Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Publication year range
1.
Immunity ; 48(2): 364-379.e8, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466759

ABSTRACT

Neutrophils are specialized innate cells that require constant replenishment from proliferative bone marrow (BM) precursors as a result of their short half-life. Although it is established that neutrophils are derived from the granulocyte-macrophage progenitor (GMP), the differentiation pathways from GMP to functional mature neutrophils are poorly defined. Using mass cytometry (CyTOF) and cell-cycle-based analysis, we identified three neutrophil subsets within the BM: a committed proliferative neutrophil precursor (preNeu) which differentiates into non-proliferating immature neutrophils and mature neutrophils. Transcriptomic profiling and functional analysis revealed that preNeu require the C/EBPε transcription factor for their generation from the GMP, and their proliferative program is substituted by a gain of migratory and effector function as they mature. preNeus expand under microbial and tumoral stress, and immature neutrophils are recruited to the periphery of tumor-bearing mice. In summary, our study identifies specialized BM granulocytic populations that ensure supply under homeostasis and stress responses.


Subject(s)
Bone Marrow Cells/physiology , Neutrophils/physiology , Animals , Bone Marrow Cells/immunology , CCAAT-Enhancer-Binding Proteins/physiology , Cell Lineage , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Expression Profiling , Humans , Mice , Neoplasms, Experimental/immunology , Neutrophils/immunology
2.
Int J Mol Sci ; 24(1)2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36614196

ABSTRACT

Large numbers of neutrophils infiltrate tumors and comprise a notable component of the inflammatory tumor microenvironment. While it is established that tumor cells exhibit the Warburg effect for energy production, the contribution of the neutrophil metabolic state to tumorigenesis is unknown. Here, we investigated whether neutrophil infiltration and metabolic status promotes tumor progression in an orthotopic mouse model of pancreatic ductal adenocarcinoma (PDAC). We observed a large increase in the proportion of neutrophils in the blood and tumor upon orthotopic transplantation. Intriguingly, these tumor-infiltrating neutrophils up-regulated glycolytic factors and hypoxia-inducible factor 1-alpha (HIF-1α) expression compared to neutrophils from the bone marrow and blood of the same mouse. This enhanced glycolytic signature was also observed in human PDAC tissue samples. Strikingly, neutrophil-specific deletion of HIF-1α (HIF-1αΔNφ) significantly reduced tumor burden and improved overall survival in orthotopic transplanted mice, by converting the pro-tumorigenic neutrophil phenotype to an anti-tumorigenic phenotype. This outcome was associated with elevated reactive oxygen species production and activated natural killer cells and CD8+ cytotoxic T cells compared to littermate control mice. These data suggest a role for HIF-1α in neutrophil metabolism, which could be exploited as a target for metabolic modulation in cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Animals , Mice , Neutrophils/metabolism , Cell Line, Tumor , Mice, Knockout , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Carcinogenesis , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Tumor Microenvironment/genetics , Pancreatic Neoplasms
3.
FASEB J ; 34(8): 11133-11142, 2020 08.
Article in English | MEDLINE | ID: mdl-32627899

ABSTRACT

Vitamin D deficiency is a major public health problem worldwide, linked to several chronic diseases including cardiovascular diseases. While immunomodulatory effects of vitamin D on monocytes have been reported in cardiovascular and metabolic diseases, there is limited understanding on monocyte phenotype in healthy individuals with suboptimal vitamin D levels and without any clinical diseases. In this work, we performed label-free, microfluidic isolation of monocytes, and characterized their functional phenotype using flow cytometry and in vitro vascular models in healthy subjects with (n = 7) and without vitamin D deficiency (n = 16). Vitamin D deficient (VitD-Def) subjects (25(OH)D3 level < 26 ng/mL) expressed significant downregulation of vitamin D receptor (VDR) on monocytes as compared to controls (P < .0001), and VDR expression was well-associated with serum 25(OH)D3 levels. Increased monocyte-platelet aggregates (MPA), a marker for platelet activation, were also observed in VitD-Def subjects (P < .05) which suggests a pro-inflammatory monocyte phenotype. Monocyte adhesion to endothelial cells, an early-stage atherosclerosis event, was also higher in VitD-Def individuals, and inversely correlated to serum 25(OH)D3 level (P < .05). Taken together, these results indicate the pro-inflammatory state and atherogenic potential of monocytes in VitD-Def healthy subjects, and propound the use of vitamin D supplementation as a prospective immunomodulatory and anti-inflammatory therapy in atherosclerosis.


Subject(s)
Blood Platelets/physiology , Cell Adhesion/physiology , Endothelial Cells/physiology , Monocytes/physiology , Vitamin D Deficiency/physiopathology , Vitamin D/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Blood Platelets/metabolism , Cells, Cultured , Dietary Supplements , Down-Regulation/physiology , Endothelial Cells/metabolism , Female , Healthy Volunteers , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Monocytes/metabolism , Platelet Activation/physiology , Receptors, Calcitriol/metabolism , Vitamin D Deficiency/metabolism
4.
Int J Mol Sci ; 22(12)2021 Jun 14.
Article in English | MEDLINE | ID: mdl-34198548

ABSTRACT

Inflammation in the tumor microenvironment has been shown to promote disease progression in pancreatic ductal adenocarcinoma (PDAC); however, the role of macrophage metabolism in promoting inflammation is unclear. Using an orthotopic mouse model of PDAC, we demonstrate that macrophages from tumor-bearing mice exhibit elevated glycolysis. Macrophage-specific deletion of Glucose Transporter 1 (GLUT1) significantly reduced tumor burden, which was accompanied by increased Natural Killer and CD8+ T cell activity and suppression of the NLRP3-IL1ß inflammasome axis. Administration of mice with a GLUT1-specific inhibitor reduced tumor burden, comparable with gemcitabine, the current standard-of-care. In addition, we observe that intra-tumoral macrophages from human PDAC patients exhibit a pronounced glycolytic signature, which reliably predicts poor survival. Our data support a key role for macrophage metabolism in tumor immunity, which could be exploited to improve patient outcomes.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cytoprotection , Glycolysis , Macrophages/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/immunology , Animals , Carcinoma, Pancreatic Ductal/immunology , Cell Proliferation/drug effects , Cytoprotection/drug effects , Drug Resistance, Neoplasm/drug effects , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Humans , Hydroxybenzoates/pharmacology , Inflammation/pathology , Interleukin-1beta/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Macrophages/drug effects , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pancreatic Neoplasms/immunology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/drug effects , Pancreatic Neoplasms
5.
Br J Cancer ; 120(4): 407-423, 2019 02.
Article in English | MEDLINE | ID: mdl-30713340

ABSTRACT

BACKGROUND: Emergence of drug-resistant cancer phenotypes is a challenge for anti-cancer therapy. Cancer stem cells are identified as one of the ways by which chemoresistance develops. METHOD: We investigated the anti-inflammatory combinatorial treatment (DA) of doxorubicin and aspirin using a preclinical microfluidic model on cancer cell lines and patient-derived circulating tumour cell clusters. The model had been previously demonstrated to predict patient overall prognosis. RESULTS: We demonstrated that low-dose aspirin with a sub-optimal dose of doxorubicin for 72 h could generate higher killing efficacy and enhanced apoptosis. Seven days of DA treatment significantly reduced the proportion of cancer stem cells and colony-forming ability. DA treatment delayed the inhibition of interleukin-6 secretion, which is mediated by both COX-dependent and independent pathways. The response of patients varied due to clinical heterogeneity, with 62.5% and 64.7% of samples demonstrating higher killing efficacy or reduction in cancer stem cell (CSC) proportions after DA treatment, respectively. These results highlight the importance of using patient-derived models for drug discovery. CONCLUSIONS: This preclinical proof of concept seeks to reduce the onset of CSCs generated post treatment by stressful stimuli. Our study will promote a better understanding of anti-inflammatory treatments for cancer and reduce the risk of relapse in patients.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Aspirin/administration & dosage , Doxorubicin/administration & dosage , Neoplasm Recurrence, Local/prevention & control , Neoplastic Stem Cells/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Drug Therapy, Combination , Epithelial-Mesenchymal Transition/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/physiology , Microfluidics , Prostaglandin-Endoperoxide Synthases/physiology , Signal Transduction/drug effects
6.
Anal Chem ; 90(24): 14535-14542, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30426739

ABSTRACT

Monocytes and platelets play key roles in atherosclerosis and thrombosis, and circulating monocyte-platelet aggregates (MPA) in blood have been widely proposed as surrogate biomarkers for cardiovascular risk stratification and monitoring antiplatelet therapies. However, conventional MPA characterization is based on whole-blood fixation and flow cytometry analysis which adversely affect cell viability and detection accuracy due to significant leukocyte and platelet contaminations. Herein, we introduce a rapid and label-free microfluidic approach for complete size-based fractionation of peripheral blood mononuclear cells (PBMCs) into monocytes, lymphocytes, and platelets. The developed biochip enables gentle sorting of intact MPA in the enriched monocytes with efficient depletion of lymphocytes and platelets for accurate MPA quantification. We first compared the developed microfluidic technology (dean flow fractionation, DFF) with standard magnetic negative isolation (MACS) and observed that DFF-sorted monocytes had similar viability, purity, and key immune functions (phagocytosis, macrophage differentiation) as MACS-sorted monocytes. As proof of concept for diabetes testing, we isolated and characterized monocytes/MPA from a cohort of healthy ( n = 5) and type 2 diabetes mellitus (T2DM) subjects ( n = 8) in PBMCs and DFF-sorted monocytes. High-speed imaging, immunofluorescence, and flow cytometry analysis clearly indicated higher levels of MPA in T2DM patients ( P < 0.05) with enhanced MPA detection sensitivity in DFF-sorted monocytes ( P < 0.05). Taken together, the developed DFF technology greatly facilitates high-throughput (∼130 µL min-1) label-free isolation of monocyte/MPA from PBMCs and can be further developed into a clinical tool for point-of-care cardiovascular risk stratification in metabolic disorders including T2DM.


Subject(s)
Blood Platelets/cytology , Microfluidics/methods , Monocytes/cytology , Blood Platelets/metabolism , Case-Control Studies , Cell Differentiation , Cytokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Flow Cytometry , Humans , Leukocytes, Mononuclear/cytology , Microscopy, Fluorescence , Monocytes/metabolism , Phagocytosis
7.
Breast Cancer Res Treat ; 163(1): 21-35, 2017 May.
Article in English | MEDLINE | ID: mdl-28233108

ABSTRACT

PURPOSE: The role of Forkhead Box Protein 3 (Foxp3) expressing regulatory T cells (Tregs) in breast cancer remains unclear. We examined the abundance and localisation of total T cells, B cells and Tregs within samples from triple-negative breast cancers (TNBCs) and asked whether these parameters were associated with clinicopathological features of the cancer or clinical outcomes. METHODS: Samples from TNBCs diagnosed between 2003 and 2010 in Singapore were divided into "high" and "low" intra-tumoural or stromal groups, based on whether they had higher or lower than median densities of specific tumour-infiltrating lymphocyte populations (CD3+ total T cells, Foxp3+CD3+ Tregs, or CD20+ B cells) in the intra-tumoural space or stroma. RESULTS: Of the 164 samples, patients bearing tumours with high Tregs within their intra-tumoural, but not stromal, areas experienced significantly longer overall and disease-free survival compared to individuals with low Treg densities. These "high intra-tumoural Treg" tumours were also characterised by relatively higher frequencies of CD8+ T cells and CD20+ B cells, and expressed significantly higher levels of some genes associated with inflammation, immune cell functions and trafficking, altogether consistent with a more "immune-activated" tumour microenvironment, in contrast to tumours bearing lower densities of Tregs. CONCLUSIONS: In summary, the combination of high densities of intra-tumoural Tregs, CD8+ T cells and CD20+ B cells represents a favourable prognostic panel in TNBCs. These data also indicate new avenues for further investigation on the interaction between immune cell types within the tumour microenvironment and highlight the potential of Treg density and localisation within tumours to affect clinical outcome.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism , Triple Negative Breast Neoplasms/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Female , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Prognosis , Triple Negative Breast Neoplasms/metabolism
8.
Allergy ; 72(3): 373-384, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27271846

ABSTRACT

BACKGROUND: Cohort studies indicated that in certain individuals the basophils do not respond toward allergens due to a desensitization of their Fc epsilon receptor pathway. Cause and functional role as well as the implications on allergic reactions, however, are not clear yet. METHODS: A cross-sectional study was carried out in the tropical urban environment of Singapore, where the allergic response is dominated by a single allergen (house dust mite; HDM). Blood samples were collected from 476 individuals and analyzed comprehensively to correlate the functional state of their basophils with the clinical state as well as the composition of the cellular and soluble plasma components. RESULTS: Inactivation of basophils ('basophil anergy') was observed in about 10% of the cohort. It was associated with a downregulation of basophil Syk and an apparent reduction in the incidence of allergic rhinitis. Correlations on the cohort level suggest that it represents a transitional state to be passed through during the interconversion of responder and nonresponder state. CONCLUSIONS: Basophil anergy thus seems to function as activation barrier to prevent unwanted reactions against minor allergens. It may therefore be relevant for diagnostic purposes or therapeutic interventions of allergic diseases.


Subject(s)
Basophils/immunology , Clonal Anergy/immunology , Allergens/immunology , Animals , Antibody Specificity/immunology , Basophils/metabolism , Biomarkers , Clonal Anergy/genetics , Cluster Analysis , Cohort Studies , Cross-Sectional Studies , Gene Expression Profiling , Gene Expression Regulation , Humans , Hypersensitivity/diagnosis , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/metabolism , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunophenotyping , Rhinitis, Allergic/immunology , Rhinitis, Allergic/metabolism , Syk Kinase/genetics , Syk Kinase/metabolism
9.
J Immunol ; 194(8): 3890-900, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25732728

ABSTRACT

Little is known about the cellular mechanisms of innate immunity against dengue virus (DV) infection. Specifically, the γδ T cell response to DV has not been characterized in detail. In this article, we demonstrate that markers of activation, proliferation, and degranulation are upregulated on γδ T cells in PBMC isolated from individuals with acute dengue fever. Primary γδ T cells responded rapidly in vitro to autologous DV-infected dendritic cells by secreting IFN-γ and upregulating CD107a. The anti-DV IFN-γ response is regulated by type I IFN and IL-18 in a TCR-independent manner, and IFN-γ secreting γδ T cells predominantly expressed IL-18Rα. Antagonizing the ATP-dependent P2X7 receptor pathway of inflammasome activation significantly inhibited the anti-DV IFN-γ response of γδ T cells. Overnight priming with IL-18 produced effector γδ T cells with significantly increased ability to lyse autologous DV-infected dendritic cells. Monocytes were identified as accessory cells that augmented the anti-DV IFN-γ response of γδ T cells. Lack of monocytes in culture is associated with lower IL-18 levels in culture supernatant and diminished production of IFN-γ by γδ T cells, whereas addition of exogenous IL-18 restored the IFN-γ response of γδ T cells in monocyte-depleted cocultures with DV-infected DC. Our results indicate that primary γδ T cells contribute to the immune response during DV infection by providing an early source of IFN-γ, as well as by killing DV-infected cells, and suggest that monocytes participate as accessory cells that sense DV infection and amplify the cellular immune response against this virus in an IL-18-dependent manner.


Subject(s)
Dendritic Cells/immunology , Dengue Virus/immunology , Dengue/immunology , Interleukin-18/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Adult , Coculture Techniques , Dendritic Cells/pathology , Dengue/pathology , Female , Humans , Interferon Type I , Interferon-gamma/immunology , Interleukin-18 Receptor alpha Subunit/immunology , Lysosomal-Associated Membrane Protein 1/immunology , Male , Monocytes/immunology , Monocytes/pathology , Receptors, Purinergic P2X7/immunology , T-Lymphocytes/pathology
10.
J Immunol ; 193(5): 2258-66, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25080484

ABSTRACT

Regulatory B cells (B-reg) produce IL-10 and suppress inflammation in both mice and humans, but limited data on the phenotype and function of these cells have precluded detailed assessment of their contribution to host immunity. In this article, we report that human B-reg cannot be defined based on a phenotype composed of conventional B cell markers, and that IL-10 production can be elicited in both the CD27(+) memory population and naive B cell subset after only a brief stimulation in vitro. We therefore sought to obtain a better definition of IL-10-producing human B-regs using a multiparameter analysis of B cell phenotype, function, and gene expression profile. Exposure to CpG and anti-Ig are the most potent stimuli for IL-10 secretion in human B cells, but microarray analysis revealed that human B cells cotreated with these reagents resulted in only ∼0.7% of genes being differentially expressed between IL-10(+) and IL-10(-) cells. Instead, connectivity map analysis revealed that IL-10-secreting B cells are those undergoing specific differentiation toward a germinal center fate, and we identified a CD11c(+) B cell subset that was not capable of producing IL-10 even under optimal conditions. Our findings will assist in the identification of a broader range of human pro-B-reg populations that may represent novel targets for immunotherapy.


Subject(s)
B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Interleukin-10/immunology , Adjuvants, Immunologic/pharmacology , Animals , B-Lymphocyte Subsets/cytology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Female , Humans , Interleukin-10/genetics , Male , Mice , Mice, Knockout , Oligodeoxyribonucleotides/pharmacology
11.
Immunology ; 145(3): 404-16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25707426

ABSTRACT

Within human blood there are two subsets of monocytes that can be identified by differential expression of CD16. Although numerous phenotypic and functional differences between the subsets have been described, little is known of the mechanisms underlying the distinctive properties of the two subsets. MicroRNAs (miRNAs) are small non-coding RNAs that can regulate gene expression through promoting mRNA degradation or repressing translation, leading to alterations in cellular processes. Their potential influence on the functions of monocyte subsets has not been investigated. In this study, we employed microarray analysis to define the miRNA expression profile of human monocyte subsets. We identified 66 miRNAs that were differentially expressed (DE) between CD16(+) and CD16(-) monocytes. Gene ontology analysis revealed that the predicted targets of the DE miRNAs were predominantly associated with cell death and cellular movement. We validated the functional impacts of selected DE miRNAs in CD16(-) monocytes, over-expression of miR-432 significantly increases apoptosis, and inhibiting miR-19a significantly reduces cell motility. Furthermore, we found that miR-345, another DE miRNA directly targets the transcription factor RelA in monocytes, which resulted in the differential expression of RelA in monocyte subsets. This implicates miR-345 indirect regulation of many genes downstream of RelA, including important inflammatory mediators. Together, our data show that DE miRNAs could contribute substantially to regulating the functions of human blood monocytes.


Subject(s)
Gene Expression Profiling , MicroRNAs/genetics , Monocytes/metabolism , Receptors, IgG/metabolism , 3' Untranslated Regions/genetics , Apoptosis/genetics , Cell Movement/genetics , Cells, Cultured , HEK293 Cells , Humans , Monocytes/classification , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/genetics
12.
J Immunol ; 190(2): 794-804, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23248262

ABSTRACT

Immune dysfunction may contribute to tumor progression in gastric cancer (GC) patients. One mechanism of immune dysfunction is the suppression of T cell activation and impairment of the efficacy of cancer immunotherapy by myeloid-derived suppressor cells (MDSCs). We assessed the phenotype and immunosuppressive function of MDSCs in GC patients. We further investigated the role of S100A8/A9 in GC and the relationship between S100A8/A9 and MDSC function. Lastly, the effect of MDSCs on survival rates and its potential as a prognostic factor in GC patients were investigated. MDSCs from PBMCs of GC patients were identified by comparing the expression of specific surface markers with PBMCs from healthy individuals. The ability of MDSCs to suppress T lymphocyte response and the effect of S100A8/A9 and RAGE blocking were tested in vitro by (autologous) MLR. GC patients had significantly more MDSCs than healthy individuals. These MDSCs suppressed both T lymphocyte proliferation and IFN-γ production and had high arginase-I expression. Levels of S100A8/A9 in plasma were higher in GC patients compared with healthy individuals, and they correlated with MDSC levels in the blood. Blocking of S100A8/A9 itself and the S100A8/A9 receptor RAGE on MDSCs from GC patients abrogated T cell effector function. We found that high levels of MDSCs correlated with more advanced cancer stage and with reduced survival (p = 0.006). S100A8/A9 has been identified as a potential target to modulate antitumor immunity by reversing MDSC-mediated immunosuppression.


Subject(s)
Calgranulin A/blood , Calgranulin B/blood , Myeloid Cells , Stomach Neoplasms/blood , Stomach Neoplasms/pathology , Antigens, CD/metabolism , Arginase/metabolism , CD11b Antigen/metabolism , Humans , Immunophenotyping , Lipopolysaccharide Receptors/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasm Staging , Nitric Oxide Synthase Type II/metabolism , Phenotype , Prognosis , Stomach Neoplasms/mortality
13.
Br J Haematol ; 165(1): 134-44, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24428589

ABSTRACT

CD137 ligand (CD137L), a member of the tumour necrosis factor family, is expressed as a cell surface molecule. Engagement of CD137L on haematopoietic progenitor cells induces monocytic differentiation, and in peripheral monocytes CD137L signalling promotes differentiation to mature dendritic cells. We hypothesized that CD137L signalling would also induce differentiation in transformed myeloid cells. Here we show that recombinant CD137 protein, which crosslinks CD137L and initiates reverse CD137L signalling in myeloid cells, induces morphological changes (adherence, spreading), loss of progenitor markers (CD117), expression of maturation markers (CD11b, CD13) and secretion of cytokines that are indicative of myeloid differentiation. Under the influence of CD137L signalling, acute myeloid leukaemia (AML) cells acquired expression of co-stimulatory molecules (CD80, CD86, CD40), the dendritic cell marker CD83 and dendritic cell activities, enabling them to stimulate T cells. CD137L signalling induced differentiation in 71% (15 of 21) of AML samples, irrespective of French-American-British classification and CD137L expression level. However, the type of response varied with the AML subtype and patient sample. In summary, this study demonstrated that CD137L signalling induced differentiation in malignant cells of AML patients, and suggests that it may be worthwhile to investigate treatment with recombinant CD137 protein as a potential novel therapeutic approach for AML.


Subject(s)
4-1BB Ligand/metabolism , Cell Differentiation , Leukemia, Myeloid, Acute/metabolism , Signal Transduction , Adult , Aged , Antigens, CD/metabolism , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/diagnosis , Middle Aged , Protein Binding , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Young Adult
14.
J Immunol ; 188(8): 3584-93, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22427642

ABSTRACT

Blood monocytes recognize Gram-negative bacteria through the TLR4, which signal via MyD88- and TRIF-dependent pathway to trigger an immune-inflammatory response. However, a dysregulated inflammatory response by these cells often leads to severe pathologies such as sepsis. We investigated the role of CD16 in the regulation of human monocyte response to Gram-negative endotoxin and sepsis. Blood monocytes from sepsis patients demonstrated an upregulation of several TRIF-dependent genes as well as a selective expansion of CD16-expressing (CD16(+)) monocytes. Gene expression and biochemical studies revealed CD16 to regulate the TRIF-dependent TLR4 pathway in monocytes by activating Syk, IFN regulatory factor 3, and STAT1, which resulted in enhanced expression of IFNB, CCL5, and CXCL10. CD16 also upregulated the expression of IL-1R-associated kinase M and IL-1 receptor antagonist, which are negative regulators of the MyD88-dependent pathway. CD16 overexpression or small interfering RNA knockdown in monocytes confirmed the above findings. Interestingly, these results were mirrored in the CD16(+) monocyte subset isolated from sepsis patients, providing an in vivo confirmation to our findings. Collectively, the results from the current study demonstrate CD16 as a key regulator of the TRIF-dependent TLR4 pathway in human monocytes and their CD16-expressing subset, with implications in sepsis.


Subject(s)
Gene Expression Regulation/immunology , Monocytes/metabolism , Receptors, IgG/genetics , Sepsis/immunology , Adaptive Immunity , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adult , Animals , Endotoxins/pharmacology , Gene Expression Regulation/drug effects , Humans , Interferons/genetics , Interferons/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Mice , Mice, Inbred C57BL , Middle Aged , Monocytes/immunology , Monocytes/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Primary Cell Culture , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , RNA, Small Interfering/genetics , Receptors, IgG/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Sepsis/genetics , Sepsis/pathology , Signal Transduction/drug effects , Signal Transduction/immunology , Syk Kinase , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transfection
15.
Eur J Immunol ; 42(1): 89-100, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22009685

ABSTRACT

High macrophage infiltration into tumours often correlates with poor prognoses; in colorectal, stomach and skin cancers, however, the opposite is observed but the mechanisms behind this phenomenon remain unclear. Here, we sought to understand how tumour-associated macrophages (TAMs) in colorectal cancer execute tumour-suppressive roles. We found that TAMs in a colorectal cancer model were pro-inflammatory and inhibited the proliferation of tumour cells. TAMs also produced chemokines that attract T cells, stimulated proliferation of allogeneic T cells and activated type-1 T cells associated with anti-tumour immune responses. Using colorectal tumour tissues, we verified that TAMs in vivo were indeed pro-inflammatory. Furthermore, the number of tumour-infiltrating T cells correlated with the number of TAMs, suggesting that TAMs could attract T cells; and indeed, type-1 T cells were present in the tumour tissues. Patient clinical data suggested that TAMs exerted tumour-suppressive effects with the help of T cells. Hence, the tumour-suppressive mechanisms of TAMs in colorectal cancer involve the inhibition of tumour cell proliferation alongside the production of pro-inflammatory cytokines, chemokines and promoting type-1 T-cell responses. These new findings would contribute to the development of future cancer immunotherapies based on enhancing the tumour-suppressive properties of TAMs to boost anti-tumour immune responses.


Subject(s)
Colorectal Neoplasms/immunology , Cytokines/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Th1 Cells/immunology , Coculture Techniques , Colorectal Neoplasms/pathology , Cytokines/genetics , Gene Expression Profiling , HT29 Cells , Humans , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/pathology , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Th1 Cells/pathology
16.
Blood ; 118(5): e16-31, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21653326

ABSTRACT

New official nomenclature subdivides human monocytes into 3 subsets: the classical (CD14(++)CD16(-)), intermediate (CD14(++)CD16(+)), and nonclassical (CD14(+)CD16(++)) monocytes. This introduces new challenges, as monocyte heterogeneity is mostly understood based on 2 subsets, the CD16(-) and CD16(+) monocytes. Here, we comprehensively defined the 3 circulating human monocyte subsets using microarray, flow cytometry, and cytokine production analysis. We find that intermediate monocytes expressed a large majority (87%) of genes and surface proteins at levels between classical and nonclassical monocytes. This establishes their intermediary nature at the molecular level. We unveil the close relationship between the intermediate and nonclassic monocytes, along with features that separate them. Intermediate monocytes expressed highest levels of major histocompatibility complex class II, GFRα2 and CLEC10A, whereas nonclassic monocytes were distinguished by cytoskeleton rearrangement genes, inflammatory cytokine production, and CD294 and Siglec10 surface expression. In addition, we identify new features for classic monocytes, including AP-1 transcription factor genes, CLEC4D and IL-13Rα1 surface expression. We also find circumstantial evidence supporting the developmental relationship between the 3 subsets, including gradual changes in maturation genes and surface markers. By comprehensively defining the 3 monocyte subsets during healthy conditions, we facilitate target identification and detailed analyses of aberrations that may occur to monocyte subsets during diseases.


Subject(s)
Gene Expression Profiling , Microarray Analysis , Monocytes/classification , Monocytes/metabolism , Cell Differentiation/immunology , Cell Separation/methods , Cluster Analysis , Flow Cytometry , Gene Expression Profiling/methods , Humans , Models, Biological , Monocytes/immunology , Monocytes/physiology , Validation Studies as Topic
17.
Trends Immunol ; 31(1): 18-23, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19836307

ABSTRACT

Dendritic cells (DC) are professional phagocytes possessing a unique ability to sense perturbations in the tissue microenvironment and promote adaptive immune responses, whilst maintaining immunological tolerance. Mouse myeloid DC progenitors with the ability to migrate through the blood and replenish the DC pool have been identified in bone marrow but the ontogeny of human DC is poorly understood. Access to lymphoid tissues for human DC isolation is severely limited and researchers have resorted to the use of in vitro derivation systems in attempts to understand DC development, which may result in misleading conclusions. The identification of a human DC progenitor in blood would greatly enhance the understanding of DC homeostasis and their role in pathogenesis.


Subject(s)
Blood Cells/cytology , Dendritic Cells/cytology , Stem Cells/cytology , Animals , Humans , Mice , Monocytes/cytology
18.
Eur J Immunol ; 41(6): 1639-51, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21469104

ABSTRACT

TNF/iNOS-producing dendritic cells (Tip-DCs) have been shown to arise during inflammation and are important mediators of immune defense. However, it is still relatively unclear which cell types contribute to their differentiation. Here we show that CD8(+) T cells, through the interaction with DCs, can induce the rapid development of human monocytes into Tip-DCs that express high levels of TNF-α and iNOS. Tip-DCs exhibited T-cell priming ability, expressed high levels of MHC class II, upregulated co-stimulatory molecules CD40, CD80, CD86, toll-like receptors TLR2, TLR3, TLR4, chemokine receptors CCR1 and CX3CR1 and expressed the classical mature DC marker, CD83. Differentiation of monocytes into Tip-DCs was partially dependent on IFN-γ as blocking the IFN-γ receptor on monocytes resulted in a significant decrease in CD40 and CD83 expression and in TNF-α production. Importantly, these Tip-DCs were capable of further driving Th1 responses by priming naive CD4(+) T cells for proliferation and IFN-γ production and this was partially dependent on Tip-DC production of TNF-α and NO. Our study hence identifies a role for CD8(+) T cells in orchestrating Th1-mediating signals through the differentiation of monocytes into Th1-inducing Tip-DCs.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Interferon-gamma/metabolism , Nitric Oxide Synthase Type II/metabolism , Tumor Necrosis Factor-alpha/metabolism , Antibodies, Blocking/pharmacology , Antigens, CD/biosynthesis , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CX3C Chemokine Receptor 1 , Cell Communication/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Histocompatibility Antigens Class II/biosynthesis , Humans , Interferon-gamma/immunology , Lymphocyte Activation/drug effects , Nitric Oxide Synthase Type II/immunology , Receptors, CCR1/biosynthesis , Receptors, Chemokine/biosynthesis , Th1 Cells/drug effects , Th1 Cells/immunology , Toll-Like Receptors/biosynthesis , Tumor Necrosis Factor-alpha/immunology
19.
J Immunol ; 184(7): 3697-709, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20190138

ABSTRACT

Integrins are type I membrane and heterodimeric (alphabeta) cell adhesion receptors. Intracellular signals triggered by ligand-bound integrins are important for cell growth, differentiation, and migration. Integrin alpha(M)beta(2) plays key roles in myeloid cell adhesion, phagocytosis, and degranulation. In this study, we show that protein kinase C (PKC) delta is involved in alpha(M)beta(2) signaling. In human monocytic U937 cells and peripheral blood monocytes, alpha(M)beta(2) clustering induced PKCdelta translocation to the plasma membrane, followed by Tyr(311) phosphorylation and activation of PKCdelta by the src family kinases Hck and Lyn. Interestingly, alpha(M)beta(2)-induced PKCdelta Tyr(311) phosphorylation was not mediated by the tyrosine kinase Syk, which is a well reported kinase in beta(2) integrin signaling. Analysis of the beta(2) cytoplasmic tail showed that the sequence Asn(727)-Ser(734) is important in alpha(M)beta(2)-induced PKCdelta Tyr(311) phosphorylation. It has been shown that alpha(M)beta(2) clustering regulates the expression the transcription factor Foxp1 that has a role in monocyte differentiation. We show that Foxp1 expression was reduced in monocytes that were allowed to adhere to human microvascular endothelial cells. However, the expression of Foxp1 was not affected in monocytes that were treated with PKCdelta-targeting small interfering RNA, suggesting that PKCdelta regulates Foxp1 expression. These results demonstrate a role of PKCdelta in alpha(M)beta(2)-mediated Foxp1 regulation in monocytes.


Subject(s)
Enzyme Activation/immunology , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation/immunology , Macrophage-1 Antigen/metabolism , Monocytes/metabolism , Protein Kinase C/metabolism , Repressor Proteins/biosynthesis , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Humans , Immunoblotting , Immunoprecipitation , Jurkat Cells , K562 Cells , Macrophage-1 Antigen/immunology , Monocytes/immunology , Phosphorylation , Protein Kinase C/immunology , Protein Transport/immunology , Signal Transduction/immunology , Transfection , U937 Cells
20.
J Immunol ; 184(10): 5903-13, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20404274

ABSTRACT

Chikungunya virus (CHIKV) is an alphavirus that causes chronic and incapacitating arthralgia in humans. To date, interactions between the immune system and the different stages of the virus life cycle remain poorly defined. We demonstrated for the first time that CHIKV Ags could be detected in vivo in the monocytes of acutely infected patients. Using in vitro experimental systems, whole blood and purified monocytes, we confirmed that monocytes could be infected and virus growth could be sustained. CHIKV interactions with monocytes, and with other blood leukocytes, induced a robust and rapid innate immune response with the production of specific chemokines and cytokines. In particular, high levels of IFN-alpha were produced rapidly after CHIKV incubation with monocytes. The identification of monocytes during the early phase of CHIKV infection in vivo is significant as infected monocyte/macrophage cells have been detected in the synovial tissues of chronically CHIKV-infected patients, and these cells may behave as the vehicles for virus dissemination. This may explain the persistence of joint symptoms despite the short duration of viremia. Our results provide a better understanding on the basic mechanisms of infection and early antiviral immune responses and will help in the development of future effective control strategies.


Subject(s)
Alphavirus Infections/immunology , Chikungunya virus/immunology , Immunity, Innate , Monocytes/immunology , Monocytes/virology , Acute Disease , Alphavirus Infections/blood , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/virology , Cells, Cultured , Chlorocebus aethiops , Chronic Disease , Dendritic Cells/immunology , Dendritic Cells/virology , Disease Models, Animal , Humans , Insect Vectors/immunology , Insect Vectors/virology , Macaca , Macrophages/immunology , Macrophages/virology , Monocytes/metabolism , Myeloid Cells/immunology , Myeloid Cells/virology , Vero Cells , Viremia/immunology
SELECTION OF CITATIONS
SEARCH DETAIL