Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Sci Transl Med ; 15(711): eadd9990, 2023 08 30.
Article in English | MEDLINE | ID: mdl-37647386

ABSTRACT

Myeloid cells in the tumor microenvironment (TME) can exist in immunosuppressive and immunostimulatory states that impede or promote antitumor immunity, respectively. Blocking suppressive myeloid cells or increasing stimulatory cells to enhance antitumor immune responses is an area of interest for therapeutic intervention. Triggering receptor expressed on myeloid cells-1 (TREM1) is a proinflammatory receptor that amplifies immune responses. TREM1 is expressed on neutrophils, subsets of monocytes and tissue macrophages, and suppressive myeloid populations in the TME, including tumor-associated neutrophils, monocytes, and tumor-associated macrophages. Depletion or inhibition of immunosuppressive myeloid cells, or stimulation by TREM1-mediated inflammatory signaling, could be used to promote an immunostimulatory TME. We developed PY159, an afucosylated humanized anti-TREM1 monoclonal antibody with enhanced FcγR binding. PY159 is a TREM1 agonist that induces signaling, leading to up-regulation of costimulatory molecules on monocytes and macrophages, production of proinflammatory cytokines and chemokines, and enhancement of T cell activation in vitro. An antibody against mouse TREM1, PY159m, promoted antitumor efficacy in syngeneic mouse tumor models. These results suggest that PY159-mediated agonism of TREM1 on tumoral myeloid cells can promote a proinflammatory TME and offer a promising strategy for immunotherapy.


Subject(s)
Monocytes , Myeloid Cells , Animals , Mice , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Immunosuppressive Agents , Macrophages , Triggering Receptor Expressed on Myeloid Cells-1
2.
Cell Rep ; 42(6): 112582, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37261951

ABSTRACT

Pre-metastatic niche formation is a critical step during the metastatic spread of cancer. One way by which primary tumors prime host cells at future metastatic sites is through the shedding of tumor-derived microparticles as a consequence of vascular sheer flow. However, it remains unclear how the uptake of such particles by resident immune cells affects their phenotype and function. Here, we show that ingestion of tumor-derived microparticles by macrophages induces a rapid metabolic and phenotypic switch that is characterized by enhanced mitochondrial mass and function, increased oxidative phosphorylation, and upregulation of adhesion molecules, resulting in reduced motility in the early metastatic lung. This reprogramming event is dependent on signaling through the mTORC1, but not the mTORC2, pathway and is induced by uptake of tumor-derived microparticles. Together, these data support a mechanism by which uptake of tumor-derived microparticles induces reprogramming of macrophages to shape their fate and function in the early metastatic lung.


Subject(s)
Lung Neoplasms , Neoplasms , Humans , Macrophages/pathology , Lung/pathology , Neoplasms/pathology , Signal Transduction , Biological Transport , Lung Neoplasms/pathology
3.
Cancer Immunol Res ; 10(4): 403-419, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35181780

ABSTRACT

The tumor immune microenvironment (TIME) is commonly infiltrated by diverse collections of myeloid cells. Yet, the complexity of myeloid-cell identity and plasticity has challenged efforts to define bona fide populations and determine their connections to T-cell function and their relationship to patient outcome. Here, we have leveraged single-cell RNA-sequencing analysis of several mouse and human tumors and found that monocyte-macrophage diversity is characterized by a combination of conserved lineage states as well as transcriptional programs accessed along the differentiation trajectory. We also found in mouse models that tumor monocyte-to-macrophage progression was profoundly tied to regulatory T cell (Treg) abundance. In human kidney cancer, heterogeneity in macrophage accumulation and myeloid composition corresponded to variance in, not only Treg density, but also the quality of infiltrating CD8+ T cells. In this way, holistic analysis of monocyte-to-macrophage differentiation creates a framework for critically different immune states.


Subject(s)
Kidney Neoplasms , Monocytes , Animals , Macrophages , Mice , Phenotype , Tumor Microenvironment
4.
Cell Rep ; 37(3): 109844, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686340

ABSTRACT

Converting checkpoint inhibitor (CPI)-resistant individuals to being responsive requires identifying suppressive mechanisms. We identify TREM2+ tumor-associated macrophages (TAMs) as being correlated with exhausted CD8+ tumor-infiltrating lymphocytes (TILs) in mouse syngeneic tumor models and human solid tumors of multiple histological types. Fc domain-enhanced anti-TREM2 monoclonal antibody (mAb) therapy promotes anti-tumor immunity by elimination and modulation of TAM populations, which leads to enhanced CD8+ TIL infiltration and effector function. TREM2+ TAMs are most enriched in individuals with ovarian cancer, where TREM2 expression corresponds to disease grade accompanied by worse recurrence-free survival. In an aggressive orthotopic ovarian cancer model, anti-TREM2 mAb therapy drives potent anti-tumor immunity. These results highlight TREM2 as a highly attractive target for immunotherapy modulation in individuals who are refractory to CPI therapy and likely have a TAM-rich tumor microenvironment.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Neoplasms/drug therapy , Receptors, Immunologic/antagonists & inhibitors , Tumor-Associated Macrophages/drug effects , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Coculture Techniques , Drug Resistance, Neoplasm , Female , HEK293 Cells , Humans , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Membrane Glycoproteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
5.
Front Immunol ; 10: 1611, 2019.
Article in English | MEDLINE | ID: mdl-31402908

ABSTRACT

The tumor microenvironment (TME) of diverse cancer types is often characterized by high levels of infiltrating myeloid cells including monocytes, macrophages, dendritic cells, and granulocytes. These cells perform a variety of functions in the TME, varying from immune suppressive to immune stimulatory roles. In this review, we summarize the different myeloid cell populations in the TME and the intratumoral myeloid targeting approaches that are being clinically investigated, and discuss strategies that identify new myeloid subpopulations within the TME. The TME therapies include agents that modulate the functional activities of myeloid populations, that impact recruitment and survival of myeloid subpopulations, and that functionally reprogram or activate myeloid populations. We discuss the benefits, limitations and potential side effects of these therapeutic approaches.


Subject(s)
Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/etiology , Neoplasms/pathology , Tumor Microenvironment , Animals , Biomarkers , Cell Differentiation , Cell Proliferation , Cell Survival , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunomodulation , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Tumor Microenvironment/immunology
6.
Cell ; 177(3): 556-571.e16, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30955881

ABSTRACT

Differentiation of proinflammatory CD4+ conventional T cells (Tconv) is critical for productive antitumor responses yet their elicitation remains poorly understood. We comprehensively characterized myeloid cells in tumor draining lymph nodes (tdLN) of mice and identified two subsets of conventional type-2 dendritic cells (cDC2) that traffic from tumor to tdLN and present tumor-derived antigens to CD4+ Tconv, but then fail to support antitumor CD4+ Tconv differentiation. Regulatory T cell (Treg) depletion enhanced their capacity to elicit strong CD4+ Tconv responses and ensuing antitumor protection. Analogous cDC2 populations were identified in patients, and as in mice, their abundance relative to Treg predicts protective ICOS+ PD-1lo CD4+ Tconv phenotypes and survival. Further, in melanoma patients with low Treg abundance, intratumoral cDC2 density alone correlates with abundant CD4+ Tconv and with responsiveness to anti-PD-1 therapy. Together, this highlights a pathway that restrains cDC2 and whose reversal enhances CD4+ Tconv abundance and controls tumor growth.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Animals , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Line, Tumor , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Diphtheria Toxin/immunology , Forkhead Transcription Factors/metabolism , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Chemokine/metabolism , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment
7.
Am J Pathol ; 189(2): 258-271, 2019 02.
Article in English | MEDLINE | ID: mdl-30448409

ABSTRACT

Recent fate-mapping studies in mice have provided substantial evidence that mature adult hepatocytes are a major source of new hepatocytes after liver injury. In other systems, integrin αvß8 has a major role in activating transforming growth factor (TGF)-ß, a potent inhibitor of hepatocyte proliferation. We hypothesized that depletion of hepatocyte integrin αvß8 would increase hepatocyte proliferation and accelerate liver regeneration after injury. Using Itgb8flox/flox;Alb-Cre mice to deplete hepatocyte αvß8, after partial hepatectomy, hepatocyte proliferation and liver-to-body weight ratio were significantly increased in Itgb8flox/flox;Alb-Cre mice compared with control mice. Antibody-mediated blockade of hepatocyte αvß8 in vitro, with assessment of TGF-ß signaling pathways by real-time quantitative PCR array, supported the hypothesis that integrin αvß8 inhibition alters hepatocyte TGF-ß signaling toward a pro-regenerative phenotype. A diethylnitrosamine-induced model of hepatocellular carcinoma, used to examine the possibility that this pro-proliferative phenotype might be oncogenic, revealed no difference in either tumor number or size between Itgb8flox/flox;Alb-Cre and control mice. Immunohistochemistry for integrin αvß8 in healthy and injured human liver demonstrated that human hepatocytes express integrin αvß8. Depletion of hepatocyte integrin αvß8 results in increased hepatocyte proliferation and accelerated liver regeneration after partial hepatectomy in mice. These data demonstrate that targeting integrin αvß8 may represent a promising therapeutic strategy to drive liver regeneration in patients with a broad range of liver diseases.


Subject(s)
Cell Proliferation , Hepatocytes/metabolism , Integrins/deficiency , Liver Regeneration , Liver/metabolism , Signal Transduction , Animals , Hepatocytes/pathology , Liver/pathology , Mice , Mice, Transgenic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
8.
Immunity ; 49(1): 33-41.e7, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30021144

ABSTRACT

In the small intestine, type 2 responses are regulated by a signaling circuit that involves tuft cells and group 2 innate lymphoid cells (ILC2s). Here, we identified the microbial metabolite succinate as an activating ligand for small intestinal (SI) tuft cells. Sequencing analyses of tuft cells isolated from the small intestine, gall bladder, colon, thymus, and trachea revealed that expression of tuft cell chemosensory receptors is tissue specific. SI tuft cells expressed the succinate receptor (SUCNR1), and providing succinate in drinking water was sufficient to induce a multifaceted type 2 immune response via the tuft-ILC2 circuit. The helminth Nippostrongylus brasiliensis and a tritrichomonad protist both secreted succinate as a metabolite. In vivo sensing of the tritrichomonad required SUCNR1, whereas N. brasiliensis was SUCNR1 independent. These findings define a paradigm wherein tuft cells monitor microbial metabolites to initiate type 2 immunity and suggest the existence of other sensing pathways triggering the response to helminths.


Subject(s)
Immunity, Mucosal/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Succinic Acid/pharmacology , Animals , Cell Line , Female , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Intestine, Small/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/drug effects , Nippostrongylus/immunology , Nippostrongylus/metabolism , Organ Specificity , Protozoan Infections/immunology , Receptors, G-Protein-Coupled/immunology , Signal Transduction/immunology , Species Specificity , Strongylida Infections/immunology , TRPM Cation Channels/metabolism , Th2 Cells/immunology , Tritrichomonas/drug effects , Tritrichomonas/immunology , Tritrichomonas/metabolism
9.
Nature ; 559(7715): 627-631, 2018 07.
Article in English | MEDLINE | ID: mdl-30022164

ABSTRACT

The thymus is responsible for generating a diverse yet self-tolerant pool of T cells1. Although the thymic medulla consists mostly of developing and mature AIRE+ epithelial cells, recent evidence has suggested that there is far greater heterogeneity among medullary thymic epithelial cells than was previously thought2. Here we describe in detail an epithelial subset that is remarkably similar to peripheral tuft cells that are found at mucosal barriers3. Similar to the periphery, thymic tuft cells express the canonical taste transduction pathway and IL-25. However, they are unique in their spatial association with cornified aggregates, ability to present antigens and expression of a broad diversity of taste receptors. Some thymic tuft cells pass through an Aire-expressing stage and depend on a known AIRE-binding partner, HIPK2, for their development. Notably, the taste chemosensory protein TRPM5 is required for their thymic function through which they support the development and polarization of thymic invariant natural killer T cells and act to establish a medullary microenvironment that is enriched in the type 2 cytokine, IL-4. These findings indicate that there is a compartmentalized medullary environment in which differentiation of a minor and highly specialized epithelial subset has a non-redundant role in shaping thymic function.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Interleukin-4/metabolism , Thymocytes/cytology , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Cellular Microenvironment , Doublecortin-Like Kinases , Female , Humans , Immune Tolerance/immunology , Interleukin-4/biosynthesis , Interleukins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism , Thymocytes/metabolism , Thymus Gland/anatomy & histology , Transcription Factors/deficiency , Transcription Factors/genetics , AIRE Protein
10.
Nat Med ; 24(8): 1178-1191, 2018 08.
Article in English | MEDLINE | ID: mdl-29942093

ABSTRACT

Intratumoral stimulatory dendritic cells (SDCs) play an important role in stimulating cytotoxic T cells and driving immune responses against cancer. Understanding the mechanisms that regulate their abundance in the tumor microenvironment (TME) could unveil new therapeutic opportunities. We find that in human melanoma, SDC abundance is associated with intratumoral expression of the gene encoding the cytokine FLT3LG. FLT3LG is predominantly produced by lymphocytes, notably natural killer (NK) cells in mouse and human tumors. NK cells stably form conjugates with SDCs in the mouse TME, and genetic and cellular ablation of NK cells in mice demonstrates their importance in positively regulating SDC abundance in tumor through production of FLT3L. Although anti-PD-1 'checkpoint' immunotherapy for cancer largely targets T cells, we find that NK cell frequency correlates with protective SDCs in human cancers, with patient responsiveness to anti-PD-1 immunotherapy, and with increased overall survival. Our studies reveal that innate immune SDCs and NK cells cluster together as an excellent prognostic tool for T cell-directed immunotherapy and that these innate cells are necessary for enhanced T cell tumor responses, suggesting this axis as a target for new therapies.


Subject(s)
Dendritic Cells/immunology , Immunotherapy , Killer Cells, Natural/immunology , Tumor Microenvironment/immunology , Antigens, Surface/metabolism , Cell Communication , Cell Survival , Humans , Lymphocytes/metabolism , Melanoma/immunology , Melanoma/pathology , Membrane Proteins/metabolism , Survival Analysis , Thrombomodulin
11.
Can J Diabetes ; 42(5): 493-499.e1, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29555342

ABSTRACT

OBJECTIVES: There is a lack of Canadian data concerning the effectiveness of diabetes interventions in the workplace. The objective of this study was to evaluate the effectiveness of Motivaction, a diabetes screening and education pilot program, in the workplace. METHODS: The Motivaction program involves a voluntary web-based diabetes health-risk assessment, the Canadian Diabetes Risk Questionnaire (CANRISK), combined with an opportunity for those eligible (i.e. having diabetes or having a CANRISK score ≥21) to attend 2 on-site biometric screening meetings with a registered nurse and 4 educational sessions by telephone with a certified diabetes educator. Biometric data, as well as information about self-efficacy, lifestyle changes, productivity, well-being, mental health and program satisfaction, were collected at baseline and at 6 months. RESULTS: Attendance at the initial and 6-month clinical visits included 293 people. At baseline, 21% were identified as having prediabetes (13%) or having diabetes (8%). Statistically significant reductions in glycated hemoglobin levels from baseline to the study's end were observed in those with prediabetes or diabetes. No statistically significant changes in glycated hemoglobin levels were observed in individuals with normal levels or in those at risk for diabetes at baseline. No statistical differences were observed in terms of productivity or mental health for the full population or across diabetes-risk categories. More than 90% of employees would recommend the Motivaction program to other employers. CONCLUSIONS: This study provides a framework for future diabetes interventions in the workplace and demonstrates that workplace interventions may reduce employees' diabetes risk levels and are valued by employees.


Subject(s)
Diabetes Mellitus, Type 2/diagnosis , Mass Screening , Workplace/statistics & numerical data , Adult , Canada/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Female , Humans , Life Style , Male , Mass Screening/methods , Mass Screening/standards , Mass Screening/statistics & numerical data , Middle Aged , Occupational Health Services/statistics & numerical data , Personal Satisfaction , Pilot Projects , Program Evaluation , Surveys and Questionnaires
12.
Am J Respir Crit Care Med ; 197(3): 313-324, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29064281

ABSTRACT

RATIONALE: Quantification of type 2 inflammation provided a molecular basis for heterogeneity in asthma. Non-type 2 pathways that contribute to asthma pathogenesis are not well understood. OBJECTIVES: To identify dysregulated pathways beyond type 2 inflammation. METHODS: We applied RNA sequencing to airway epithelial brushings obtained from subjects with stable mild asthma not on corticosteroids (n = 19) and healthy control subjects (n = 16). Sequencing reads were mapped to human and viral genomes. In the same cohort, and in a separate group with severe asthma (n = 301), we profiled blood gene expression with microarrays. MEASUREMENTS AND MAIN RESULTS: In airway brushings from mild asthma on inhaled corticosteroids, RNA sequencing yielded 1,379 differentially expressed genes (false discovery rate < 0.01). Pathway analysis revealed increased expression of type 2 markers, IFN-stimulated genes (ISGs), and endoplasmic reticulum (ER) stress-related genes. Airway epithelial ISG expression was not associated with type 2 inflammation in asthma or with viral transcripts but was associated with reduced lung function by FEV1 (ρ = -0.72; P = 0.0004). ER stress was confirmed by an increase in XBP1 (X-box binding protein 1) splicing in mild asthma and was associated with both type 2 inflammation and ISG expression. ISGs were also the most activated genes in blood cells in asthma and were correlated with airway ISG expression (ρ = 0.55; P = 0.030). High blood ISG expression in severe asthma was similarly unrelated to type 2 inflammation. CONCLUSIONS: ISG activation is prominent in asthma, independent of viral transcripts, orthogonal to type 2 inflammation, and associated with distinct clinical features. ER stress is associated with both type 2 inflammation and ISG expression.


Subject(s)
Asthma/genetics , Asthma/physiopathology , Endoplasmic Reticulum/genetics , Gene Expression Regulation , Interferon Regulatory Factor-3/genetics , Adult , Case-Control Studies , Eosinophils/immunology , Female , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Oxidative Stress/genetics , RNA/genetics , Reference Values , Sensitivity and Specificity , Signal Transduction
13.
Can J Diabetes ; 42(3): 289-295, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28958785

ABSTRACT

OBJECTIVES: Type 2 diabetes mellitus (DM) is associated with substantial impairments in workplace function; however, the mediators of workplace performance in DM are not well characterized. Herein, we investigate depressive symptomatology and cognitive dysfunction as mediators of workplace productivity and hypothesize that workplace impairment is mediated principally by cognitive dysfunction in DM. METHODS: The Motivaction study screened individuals employed across Canada for diabetes. Subjects with impairments in glucose regulation indicative of risk for (i.e. glycated hemoglobin [A1C] levels 5.5% to 5.9%) or the presence of (i.e. A1C≥6.0%) DM were assessed on measures of depressive symptom severity [Patient Health Questionnaire, 9-item (PHQ-9)], self-rated cognitive impairment [Perceived Deficits Questionnaire, 5-item (PDQ-5)], and workplace impairment [Endicott Work Productivity Scale (EWPS)]. Multivariate regression and mediational analyses assessed for correlations between PHQ-9, PDQ-5 and EWPS total scores and the mediational role of self-reported cognitive dysfunction on the effect of depressive symptom severity on workplace impairment, respectively. RESULTS: A total of 3627 individuals were screened, 1738 met eligibility criteria and 724 consented to the Motivaction study; 205 subjects with impaired glucose regulation were included in the analysis. Self-rated depressive and cognitive symptoms were positively correlated with workplace impairment among subjects with or at risk for DM. The deleterious effects of depressive symptomatology on workplace effectiveness was mediated principally by cognitive dysfunction in subjects with impaired glucose tolerance. CONCLUSIONS: Among employed Canadians, impaired glucose tolerance is highly associated with impaired workplace performance. We report a novel finding insofar as the association between depressive symptoms and workplace impairment in individuals with impaired glucose regulation is mediated principally by self-rated cognitive dysfunction.


Subject(s)
Cognitive Dysfunction , Depression , Diabetes Mellitus, Type 2 , Work Performance/statistics & numerical data , Canada/epidemiology , Cognitive Dysfunction/epidemiology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Cross-Sectional Studies , Depression/epidemiology , Depression/etiology , Depression/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Female , Humans , Male , Mass Screening , Middle Aged , Surveys and Questionnaires
14.
Nat Immunol ; 17(12): 1381-1387, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27749840

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) and CD4+ type 2 helper T cells (TH2 cells) are defined by their similar effector cytokines, which together mediate the features of allergic immunity. We found that tissue ILC2s and TH2 cells differentiated independently but shared overlapping effector function programs that were mediated by exposure to the tissue-derived cytokines interleukin 25 (IL-25), IL-33 and thymic stromal lymphopoietin (TSLP). Loss of these three tissue signals did not affect lymph node priming, but abrogated the terminal differentiation of effector TH2 cells and adaptive lung inflammation in a T cell-intrinsic manner. Our findings suggest a mechanism by which diverse perturbations can activate type 2 immunity and reveal a shared local-tissue-elicited checkpoint that can be exploited to control both innate and adaptive allergic inflammation.


Subject(s)
Cytokines/metabolism , Hypersensitivity/immunology , Immunity, Innate , Interleukin-17/metabolism , Interleukin-33/metabolism , Lymphocytes/immunology , Th2 Cells/immunology , Adaptive Immunity , Allergens/immunology , Animals , Aspergillus niger , Bee Venoms/immunology , Bees , Cell Differentiation , Cells, Cultured , Cytokines/genetics , Dermatophagoides farinae , Interleukin-17/genetics , Interleukin-33/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Thymic Stromal Lymphopoietin
15.
PLoS One ; 10(8): e0135440, 2015.
Article in English | MEDLINE | ID: mdl-26270036

ABSTRACT

Thymic epithelial cells (TECs) support T cell development in the thymus. Cortical thymic epithelial cells (cTECs) facilitate positive selection of developing thymocytes whereas medullary thymic epithelial cells (mTECs) facilitate the deletion of self-reactive thymocytes in order to prevent autoimmunity. The mTEC compartment is highly dynamic with continuous maturation and turnover, but the genetic regulation of these processes remains poorly understood. MicroRNAs (miRNAs) are important regulators of TEC genetic programs since miRNA-deficient TECs are severely defective. However, the individual miRNAs important for TEC maintenance and function and their mechanisms of action remain unknown. Here, we demonstrate that miR-205 is highly and preferentially expressed in mTECs during both thymic ontogeny and in the postnatal thymus. This distinct expression is suggestive of functional importance for TEC biology. Genetic ablation of miR-205 in TECs, however, neither revealed a role for miR-205 in TEC function during homeostatic conditions nor during recovery from thymic stress conditions. Thus, despite its distinct expression, miR-205 on its own is largely dispensable for mTEC biology.


Subject(s)
Epithelial Cells/metabolism , Gene Expression Regulation/physiology , MicroRNAs/biosynthesis , Thymus Gland/metabolism , Animals , Epithelial Cells/cytology , Mice , Mice, Transgenic , MicroRNAs/genetics , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology
16.
Cancer Cell ; 26(5): 638-52, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25446897

ABSTRACT

It is well understood that antigen-presenting cells (APCs) within tumors typically do not maintain cytotoxic T cell (CTL) function, despite engaging them. Across multiple mouse tumor models and human tumor biopsies, we have delineated the intratumoral dendritic cell (DC) populations as distinct from macrophage populations. Within these, CD103(+) DCs are extremely sparse and yet remarkably capable CTL stimulators. These are uniquely dependent on IRF8, Zbtb46, and Batf3 transcription factors and are generated by GM-CSF and FLT3L cytokines. Regressing tumors have higher proportions of these cells, T-cell-dependent immune clearance relies on them, and abundance of their transcripts in human tumors correlates with clinical outcome. This cell type presents opportunities for prognostic and therapeutic approaches across multiple cancer types.


Subject(s)
Dendritic Cells/immunology , Mammary Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/metabolism , Coculture Techniques , Dendritic Cells/metabolism , Female , Humans , Immunotherapy, Adoptive , Macrophages/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice, Inbred C57BL , Myeloid Cells/immunology , Myeloid Cells/metabolism , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Tumor Microenvironment
17.
Nat Biotechnol ; 32(4): 387-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24633241

ABSTRACT

Functional characterization of noncoding sequences is crucial for understanding the human genome and learning how genetic variation contributes to disease. 3' untranslated regions (UTRs) are an important class of noncoding sequences, but their functions remain largely uncharacterized. We developed a method for massively parallel functional annotation of sequences from 3' UTRs (fast-UTR) and used this approach to measure the effects of a total of >450 kilobases of 3' UTR sequences from >2,000 human genes on steady-state mRNA abundance, mRNA stability and protein production. We found widespread regulatory effects on mRNA that were coupled to effects on mRNA stability and protein production. Furthermore, we discovered 87 novel cis-regulatory elements and measured the effects of genetic variation within known and novel 3' UTR motifs. This work shows how massively parallel approaches can improve the functional annotation of noncoding sequences, advance our understanding of cis-regulatory mechanisms and quantify the effects of human genetic variation.


Subject(s)
3' Untranslated Regions/genetics , Genomics/methods , Molecular Sequence Annotation/methods , Sequence Analysis, RNA/methods , Genome, Human , Humans , MicroRNAs/genetics , RNA, Messenger/analysis , RNA, Messenger/chemistry , RNA, Messenger/genetics
18.
Nat Immunol ; 15(3): 258-65, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24464130

ABSTRACT

The maintenance of immunological tolerance requires the deletion of self-reactive T cells in the thymus. The expression of genes encoding tissue-specific antigens (TSAs) by thymic epithelial cells is critical for this process and depends on activity of the transcriptional regulator Aire; however, the molecular mechanisms Aire uses to target loci encoding TSAs are unknown. Here we identified two Aire-interacting proteins known to be involved in gene repression, ATF7ip and MBD1, that were required for Aire's targeting of loci encoding TSAs. Moreover, Mbd1(-/-) mice developed pathological autoimmunity and had a defect in Aire-dependent thymic expression of genes encoding TSAs, which underscores the importance of Aire's interaction with the ATF7ip-MBD1 protein complex in maintaining central tolerance.


Subject(s)
Central Tolerance/immunology , DNA-Binding Proteins/immunology , Gene Expression Regulation/immunology , Immune Tolerance , Repressor Proteins/immunology , Transcription Factors/immunology , Animals , Autoantigens/immunology , Central Tolerance/genetics , DNA-Binding Proteins/genetics , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Protein Binding , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transfection , Two-Hybrid System Techniques , AIRE Protein
20.
PLoS One ; 8(5): e61956, 2013.
Article in English | MEDLINE | ID: mdl-23658702

ABSTRACT

The medial ganglionic eminence (MGE) is an embryonic forebrain structure that generates the majority of cortical interneurons. MGE transplantation into specific regions of the postnatal central nervous system modifies circuit function and improves deficits in mouse models of epilepsy, Parkinson's disease, pain, and phencyclidine-induced cognitive deficits. Herein, we describe approaches to generate MGE-like progenitor cells from mouse embryonic stem (ES) cells. Using a modified embryoid body method, we provided gene expression evidence that mouse ES-derived Lhx6(+) cells closely resemble immature interneurons generated from authentic MGE-derived Lhx6(+) cells. We hypothesized that enhancers that are active in the mouse MGE would be useful tools in detecting when ES cells differentiate into MGE cells. Here we demonstrate the utility of enhancer elements [422 (DlxI12b), Lhx6, 692, 1056, and 1538] as tools to mark MGE-like cells in ES cell differentiation experiments. We found that enhancers DlxI12b, 692, and 1538 are active in Lhx6-GFP(+) cells, while enhancer 1056 is active in Olig2(+) cells. These data demonstrate unique techniques to follow and purify MGE-like derivatives from ES cells, including GABAergic cortical interneurons and oligodendrocytes, for use in stem cell-based therapeutic assays and treatments.


Subject(s)
Embryoid Bodies/physiology , Enhancer Elements, Genetic , Neural Stem Cells/physiology , Prosencephalon/cytology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Separation , Cells, Cultured , Embryoid Bodies/transplantation , Female , Flow Cytometry , GABAergic Neurons/metabolism , Gene Expression , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic , Staining and Labeling , Transcriptome , Transduction, Genetic , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL