Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
J Clin Invest ; 132(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36377658

ABSTRACT

Myeloid lineage cells suppress T cell viability through arginine depletion via arginase 1 (ARG1). Despite numerous studies exploring the mechanisms by which ARG1 perturbs lymphocyte function, the cellular populations responsible for its generation and release remain poorly understood. Here, we showed that neutrophil lineage cells and not monocytes or macrophages expressed ARG1 in human non-small cell lung cancer (NSCLC). Importantly, we showed that approximately 40% of tumor-associated neutrophils (TANs) actively transcribed ARG1 mRNA. To determine the mechanism by which ARG1 mRNA is induced in TANs, we utilized FPLC followed by MS/MS to screen tumor-derived factors capable of inducing ARG1 mRNA expression in neutrophils. These studies identified ANXA2 as the major driver of ARG1 mRNA expression in TANs. Mechanistically, ANXA2 signaled through the TLR2/MYD88 axis in neutrophils to induce ARG1 mRNA expression. The current study describes what we believe to be a novel mechanism by which ARG1 mRNA expression is regulated in neutrophils in cancer and highlights the central role that neutrophil lineage cells play in the suppression of tumor-infiltrating lymphocytes.


Subject(s)
Annexin A2 , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Annexin A2/genetics , Arginase/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Neutrophils/metabolism , RNA, Messenger , Tandem Mass Spectrometry , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism
2.
Clin Cancer Res ; 28(20): 4551-4564, 2022 10 14.
Article in English | MEDLINE | ID: mdl-35920742

ABSTRACT

PURPOSE: The addition of immune checkpoint blockade (ICB) to platinum/etoposide chemotherapy changed the standard of care for small cell lung cancer (SCLC) treatment. However, ICB addition only modestly improved clinical outcomes, likely reflecting the high prevalence of an immunologically "cold" tumor microenvironment in SCLC, despite high mutational burden. Nevertheless, some patients clearly benefit from ICB and recent reports have associated clinical responses to ICB in SCLC with (i) decreased neuroendocrine characteristics and (ii) activation of NOTCH signaling. We previously showed that inhibition of the lysine-specific demethylase 1a (LSD1) demethylase activates NOTCH and suppresses neuroendocrine features of SCLC, leading us to investigate whether LSD1 inhibition would enhance the response to PD-1 inhibition in SCLC. EXPERIMENTAL DESIGN: We employed a syngeneic immunocompetent model of SCLC, derived from a genetically engineered mouse model harboring Rb1/Trp53 inactivation, to investigate combining the LSD1 inhibitor bomedemstat with anti-PD-1 therapy. In vivo experiments were complemented by cell-based studies in murine and human models. RESULTS: Bomedemstat potentiated responses to PD-1 inhibition in a syngeneic model of SCLC, resulting in increased CD8+ T-cell infiltration and strong tumor growth inhibition. Bomedemstat increased MHC class I expression in mouse SCLC tumor cells in vivo and augmented MHC-I induction by IFNγ and increased killing by tumor-specific T cells in cell culture. CONCLUSIONS: LSD1 inhibition increased MHC-I expression and enhanced responses to PD-1 inhibition in vivo, supporting a new clinical trial to combine bomedemstat with standard-of-care PD-1 axis inhibition in SCLC.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Animals , Cell Death , Enzyme Inhibitors/therapeutic use , Etoposide/therapeutic use , Histone Demethylases/metabolism , Humans , Immune Checkpoint Inhibitors , Lung Neoplasms/pathology , Lysine/therapeutic use , Mice , Platinum/therapeutic use , Small Cell Lung Carcinoma/pathology , Tumor Microenvironment
3.
JCI Insight ; 4(24)2019 12 19.
Article in English | MEDLINE | ID: mdl-31852845

ABSTRACT

Immune checkpoint inhibitor (ICI) treatment has recently become a first-line therapy for many non-small cell lung cancer (NSCLC) patients. Unfortunately, most NSCLC patients are refractory to ICI monotherapy, and initial attempts to address this issue with secondary therapeutics have proven unsuccessful. To identify entities precluding CD8+ T cell accumulation in this process, we performed unbiased analyses on flow cytometry, gene expression, and multiplexed immunohistochemical data from a NSCLC patient cohort. The results revealed the presence of a myeloid-rich subgroup, which was devoid of CD4+ and CD8+ T cells. Of all myeloid cell types assessed, neutrophils were the most highly associated with the myeloid phenotype. Additionally, the ratio of CD8+ T cells to neutrophils (CD8/PMN) within the tumor mass optimally distinguished between active and myeloid cases. This ratio was also capable of showing the separation of patients responsive to ICI therapy from those with stable or progressive disease in 2 independent cohorts. Tumor-bearing mice treated with a combination of anti-PD1 and SX-682 (CXCR1/2 inhibitor) displayed relocation of lymphocytes from the tumor periphery into a malignant tumor, which was associated with induction of IFN-γ-responsive genes. These results suggest that neutrophil antagonism may represent a viable secondary therapeutic strategy to enhance ICI treatment outcomes.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/immunology , Neutrophils/immunology , Aged , Animals , Antineoplastic Agents, Immunological/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/immunology , Cohort Studies , Datasets as Topic , Disease Models, Animal , Female , Flow Cytometry , Gene Expression Profiling , Humans , Immunohistochemistry , Leukocyte Count , Lung Neoplasms/blood , Lung Neoplasms/immunology , Male , Mice , Middle Aged , Neutrophils/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Treatment Failure
4.
J Autoimmun ; 84: 65-74, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28711285

ABSTRACT

Immunotherapy for type 1 diabetes (T1D) has previously focused on suppressing the autoimmune response against pancreatic beta cells to preserve endogenous insulin production and regulate glucose levels. With increased attention toward combination therapy strategies, studies indicate the multifunctional cytokine interleukin-21 (IL-21) may be a suitable target as an immuno-modulatory arm, while glucagon-like peptide-1 receptor (GLP-1R) agonists may be appropriate as a beta cell protective arm in combination therapy for T1D. We report here that treatment with anti-IL-21 monoclonal antibody delays diabetes onset in the spontaneous non-obese diabetic (NOD) and NOD.scid adoptive transfer models, while its effect in reversing recent-onset hyperglycemia is limited. However, the combination of anti-IL-21 plus the GLP-1R agonist liraglutide is effective in reversing established disease compared to either monotherapy in both the NOD and rat insulin promotor-lymphocytic choriomeningitis virus glycoprotein (RIP-LCMV-GP) models of autoimmune diabetes. Enhanced efficacy is particularly evident in severely hyperglycemic mice, with return to normoglycemia remaining stable for the majority of mice even after therapy is withdrawn. Importantly, increased beta cell proliferation does not appear to be the predominant mechanism. In conclusion, combination therapy with anti-IL-21 and liraglutide is able to consistently reverse disease in mouse models of T1D. The observed effects rival the most effective experimental disease-modifying treatments tested in preclinical studies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus, Type 1/therapy , Hyperglycemia/therapy , Immunotherapy/methods , Insulin-Secreting Cells/immunology , Interleukins/immunology , Liraglutide/therapeutic use , Animals , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Drug Therapy, Combination , Female , Humans , Hyperglycemia/immunology , Insulin/genetics , Insulin/metabolism , Mice , Mice, Inbred NOD , Mice, Transgenic
5.
J Exp Med ; 213(6): 913-20, 2016 05 30.
Article in English | MEDLINE | ID: mdl-27139493

ABSTRACT

T cell development requires a period of postthymic maturation. Why this is the case has remained a mystery, particularly given the rigors of intrathymic developmental checkpoints, successfully traversed by only ∼5% of thymocytes. We now show that the first few weeks of T cell residence in the lymphoid periphery define a period of heightened susceptibility to tolerance induction to tissue-restricted antigens (TRAs), the outcome of which depends on the context in which recent thymic emigrants (RTEs) encounter antigen. After encounter with TRAs in the absence of inflammation, RTEs exhibited defects in proliferation, diminished cytokine production, elevated expression of anergy-associated genes, and diminished diabetogenicity. These properties were mirrored in vitro by enhanced RTE susceptibility to regulatory T cell-mediated suppression. In the presence of inflammation, RTEs and mature T cells were, in contrast, equally capable of inducing diabetes, proliferating, and producing cytokines. Thus, recirculating RTEs encounter TRAs during a transitional developmental stage that facilitates tolerance induction, but inflammation converts antigen-exposed, tolerance-prone RTEs into competent effector cells.


Subject(s)
Cell Movement/immunology , Immune Tolerance/physiology , Immunity, Cellular/physiology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Inflammation/immunology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology
6.
Proc Natl Acad Sci U S A ; 111(15): 5652-7, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24706795

ABSTRACT

Peripheral CD4 T cells in Vß5 transgenic (Tg) C57BL/6J mice undergo tolerance to an endogenous superantigen encoded by mouse mammary tumor virus 8 (Mtv-8) by either deletion or T-cell receptor (TCR) revision. Revision is a process by which surface expression of the Vß5(+) TCR is down-regulated in response to Mtv-8 and recombination activating genes are expressed to drive rearrangement of the endogenous TCRß locus, effecting cell rescue through the expression of a newly generated, non-self-reactive TCR. In an effort to identify the microenvironment in which revision takes place, we show here that the proportion of T follicular helper cells (Tfh) and production of high-affinity antibody during a primary response are increased in Vß5 Tg mice in an Mtv-8-dependent manner. Revising T cells have a Tfh-like surface phenotype and transcription factor profile, with elevated expression of B-cell leukemia/lymphoma 6 (Bcl-6), CXC chemokine receptor 5, programmed death-1, and other Tfh-associated markers. Efficient revision requires Bcl-6 and is inhibited by B lymphocyte-induced maturation protein-1. Revision completes less efficiently in the absence of signaling lymphocytic activation molecule-associated protein although initiation proceeds normally. These data indicate that Tfh formation is required for the initiation of revision and germinal-center interactions for its completion. The germinal center is known to provide a confined space in which B-cell antigen receptors undergo selection. Our data extend the impact of this selective microenvironment into the arena of T cells, suggesting that this fluid structure also provides a regulatory environment in which TCR revision can safely take place.


Subject(s)
Gene Rearrangement, T-Lymphocyte/immunology , Germinal Center/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , DNA Primers/genetics , Flow Cytometry , Mice , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/metabolism , Recombination, Genetic/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Helper-Inducer/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...