Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.732
Filter
1.
Biomed Res ; 45(3): 115-123, 2024.
Article in English | MEDLINE | ID: mdl-38839354

ABSTRACT

Mixed lymphocyte culture under the blockade of CD80/CD86-CD28 co-stimulation induces anergic (completely hyporesponsive) T cells with immune suppressive function (inducible suppressing T cells: iTS cells). Previously, iTS cell therapy has demonstrated outstanding benefits in clinical trials for organ transplantation. Here, we examined whether peptide antigen-specific iTS cells are inducible. DO 11.10 iTS cells were obtained from splenocytes of BALB/c DO 11.10 mice by stimulation with OVA peptide and antagonistic anti-CD80/CD86 mAbs. When DO 11.10 iTS or Foxp3- DO 11.10 iTS cells were stimulated with OVA, these cells produced IL-13, but not IL-4. DO 11.10 iTS cells decreased IL-4 and increased IL-13 production from OVA-stimulated naïve DO 11.10 splenocytes. When Foxp3+ DO 11.10 iTS cells were prepared, these cells significantly inhibited the production of IL-4 and IL-13 compared with freshly isolated Foxp3+ DO 11.10 T cells. Moreover, an increase in the population expressing OX40, ICOS, and 4-1BB suggested activation of Foxp3+ DO 11.10 iTS cells. Thus, blockade of CD80/CD86-CD28 co-stimulation during peptide antigen stimulation augments the inhibitory function of Foxp3+ regulatory T cells, and does not induce anergic Foxp3- conventional T cells. Peptide-specific Foxp3+ regulatory iTS cells could be useful for the treatment of allergic and autoimmune diseases without adverse effects.


Subject(s)
B7-1 Antigen , B7-2 Antigen , CD28 Antigens , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , Mice , B7-1 Antigen/metabolism , B7-1 Antigen/immunology , B7-2 Antigen/metabolism , B7-2 Antigen/immunology , Mice, Inbred BALB C , Forkhead Transcription Factors/metabolism , Peptides/pharmacology , Peptides/immunology , Lymphocyte Activation/immunology , Interleukin-4/metabolism , Interleukin-4/immunology , Interleukin-13/metabolism , Interleukin-13/immunology , Ovalbumin/immunology , Spleen/immunology , Spleen/cytology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/immunology
2.
Hum Immunol ; 85(4): 110831, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38870593

ABSTRACT

Surface expression of programmed death-ligand 1 (PD-L1) is mainly observed on antigen presenting cells (APC) such as monocytes or dendritic cells (DCs). Our results showing a high expression of PD-L1 on human naïve CD4+ effector T-cells (TEFFs) and CD4+ regulatory T cells (TREGs) after activation with human DCs, allow us to propose a new role for PD-L1 and its ligands and their potential impact on new signaling pathways. Indeed, expression of PD-L1 on activated CD4+T cells could allow cis interaction with its ligands such as PD-1 and CD80, thus disrupting interactions with other signaling receptors, such as cytotoxic T-lymphocyte antigen-4 (CTLA-4) or CD28, which interact with CD80. The ability to compete with hypothetical configuration modifications that may cause a change in affinity/avidity for the trans and cis interactions between these proteins expressed on T cells and/or DCs is discussed. As the study of cancer is strongly influenced by the role of the PD-L1/PD-1 pathway and CD4+T cells, new interactions, cis and/or trans, between TEFFs, TREGs and tumor cells are also proposed. The presence of PD-L1 on activated CD4+ T cells could influence the quality of the cytotoxic T lymphocyte response during priming to provide other help signals.


Subject(s)
B7-H1 Antigen , CD4-Positive T-Lymphocytes , Cell Communication , Dendritic Cells , Lymphocyte Activation , Programmed Cell Death 1 Receptor , Signal Transduction , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Lymphocyte Activation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Programmed Cell Death 1 Receptor/metabolism , Cell Communication/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , B7-1 Antigen/metabolism , B7-1 Antigen/immunology , CTLA-4 Antigen/metabolism , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , CD28 Antigens/metabolism , CD28 Antigens/immunology
3.
Bull Exp Biol Med ; 176(6): 806-810, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38907062

ABSTRACT

When developing a program of preclinical studies of human cell-based drugs intended for adoptive immunotherapy of cancer patients, the biological effect should be substantiated by data describing their immunological action. Administration and study of human autologous dendritic cell vaccine to immunocompetent animals are not adequate in terms of immunological compatibility. It is possible to use immunocompromised, knockout, or transgenic animals or to obtain a homologous cellular product, namely, a preparation based on animal cells using a technology similar to obtaining the original preparation for clinical practice in humans. Within the framework of this study, we have developed a protocol for obtaining a homologous cell product based on animal dendritic cells (mice, rats) according to a similar technology for obtaining human vaccine dendritic cells, and demonstrated the comparability of morphological characteristics and expression of differentiation antigens of dendritic cells (CD11c, CD80, CD86, and CD83) of animals (mice) and humans.


Subject(s)
Cancer Vaccines , Dendritic Cells , Immunotherapy, Adoptive , Animals , Dendritic Cells/immunology , Dendritic Cells/drug effects , Cancer Vaccines/immunology , Mice , Humans , Rats , Immunotherapy, Adoptive/methods , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-1 Antigen/genetics , CD11c Antigen/metabolism , CD11c Antigen/immunology , B7-2 Antigen/metabolism , B7-2 Antigen/immunology , B7-2 Antigen/genetics
4.
Am J Physiol Cell Physiol ; 326(6): C1563-C1572, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38586879

ABSTRACT

Atherosclerosis is an inflammatory disease of blood vessels involving the immune system. Natural killer T (NKT) cells, as crucial components of the innate and acquired immune systems, play critical roles in the development of atherosclerosis. However, the mechanism and clinical relevance of NKT cells in early atherosclerosis are largely unclear. The study investigated the mechanism influencing NKT cell function in apoE deficiency-induced early atherosclerosis. Our findings demonstrated that there were higher populations of NKT cells and interferon-gamma (IFN-γ)-producing NKT cells in the peripheral blood of patients with hyperlipidemia and in the aorta, blood, spleen, and bone marrow of early atherosclerotic mice compared with the control groups. Moreover, we discovered that the infiltration of CD80+ macrophages and CD1d expression on CD80+ macrophages in atherosclerotic mice climbed remarkably. CD1d expression increased in CD80+ macrophages stimulated by oxidized low-density lipoprotein (ox-LDL) ex vivo and in vitro. Ex vivo coculture of macrophages with NKT cells revealed that ox-LDL-induced CD80+ macrophages presented lipid antigen α-Galcer (alpha-galactosylceramide) to NKT cells via CD1d, enabling NKT cells to express more IFN-γ. Furthermore, a greater proportion of CD1d+ monocytes and CD1d+CD80+ monocytes were found in peripheral blood of hyperlipidemic patients compared with that of healthy donors. Positive correlations were found between CD1d+CD80+ monocytes and NKT cells or IFN-γ+ NKT cells in hyperlipidemic patients. Our findings illustrated that CD80+ macrophages stimulated NKT cells to secrete IFN-γ via CD1d-presenting α-Galcer, which may accelerate the progression of early atherosclerosis. Inhibiting lipid antigen presentation by CD80+ macrophages to NKT cells may be a promising immune target for the treatment of early atherosclerosis.NEW & NOTEWORTHY This work proposed the ox-LDL-CD80+ monocyte/macrophage-CD1d-NKT cell-IFN-γ axis in the progression of atherosclerosis. The proinflammatory IFN-γ+ NKT cells are closely related to CD1d+CD80+ monocytes in hyperlipidemic patients. Inhibiting CD80+ macrophages to present lipid antigens to NKT cells through CD1d blocking may be a new therapeutic target for atherosclerosis.


Subject(s)
Antigens, CD1d , Atherosclerosis , B7-1 Antigen , Hyperlipidemias , Lipoproteins, LDL , Macrophages , Natural Killer T-Cells , Animals , Humans , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Antigens, CD1d/metabolism , Antigens, CD1d/immunology , Antigens, CD1d/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Hyperlipidemias/immunology , Hyperlipidemias/metabolism , Lipoproteins, LDL/immunology , Lipoproteins, LDL/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , B7-1 Antigen/metabolism , B7-1 Antigen/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Mice, Inbred C57BL , Female , Middle Aged
5.
Nat Cancer ; 5(5): 760-773, 2024 May.
Article in English | MEDLINE | ID: mdl-38503896

ABSTRACT

Chimeric antigen receptor T cells have dramatically improved the treatment of hematologic malignancies. T cell antigen receptor (TCR)-based cell therapies are yet to achieve comparable outcomes. Importantly, chimeric antigen receptors not only target selected antigens but also reprogram T cell functions through the co-stimulatory pathways that they engage upon antigen recognition. We show here that a fusion receptor comprising the CD80 ectodomain and the 4-1BB cytoplasmic domain, termed 80BB, acts as both a ligand and a receptor to engage the CD28 and 4-1BB pathways, thereby increasing the antitumor potency of human leukocyte antigen-independent TCR (HIT) receptor- or TCR-engineered T cells and tumor-infiltrating lymphocytes. Furthermore, 80BB serves as a switch receptor that provides agonistic 4-1BB co-stimulation upon its ligation by the inhibitory CTLA4 molecule. By combining multiple co-stimulatory features in a single antigen-agnostic synthetic receptor, 80BB is a promising tool to sustain CD3-dependent T cell responses in a wide range of targeted immunotherapies.


Subject(s)
CD28 Antigens , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Humans , Receptors, Chimeric Antigen/immunology , Receptors, Antigen, T-Cell/immunology , CD28 Antigens/immunology , Animals , Mice , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , B7-1 Antigen/immunology , T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Cell- and Tissue-Based Therapy/methods
6.
Sci Rep ; 11(1): 17214, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34446765

ABSTRACT

Salmonella enterica serovar Gallinarum is a host-restricted bacterial pathogen that causes a serious systemic disease exclusively in birds of all ages. Salmonella enterica serovar Typhimurium is a host-generalist serovar. Dendritic cells (DCs) are key antigen-presenting cells that play an important part in Salmonella host-restriction. We evaluated the differential response of chicken blood monocyte-derived dendritic cells (chMoDCs) exposed to S. Gallinarum or S. Typhimurium. S. Typhimurium was found to be more invasive while S. Gallinarum was more cytotoxic at the early phase of infection and later showed higher resistance against chMoDCs killing. S. Typhimurium promoted relatively higher upregulation of costimulatory and other immune function genes on chMoDCs in comparison to S. Gallinarum during early phase of infection (6 h) as analyzed by real-time PCR. Both Salmonella serovars strongly upregulated the proinflammatory transcripts, however, quantum was relatively narrower with S. Gallinarum. S. Typhimurium-infected chMoDCs promoted relatively higher proliferation of naïve T-cells in comparison to S. Gallinarum as assessed by mixed lymphocyte reaction. Our findings indicated that host restriction of S. Gallinarum to chicken is linked with its profound ability to interfere the DCs function. Present findings provide a valuable roadmap for future work aimed at improved vaccine strategies against this pathogen.


Subject(s)
Dendritic Cells/immunology , Monocytes/immunology , Salmonella typhimurium/immunology , Salmonella/immunology , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , CD40 Antigens/genetics , CD40 Antigens/immunology , Chickens , Cytokines/genetics , Cytokines/immunology , Cytotoxicity, Immunologic/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Gene Expression/immunology , Host-Pathogen Interactions/immunology , Microbial Viability/immunology , Monocytes/cytology , Salmonella/physiology , Salmonella typhimurium/physiology , Species Specificity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
7.
J Immunol ; 207(1): 110-114, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34135059

ABSTRACT

Macrophages play a central role in lung physiology and pathology. In this study, we show in mice that alveolar macrophages (AMs), unlike other macrophage types (interstitial, peritoneal, and splenic macrophages), constitutively express programmed death-1 ligand 1 (PD-L1), thereby possessing a superior phagocytic ability and the capacity to repress CTLs by cis- and trans-interacting with CD80 and programmed death-1 (PD-1), respectively. This extraordinary ability of AMs assures optimal protective immunity and tolerance within the lung. These findings uncover a unique characteristic of AMs and an innate immune function of PD-L1 and CD80 and therefore help in the understanding of lung physiology, diseases, and PD-L1/PD-1-based immunotherapy.


Subject(s)
B7-H1 Antigen/immunology , Macrophages, Alveolar/immunology , Animals , B7-1 Antigen/immunology , Mice , Mice, Inbred Strains , Mice, Knockout
8.
Front Immunol ; 12: 666356, 2021.
Article in English | MEDLINE | ID: mdl-34054836

ABSTRACT

Type II interferon gamma (IFNγ) is a pleiotropic cytokine capable of modulating the innate and adaptive immune responses which has been widely characterized in several teleost families. In fish, IFNγ stimulates the expression of cytokines and chemokines associated with the pro-inflammatory response and enhances the production of nitrogen and oxygen reactive species in phagocytic cells. This work studied the effect of IFNγ on the expression of cell-surface markers on splenocytes of Atlantic salmon (Salmo salar). In vitro results showed that subpopulations of mononuclear splenocytes cultured for 15 days were capable of increasing gene expression and protein availability of cell-surface markers such as CD80/86, CD83 and MHC II, after being stimulated with recombinant IFNγ. These results were observed for subpopulations with characteristics associated with monocytes (51%), and features that could be related to lymphocytes (46.3%). In addition, a decrease in the expression of zbtb46 was detected in IFNγ-stimulated splenocytes. Finally, the expression of IFNγ and cell-surface markers was assessed in Atlantic salmon under field conditions. In vivo results showed that the expression of ifnγ increased simultaneously with the up-regulation of cd80/86, cd83 and mhcii during a natural outbreak of Piscirickettsia salmonis. Overall, the results obtained in this study allow us to propose IFNγ as a candidate molecule to stimulate the phenotypic progression of a small population of immune cells, which will increase antigen presenting cells markers. Thereby, modulatory strategies using IFNγ may generate a robust and coordinated immune response in fish against pathogens that affect aquaculture.


Subject(s)
Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Histocompatibility Antigens Class II/metabolism , Immunoglobulins/metabolism , Interferon-gamma/immunology , Membrane Glycoproteins/metabolism , Salmo salar/immunology , Spleen/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, CD/genetics , Antigens, CD/immunology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-2 Antigen/genetics , B7-2 Antigen/immunology , Biomarkers/metabolism , Fish Diseases/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Immunoglobulins/genetics , Immunoglobulins/immunology , Interferon-gamma/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Piscirickettsia , Piscirickettsiaceae Infections/immunology , Piscirickettsiaceae Infections/veterinary , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism , CD83 Antigen
9.
Trends Mol Med ; 27(3): 207-219, 2021 03.
Article in English | MEDLINE | ID: mdl-33199209

ABSTRACT

Immune checkpoints negatively regulate immune cell responses. Programmed cell death protein 1:programmed death ligand 1 (PD-1:PD-L1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4):B7-1 are among the most important immune checkpoint pathways, and are key targets for immunotherapies that seek to modulate the balance between stimulatory and inhibitory signals to lead to favorable therapeutic outcomes. The current dogma of these two immune checkpoint pathways has regarded them as independent with no interactions. However, the newly characterized PD-L1:B7-1 ligand-ligand cis-interaction and its ability to bind CTLA-4 and CD28, but not PD-1, suggests that these pathways have significant crosstalk. Here, we propose that the PD-L1:B7-1 cis-interaction brings novel mechanistic understanding of these pathways, new insights into mechanisms of current immunotherapies, and fresh ideas to develop better treatments in a variety of therapeutic settings.


Subject(s)
B7-1 Antigen , B7-H1 Antigen , Immunity , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , B7-1 Antigen/chemistry , B7-1 Antigen/immunology , B7-H1 Antigen/chemistry , B7-H1 Antigen/immunology , Humans , Immune Checkpoint Inhibitors , Immune Evasion , Immunity/immunology , Immunity/physiology , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Organ Transplantation , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/immunology
10.
Med Oncol ; 37(11): 107, 2020 Nov 02.
Article in English | MEDLINE | ID: mdl-33136217

ABSTRACT

The treatment of castration-resistant prostate cancer (CRPC) is always a difficulty in the clinic. Most patients with localized tumor eventually develop CRPC, even if hormone therapy is initially effective. Increasing evidence shows immunotherapy has special advantages compared with traditional therapy in cancer treatment. In this study, we constructed the DC-PC-3 fusion vaccine with B7-1- and GM-CSF-specific modification, and studied its ability to stimulate specific immune response and anti-tumor effect in vitro. The results showed that fusion of DC and tumor cells can improve the expression of associated antigens of DCs. DC-tumor fusion vaccine can strongly promote T cell proliferation and IFN-γ secretion and induce a significant tumor-specific cytotoxic T lymphocyte response. In addition, the B7-1/GM-CSF-modified fusion vaccine showed a more significant anti-tumor effect and greater ability to stimulate the immune response than that without specific modification in vitro. Thus, GM-CSF/B7-1-modified fusion vaccine might be used as a potential therapy strategy for prostate cancer.


Subject(s)
B7-1 Antigen/immunology , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunotherapy/methods , Prostatic Neoplasms, Castration-Resistant/therapy , T-Lymphocytes, Cytotoxic/immunology , B7-1 Antigen/metabolism , Cancer Vaccines/immunology , Cell Fusion/methods , Cell Line, Tumor , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunologic Factors/immunology , Immunologic Factors/metabolism , Male , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology
11.
J Immunol ; 205(9): 2362-2374, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32978280

ABSTRACT

The roles distinct B cell subsets play in clonal expansion, isotype switching, and memory B cell differentiation in response to T cell-independent type 2 Ags (TI-2 Ags) has been understudied. Using sorted B cells from VHB1-8 knock-in mice, we evaluated B-1b, marginal zone, and follicular B cell responses to the TI-2 Ag, NP-Ficoll. All subsets extensively divided in response to NP-Ficoll. Nonetheless, B-1b cells exhibited significantly increased IgG switching and differentiation into Ab-secreting cells (ASC)-a finding that coincided with increased AgR signaling capacity and Blimp1 expression by B-1b cells. All subsets formed memory cells and expressed markers previously identified for T cell-dependent memory B cells, including CD80, PDL2, and CD73, although B-1b cells generated the greatest number of memory cells with higher frequencies of IgG- and CD80-expressing cells. Despite memory formation, secondary immunization 4 wk after primary immunization did not increase NP-specific IgG. However, boosting occurred in B-1b cell-recipient mice when IgG levels declined. CD80+ memory B-1b cells divided, class switched, and differentiated into ASC in response to Ag in vivo, but this was inhibited in the presence of NP-specific IgG. Furthermore, CD80 blockade significantly increased memory B-1b cell division and differentiation to ASC upon Ag restimulation. Collectively, these findings demonstrate B-1b, marginal zone B, and follicular B subsets significantly contribute to the TI-2 Ag-specific memory B cell pool. In particular, we show B-1b cells generate a functional CD80-regulated memory population that can be stimulated to divide and differentiate into ASC upon Ag re-encounter when Ag-specific IgG levels decline.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunologic Memory/immunology , T-Lymphocytes/immunology , Animals , Antigens, T-Independent/immunology , B7-1 Antigen/immunology , Cell Differentiation/immunology , Cell Division/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology
12.
Toxicol In Vitro ; 67: 104918, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32565220

ABSTRACT

The effect of cryopreservation on antigen-presenting cells (APCs) is understudied. It is important to understand the effects of cryopreservation on these cells as they play a major role in immune responses, and they could be utilized in different clinical applications. In this study, we compared fresh and cryopreserved PBMCs in regards of their general immune responsiveness and, furthermore, the effect of cryopreservation on the circulating APCs among PBMCs. We stimulated fresh and cryopreserved PBMCs (N = 6) with LPS or Poly(I:C).Cytokine production of PBMCs and expression of functional markers CD80 and ILT4 on major types of APCs, dendritic cells (DCs) and monocytes, were analysed. We also analysed whether cryopreservation affects different subtypes of DCs (plasmacytoid and myeloid DCs) differently. Cryopreserved PBMCs produced less cytokines than fresh cells in response to stimulation, but the response profiles were comparable. Cryopreservation had also an effect on the relative proportions of APCs. Stimuli-induced responses were somewhat parallel but weaker than those observed in fresh cells. This study suggests that the use of cryopreserved cells is more suitable in studies that assess general responses to stimuli instead of measuring exact levels of reactions. Thus, the interpretation and comparison of the results of different studies should not be done without considering the differences in cryopreservation techniques and their effects on PBMCs and, more specifically, on APCs.


Subject(s)
Antigen-Presenting Cells , Cryopreservation , Immunity , Leukocytes, Mononuclear , Adult , B7-1 Antigen/immunology , Cells, Cultured , Cytokines/immunology , Humans , Immunity/drug effects , Immunophenotyping , Lipopolysaccharides/pharmacology , Poly I-C/pharmacology
13.
Genome Med ; 12(1): 55, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32580776

ABSTRACT

BACKGROUND: Traditionally, the transcriptomic and proteomic characterisation of CD4+ T cells at the single-cell level has been performed by two largely exclusive types of technologies: single-cell RNA sequencing (scRNA-seq) and antibody-based cytometry. Here, we present a multi-omics approach allowing the simultaneous targeted quantification of mRNA and protein expression in single cells and investigate its performance to dissect the heterogeneity of human immune cell populations. METHODS: We have quantified the single-cell expression of 397 genes at the mRNA level and up to 68 proteins using oligo-conjugated antibodies (AbSeq) in 43,656 primary CD4+ T cells isolated from the blood and 31,907 CD45+ cells isolated from the blood and matched duodenal biopsies. We explored the sensitivity of this targeted scRNA-seq approach to dissect the heterogeneity of human immune cell populations and identify trajectories of functional T cell differentiation. RESULTS: We provide a high-resolution map of human primary CD4+ T cells and identify precise trajectories of Th1, Th17 and regulatory T cell (Treg) differentiation in the blood and tissue. The sensitivity provided by this multi-omics approach identified the expression of the B7 molecules CD80 and CD86 on the surface of CD4+ Tregs, and we further demonstrated that B7 expression has the potential to identify recently activated T cells in circulation. Moreover, we identified a rare subset of CCR9+ T cells in the blood with tissue-homing properties and expression of several immune checkpoint molecules, suggestive of a regulatory function. CONCLUSIONS: The transcriptomic and proteomic hybrid technology described in this study provides a cost-effective solution to dissect the heterogeneity of immune cell populations at extremely high resolution. Unexpectedly, CD80 and CD86, normally expressed on antigen-presenting cells, were detected on a subset of activated Tregs, indicating a role for these co-stimulatory molecules in regulating the dynamics of CD4+ T cell responses.


Subject(s)
B7-1 Antigen/immunology , B7-2 Antigen/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Female , Forkhead Transcription Factors/genetics , Humans , Male , Proteome , RNA , RNA-Seq , Single-Cell Analysis , Transcriptome
14.
Front Immunol ; 11: 327, 2020.
Article in English | MEDLINE | ID: mdl-32161596

ABSTRACT

Overcoming tolerance to tumor-associated antigens remains a hurdle for cancer vaccine-based immunotherapy. A strategy to enhance the anti-tumor immune response is the inclusion of adjuvants to cancer vaccine protocols. In this report, we generated and systematically screened over twenty gene-based molecular adjuvants composed of cytokines, chemokines, and T cell co-stimulators for the ability to increase anti-tumor antigen T cell immunity. We identified several robust adjuvants whose addition to vaccine formulations resulted in enhanced T cell responses targeting the cancer antigens STEAP1 and TERT. We further characterized direct T cell stimulation through CD80-Fc and indirect T cell targeting via the dendritic cell activator Flt3L-Fc. Mechanistically, intramuscular delivery of Flt3L-Fc into mice was associated with a significant increase in infiltration of dendritic cells at the site of administration and trafficking of activated dendritic cells to the draining lymph node. Gene expression analysis of the muscle tissue confirmed a significant up-regulation in genes associated with dendritic cell signaling. Addition of CD80-Fc to STEAP1 vaccine formulation mimicked the engagement provided by DCs and increased T cell responses to STEAP1 by 8-fold, significantly increasing the frequency of antigen-specific cells expressing IFNγ, TNFα, and CD107a for both CD8+ and CD4+ T cells. CD80-Fc enhanced T cell responses to multiple tumor-associated antigens including Survivin and HPV, indicating its potential as a universal adjuvant for cancer vaccines. Together, the results of our study highlight the adjuvanting effect of T cell engagement either directly, CD80-Fc, or indirectly, Flt3L-Fc, for cancer vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , B7-1 Antigen/immunology , Cancer Vaccines/immunology , Membrane Proteins/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Tetraspanin 28/immunology , Animals , Antigens, Neoplasm , B7-1 Antigen/genetics , Cell Movement/immunology , Cytokines/metabolism , DNA/genetics , Dendritic Cells/immunology , Female , Humans , Immunotherapy/methods , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Plasmids/genetics , Vaccines, DNA/genetics , Vaccines, DNA/immunology
15.
Eur J Immunol ; 50(5): 685-694, 2020 05.
Article in English | MEDLINE | ID: mdl-32012247

ABSTRACT

Syncytin-1 is the envelope protein of the human endogenous retrovirus W (HERV-W). It has been related to multiple sclerosis (MS) but its role in cellular immunity and its pathogenic mechanism in the autoimmune context are not fully understood. We analyzed syncytin-1 levels in peripheral blood mononuclear cells (PBMC) subsets from healthy donors, MS patients in relapse or remission, and patients with acute infections by flow cytometry. PBMC cultures were also prepared to analyze protein expression kinetics. MS patients had higher levels of syncytin-1 levels than controls. We found that syncytin-1 is elevated in monocytes during MS relapses and infections. Cells expressing syncytin-1, including monocytes, T and B lymphocytes, and NKs presented mainly an activated phenotype and, upon stimulation with LPS, its levels increased rapidly on antigen-presenting cells. Syncytin-1 ligation promoted the activation of monocytes, as demonstrated by the upregulation of CD80 and the nonclassical subset CD14low CD16+ . Our results suggest an important role for syncytin-1 in the activation of leukocytes. Given that the expression of syncytin-1 is upregulated in MS patients, this protein might be contributing to the autoimmune cascade in the disease.


Subject(s)
Endogenous Retroviruses/immunology , Gene Products, env/genetics , Monocytes/virology , Multiple Sclerosis/genetics , Multiple Sclerosis/virology , Pregnancy Proteins/genetics , Adult , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/virology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Case-Control Studies , Endogenous Retroviruses/genetics , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Regulation , Gene Products, env/immunology , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/pharmacology , Male , Middle Aged , Monocytes/drug effects , Monocytes/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Pregnancy Proteins/immunology , Primary Cell Culture , Receptors, IgG/genetics , Receptors, IgG/immunology , Recurrence , Remission Induction , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology
16.
Nat Commun ; 11(1): 931, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32071302

ABSTRACT

Intrinsic malignant brain tumors, such as glioblastomas are frequently resistant to immune checkpoint blockade (ICB) with few hypermutated glioblastomas showing response. Modeling patient-individual resistance is challenging due to the lack of predictive biomarkers and limited accessibility of tissue for serial biopsies. Here, we investigate resistance mechanisms to anti-PD-1 and anti-CTLA-4 therapy in syngeneic hypermutated experimental gliomas and show a clear dichotomy and acquired immune heterogeneity in ICB-responder and non-responder tumors. We made use of this dichotomy to establish a radiomic signature predicting tumor regression after pseudoprogression induced by ICB therapy based on serial magnetic resonance imaging. We provide evidence that macrophage-driven ICB resistance is established by CD4 T cell suppression and Treg expansion in the tumor microenvironment via the PD-L1/PD-1/CD80 axis. These findings uncover an unexpected heterogeneity of response to ICB in strictly syngeneic tumors and provide a rationale for targeting PD-L1-expressing tumor-associated macrophages to overcome resistance to ICB.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Glioma/drug therapy , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Immunological/therapeutic use , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Cell Line, Tumor/transplantation , Disease Models, Animal , Drug Resistance, Neoplasm/immunology , Female , Glioma/diagnostic imaging , Glioma/genetics , Glioma/immunology , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Magnetic Resonance Imaging , Male , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
17.
Elife ; 92020 01 21.
Article in English | MEDLINE | ID: mdl-31959281

ABSTRACT

Focal Adhesion Kinase (FAK) inhibitors are currently undergoing clinical testing in combination with anti-PD-1 immune checkpoint inhibitors. However, which patients are most likely to benefit from FAK inhibitors, and what the optimal FAK/immunotherapy combinations are, is currently unknown. We identify that cancer cell expression of the T-cell co-stimulatory ligand CD80 sensitizes murine tumors to a FAK inhibitor and show that CD80 is expressed by human cancer cells originating from both solid epithelial cancers and some hematological malignancies in which FAK inhibitors have not been tested clinically. In the absence of CD80, we identify that targeting alternative T-cell co-stimulatory receptors, in particular OX-40 and 4-1BB in combination with FAK, can drive enhanced anti-tumor immunity and even complete regression of murine tumors. Our findings provide rationale supporting the clinical development of FAK inhibitors in combination with patient selection based on cancer cell CD80 expression, and alternatively with therapies targeting T-cell co-stimulatory pathways.


Subject(s)
Focal Adhesion Kinase 1 , Immune Checkpoint Inhibitors , T-Lymphocytes , Animals , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/immunology , Humans , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/pharmacology , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
18.
J Virol ; 94(6)2020 02 28.
Article in English | MEDLINE | ID: mdl-31852788

ABSTRACT

High rates of wild-type (WT) herpes simplex virus 1 (HSV-1) latency reactivation depend on the anti-apoptotic activities of latency-associated transcript (LAT). Replacing LAT with the baculovirus inhibitor of apoptosis protein (cpIAP) or cellular FLIP (FLICE-like inhibitory protein) gene restored the WT latency reactivation phenotype to that of a LAT-minus [LAT(-)] virus, while similar recombinant viruses expressing interleukin-4 (IL-4) or interferon gamma (IFN-γ) did not. However, HSV-1 recombinant virus expressing cpIAP did not restore all LAT functions. Recently, we reported that a similar recombinant virus expressing CD80 in place of LAT had higher latency reactivation than a LAT-null virus. The present study was designed to determine if this CD80-expressing recombinant virus can restore all LAT functions as observed with WT virus. Our results suggest that overexpression of CD80 fully rescues LAT function in latency reactivation, apoptosis, and immune exhaustion, suggesting that LAT and CD80 have multiple overlapping functions.IMPORTANCE Recurring ocular infections caused by HSV-1 can cause corneal scarring and blindness. A major function of the HSV-1 latency-associated transcript (LAT) is to establish high levels of latency and reactivation, thus contributing to the development of eye disease. Here, we show that the host CD80 T cell costimulatory molecule functions similarly to LAT and can restore the ability of LAT to establish latency, reactivation, and immune exhaustion as well as induce the expression of caspase 3, caspase 8, caspase 9, and Bcl2. Our results suggest that, in contrast to several other previously tested genes, CD80-expressing virus can completely compensate for all known and tested LAT functions.


Subject(s)
Apoptosis/immunology , B7-1 Antigen/immunology , Herpesvirus 1, Human/physiology , MicroRNAs/immunology , RNA, Viral/immunology , Virus Activation/immunology , Virus Latency/immunology , Animals , Apoptosis/genetics , B7-1 Antigen/genetics , Mice , MicroRNAs/genetics , RNA, Viral/genetics , Virus Activation/genetics , Virus Latency/genetics
19.
Drugs ; 80(1): 33-46, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31749062

ABSTRACT

Costimulation between T cells and antigen-presenting cells is essential for the regulation of an effective alloimmune response and is not targeted with the conventional immunosuppressive therapy after kidney transplantation. Costimulation blockade therapy with biologicals allows precise targeting of the immune response but without non-immune adverse events. Multiple costimulation blockade approaches have been developed that inhibit the alloimmune response in kidney transplant recipients with varying degrees of success. Belatacept, an immunosuppressive drug that selectively targets the CD28-CD80/CD86 pathway, is the only costimulation blockade therapy that is currently approved for kidney transplant recipients. In the last decade, belatacept therapy has been shown to be a promising therapy in subgroups of kidney transplant recipients; however, the widespread use of belatacept has been tempered by an increased risk of acute kidney transplant rejection. The purpose of this review is to provide an overview of the costimulation blockade therapies that are currently in use or being developed for kidney transplant indications.


Subject(s)
Abatacept/administration & dosage , Immunosuppressive Agents/administration & dosage , Kidney Transplantation/methods , Abatacept/immunology , Animals , B7-1 Antigen/immunology , B7-2 Antigen/immunology , CD28 Antigens/immunology , Graft Rejection/immunology , Humans , Immunosuppressive Agents/immunology
20.
J Virol ; 94(2)2020 01 06.
Article in English | MEDLINE | ID: mdl-31619558

ABSTRACT

We recently reported that herpes simplex virus 1 (HSV-1) infection suppresses CD80 but not CD86 expression in vitro and in vivo This suppression required the HSV-1 ICP22 gene. We also reported that overexpression of CD80 by HSV-1 exacerbated corneal scarring in BALB/c mice. We now show that this recombinant virus (HSV-CD80) expressed high levels of CD80 both in vitro in cultured rabbit skin cells and in vivo in infected mouse corneas. CD80 protein was detected on the surface of infected cells. The virulence of the recombinant HSV-CD80 virus was similar to that of the parental strain, and the replication of HSV-CD80 was similar to that of control virus in vitro and in vivo Transcriptome analysis detected 75 known HSV-1 genes in the corneas of mice infected with HSV-CD80 or parental virus on day 4 postinfection. Except for significantly higher CD80 expression in HSV-CD80-infected mice, levels of HSV-1 gene expression were similar in corneas from HSV-CD80-infected and parental virus-infected mice. The number of CD8+ T cells was higher, and the number of CD4+ T cells was lower, in the corneas of HSV-CD80-infected mice than in mice infected with parental virus. HSV-CD80-infected mice displayed a transient increase in dendritic cells. Transcriptome analysis revealed mild differences in dendritic cell maturation and interleukin-1 signaling pathways and increased expression of interferon-induced protein with tetratricopeptide repeats 2 (Ifit2). Together, these results suggest that increased CD80 levels promote increased CD8+ T cells, leading to exacerbated eye disease in HSV-1-infected mice.IMPORTANCE HSV-1 ocular infections are the leading cause of corneal blindness. Eye disease is the result of a prolonged immune response to the replicating virus. HSV-1, on the other hand, has evolved several mechanisms to evade clearance by the host immune system. We describe a novel mechanism of HSV-1 immune evasion via ICP22-dependent downregulation of the host T cell costimulatory molecule CD80. However, the exact role of CD80 in HSV-1 immune pathology is not clear. In this study, we show that eye disease is independent of the level of HSV-1 replication and that viral expression of CD80 has a detrimental role in corneal scarring, likely by increasing CD8+ T cell recruitment and activation.


Subject(s)
B7-1 Antigen , Cornea , Herpesvirus 1, Human , Keratitis, Herpetic , Signal Transduction , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cornea/immunology , Cornea/pathology , Cornea/virology , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Interleukin-1/genetics , Interleukin-1/immunology , Keratitis, Herpetic/genetics , Keratitis, Herpetic/immunology , Keratitis, Herpetic/pathology , Mice , Mice, Inbred BALB C , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Rabbits , Signal Transduction/genetics , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL