Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 646
Filter
1.
Nat Immunol ; 18(6): 642-653, 2017 06.
Article in English | MEDLINE | ID: mdl-28436955

ABSTRACT

It remains unclear whether activated inflammatory macrophages can adopt features of tissue-resident macrophages, or what mechanisms might mediate such a phenotypic conversion. Here we show that vitamin A is required for the phenotypic conversion of interleukin 4 (IL-4)-activated monocyte-derived F4/80intCD206+PD-L2+MHCII+ macrophages into macrophages with a tissue-resident F4/80hiCD206-PD-L2-MHCII-UCP1+ phenotype in the peritoneal cavity of mice and during the formation of liver granulomas in mice infected with Schistosoma mansoni. The phenotypic conversion of F4/80intCD206+ macrophages into F4/80hiCD206- macrophages was associated with almost complete remodeling of the chromatin landscape, as well as alteration of the transcriptional profiles. Vitamin A-deficient mice infected with S. mansoni had disrupted liver granuloma architecture and increased mortality, which indicates that failure to convert macrophages from the F4/80intCD206+ phenotype to F4/80hiCD206- may lead to dysregulated inflammation during helminth infection.


Subject(s)
Granuloma/immunology , Liver/immunology , Macrophages/immunology , Schistosomiasis mansoni/immunology , Vitamin A Deficiency/immunology , Animals , Antigens, Differentiation/metabolism , Flow Cytometry , Histocompatibility Antigens Class II/metabolism , Interleukin-4/immunology , Lectins, C-Type/metabolism , Liver/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Peritoneal Cavity/cytology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/metabolism , Schistosoma mansoni , Schistosomiasis mansoni/pathology , Tretinoin/pharmacology , Uncoupling Protein 1/metabolism , Vitamins/pharmacology
2.
Immunity ; 51(6): 1088-1101.e5, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31732168

ABSTRACT

The B cell response to Ehrlichia muris is dominated by plasmablasts (PBs), with few-if any-germinal centers (GCs), yet it generates protective immunoglobulin M (IgM) memory B cells (MBCs) that express the transcription factor T-bet and harbor V-region mutations. Because Ehrlichia prominently infects the liver, we investigated the nature of liver B cell response and that of the spleen. B cells within infected livers proliferated and underwent somatic hypermutation (SHM). Vh-region sequencing revealed trafficking of clones between the spleen and liver and often subsequent local clonal expansion and intraparenchymal localization of T-bet+ MBCs. T-bet+ MBCs expressed MBC subset markers CD80 and PD-L2. Many T-bet+ MBCs lacked CD11b or CD11c expression but had marginal zone (MZ) B cell phenotypes and colonized the splenic MZ, revealing T-bet+ MBC plasticity. Hence, liver and spleen are generative sites of B cell responses, and they include V-region mutation and result in liver MBC localization.


Subject(s)
B-Lymphocytes/immunology , Ehrlichia/immunology , Ehrlichiosis/immunology , Immunoglobulin M/immunology , Liver/immunology , Spleen/immunology , Animals , B7-1 Antigen/biosynthesis , Immunoglobulin Variable Region/genetics , Immunologic Memory/immunology , Liver/cytology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Ligand 2 Protein/biosynthesis , Somatic Hypermutation, Immunoglobulin/genetics , Spleen/cytology , T-Box Domain Proteins/metabolism
3.
Mol Cell ; 78(6): 1207-1223.e8, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32504554

ABSTRACT

Tumor interferon (IFN) signaling promotes PD-L1 expression to suppress T cell-mediated immunosurveillance. We identify the IFN-stimulated non-coding RNA 1 (INCR1) as a long noncoding RNA (lncRNA) transcribed from the PD-L1 locus and show that INCR1 controls IFNγ signaling in multiple tumor types. Silencing INCR1 decreases the expression of PD-L1, JAK2, and several other IFNγ-stimulated genes. INCR1 knockdown sensitizes tumor cells to cytotoxic T cell-mediated killing, improving CAR T cell therapy. We discover that PD-L1 and JAK2 transcripts are negatively regulated by binding to HNRNPH1, a nuclear ribonucleoprotein. The primary transcript of INCR1 binds HNRNPH1 to block its inhibitory effects on the neighboring genes PD-L1 and JAK2, enabling their expression. These findings introduce a mechanism of tumor IFNγ signaling regulation mediated by the lncRNA INCR1 and suggest a therapeutic target for cancer immunotherapy.


Subject(s)
B7-H1 Antigen/genetics , Interferon-gamma/metabolism , RNA, Long Noncoding/genetics , Aged , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Immunotherapy, Adoptive/methods , Interferon-gamma/genetics , Interferons/genetics , Interferons/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Mice , Mice, Inbred NOD , Middle Aged , Programmed Cell Death 1 Ligand 2 Protein/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Cytotoxic
4.
Nat Immunol ; 15(7): 631-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24880458

ABSTRACT

Memory B cells (MBCs) are long-lived sources of rapid, isotype-switched secondary antibody-forming cell (AFC) responses. Whether MBCs homogeneously retain the ability to self-renew and terminally differentiate or if these functions are compartmentalized into MBC subsets has remained unclear. It has been suggested that antibody isotype controls MBC differentiation upon restimulation. Here we demonstrate that subcategorizing MBCs on the basis of their expression of CD80 and PD-L2, independently of isotype, identified MBC subsets with distinct functions upon rechallenge. CD80(+)PD-L2(+) MBCs differentiated rapidly into AFCs but did not generate germinal centers (GCs); conversely, CD80(-)PD-L2(-) MBCs generated few early AFCs but robustly seeded GCs. The gene-expression patterns of the subsets supported both the identity and function of these distinct MBC types. Hence, the differentiation and regeneration of MBCs are compartmentalized.


Subject(s)
B-Lymphocyte Subsets/immunology , B7-1 Antigen/physiology , Immunoglobulin Isotypes/physiology , Immunologic Memory , Programmed Cell Death 1 Ligand 2 Protein/physiology , Amino Acid Sequence , Animals , Antibody-Producing Cells/physiology , Germinal Center/immunology , Immunization , Mice , Mice, Inbred BALB C , Molecular Sequence Data , T-Lymphocytes/physiology
5.
Immunity ; 45(2): 231-3, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27533008

ABSTRACT

How early interactions between innate and adaptive immune cells influence outcomes of acute infections is incompletely understood. In this issue of Immunity, Karunarathne et al. (2016) show that dendritic cells help CD4(+) T helper 1 cell immunity against malaria through PD-L2's competition with PD-L1.


Subject(s)
B7-H1 Antigen/immunology , Programmed Cell Death 1 Ligand 2 Protein , Dendritic Cells/immunology , Humans , Lymphocyte Activation/immunology , Malaria
6.
Immunity ; 45(2): 333-45, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27533014

ABSTRACT

Many pathogens, including Plasmodium spp., exploit the interaction of programmed death-1 (PD-1) with PD-1-ligand-1 (PD-L1) to "deactivate" T cell functions, but the role of PD-L2 remains unclear. We studied malarial infections to understand the contribution of PD-L2 to immunity. Here we have shown that higher PD-L2 expression on blood dendritic cells, from Plasmodium falciparum-infected individuals, correlated with lower parasitemia. Mechanistic studies in mice showed that PD-L2 was indispensable for establishing effective CD4(+) T cell immunity against malaria, because it not only inhibited PD-L1 to PD-1 activity but also increased CD3 and inducible co-stimulator (ICOS) expression on T cells. Importantly, administration of soluble multimeric PD-L2 to mice with lethal malaria was sufficient to dramatically improve immunity and survival. These studies show immuno-regulation by PD-L2, which has the potential to be translated into an effective treatment for malaria and other diseases where T cell immunity is ineffective or short-lived due to PD-1-mediated signaling.


Subject(s)
B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/metabolism , Adamantane/analogs & derivatives , Adamantane/therapeutic use , Adult , Animals , Antimalarials/therapeutic use , B7-H1 Antigen/genetics , Cells, Cultured , Clinical Trials as Topic , Dendritic Cells/parasitology , Female , Humans , Immunity, Cellular , Lymphocyte Activation , Malaria, Falciparum/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Parasitemia/immunology , Peroxides/therapeutic use , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Receptor/genetics , Pyrimidines/therapeutic use , Triazoles/therapeutic use , Young Adult
7.
N Engl J Med ; 384(12): 1125-1135, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33577729

ABSTRACT

BACKGROUND: Patients with advanced urothelial carcinoma have poor overall survival after platinum-containing chemotherapy and programmed cell death protein 1 (PD-1) or programmed death ligand 1 (PD-L1) inhibitor treatment. METHODS: We conducted a global, open-label, phase 3 trial of enfortumab vedotin for the treatment of patients with locally advanced or metastatic urothelial carcinoma who had previously received platinum-containing chemotherapy and had had disease progression during or after treatment with a PD-1 or PD-L1 inhibitor. Patients were randomly assigned in a 1:1 ratio to receive enfortumab vedotin (at a dose of 1.25 mg per kilogram of body weight on days 1, 8, and 15 of a 28-day cycle) or investigator-chosen chemotherapy (standard docetaxel, paclitaxel, or vinflunine), administered on day 1 of a 21-day cycle. The primary end point was overall survival. RESULTS: A total of 608 patients underwent randomization; 301 were assigned to receive enfortumab vedotin and 307 to receive chemotherapy. As of July 15, 2020, a total of 301 deaths had occurred (134 in the enfortumab vedotin group and 167 in the chemotherapy group). At the prespecified interim analysis, the median follow-up was 11.1 months. Overall survival was longer in the enfortumab vedotin group than in the chemotherapy group (median overall survival, 12.88 vs. 8.97 months; hazard ratio for death, 0.70; 95% confidence interval [CI], 0.56 to 0.89; P = 0.001). Progression-free survival was also longer in the enfortumab vedotin group than in the chemotherapy group (median progression-free survival, 5.55 vs. 3.71 months; hazard ratio for progression or death, 0.62; 95% CI, 0.51 to 0.75; P<0.001). The incidence of treatment-related adverse events was similar in the two groups (93.9% in the enfortumab vedotin group and 91.8% in the chemotherapy group); the incidence of events of grade 3 or higher was also similar in the two groups (51.4% and 49.8%, respectively). CONCLUSIONS: Enfortumab vedotin significantly prolonged survival as compared with standard chemotherapy in patients with locally advanced or metastatic urothelial carcinoma who had previously received platinum-based treatment and a PD-1 or PD-L1 inhibitor. (Funded by Astellas Pharma US and Seagen; EV-301 ClinicalTrials.gov number, NCT03474107.).


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Adhesion Molecules/antagonists & inhibitors , Urologic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Drug Resistance, Neoplasm , Female , Humans , Intention to Treat Analysis , Male , Middle Aged , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Progression-Free Survival , Survival Analysis , Urologic Neoplasms/mortality , Urologic Neoplasms/pathology , Urothelium/pathology
8.
Lupus ; 33(8): 816-827, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38622764

ABSTRACT

OBJECTIVE: This study aimed to investigate the role of the programmed cell death protein 1 (PD-1) pathway and T peripheral helper (Tph) cells in the pathogenesis of lupus nephritis using lupus-prone BXSB-Yaa mice. METHODS: Male BXSB-Yaa mice and age-matched male C57BL/6 mice were used. The expression of PD-1 and its ligands (programmed cell death 1 ligand-1, PD-L1 and programmed cell death 1 ligand-2, PD-L2) and the phenotypes of kidney-derived cells and splenocytes expressing these molecules were analyzed by immunofluorescence and flow cytometry. RESULTS: Nephritis spontaneously developed in 16-week-old but not in 8-week-old BXSB-Yaa or C57BL/6 mice. PD-1 was expressed on CD4+ mononuclear cells (MNCs) that infiltrated the glomeruli of 16-week-old BXSB-Yaa mice. The frequency of CD4+PD-1+CXCR5-ICOS+ kidney-derived Tph cells was higher in 16-week-old than in 8-week-old BXSB-Yaa and C57BL/6 mice, whereas the frequency of CD4+PD-1+CXCR5+ICOS+ kidney-derived T follicular helper (Tfh) cells was not significantly different between the mice. PD-L1 was constitutively expressed in the renal tubules. PD-L2 was expressed in the glomeruli of 16-week-old BXSB-Yaa mice. The frequency of PD-L1highCD11c+CD3-CD19- and PD-L2+CD11c+CD3-CD19- kidney-derived MNCs in 16-week-old BXSB-Yaa mice was significantly higher than that of the control mice. The percentage of kidney-derived Tph cells but not Tfh cells was correlated with the urinary protein levels in the nephritic mice. CONCLUSION: The results of this study suggest that kidney-infiltrating PD-1+ Tph cells expanded concomitantly with the upregulation of PD-L1 and PD-L2 in the kidneys and the progression of lupus nephritis.


Subject(s)
B7-H1 Antigen , Kidney , Lupus Nephritis , Mice, Inbred C57BL , Programmed Cell Death 1 Ligand 2 Protein , Programmed Cell Death 1 Receptor , T-Lymphocytes, Helper-Inducer , Up-Regulation , Animals , Programmed Cell Death 1 Receptor/metabolism , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Mice , Male , Programmed Cell Death 1 Ligand 2 Protein/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , B7-H1 Antigen/metabolism , Kidney/pathology , Kidney/metabolism , Kidney/immunology , Disease Models, Animal
9.
Zhonghua Jie He He Hu Xi Za Zhi ; 47(5): 485-489, 2024 May 12.
Article in Zh | MEDLINE | ID: mdl-38706074

ABSTRACT

Programmed cell death 1 (PD-1) and its ligands, PD-L1 and PD-L2, expressed on a variety of immune cells, play multiple regulatory roles in the host immune response to Mycobacterium tuberculosis infection. In this study, we reviewed that the regulatory roles of PD-1/PD-L1, PD-L2 signaling in the host adaptive immune response, such as the innate response of macrophages, and the interaction between T cells and macrophages in response to MTB. In addition, during MTB infection, PD-1/PD-L1, PD-L2 signaling is also involved in the host inflammatory response, as well as the potential roles of PD-1/PD-L1, PD-L2 in the diagnosis and treatment of tuberculosis.


Subject(s)
B7-H1 Antigen , Macrophages , Mycobacterium tuberculosis , Programmed Cell Death 1 Ligand 2 Protein , Programmed Cell Death 1 Receptor , Signal Transduction , Tuberculosis , Humans , Tuberculosis/immunology , Tuberculosis/microbiology , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Mycobacterium tuberculosis/immunology , Macrophages/immunology , Macrophages/metabolism , Immunity, Innate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Adaptive Immunity
10.
Cancer Immunol Immunother ; 72(12): 4065-4075, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37768345

ABSTRACT

Targeting PD-1/PD-L1 has shown substantial therapeutic response and unprecedented long-term durable responses in the clinic. However, several challenges persist, encompassing the prediction of treatment effectiveness and patient responses, the emergence of treatment resistance, and the necessity for additional biomarkers. Consequently, we comprehensively explored the often-overlooked isoforms of crucial immunotherapy players, leveraging transcriptomic analysis, structural modeling, and immunohistochemistry (IHC) data. Our investigation has led to the identification of an alternatively spliced isoform of PD-L1 that lacks exon 3 (PD-L1∆3) and the IgV domain required to interact with PD-1. PD-L1∆3 is expressed more than the canonical isoform in a subset of breast cancers and other TCGA tumors. Using the deep learning-based protein modeling tool AlphaFold2, we show the lack of a possible interaction between PD-L1∆3 and PD-1. In addition, we present data on the expression of an additional ligand for PD-1, PD-L2. PD-L2 expression is widespread and positively correlates with PD-L1 levels in breast and other tumors. We report enriched epithelial-mesenchymal transition (EMT) signature in high PD-L2 transcript expressing (PD-L2 > PD-L1) tumors in all breast cancer subtypes, highlighting potential crosstalk between EMT and immune evasion. Notably, the estrogen gene signature is downregulated in ER + breast tumors with high PD-L2. The data on PD-L2 IHC positivity but PD-L1 negativity in breast tumors, together with our results on PD-L1∆3, highlight the need to utilize PD-L2 and PD-L1 isoform-specific antibodies for staining patient tissue sections to offer a more precise prediction of the outcomes of PD-1/PD-L1 immunotherapy.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Breast Neoplasms/therapy , B7-H1 Antigen/metabolism , Programmed Cell Death 1 Receptor/genetics , Immunotherapy , Protein Isoforms/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism
11.
Blood ; 137(11): 1491-1502, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33512416

ABSTRACT

Intravascular large B-cell lymphoma (IVLBCL) is a unique type of extranodal lymphoma characterized by selective growth of tumor cells in small vessels without lymphadenopathy. Greater understanding of the molecular pathogenesis of IVLBCL is hampered by the paucity of lymphoma cells in biopsy specimens, creating a limitation in obtaining sufficient tumor materials. To uncover the genetic landscape of IVLBCL, we performed whole-exome sequencing (WES) of 21 patients with IVLBCL using plasma-derived cell-free DNA (cfDNA) (n = 18), patient-derived xenograft tumors (n = 4), and tumor DNA from bone marrow (BM) mononuclear cells (n = 2). The concentration of cfDNA in IVLBCL was significantly higher than that in diffuse large B-cell lymphoma (DLBCL) (P < .0001) and healthy donors (P = .0053), allowing us to perform WES; most mutations detected in BM tumor DNA were successfully captured in cfDNA and xenograft. IVLBCL showed a high frequency of genetic lesions characteristic of activated B-cell-type DLBCL, with the former showing conspicuously higher frequencies (compared with nodal DLBCL) of mutations in MYD88 (57%), CD79B (67%), SETD1B (57%), and HLA-B (57%). We also found that 8 IVLBCL (38%) harbored rearrangements of programmed cell death 1 ligand 1 and 2 (PD-L1/PD-L2) involving the 3' untranslated region; such rearrangements are implicated in immune evasion via PD-L1/PD-L2 overexpression. Our data demonstrate the utility of cfDNA and imply important roles for immune evasion in IVLBCL pathogenesis and PD-1/PD-L1/PD-L2 blockade in therapeutics for IVLBCL.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/genetics , Mutation , Tumor Escape , Vascular Neoplasms/genetics , Aged , Aged, 80 and over , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell-Free Nucleic Acids/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, Large B-Cell, Diffuse/immunology , Male , Middle Aged , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Vascular Neoplasms/immunology , Exome Sequencing
12.
Blood ; 137(20): 2785-2799, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33232972

ABSTRACT

Aberrant B-cell receptor/NF-κB signaling is a hallmark feature of B-cell non-Hodgkin lymphomas, especially in diffuse large B-cell lymphoma (DLBCL). Recurrent mutations in this cascade, for example, in CD79B, CARD11, or NFKBIZ, and also in the Toll-like receptor pathway transducer MyD88, all deregulate NF-κB, but their differential impact on lymphoma development and biology remains to be determined. Here, we functionally investigate primary mouse lymphomas that formed in recipient mice of Eµ-myc transgenic hematopoietic stem cells stably transduced with naturally occurring NF-κB mutants. Although most mutants supported Myc-driven lymphoma formation through repressed apoptosis, CARD11- or MyD88-mutant lymphoma cells selectively presented with a macrophage-activating secretion profile, which, in turn, strongly enforced transforming growth factor ß (TGF-ß)-mediated senescence in the lymphoma cell compartment. However, MyD88- or CARD11-mutant Eµ-myc lymphomas exhibited high-level expression of the immune-checkpoint mediator programmed cell death ligand 1 (PD-L1), thus preventing their efficient clearance by adaptive host immunity. Conversely, these mutant-specific dependencies were therapeutically exploitable by anti-programmed cell death 1 checkpoint blockade, leading to direct T-cell-mediated lysis of predominantly but not exclusively senescent lymphoma cells. Importantly, mouse-based mutant MyD88- and CARD11-derived signatures marked DLBCL subgroups exhibiting mirroring phenotypes with respect to the triad of senescence induction, macrophage attraction, and evasion of cytotoxic T-cell immunity. Complementing genomic subclassification approaches, our functional, cross-species investigation unveils pathogenic principles and therapeutic vulnerabilities applicable to and testable in human DLBCL subsets that may inform future personalized treatment strategies.


Subject(s)
Adaptive Immunity , CARD Signaling Adaptor Proteins/genetics , Cellular Senescence/physiology , Guanylate Cyclase/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Myeloid Differentiation Factor 88/genetics , Neoplasm Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , B7-H1 Antigen/antagonists & inhibitors , CD79 Antigens/genetics , Cell Line, Tumor , Chemotaxis , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genes, Reporter , Genes, myc , Humans , Immune Checkpoint Inhibitors , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/therapy , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , NF-kappa B/genetics , NF-kappa B/metabolism , Point Mutation , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Transcriptome
13.
BMC Cancer ; 23(1): 971, 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37828429

ABSTRACT

BACKGROUND: Cancer cells express immunosuppressive molecules, such as programmed death ligands (PD-L)1 and PD-L2, enabling evasion from the host's immune system. Cancer cells synthesize and secrete acetylcholine (ACh), acting as an autocrine or paracrine hormone to promote their proliferation, differentiation, and migration. METHODS: We correlated the expression of PD-L1, PD-L2, cholinergic muscarinic receptor 3 (M3R), alpha 7 nicotinic receptor (α7nAChR), and choline acetyltransferase (ChAT) in colorectal cancer (CRC) tissues with the stage of disease, gender, age, risk, and patient survival. The effects of a muscarinic receptor blocker, atropine, and a selective M3R blocker, 4-DAMP, on the expression of immunosuppressive and cholinergic markers were evaluated in human CRC (LIM-2405, HT-29) cells. RESULTS: Increased expression of PD-L1, M3R, and ChAT at stages III-IV was associated with a high risk of CRC and poor survival outcomes independent of patients' gender and age. α7nAChR and PD-L2 were not changed at any CRC stages. Atropine and 4-DAMP suppressed the proliferation and migration of human CRC cells, induced apoptosis, and decreased PD-L1, PD-L2, and M3R expression in CRC cells via inhibition of EGFR and phosphorylation of ERK. CONCLUSIONS: The expression of immunosuppressive and cholinergic markers may increase the risk of recurrence of CRC. These markers might be used in determining prognosis and treatment regimens for CRC patients. Blocking cholinergic signaling may be a potential therapeutic for CRC through anti-proliferation and anti-migration via inhibition of EGFR and phosphorylation of ERK. These effects allow the immune system to recognize and eliminate cancer cells.


Subject(s)
Colorectal Neoplasms , Immune Checkpoint Inhibitors , Humans , alpha7 Nicotinic Acetylcholine Receptor/genetics , Atropine , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cholinergic Agents , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , ErbB Receptors/metabolism , HT29 Cells , Receptors, Muscarinic/metabolism , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism
14.
J Immunol ; 207(8): 1978-1989, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34535576

ABSTRACT

The inability of T cell-independent type 2 (TI-2) Ags to induce recall responses is a poorly understood facet of humoral immunity, yet critically important for improving vaccines. Using normal and VHB1-8 transgenic mice, we demonstrate that B cell-intrinsic PD-1 expression negatively regulates TI-2 memory B cell (Bmem) generation and reactivation in part through interacting with PDL1 and PDL2 on non-Ag-specific cells. We also identified a significant role for PDL2 expression on Bmems in inhibiting reactivation and Ab production, thereby revealing a novel self-regulatory mechanism exists for TI-2 Bmems This regulation impacts responses to clinically relevant vaccines, because PD-1 deficiency was associated with significantly increased Ab boosting to the pneumococcal vaccine after both vaccination and infection. Notably, we found a B cell-activating adjuvant enabled even greater boosting of protective pneumococcal polysaccharide-specific IgG responses when PD-1 inhibition was relieved. This work highlights unique self-regulation by TI-2 Bmems and reveals new opportunities for significantly improving TI-2 Ag-based vaccine responses.


Subject(s)
B-Lymphocytes/immunology , Pneumococcal Infections/immunology , Pneumococcal Vaccines/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , Animals , B7-H1 Antigen/metabolism , Homeostasis , Immunity, Humoral , Immunogenicity, Vaccine , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Protein Binding , Signal Transduction
15.
Nature ; 543(7645): 378-384, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28112728

ABSTRACT

Cervical cancer remains one of the leading causes of cancer-related deaths worldwide. Here we report the extensive molecular characterization of 228 primary cervical cancers, one of the largest comprehensive genomic studies of cervical cancer to date. We observed notable APOBEC mutagenesis patterns and identified SHKBP1, ERBB3, CASP8, HLA-A and TGFBR2 as novel significantly mutated genes in cervical cancer. We also discovered amplifications in immune targets CD274 (also known as PD-L1) and PDCD1LG2 (also known as PD-L2), and the BCAR4 long non-coding RNA, which has been associated with response to lapatinib. Integration of human papilloma virus (HPV) was observed in all HPV18-related samples and 76% of HPV16-related samples, and was associated with structural aberrations and increased target-gene expression. We identified a unique set of endometrial-like cervical cancers, comprised predominantly of HPV-negative tumours with relatively high frequencies of KRAS, ARID1A and PTEN mutations. Integrative clustering of 178 samples identified keratin-low squamous, keratin-high squamous and adenocarcinoma-rich subgroups. These molecular analyses reveal new potential therapeutic targets for cervical cancers.


Subject(s)
Uterine Cervical Neoplasms/genetics , APOBEC-1 Deaminase/genetics , Adenocarcinoma/genetics , B7-H1 Antigen/genetics , Carcinoma, Squamous Cell/genetics , Caspase 8/genetics , DNA-Binding Proteins , Female , Genomics , HLA-A Antigens/genetics , Human papillomavirus 16/genetics , Human papillomavirus 16/isolation & purification , Humans , Keratins/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Targeted Therapy , Mutation , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Programmed Cell Death 1 Ligand 2 Protein/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteomics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Long Noncoding/genetics , Receptor, ErbB-3/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transcription Factors/genetics , Uterine Cervical Neoplasms/classification , Uterine Cervical Neoplasms/drug therapy , Virus Integration
16.
J Allergy Clin Immunol ; 149(2): 610-623, 2022 02.
Article in English | MEDLINE | ID: mdl-34224786

ABSTRACT

BACKGROUND: Local immunoglobulin hyperproduction is observed in nasal polyps (NPs) with and without ectopic lymphoid tissues (eLTs). OBJECTIVE: Our aim was to identify the T-cell subsets involved in local immunoglobulin production independent of eLTs in NPs. METHODS: The localization, abundance, and phenotype of CD4+ T-cell subsets were studied by immunofluorescence, flow cytometry, and single-cell RNA sequencing. Purified nasal T-cell subsets were cultured with autologous peripheral naive B cells to explore their function. Programmed death ligand 1 and programmed death ligand 2 expression in NPs was investigated by immunofluorescence staining and flow cytometry. RESULTS: Accumulation of PD-1highCXCR5-CD4+ T cells outside lymphoid aggregates was found in NPs. Nasal PD-1highCXCR5-CD4+ T cells were characterized by a unique phenotype that was related to B-cell help and tissue residency and distinct from PD-1-/intCXCR5- and CXCR5+ CD4+ T cells in NPs as well as PD-1highCXCR5highCD4+ follicular helper T cells in tonsils. Compared with the frequencies of PD-1highCXCR5-CD4+ T cells and their IFN-γ+, IL-17A+, and IL-21+ subsets in the control inferior turbinate tissues, the frequencies of these cells and their subsets were increased in both eosinophilic and noneosinophilic NPs, whereas the frequencies of the IL-4+ and IL-4+IL-21+ subsets were increased only in eosinophilic NPs. Nasal PD-1highCXCR5-CD4+ T cells induced immunoglobulin production from B cells in a potency comparable to that induced by tonsillar follicular helper T cells. PD-1highCXCR5-CD4+ T-cell frequencies were correlated with IgE levels in eosinophilic NPs. PD-L1 and PD-L2 suppressed the function of PD-1highCXCR5-CD4+ T cells, and their levels were reduced in NPs. PD-1highCXCR5-CD4+ T-cell abundance was associated with the postsurgical relapse of NPs. CONCLUSION: PD-1highCXCR5-CD4+ T cells participate in local immunoglobulin production independent of eLTs in NPs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunoglobulins/biosynthesis , Nasal Polyps/immunology , Programmed Cell Death 1 Receptor/analysis , Receptors, CXCR5/analysis , B7-H1 Antigen/analysis , Cells, Cultured , Humans , Interleukin-4/biosynthesis , Programmed Cell Death 1 Ligand 2 Protein/analysis
17.
Cancer Sci ; 113(2): 399-410, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34773342

ABSTRACT

Immune checkpoint inhibitors have shown efficacy in various cancers. Although programmed death ligand 1/2 (PD-L1/L2) expressions have been demonstrated as predictive biomarkers of response to immune checkpoint inhibitors and prognostic markers, whether PD-L1/L2 expression is altered in esophageal squamous cell carcinoma during the therapeutic course is unclear. Whether PD-L1/L2 expression in metastatic or recurrent lesions is consistent with that in primary tumors is also unknown. This study included 561 surgically resected esophageal squamous cell carcinomas and PD-L1/L2 expression was evaluated by immunohistochemistry. We investigated the influence of chemotherapeutic drugs (cisplatin and fluorouracil) on PD-L1/L2 expression and PD-L1/L2-related pathways in vitro. We also examined PD-L1/L2 expression in 18 surgically resected lymph node metastases and 10 recurrent lesions compared with primary lesions. The positive rate of PD-L1 was significantly higher in patients with preoperative chemotherapy than in those without preoperative therapy. The positive rate of PD-L2 expression showed no significant difference between patient groups. Cisplatin increased PD-L1 expression in cancer cell lines in vitro, but decreased PD-L2 in some cell lines. The effects of cisplatin on phosphorylated signal transducer and activator of transcription 1/3 (pSTAT1/3) also differed depending on cell lines. Fluorouracil increased PD-L1 and PD-L2 expression. PD-L1/L2 expression in lymph node metastases and recurrent lesions did not always match expression in primary lesions. PD-L1/L2 expression may be altered by preoperative chemotherapy, and PD-L1 /L2 expression in primary lesions does not always match that of metastatic/recurrent lesions. Thus, one-time evaluation is not sufficient to evaluate PD-L1/L2 expression as a biomarker in esophageal cancer.


Subject(s)
B7-H1 Antigen/metabolism , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cisplatin/therapeutic use , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/pathology , Fluorouracil/therapeutic use , Humans , Lymphatic Metastasis , Neoadjuvant Therapy , Neoplasm Recurrence, Local
18.
Cancer Immunol Immunother ; 71(2): 445-459, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34228218

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive tumor type with early dissemination and distant metastasis capacity. Even though optimal chemotherapy responses are observed initially in many patients, therapy resistance is almost inevitable. Accordingly, SCLC has been regarded as an archetype for cancer stem cell (CSC) dynamics. To determine the immune-modulatory influence of CSC in SCLC, this study focused on the characterization of CD44+CD90+ CSC-like subpopulations in SCLC. These cells displayed mesenchymal properties, differentiated into different lineages and further contributed to CD8+ cytotoxic T lymphocytes (CTL) responses. The interaction between CD44+CD90+ CSC-like cells and T cells led to the upregulation of checkpoint molecules PD-1, CTLA-4, TIM-3, and LAG3. In the patient-derived lymph nodes, CD44+ SCLC metastases were also observed with T cells expressing PD-1, TIM-3, or LAG3. Proliferation and IFN-γ expression capacity of TIM-3 and LAG3 co-expressing CTLs are adversely affected over long-time co-culture with CD44+CD90+ CSC-like cells. Moreover, especially through IFN-γ secreted by the T cells, the CSC-like SCLC cells highly expressed PD-L1 and PD-L2. Upon a second encounter with immune-experienced, IFN-γ-stimulated CSC-like SCLC cells, both cytotoxic and proliferation capacities of T cells were hampered. In conclusion, our data provide evidence for the superior potential of the SCLC cells with stem-like and mesenchymal properties to gain immune regulatory capacities and cope with cytotoxic T cell responses. With their high metastatic and immune-modulatory assets, the CSC subpopulation in SCLC may serve as a preferential target for checkpoint blockade immunotherapy .


Subject(s)
B7-H1 Antigen/metabolism , Lung Neoplasms/pathology , Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/pathology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Small Cell Lung Carcinoma/pathology , T-Lymphocytes, Cytotoxic/immunology , Apoptosis , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Humans , Hyaluronan Receptors/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/metabolism , Tumor Cells, Cultured
19.
Rheumatology (Oxford) ; 61(2): 815-825, 2022 02 02.
Article in English | MEDLINE | ID: mdl-33930105

ABSTRACT

OBJECTIVE: To investigate the role of programmed cell death protein 1 (PD-1) and its two ligands, PD-L1 and PD-L2, in the pathogenesis of IgG4-related disease (IgG4-RD). METHODS: Patients with IgG4-RD (n = 43) and healthy controls (n = 34) were recruited. Expression levels of PD-1, PD-L1 and PD-L2 in plasma, submandibular gland and T cell subsets were determined by ELISA, immunohistochemistry and flow cytometry. Naïve T cells were stimulated with or without PD-L1/PD-L2 or anti-PD-L1/anti-PD-L2 for 7 days and the proportion of CD4+CD25+ Treg cells was detected by flow cytometry. RESULTS: The expression of PD-1, PD-L1 and PD-L2 in the plasma, submandibular gland and on the surface of Treg cells was increased in IgG4-RD patients. Plasma soluble (s)PD-1 was positively correlated with serum IgG, IgG1, IgG3, IgG4, IgG4-RD responder index and numbers of organs involved, and negatively correlated with serum IgM, IgA, C3 and C4. Plasma sPD-L2 was positively correlated with serum IgG1, and plasma sPD-L1 was positively correlated with sPD-L2 and negatively correlated with C3. Stimulation of PD-L1 but not PD-L2 promoted the differentiation of naïve T cells from IgG4-RD patients into CD4+CD25+ Treg cells. CONCLUSION: Plasma concentrations of sPD-1, sPD-L1 and sPD-L2 were significantly increased in patients with IgG4-RD, and the expression of PD-1 and PD-L2 on Treg cells was upregulated. PD-1-PD-L1 can promote the differentiation of naïve T cells into Treg cells and thus participate in the pathogenesis of IgG4-RD.


Subject(s)
B7-H1 Antigen/metabolism , Immunoglobulin G4-Related Disease/etiology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/blood , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoglobulin G4-Related Disease/blood , Immunoglobulin G4-Related Disease/metabolism , Programmed Cell Death 1 Ligand 2 Protein/blood , Programmed Cell Death 1 Receptor/blood , Submandibular Gland/metabolism , T-Lymphocytes, Regulatory/metabolism
20.
BMC Cancer ; 22(1): 9, 2022 Jan 03.
Article in English | MEDLINE | ID: mdl-34980000

ABSTRACT

PURPOSE: We sought to understand the clinical course and molecular phenotype of patients who showed disease progression after programmed cell death ligand 1 (PD-L1) inhibitor treatment but subsequently responded to PD-1 inhibitor treatment. We also explored the response to PD-1-axis targeted therapy of classical Hodgkin lymphoma (cHL) according to genetically driven PD-L1 and programmed cell death ligand 2 (PD-L2) expression. METHODS: Five patients in a phase II clinical trial of CS1001 (PD-L1 inhibitor) for relapsed or refractory (R/R) cHL were retrospectively reviewed. Formalin-fixed, paraffin-embedded whole tissues from the five patients were evaluated for 9p24.1 genetic alterations based on FISH and the expression of PD-L1, PD-L2, PD-1, major histocompatibility complex (MHC) class I-II, and the tumor microenvironment factorsCD163 and FOXP3 in the microenvironmental niche, as revealed by multiplex immunofluorescence. RESULTS: All five patients showed primary refractory disease during first-line treatment. Four patients received PD-1 inhibitor after dropping out of the clinical trial, and all demonstrated at least a partial response. The progression-free survival ranged from 7 to 28 months (median = 18 months), and 9p24.1 amplification was observed in all five patients at the PD-L1/PD-L2 locus. PD-L1 and PD-L2 were colocalized on Hodgkin Reed-Sternberg (HRS) cells in four of the five (80%) patients. There was differential expression of PD-L1 and PD-L2 in cells in the tumor microenvironment in cHL, especially in HRS cells, background cells and tumor-associated macrophages. CONCLUSIONS: PD-L1 monotherapy may not be sufficient to block the PD-1 pathway; PD-L2 was expressed in HRS and background cells in cHL. The immunologic function of the PD-L2 pathway in anti-tumor activity may be underestimated in R/R cHL. Further study is needed to elucidate the anti-tumor mechanism of PD-1 inhibitor and PD-L1 inhibitor treatment.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Hodgkin Disease/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , Adult , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Clinical Trials, Phase II as Topic , Female , Forkhead Transcription Factors/immunology , Histocompatibility Antigens/immunology , Hodgkin Disease/immunology , Humans , Male , Middle Aged , Programmed Cell Death 1 Receptor/immunology , Progression-Free Survival , Receptors, Cell Surface/immunology , Recurrence , Retrospective Studies , Treatment Outcome , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL