Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters








Publication year range
2.
Cell Death Differ ; 31(3): 265-279, 2024 03.
Article in English | MEDLINE | ID: mdl-38383888

ABSTRACT

PirB is an inhibitory cell surface receptor particularly prominent on myeloid cells. PirB curtails the phenotypes of activated macrophages during inflammation or tumorigenesis, but its functions in macrophage homeostasis are obscure. To elucidate PirB-related functions in macrophages at steady-state, we generated and compared single-cell RNA-sequencing (scRNAseq) datasets obtained from myeloid cell subsets of wild type (WT) and PirB-deficient knockout (PirB KO) mice. To facilitate this analysis, we developed a novel approach to clustering parameter optimization called "Cluster Similarity Scoring and Distinction Index" (CaSSiDI). We demonstrate that CaSSiDI is an adaptable computational framework that facilitates tandem analysis of two scRNAseq datasets by optimizing clustering parameters. We further show that CaSSiDI offers more advantages than a standard Seurat analysis because it allows direct comparison of two or more independently clustered datasets, thereby alleviating the need for batch-correction while identifying the most similar and different clusters. Using CaSSiDI, we found that PirB is a novel regulator of Cebpb expression that controls the generation of Ly6Clo patrolling monocytes and the expansion properties of peritoneal macrophages. PirB's effect on Cebpb is tissue-specific since it was not observed in splenic red pulp macrophages (RPMs). However, CaSSiDI revealed a segregation of the WT RPM population into a CD68loIrf8+ "neuronal-primed" subset and an CD68hiFtl1+ "iron-loaded" subset. Our results establish the utility of CaSSiDI for single-cell assay analyses and the determination of optimal clustering parameters. Our application of CaSSiDI in this study has revealed previously unknown roles for PirB in myeloid cell populations. In particular, we have discovered homeostatic functions for PirB that are related to Cebpb expression in distinct macrophage subsets.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Macrophages , Receptors, Immunologic , Single-Cell Analysis , Animals , Mice , Macrophages/metabolism , Monocytes/metabolism , Myeloid Cells/metabolism , Receptors, Cell Surface , Receptors, Immunologic/metabolism , Single-Cell Analysis/methods , CCAAT-Enhancer-Binding Protein-beta/metabolism
3.
Proc Natl Acad Sci U S A ; 120(39): e2307999120, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37729199

ABSTRACT

Asbestos is the main cause of malignant mesothelioma. Previous studies have linked asbestos-induced mesothelioma to the release of HMGB1 from the nucleus to the cytoplasm, and from the cytoplasm to the extracellular space. In the cytoplasm, HMGB1 induces autophagy impairing asbestos-induced cell death. Extracellularly, HMGB1 stimulates the secretion of TNFα. Jointly, these two cytokines kick-start a chronic inflammatory process that over time promotes mesothelioma development. Whether the main source of extracellular HMGB1 were the mesothelial cells, the inflammatory cells, or both was unsolved. This information is critical to identify the targets and design preventive/therapeutic strategies to interfere with asbestos-induced mesothelioma. To address this issue, we developed the conditional mesothelial HMGB1-knockout (Hmgb1ΔpMeso) and the conditional myelomonocytic-lineage HMGB1-knockout (Hmgb1ΔMylc) mouse models. We establish here that HMGB1 is mainly produced and released by the mesothelial cells during the early phases of inflammation following asbestos exposure. The release of HMGB1 from mesothelial cells leads to atypical mesothelial hyperplasia, and in some animals, this evolves over the years into mesothelioma. We found that Hmgb1ΔpMeso, whose mesothelial cells cannot produce HMGB1, show a greatly reduced inflammatory response to asbestos, and their mesothelial cells express and secrete significantly reduced levels of TNFα. Moreover, the tissue microenvironment in areas of asbestos deposits displays an increased fraction of M1-polarized macrophages compared to M2 macrophages. Supporting the biological significance of these findings, Hmgb1ΔpMeso mice showed a delayed and reduced incidence of mesothelioma and an increased mesothelioma-specific survival. Altogether, our study provides a biological explanation for HMGB1 as a driver of asbestos-induced mesothelioma.


Subject(s)
Asbestos , HMGB1 Protein , Mesothelioma, Malignant , Mesothelioma , Animals , Mice , Tumor Necrosis Factor-alpha/genetics , HMGB1 Protein/genetics , Mesothelioma/chemically induced , Mesothelioma/genetics , Asbestos/toxicity , Inflammation , Tumor Microenvironment
4.
Nat Cancer ; 4(10): 1437-1454, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37640929

ABSTRACT

Cholinergic nerves are involved in tumor progression and dissemination. In contrast to other visceral tissues, cholinergic innervation in the hepatic parenchyma is poorly detected. It remains unclear whether there is any form of cholinergic regulation of liver cancer. Here, we show that cholinergic T cells curtail the development of liver cancer by supporting antitumor immune responses. In a mouse multihit model of hepatocellular carcinoma (HCC), we observed activation of the adaptive immune response and induction of two populations of CD4+ T cells expressing choline acetyltransferase (ChAT), including regulatory T cells and dysfunctional PD-1+ T cells. Tumor antigens drove the clonal expansion of these cholinergic T cells in HCC. Genetic ablation of Chat in T cells led to an increased prevalence of preneoplastic cells and exacerbated liver cancer due to compromised antitumor immunity. Mechanistically, the cholinergic activity intrinsic in T cells constrained Ca2+-NFAT signaling induced by T cell antigen receptor engagement. Without this cholinergic modulation, hyperactivated CD25+ T regulatory cells and dysregulated PD-1+ T cells impaired HCC immunosurveillance. Our results unveil a previously unappreciated role for cholinergic T cells in liver cancer immunobiology.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Programmed Cell Death 1 Receptor/genetics , Monitoring, Immunologic , T-Lymphocytes, Regulatory/pathology
5.
J Immunol ; 211(6): 981-993, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37493438

ABSTRACT

Current vaccine efforts to combat SARS-CoV-2 are focused on the whole spike protein administered as mRNA, viral vector, or protein subunit. However, the SARS-CoV-2 receptor-binding domain (RBD) is the immunodominant portion of the spike protein, accounting for 90% of serum neutralizing activity. In this study, we constructed several versions of RBD and together with aluminum hydroxide or DDA (dimethyldioctadecylammonium bromide)/TDB (d-(+)-trehalose 6,6'-dibehenate) adjuvant evaluated immunogenicity in mice. We generated human angiotensin-converting enzyme 2 knock-in mice to evaluate vaccine efficacy in vivo following viral challenge. We found that 1) subdomain (SD)1 was essential for the RBD to elicit maximal immunogenicity; 2) RBDSD1 produced in mammalian HEK cells elicited better immunogenicity than did protein produced in insect or yeast cells; 3) RBDSD1 combined with the CD4 Th1 adjuvant DDA/TDB produced higher neutralizing Ab responses and stronger CD4 T cell responses than did aluminum hydroxide; 4) addition of monomeric human Fc receptor to RBDSD1 (RBDSD1Fc) significantly enhanced immunogenicity and neutralizing Ab titers; 5) the Beta version of RBDSD1Fc provided a broad range of cross-neutralization to multiple antigenic variants of concern, including Omicron; and 6) the Beta version of RBDSD1Fc with DDA/TDB provided complete protection against virus challenge in the knock-in mouse model. Thus, we have identified an optimized RBD-based subunit vaccine suitable for clinical trials.


Subject(s)
COVID-19 , Viral Vaccines , Humans , Animals , Mice , SARS-CoV-2 , COVID-19 Vaccines , Aluminum Hydroxide , Spike Glycoprotein, Coronavirus , Vaccines, Subunit , Antibodies, Viral , Antibodies, Neutralizing , Mammals
6.
Cell Death Differ ; 30(2): 407-416, 2023 02.
Article in English | MEDLINE | ID: mdl-36528755

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a mouse model of multiple sclerosis (MS) in which Th17 cells have a crucial but unclear function. Here we show that choline acetyltransferase (ChAT), which synthesizes acetylcholine (ACh), is a critical driver of pathogenicity in EAE. Mice with ChAT-deficient Th17 cells resist disease progression and show reduced brain-infiltrating immune cells. ChAT expression in Th17 cells is linked to strong TCR signaling, expression of the transcription factor Bhlhe40, and increased Il2, Il17, Il22, and Il23r mRNA levels. ChAT expression in Th17 cells is independent of IL21r signaling but dampened by TGFß, implicating ChAT in controlling the dichotomous nature of Th17 cells. Our study establishes a cholinergic program in which ACh signaling primes chronic activation of Th17 cells, and thereby constitutes a pathogenic determinant of EAE. Our work may point to novel targets for therapeutic immunomodulation in MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Mice , Animals , Th17 Cells , Virulence , Cholinergic Agents , Multiple Sclerosis/genetics , Acetylcholine/metabolism , Mice, Inbred C57BL , Cell Differentiation
7.
Immunity ; 55(12): 2369-2385.e10, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36370712

ABSTRACT

Type I and II interferons (IFNs) stimulate pro-inflammatory programs that are critical for immune activation, but also induce immune-suppressive feedback circuits that impede control of cancer growth. Here, we sought to determine how these opposing programs are differentially induced. We demonstrated that the transcription factor interferon regulatory factor 2 (IRF2) was expressed by many immune cells in the tumor in response to sustained IFN signaling. CD8+ T cell-specific deletion of IRF2 prevented acquisition of the T cell exhaustion program within the tumor and instead enabled sustained effector functions that promoted long-term tumor control and increased responsiveness to immune checkpoint and adoptive cell therapies. The long-term tumor control by IRF2-deficient CD8+ T cells required continuous integration of both IFN-I and IFN-II signals. Thus, IRF2 is a foundational feedback molecule that redirects IFN signals to suppress T cell responses and represents a potential target to enhance cancer control.


Subject(s)
Interferon Type I , Neoplasms , Humans , Interferon Regulatory Factor-2/genetics , CD8-Positive T-Lymphocytes , Transcription Factors , T-Cell Exhaustion , Neoplasms/pathology
8.
Cell Death Differ ; 28(11): 3036-3051, 2021 11.
Article in English | MEDLINE | ID: mdl-34059798

ABSTRACT

The tumor suppressor PTEN is disrupted in a large proportion of cancers, including in HER2-positive breast cancer, where its loss is associated with resistance to therapy. Upon genotoxic stress, ataxia telangiectasia mutated (ATM) is activated and phosphorylates PTEN on residue 398. To elucidate the physiological role of this molecular event, we generated and analyzed knock-in mice expressing a mutant form of PTEN that cannot be phosphorylated by ATM (PTEN-398A). This mutation accelerated tumorigenesis in a model of HER2-positive breast cancer. Mammary tumors in bi-transgenic mice carrying MMTV-neu and Pten398A were characterized by DNA damage accumulation but reduced apoptosis. Mechanistically, phosphorylation of PTEN at position 398 is essential for the proper activation of the S phase checkpoint controlled by the PI3K-p27Kip1-CDK2 axis. Moreover, we linked these defects to the impaired ability of the PTEN-398A protein to relocalize to the plasma membrane in response to genotoxic stress. Altogether, our results uncover a novel role for ATM-dependent PTEN phosphorylation in the control of genomic stability, cell cycle progression, and tumorigenesis.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Breast Neoplasms/genetics , Cell Cycle Checkpoints/genetics , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Carcinogenesis , Cell Cycle , Female , Humans , Mammary Neoplasms, Animal , Mice
9.
Cell Rep ; 35(1): 108931, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33826883

ABSTRACT

Bacterial genotoxins cause DNA damage in eukaryotic cells, resulting in activation of the DNA damage response (DDR) in vitro. These toxins are produced by Gram-negative bacteria, enriched in the microbiota of inflammatory bowel disease (IBD) and colorectal cancer (CRC) patients. However, their role in infection remains poorly characterized. We address the role of typhoid toxin in modulation of the host-microbial interaction in health and disease. Infection with a genotoxigenic Salmonella protects mice from intestinal inflammation. We show that the presence of an active genotoxin promotes DNA fragmentation and senescence in vivo, which is uncoupled from an inflammatory response and unexpectedly associated with induction of an anti-inflammatory environment. The anti-inflammatory response is lost when infection occurs in mice with acute colitis. These data highlight a complex context-dependent crosstalk between bacterial-genotoxin-induced DDR and the host immune response, underlining an unexpected role for bacterial genotoxins.


Subject(s)
Cellular Microenvironment , Host-Pathogen Interactions/immunology , Toxins, Biological/toxicity , Typhoid Fever/immunology , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/metabolism , Cellular Microenvironment/drug effects , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Host-Pathogen Interactions/drug effects , Immunity/drug effects , Inflammation/pathology , Mice, Inbred C57BL , Mutagens/toxicity , Salmonella/physiology
10.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33444287

ABSTRACT

In order to sustain proficient life-long hematopoiesis, hematopoietic stem cells (HSCs) must possess robust mechanisms to preserve their quiescence and genome integrity. DNA-damaging stress can perturb HSC homeostasis by affecting their survival, self-renewal, and differentiation. Ablation of the kinase ataxia telangiectasia mutated (ATM), a master regulator of the DNA damage response, impairs HSC fitness. Paradoxically, we show here that loss of a single allele of Atm enhances HSC functionality in mice. To explain this observation, we explored a possible link between ATM and the tumor suppressor phosphatase and tensin homolog (PTEN), which also regulates HSC function. We generated and analyzed a knockin mouse line (PtenS398A/S398A), in which PTEN cannot be phosphorylated by ATM. Similar to Atm+/-, PtenS398A/S398A HSCs have enhanced hematopoietic reconstitution ability, accompanied by resistance to apoptosis induced by genotoxic stress. Single-cell transcriptomic analyses and functional assays revealed that dormant PtenS398A/S398A HSCs aberrantly tolerate elevated mitochondrial activity and the accumulation of reactive oxygen species, which are normally associated with HSC priming for self-renewal or differentiation. Our results unveil a molecular connection between ATM and PTEN, which couples the response to genotoxic stress and dormancy in HSCs.


Subject(s)
Apoptosis , Cell Differentiation , DNA Damage , Hematopoietic Stem Cells/metabolism , PTEN Phosphohydrolase/metabolism , Amino Acid Substitution , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Mice , Mice, Transgenic , Mutation, Missense , PTEN Phosphohydrolase/genetics
11.
Blood ; 137(7): 945-958, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33254233

ABSTRACT

Isocitrate dehydrogenase (IDH) mutations are common genetic alterations in myeloid disorders, including acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Epigenetic changes, including abnormal histone and DNA methylation, have been implicated in the pathogenic build-up of hematopoietic progenitors, but it is still unclear whether and how IDH mutations themselves affect hematopoiesis. Here, we show that IDH1-mutant mice develop myeloid dysplasia in that these animals exhibit anemia, ineffective erythropoiesis, and increased immature progenitors and erythroblasts. In erythroid cells of these mice, D-2-hydroxyglutarate, an aberrant metabolite produced by the mutant IDH1 enzyme, inhibits oxoglutarate dehydrogenase activity and diminishes succinyl-coenzyme A (CoA) production. This succinyl-CoA deficiency attenuates heme biosynthesis in IDH1-mutant hematopoietic cells, thus blocking erythroid differentiation at the late erythroblast stage and the erythroid commitment of hematopoietic stem cells, while the exogenous succinyl-CoA or 5-ALA rescues erythropoiesis in IDH1-mutant erythroid cells. Heme deficiency also impairs heme oxygenase-1 expression, which reduces levels of important heme catabolites such as biliverdin and bilirubin. These deficits result in accumulation of excessive reactive oxygen species that induce the cell death of IDH1-mutant erythroid cells. Our results clearly show the essential role of IDH1 in normal erythropoiesis and describe how its mutation leads to myeloid disorders. These data thus have important implications for the devising of new treatments for IDH-mutant tumors.


Subject(s)
Erythropoiesis/genetics , Hematopoietic Stem Cells/metabolism , Heme/biosynthesis , Isocitrate Dehydrogenase/genetics , Mutation, Missense , Point Mutation , Preleukemia/genetics , Acyl Coenzyme A/biosynthesis , Acyl Coenzyme A/deficiency , Anemia/genetics , Animals , Bone Marrow/pathology , Erythroblasts/metabolism , Gene Knock-In Techniques , Glutarates/metabolism , Heme/deficiency , Heme Oxygenase-1/metabolism , Isocitrate Dehydrogenase/physiology , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/pathology , Myelopoiesis/genetics , Preleukemia/metabolism , Preleukemia/pathology , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Splenomegaly/etiology , Thrombocytopenia/genetics
12.
Cancer Cell ; 37(3): 308-323.e12, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32142668

ABSTRACT

Diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors for which there is currently no effective treatment. Some of these tumors combine gain-of-function mutations in ACVR1, PIK3CA, and histone H3-encoding genes. The oncogenic mechanisms of action of ACVR1 mutations are currently unknown. Using mouse models, we demonstrate that Acvr1G328V arrests the differentiation of oligodendroglial lineage cells, and cooperates with Hist1h3bK27M and Pik3caH1047R to generate high-grade diffuse gliomas. Mechanistically, Acvr1G328V upregulates transcription factors which control differentiation and DIPG cell fitness. Furthermore, we characterize E6201 as a dual inhibitor of ACVR1 and MEK1/2, and demonstrate its efficacy toward tumor cells in vivo. Collectively, our results describe an oncogenic mechanism of action for ACVR1 mutations, and suggest therapeutic strategies for DIPGs.


Subject(s)
Activin Receptors, Type I/chemistry , Activin Receptors, Type I/genetics , Brain Neoplasms/pathology , Glioma/pathology , Mutation , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Female , Glioma/drug therapy , Glioma/genetics , Histones/genetics , Histones/metabolism , Humans , Lactones/pharmacology , Male , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neuroglia/metabolism , Neuroglia/pathology , Oligodendroglia/pathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
13.
Nat Cancer ; 1(6): 603-619, 2020 06.
Article in English | MEDLINE | ID: mdl-34124690

ABSTRACT

Primary tumor-derived factors (TDFs) act upon normal cells to generate a pre-metastatic niche, which promotes colonization of target organs by disseminated malignant cells. Here we report that TDFs-induced activation of the p38α kinase in lung fibroblasts plays a critical role in the formation of a pre-metastatic niche in the lungs and subsequent pulmonary metastases. Activation of p38α led to inactivation of type I interferon signaling and stimulation of expression of fibroblast activation protein (FAP). FAP played a key role in remodeling of the extracellular matrix as well as inducing the expression of chemokines that enable lung infiltration by neutrophils. Increased activity of p38 in normal cells was associated with metastatic disease and poor prognosis in human melanoma patients whereas inactivation of p38 suppressed lung metastases. We discuss the p38α-driven mechanisms stimulating the metastatic processes and potential use of p38 inhibitors in adjuvant therapy of metastatic cancers.


Subject(s)
Lung Neoplasms , Signal Transduction , Fibroblasts/pathology , Humans , Lung/pathology , Lung Neoplasms/pathology , Protein Kinases
15.
Nat Commun ; 10(1): 2678, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31213601

ABSTRACT

Myeloid cells contribute to tumor progression, but how the constellation of receptors they express regulates their functions within the tumor microenvironment (TME) is unclear. We demonstrate that Fcmr (Toso), the putative receptor for soluble IgM, modulates myeloid cell responses to cancer. In a syngeneic melanoma model, Fcmr ablation in myeloid cells suppressed tumor growth and extended mouse survival. Fcmr deficiency increased myeloid cell population density in this malignancy and enhanced anti-tumor immunity. Single-cell RNA sequencing of Fcmr-deficient tumor-associated mononuclear phagocytes revealed a unique subset with enhanced antigen processing/presenting properties. Conversely, Fcmr activity negatively regulated the activation and migratory capacity of myeloid cells in vivo, and T cell activation by bone marrow-derived dendritic cells in vitro. Therapeutic targeting of Fcmr during oncogenesis decreased tumor growth when used as a single agent or in combination with anti-PD-1. Thus, Fcmr regulates myeloid cell activation within the TME and may be a potential therapeutic target.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carrier Proteins/metabolism , Melanoma, Experimental/immunology , Membrane Proteins/metabolism , Monocytes/immunology , Skin Neoplasms/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Line, Tumor/transplantation , Cell Movement/drug effects , Cell Movement/immunology , Female , Lymphocyte Activation/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Analysis , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
16.
Proc Natl Acad Sci U S A ; 116(10): 4326-4335, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30770442

ABSTRACT

The combination of immune checkpoint blockade with chemotherapy is currently under investigation as a promising strategy for the treatment of triple negative breast cancer (TNBC). Tumor-associated macrophages (TAMs) are the most prominent component of the breast cancer microenvironment because they influence tumor progression and the response to therapies. Here we show that macrophages acquire an immunosuppressive phenotype and increase the expression of programmed death ligand-1 (PD-L1) when treated with reactive oxygen species (ROS) inducers such as the glutathione synthesis inhibitor, buthionine sulphoximine (BSO), and paclitaxel. Mechanistically, these agents cause accumulation of ROS that in turn activate NF-κB signaling to promote PD-L1 transcription and the release of immunosuppressive chemokines. Systemic in vivo administration of paclitaxel promotes PD-L1 accumulation on the surface of TAMS in a mouse model of TNBC, consistent with in vitro results. Combinatorial treatment with paclitaxel and an anti-mouse PD-L1 blocking antibody significantly improved the therapeutic efficacy of paclitaxel by reducing tumor burden and increasing the number of tumor-associated cytotoxic T cells. Our results provide a strong rationale for the use of anti-PD-L1 blockade in the treatment of TNBC patients. Furthermore, interrogation of chemotherapy-induced PD-L1 expression in TAMs is warranted to define appropriate patient selection in the use of PD-L1 blockade.


Subject(s)
B7-H1 Antigen/metabolism , Immunosuppressive Agents/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Animals , B7-H1 Antigen/genetics , Breast Neoplasms/metabolism , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Chemokines , Drug Therapy , Female , Glutathione/metabolism , Humans , Mice , Paclitaxel/pharmacology , Phenotype , RNA, Messenger/metabolism , Triple Negative Breast Neoplasms , Tumor Microenvironment , Up-Regulation
17.
Proc Natl Acad Sci U S A ; 116(9): 3604-3613, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30733286

ABSTRACT

Cancer cells have higher reactive oxygen species (ROS) than normal cells, due to genetic and metabolic alterations. An emerging scenario is that cancer cells increase ROS to activate protumorigenic signaling while activating antioxidant pathways to maintain redox homeostasis. Here we show that, in basal-like and BRCA1-related breast cancer (BC), ROS levels correlate with the expression and activity of the transcription factor aryl hydrocarbon receptor (AhR). Mechanistically, ROS triggers AhR nuclear accumulation and activation to promote the transcription of both antioxidant enzymes and the epidermal growth factor receptor (EGFR) ligand, amphiregulin (AREG). In a mouse model of BRCA1-related BC, cancer-associated AhR and AREG control tumor growth and production of chemokines to attract monocytes and activate proangiogenic function of macrophages in the tumor microenvironment. Interestingly, the expression of these chemokines as well as infiltration of monocyte-lineage cells (monocyte and macrophages) positively correlated with ROS levels in basal-like BC. These data support the existence of a coordinated link between cancer-intrinsic ROS regulation and the features of tumor microenvironment. Therapeutically, chemical inhibition of AhR activity sensitizes human BC models to Erlotinib, a selective EGFR tyrosine kinase inhibitor, suggesting a promising combinatorial anticancer effect of AhR and EGFR pathway inhibition. Thus, AhR represents an attractive target to inhibit redox homeostasis and modulate the tumor promoting microenvironment of basal-like and BRCA1-associated BC.


Subject(s)
Amphiregulin/genetics , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Receptors, Aryl Hydrocarbon/genetics , Adult , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , ErbB Receptors/genetics , Erlotinib Hydrochloride/administration & dosage , Female , Gene Expression Regulation, Neoplastic , Homeostasis/genetics , Humans , Mice , Middle Aged , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Tumor Microenvironment/genetics
18.
Science ; 363(6427): 639-644, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30733420

ABSTRACT

Although widely studied as a neurotransmitter, T cell-derived acetylcholine (ACh) has recently been reported to play an important role in regulating immunity. However, the role of lymphocyte-derived ACh in viral infection is unknown. Here, we show that the enzyme choline acetyltransferase (ChAT), which catalyzes the rate-limiting step of ACh production, is robustly induced in both CD4+ and CD8+ T cells during lymphocytic choriomeningitis virus (LCMV) infection in an IL-21-dependent manner. Deletion of Chat within the T cell compartment in mice ablated vasodilation in response to infection, impaired the migration of antiviral T cells into infected tissues, and ultimately compromised the control of chronic LCMV clone 13 infection. Our results reveal a genetic proof of function for ChAT in T cells during viral infection and identify a pathway of T cell migration that sustains antiviral immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Choline O-Acetyltransferase/immunology , Interleukins/immunology , Lymphocytic Choriomeningitis/immunology , Animals , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/enzymology , Cell Movement , Choline O-Acetyltransferase/genetics , Female , Lymphocyte Activation , Lymphocytic choriomeningitis virus , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Vasodilation
20.
Nat Commun ; 8: 14003, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28084302

ABSTRACT

T-cell proliferation is regulated by ubiquitination but the underlying molecular mechanism remains obscure. Here we report that Lys-48-linked ubiquitination of the transcription factor KLF4 mediated by the E3 ligase Mule promotes T-cell entry into S phase. Mule is elevated in T cells upon TCR engagement, and Mule deficiency in T cells blocks proliferation because KLF4 accumulates and drives upregulation of its transcriptional targets E2F2 and the cyclin-dependent kinase inhibitors p21 and p27. T-cell-specific Mule knockout (TMKO) mice develop exacerbated experimental autoimmune encephalomyelitis (EAE), show impaired generation of antigen-specific CD8+ T cells with reduced cytokine production, and fail to clear LCMV infections. Thus, Mule-mediated ubiquitination of the novel substrate KLF4 regulates T-cell proliferation, autoimmunity and antiviral immune responses in vivo.


Subject(s)
Cell Proliferation , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Animals , Female , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL