Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 8(4): 436-450, 2020 04.
Article in English | MEDLINE | ID: mdl-32075803

ABSTRACT

Cancer-associated fibroblasts (CAF) represent a functionally heterogeneous population of activated fibroblasts that constitutes a major component of tumor stroma. Although CAFs have been shown to promote tumor growth and mediate resistance to chemotherapy, the mechanisms by which they may contribute to immune suppression within the tumor microenvironment (TME) in lung squamous cell carcinoma (LSCC) remain largely unexplored. Here, we identified a positive correlation between CAF and monocytic myeloid cell abundances in 501 primary LSCCs by mining The Cancer Genome Atlas data sets. We further validated this finding in an independent cohort using imaging mass cytometry and found a significant spatial interaction between CAFs and monocytic myeloid cells in the TME. To delineate the interplay between CAFs and monocytic myeloid cells, we used chemotaxis assays to show that LSCC patient-derived CAFs promoted recruitment of CCR2+ monocytes via CCL2, which could be reversed by CCR2 inhibition. Using a three-dimensional culture system, we found that CAFs polarized monocytes to adopt a myeloid-derived suppressor cell (MDSC) phenotype, characterized by robust suppression of autologous CD8+ T-cell proliferation and IFNγ production. We further demonstrated that inhibiting IDO1 and NADPH oxidases, NOX2 and NOX4, restored CD8+ T-cell proliferation by reducing reactive oxygen species (ROS) generation in CAF-induced MDSCs. Taken together, our study highlights a pivotal role of CAFs in regulating monocyte recruitment and differentiation and demonstrated that CCR2 inhibition and ROS scavenging abrogate the CAF-MDSC axis, illuminating a potential therapeutic path to reversing the CAF-mediated immunosuppressive microenvironment.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Carcinoma, Squamous Cell/immunology , Lung Neoplasms/immunology , Monocytes/immunology , Myeloid-Derived Suppressor Cells/immunology , Reactive Oxygen Species/metabolism , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/immunology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Cells, Cultured , Female , Humans , Immunosuppression Therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , NADPH Oxidase 2/immunology , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/immunology , NADPH Oxidase 4/metabolism , Receptors, CCR2/immunology , Receptors, CCR2/metabolism , Signal Transduction , Tumor Microenvironment
2.
Arthritis Rheumatol ; 67(12): 3146-57, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26315890

ABSTRACT

OBJECTIVE: Polymorphisms in the transcription factor interferon regulatory factor 5 (IRF5) are associated with an increased risk of developing rheumatoid arthritis (RA). This study was undertaken to determine the role of IRF5 in a mouse model of arthritis development. METHODS: K/BxN serum-transfer arthritis was induced in mice deficient in IRF5, or lacking IRF5 only in myeloid cells, and arthritis severity was evaluated. K/BxN arthritis was also induced in mice deficient in TRIF, Toll-like receptor 2 (TLR2), TLR3, TLR4, and TLR7 to determine the pathways through which IRF5 might promote arthritis. In vitro studies were performed to determine the role of IRF5 in interleukin-1 (IL-1) receptor and TLR signaling. RESULTS: Arthritis severity was reduced in IRF5-deficient, TRIF-deficient, TLR3-deficient, and TLR7-deficient mice. The expression of multiple genes regulating neutrophil recruitment or function and bioactive IL-1ß formation was reduced in the joints during active arthritis in IRF5-deficient mice. In vitro studies showed that TLR7 and the TRIF-dependent TLR3 pathway induce proinflammatory cytokine production in disease-relevant cell types in an IRF5-dependent manner. CONCLUSION: Our findings indicate that IRF5 contributes to disease pathogenesis in inflammatory arthritis. This is likely due at least in part to the role of IRF5 in mediating proinflammatory cytokine production downstream of TLR7 and TLR3. Since TLR7 and TLR3 are both RNA-sensing TLRs, this suggests that endogenous RNA ligands present in the inflamed joint promote arthritis development. These findings may be relevant to human RA, since RNA capable of activating TLR7 and TLR3 is present in synovial fluid and TLR7 and TLR3 are up-regulated in the joints of RA patients.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , Interferon Regulatory Factors/genetics , Membrane Glycoproteins/genetics , Myeloid Cells/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 7/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Disease Models, Animal , Gene Knockdown Techniques , In Vitro Techniques , Interferon Regulatory Factors/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Severity of Illness Index , Signal Transduction , Synovial Fluid/immunology , Synovial Fluid/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 7/immunology
3.
J Immunol ; 194(4): 1467-79, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25595782

ABSTRACT

Premature atherosclerosis is a severe complication of lupus and other systemic autoimmune disorders. Gain-of-function polymorphisms in IFN regulatory factor 5 (IRF5) are associated with an increased risk of developing lupus, and IRF5 deficiency in lupus mouse models ameliorates disease. However, whether IRF5 deficiency also protects against atherosclerosis development in lupus is not known. In this study, we addressed this question using the gld.apoE(-/-) mouse model. IRF5 deficiency markedly reduced lupus disease severity. Unexpectedly, despite the reduction in systemic immune activation, IRF5-deficient mice developed increased atherosclerosis and also exhibited metabolic dysregulation characterized by hyperlipidemia, increased adiposity, and insulin resistance. Levels of the atheroprotective cytokine IL-10 were reduced in aortae of IRF5-deficient mice, and in vitro studies demonstrated that IRF5 is required for IL-10 production downstream of TLR7 and TLR9 signaling in multiple immune cell types. Chimera studies showed that IRF5 deficiency in bone marrow-derived cells prevents lupus development and contributes in part to the increased atherosclerosis. Notably, IRF5 deficiency in non-bone marrow-derived cells also contributes to the increased atherosclerosis through the generation of hyperlipidemia and increased adiposity. Together, our results reveal a protective role for IRF5 in lupus-associated atherosclerosis that is mediated through the effects of IRF5 in both immune and nonimmune cells. These findings have implications for the proposed targeting of IRF5 in the treatment of autoimmune disease as global IRF5 inhibition may exacerbate cardiovascular disease in these patients.


Subject(s)
Atherosclerosis/etiology , Interferon Regulatory Factors/immunology , Lupus Erythematosus, Systemic/immunology , Metabolic Syndrome/etiology , Animals , Atherosclerosis/immunology , Disease Models, Animal , Female , Flow Cytometry , Interferon Regulatory Factors/deficiency , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/pathology , Male , Metabolic Syndrome/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
4.
J Immunol ; 194(1): 101-12, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25416804

ABSTRACT

A subset of chronic lymphocytic leukemia (CLL) BCRs interacts with Ags expressed on apoptotic cells, suggesting that CLL BCRs have the potential to internalize apoptotic cell RNA- or DNA-containing fragments with resultant activation of TLR7 or TLR9, respectively. By blocking cAMP degradation, type 4 cAMP phosphodiesterase (PDE4) inhibitors activate cAMP-mediated signaling and induce apoptosis in CLL cells. In this study, we show that autologous irradiated leukemic cells induce proliferation in CLL cells and that such proliferation is blocked by a TLR7/8/9 inhibitor, by DNase, and by the PDE4 inhibitor rolipram. Rolipram also inhibited CLL cell proliferation induced by synthetic TLR7 and TLR9 agonists, as well as TLR agonist-induced costimulatory molecule expression and TNF-a (but not IL-6 or IL-10) production. Whereas treatment with a TLR9 agonist protected IgH V region unmutated, but not mutated, CLL cells from apoptosis, PDE4 inhibitors augmented apoptosis in both subtypes, suggesting that cAMP-mediated signaling may abrogate a TLR9-mediated survival signal in prognostically unfavorable IGHV unmutated CLL cells. Rolipram inhibited both TLR7/8- and TLR9-induced IFN regulatory factor 5 and NF-kB p65 nuclear translocation. PDE4 inhibitors also blocked TLR signaling in normal human immune cells. In PBMC and CD14-positive monocytes, PDE4 inhibitors blocked IFN-a or TNF-a (but not IL-6) production, respectively, following stimulation with synthetic TLR agonists or RNA-containing immune complexes. These results suggest that PDE4 inhibitors may be of clinical utility in CLL or autoimmune diseases that are driven by TLR-mediated signaling.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Phosphodiesterase 4 Inhibitors/therapeutic use , Rolipram/pharmacology , Active Transport, Cell Nucleus , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Apoptosis/radiation effects , Base Sequence , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Deoxyribonucleases/pharmacology , Female , Humans , Interferon Regulatory Factors/metabolism , Interferon-alpha/biosynthesis , Interleukin-10/biosynthesis , Interleukin-6/biosynthesis , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lipopolysaccharide Receptors/metabolism , Male , Middle Aged , Monocytes/immunology , Sequence Analysis, DNA , Signal Transduction , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 8/antagonists & inhibitors , Toll-Like Receptor 9/antagonists & inhibitors , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
5.
PLoS One ; 9(7): e103478, 2014.
Article in English | MEDLINE | ID: mdl-25076492

ABSTRACT

Interferon regulatory factor 5 (IRF5) polymorphisms are strongly associated with an increased risk of developing the autoimmune disease systemic lupus erythematosus. In mouse lupus models, IRF5-deficiency was shown to reduce disease severity consistent with an important role for IRF5 in disease pathogenesis. However these mouse studies were confounded by the recent demonstration that the IRF5 knockout mouse line contained a loss-of-function mutation in the dedicator of cytokinesis 2 (DOCK2) gene. As DOCK2 regulates lymphocyte trafficking and Toll-like receptor signaling, this raised the possibility that some of the protective effects attributed to IRF5 deficiency in the mouse lupus models may instead have been due to DOCK2 deficiency. We have therefore here evaluated the effect of IRF5-deficiency in the MRL/lpr mouse lupus model in the absence of the DOCK2 mutation. We find that IRF5-deficient (IRF5-/-) MRL/lpr mice develop much less severe disease than their IRF5-sufficient (IRF5+/+) littermates. Despite markedly lower serum levels of anti-nuclear autoantibodies and reduced total splenocyte and CD4+ T cell numbers, IRF5-/- MRL/lpr mice have similar numbers of all splenic B cell subsets compared to IRF5+/+ MRL/lpr mice, suggesting that IRF5 is not involved in B cell development up to the mature B cell stage. However, IRF5-/- MRL/lpr mice have greatly reduced numbers of spleen plasmablasts and bone marrow plasma cells. Serum levels of B lymphocyte stimulator (BLyS) were markedly elevated in the MRL/lpr mice but no effect of IRF5 on serum BLyS levels was seen. Overall our data demonstrate that IRF5 contributes to disease pathogenesis in the MRL/lpr lupus model and that this is due, at least in part, to the role of IRF5 in plasma cell formation. Our data also suggest that combined therapy targeting both IRF5 and BLyS might be a particularly effective therapeutic approach in lupus.


Subject(s)
GTPase-Activating Proteins/genetics , Interferon Regulatory Factors/genetics , Lupus Erythematosus, Systemic/pathology , Animals , Autoantibodies/blood , B-Cell Activating Factor/blood , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors , Immunoglobulin Isotypes/metabolism , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/mortality , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Mutation , Severity of Illness Index , Spleen/cytology , Survival Analysis , Th1 Cells/metabolism
6.
Methods Mol Biol ; 1169: 143-58, 2014.
Article in English | MEDLINE | ID: mdl-24957237

ABSTRACT

Impaired apoptotic cell clearance is thought to contribute to the pathogenesis of systemic autoimmune disease, in particular systemic lupus erythematosus (SLE). Endogenous RNA- and DNA-containing autoantigens released from dying cells can engage Toll-like receptors (TLR) 7/8 and TLR9, respectively in a number of immune cell types, thereby promoting innate and adaptive immune responses. Mouse models of lupus reliably phenocopy many of the characteristic features of SLE in humans and these models have proved invaluable in defining disease mechanisms. TLR7 signaling is essential for the development of autoantibodies to RNA and RNA-associated proteins like Sm and RNP, while TLR9 signaling is important for the development of antibodies to DNA and chromatin. TLR7 deficiency ameliorates end-organ disease, but, surprisingly, TLR9 deficiency exacerbates disease, possibly as a result of TLR7 overactivity in TLR9-deficient mice. Deficiency of interferon regulatory factor 5 (IRF5) inhibits autoantibody production and ameliorates disease likely due to its role in both TLR7 and TLR9 signaling. In this report we describe methods to analyze two commonly used mouse models of SLE in which TLRs and/or IRF5 have been shown to play a role in disease pathogenesis.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Nucleic Acids/immunology , Animals , Disease Models, Animal , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Lupus Erythematosus, Systemic/genetics , Mice , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
7.
Proc Natl Acad Sci U S A ; 110(13): 5133-8, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23479618

ABSTRACT

Patients with type 2 diabetes (T2D) have disease-associated changes in B-cell function, but the role these changes play in disease pathogenesis is not well established. Data herein show B cells from obese mice produce a proinflammatory cytokine profile compared with B cells from lean mice. Complementary in vivo studies show that obese B cell-null mice have decreased systemic inflammation, inflammatory B- and T-cell cytokines, adipose tissue inflammation, and insulin resistance (IR) compared with obese WT mice. Reduced inflammation in obese/insulin resistant B cell-null mice associates with an increased percentage of anti-inflammatory regulatory T cells (Tregs). This increase contrasts with the sharply decreased percentage of Tregs in obese compared with lean WT mice and suggests that B cells may be critical regulators of T-cell functions previously shown to play important roles in IR. We demonstrate that B cells from T2D (but not non-T2D) subjects support proinflammatory T-cell function in obesity/T2D through contact-dependent mechanisms. In contrast, human monocytes increase proinflammatory T-cell cytokines in both T2D and non-T2D analyses. These data support the conclusion that B cells are critical regulators of inflammation in T2D due to their direct ability to promote proinflammatory T-cell function and secrete a proinflammatory cytokine profile. Thus, B cells are potential therapeutic targets for T2D.


Subject(s)
B-Lymphocytes/immunology , Cytokines/immunology , Diabetes Mellitus, Type 2/immunology , Obesity/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes/pathology , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/therapy , Female , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Male , Mice , Mice, Obese , Obesity/pathology , Obesity/therapy , T-Lymphocytes, Regulatory/pathology
8.
Int Immunol ; 25(5): 295-306, 2013 May.
Article in English | MEDLINE | ID: mdl-23291967

ABSTRACT

Interferon regulatory factor 5-deficient (IRF5 (-/-) ) mice have been used for many studies of IRF5 biology. A recent report identifies a mutation in dedicator of cytokinesis 2 (DOCK2) as being responsible for the abnormal B-cell development phenotype observed in the IRF5 (-/-) line. Both dedicator of cytokinesis 2 (DOCK2) and IRF5 play important roles in immune cell function, raising the issue of whether immune effects previously associated with IRF5 are due to IRF5 or DOCK2. Here, we defined the insertion end-point of the DOCK2 mutation and designed a novel PCR to detect the mutation in genomic DNA. We confirmed the association of the DOCK2 mutation and the abnormal B-cell phenotype in our IRF5 (-/-) line and also established another IRF5 (-/-) line without the DOCK2 mutation. These two lines were used to compare the role of IRF5 in dendritic cells (DCs) and B cells in the presence or absence of the DOCK2 mutation. IRF5 deficiency reduces IFN-α, IFN-ß and IL-6 production by Toll-like receptor 9 (TLR9)- and TLR7-stimulated DCs and reduces TLR7- and TLR9-induced IL-6 production by B cells to a similar extent in the two lines. Importantly however, IRF5 (-/-) mice with the DOCK2 mutation have higher serum levels of IgG1 and lower levels of IgG2b, IgG2a/c and IgG3 than IRF5 (-/-) mice without the DOCK2 mutation, suggesting that the DOCK2 mutation confers additional Th2-type effects. Overall, these studies help clarify the function of IRF5 in B cells and DCs in the absence of the DOCK2 mutation. In addition, the PCR described will be useful for other investigators using the IRF5 (-/-) mouse line.


Subject(s)
B-Lymphocytes/metabolism , Dendritic Cells/metabolism , GTPase-Activating Proteins/genetics , Interferon Regulatory Factors/deficiency , Animals , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors , Interferon Regulatory Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Phenotype
9.
J Immunol ; 184(2): 796-806, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20007534

ABSTRACT

Polymorphisms in the transcription factor IFN regulatory factor 5 (IRF5) are strongly associated in human genetic studies with an increased risk of developing the autoimmune disease systemic lupus erythematosus. However, the biological role of IRF5 in lupus pathogenesis has not previously been tested in an animal model. In this study, we show that IRF5 is absolutely required for disease development in the FcgammaRIIB(-/-)Yaa and FcgammaRIIB(-/-) lupus models. In contrast to IRF5-sufficient FcgammaRIIB(-/-)Yaa mice, IRF5-deficient FcgammaRIIB(-/-)Yaa mice do not develop lupus manifestations and have a phenotype comparable to wild-type mice. Strikingly, full expression of IRF5 is required for the development of autoimmunity, as IRF5 heterozygotes had dramatically reduced disease. One effect of IRF5 is to induce the production of the type I IFN, IFN-alpha, a cytokine implicated in lupus pathogenesis. To address the mechanism by which IRF5 promotes disease, we evaluated FcgammaRIIB(-/-)Yaa mice lacking the type I IFN receptor subunit 1. Unlike the IRF5-deficient and IRF5-heterozygous FcgammaRIIB(-/-)Yaa mice, type I IFN receptor subunit 1-deficient FcgammaRIIB(-/-)Yaa mice maintained a substantial level of residual disease. Furthermore, in FcgammaRIIB(-/-) mice lacking Yaa, IRF5-deficiency also markedly reduced disease manifestations, indicating that the beneficial effects of IRF5 deficiency in FcgammaRIIB(-/-)Yaa mice are not due only to inhibition of the enhanced TLR7 signaling associated with the Yaa mutation. Overall, we demonstrate that IRF5 plays an essential role in lupus pathogenesis in murine models and that this is mediated through pathways beyond that of type I IFN production.


Subject(s)
Interferon Regulatory Factors/physiology , Lupus Erythematosus, Systemic/etiology , Receptors, IgG/deficiency , Animals , Autoimmunity , Disease Models, Animal , Genotype , Interferon-alpha/biosynthesis , Mice , Mice, Knockout , Mutation
10.
J Immunol ; 182(2): 820-8, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19124725

ABSTRACT

Exacerbation of disease in systemic lupus erythematosus (SLE) is associated with bacterial infection. In conventional dendritic cells (cDCs), the TLR4 ligand bacterial LPS induces IFN-beta gene expression but does not induce IFN-alpha. We hypothesized that when cDCs are primed by cytokines, as may frequently be the case in SLE, LPS would then induce the production of IFN-alpha, a cytokine believed to be important in lupus pathogenesis. In this study we show that mouse cDCs and human monocytes produce abundant IFN-alpha following TLR4 engagement whether the cells have been pretreated either with IFN-beta or with a supernatant from DCs activated by RNA-containing immune complexes from lupus patients. This TLR4-induced IFN-alpha induction is mediated by both an initial TRIF-dependent pathway and a subsequent MyD88-dependent pathway, in contrast to TLR3-induced IFN-alpha production, which is entirely TRIF-dependent. There is also a distinct requirement for IFN regulatory factors (IRFs), with LPS-induced IFN-alpha induction being entirely IRF7- and partially IRF5-dependent, in contrast to LPS-induced IFN-beta gene induction which is known to be IRF3-dependent but largely IRF7-independent. This data demonstrates a novel pathway for IFN-alpha production by cDCs and provides one possible explanation for how bacterial infection might precipitate disease flares in SLE.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon-alpha/biosynthesis , Interferon-beta/physiology , Lipopolysaccharides/metabolism , Monocytes/immunology , Monocytes/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cell-Free System/immunology , Cell-Free System/metabolism , Cell-Free System/pathology , Cells, Cultured , Humans , Immunoglobulin G/pharmacology , Interferon-alpha/physiology , Ligands , Lipopolysaccharides/pharmacology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...