV)-immortalized murine tumor model (TC-1) following the administration of a L. monocytogenes-based immunotherapeutic agent that expresses E7 from HPV-16. Here, we report that the administration of L. monocytogenes-based anticancer vaccines increases the secretion of chemokine (C-X-C motif) ligand 9 (CXCL9), and CXCL10 by tumors, hence favoring the recruitment of T cells bearing the cognate chemokine (C-X-C motif) receptor 3 (CXCR3). Furthermore, the expression of CXCL9, but not CXCL10, in TC-1 tumors was significantly reduced upon anti-IFNγ antibody treatment. CXCL9 was highly expressed by TC-1 cells following the administration of IFNγ and tumor necrosis factor α (TNFα), in vitro. Moreover, the inhibition of CXCL9 in TC-1 cells reduced the proportion of CD8+ T cells infiltrating tumors in vaccinated mice, while increasing that of CD4+ T cells, thus altering T-cell subset distribution. We conclude that the administration of L. monocytogenes-based anticancer vaccines regulates TH1 chemokine responses and that malignant cells are an important source of these chemokines.

10.
Curr Opin Immunol ; 25(2): 214-21, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23298609

ABSTRACT

Human tumors progress despite the presence of tumor associated antigen (TAA)-specific T cells. Many different molecular and cellular mechanisms contribute to the failure of T cells to eradicate the tumor. These include immune suppressive networks that impair ongoing T cell function and enable tumor escape. Recent studies have started to reveal the nature of effector T cells in the tumor microenvironment. In this article we discuss T cell anergy, exhaustion, senescence, and stemness, and review the phenotype of dysfunctional T cell subsets and the underlying molecular mechanisms in the tumor microenvironments. We suggest that targeting T cell dysfunctional mechanisms and introducing/promoting T cell stemness are important approaches to treat patients with cancer.


Subject(s)
Cellular Senescence , Clonal Anergy/immunology , Neoplasms/immunology , Stem Cells , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Microenvironment/immunology , Animals , Humans , Neoplasms/therapy , Stem Cells/immunology , T-Lymphocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...