Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
iScience ; 26(10): 108079, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37860753

ABSTRACT

Glucocorticoids (GCs) are potent anti-inflammatory agents and are broadly used in treating rheumatoid arthritis (RA) patients, albeit with adverse side effects associated with long-term usage. The negative consequences of GC therapy provide an impetus for research into gaining insights into the molecular mechanisms of GC action. We have previously reported that granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced CCL17 has a non-redundant role in inflammatory arthritis. Here, we provide molecular evidence that GCs can suppress GM-CSF-mediated upregulation of IRF4 and CCL17 expression via downregulating JMJD3 expression and activity. In mouse models of inflammatory arthritis, GC treatment inhibited CCL17 expression and ameliorated arthritic pain-like behavior and disease. Significantly, GC treatment of RA patient peripheral blood mononuclear cells ex vivo resulted in decreased CCL17 production. This delineated pathway potentially provides new therapeutic options for the treatment of many inflammatory conditions, where GCs are used as an anti-inflammatory drug but without the associated adverse side effects.

2.
Cell Rep ; 41(8): 111674, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36417867

ABSTRACT

A possible explanation for chronic inflammation in HIV-infected individuals treated with anti-retroviral therapy is hyperreactivity of myeloid cells due to a phenomenon called "trained immunity." Here, we demonstrate that human monocyte-derived macrophages originating from monocytes initially treated with extracellular vesicles containing HIV-1 protein Nef (exNef), but differentiating in the absence of exNef, release increased levels of pro-inflammatory cytokines after lipopolysaccharide stimulation. This effect is associated with chromatin changes at the genes involved in inflammation and cholesterol metabolism pathways and upregulation of the lipid rafts and is blocked by methyl-ß-cyclodextrin, statin, and an inhibitor of the lipid raft-associated receptor IGF1R. Bone-marrow-derived macrophages from exNef-injected mice, as well as from mice transplanted with bone marrow from exNef-injected animals, produce elevated levels of tumor necrosis factor α (TNF-α) upon stimulation. These phenomena are consistent with exNef-induced trained immunity that may contribute to persistent inflammation and associated co-morbidities in HIV-infected individuals with undetectable HIV load.


Subject(s)
Extracellular Vesicles , HIV Infections , HIV Seropositivity , HIV-1 , Humans , Mice , Animals , HIV-1/genetics , nef Gene Products, Human Immunodeficiency Virus/genetics , Extracellular Vesicles/metabolism , Macrophages/metabolism , Inflammation/metabolism
3.
Cell Rep ; 39(3): 110719, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35443173

ABSTRACT

Metabolic adaptations can directly influence the scope and scale of macrophage activation and polarization. Here we explore the impact of type I interferon (IFNß) on macrophage metabolism and its broader impact on cytokine signaling pathways. We find that IFNß simultaneously increased the expression of immune-responsive gene 1 and itaconate production while inhibiting isocitrate dehydrogenase activity and restricting α-ketoglutarate accumulation. IFNß also increased the flux of glutamine-derived carbon into the tricarboxylic acid cycle to boost succinate levels. Combined, we identify that IFNß controls the cellular α-ketoglutarate/succinate ratio. We show that by lowering the α-ketoglutarate/succinate ratio, IFNß potently blocks the JMJD3-IRF4-dependent pathway in GM-CSF and IL-4 activated macrophages. The suppressive effects of IFNß on JMJD3-IRF4-dependent responses, including M2 polarization and GM-CSF-induced inflammatory pain, were reversed by supplementation with α-ketoglutarate. These results reveal that IFNß modulates macrophage activation and polarization through control of the cellular α-ketoglutarate/succinate ratio.


Subject(s)
Interferon Type I , Macrophage Activation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/pharmacology , Succinic Acid
4.
Chem Res Toxicol ; 35(3): 475-489, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35212515

ABSTRACT

The potential for N-nitrosamine impurities in pharmaceutical products presents a challenge for the quality management of medicinal products. N-Nitrosamines are considered cohort-of-concern compounds due to the potent carcinogenicity of many of the structurally simple chemicals within this structural class. In the past 2 years, a number of drug products containing certain active pharmaceutical ingredients have been withdrawn or recalled from the market due to the presence of carcinogenic low-molecular-weight N,N-dialkylnitrosamine impurities. Regulatory authorities have issued guidance to market authorization holders to review all commercial drug substances/products for the potential risk of N-nitrosamine impurities, and in cases where a significant risk of N-nitrosamine impurity is identified, analytical confirmatory testing is required. A key factor to consider prior to analytical testing is the estimation of the daily acceptable intake (AI) of the N-nitrosamine impurity. A significant proportion of N-nitrosamine drug product impurities are unique/complex structures for which the development of low-level analytical methods is challenging. Moreover, these unique/complex impurities may be less potent carcinogens compared to simple nitrosamines. In the present work, our objective was to derive AIs for a large number of complex N-nitrosamines without carcinogenicity data that were identified as potential low-level impurities. The impurities were first cataloged and grouped according to common structural features, with a total of 13 groups defined with distinct structural features. Subsequently, carcinogenicity data were reviewed for structurally related N-nitrosamines relevant to each of the 13 structural groups and group AIs were derived conservatively based on the most potent N-nitrosamine within each group. The 13 structural group AIs were used as the basis for assigning AIs to each of the structurally related complex N-nitrosamine impurities. The AIs of several N-nitrosamine groups were found to be considerably higher than those for the simple N,N-dialkylnitrosamines, which translates to commensurately higher analytical method detection limits.


Subject(s)
Nitrosamines , Carcinogens , Drug Contamination , Humans
5.
Int J Mol Sci ; 23(4)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35216355

ABSTRACT

It is now well understood that the bone marrow (BM) compartment can sense systemic inflammatory signals and adapt through increased proliferation and lineage skewing. These coordinated and dynamic alterations in responding hematopoietic stem and progenitor cells (HSPCs), as well as in cells of the bone marrow niche, are increasingly viewed as key contributors to the inflammatory response. Growth factors, cytokines, metabolites, microbial products, and other signals can cause dysregulation across the entire hematopoietic hierarchy, leading to lineage-skewing and even long-term functional adaptations in bone marrow progenitor cells. These alterations may play a central role in the chronicity of disease as well as the links between many common chronic disorders. The possible existence of a form of "memory" in bone marrow progenitor cells is thought to contribute to innate immune responses via the generation of trained immunity (also called innate immune memory). These findings highlight how hematopoietic progenitors dynamically adapt to meet the demand for innate immune cells and how this adaptive response may be beneficial or detrimental depending on the context. In this review, we will discuss the role of bone marrow progenitor cells and their microenvironment in shaping the scope and scale of the immune response in health and disease.


Subject(s)
Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Inflammation/pathology , Stem Cell Niche/physiology , Stem Cells/pathology , Animals , Chronic Disease , Humans , Immunity, Innate/physiology
6.
Clin Transl Immunology ; 9(12): e1222, 2020.
Article in English | MEDLINE | ID: mdl-33363732

ABSTRACT

Macrophages are the first immune cells in the developing embryo and have a central role in organ development, homeostasis, immunity and repair. Over the last century, our understanding of these cells has evolved from being thought of as simple phagocytic cells to master regulators involved in governing a myriad of cellular processes. A better appreciation of macrophage biology has been matched with a clearer understanding of their diverse origins and the flexibility of their metabolic and transcriptional machinery. The understanding of the classical mononuclear phagocyte system in its original form has now been expanded to include the embryonic origin of tissue-resident macrophages. A better knowledge of the intrinsic similarities and differences between macrophages of embryonic or monocyte origin has highlighted the importance of ontogeny in macrophage dysfunction in disease. In this review, we provide an update on origin and classification of tissue macrophages, the mechanisms of macrophage specialisation and their role in health and disease. The importance of the macrophage niche in providing trophic factors and a specialised environment for macrophage differentiation and specialisation is also discussed.

7.
J Immunol ; 205(1): 213-222, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32461237

ABSTRACT

It has been reported that a GM-CSF→CCL17 pathway, originally identified in vitro in macrophage lineage populations, is implicated in the control of inflammatory pain, as well as arthritic pain and disease. We explore, in this study and in various inflammation models, the cellular CCL17 expression and its GM-CSF dependence as well as the function of CCL17 in inflammation and pain. This study used models allowing the convenient cell isolation from Ccl17E/+ reporter mice; it also exploited both CCL17-dependent and unique CCL17-driven inflammatory pain and arthritis models, the latter permitting a radiation chimera approach to help identify the CCL17 responding cell type(s) and the mediators downstream of CCL17 in the control of inflammation and pain. We present evidence that 1) in the particular inflammation models studied, CCL17 expression is predominantly in macrophage lineage populations and is GM-CSF dependent, 2) for its action in arthritic pain and disease development, CCL17 acts on CCR4+ non-bone marrow-derived cells, and 3) for inflammatory pain development in which a GM-CSF→CCL17 pathway appears critical, nerve growth factor, CGRP, and substance P all appear to be required.


Subject(s)
Arthritis, Experimental/immunology , Chemokine CCL17/metabolism , Pain/immunology , Peritonitis/immunology , Pneumonia/immunology , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/pathology , Calcitonin Gene-Related Peptide/metabolism , Chemokine CCL17/genetics , Genes, Reporter/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Mice , Mice, Transgenic , Nerve Growth Factor/metabolism , Pain/diagnosis , Pain/pathology , Pain Measurement , Peritonitis/complications , Peritonitis/pathology , Pneumonia/complications , Pneumonia/pathology , Signal Transduction/immunology , Substance P/metabolism
8.
Arthritis Res Ther ; 22(1): 123, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32471485

ABSTRACT

BACKGROUND: The cytokine, interleukin-23 (IL-23), can be critical for the progression of inflammatory diseases, including arthritis, and is often associated with T lymphocyte biology. We previously showed that certain lymphocyte-independent, inflammatory arthritis and pain models have a similar requirement for tumour necrosis factor (TNF), granulocyte macrophage-colony stimulating factor (GM-CSF), and C-C motif ligand 17 (CCL17). Given this correlation in cytokine requirements, we explored whether IL-23 might interact with this cytokine cluster in the control of arthritic and inflammatory pain. METHODS: The role of IL-23 in the development of pain-like behaviour was investigated using mouse arthritis models (zymosan-induced arthritis and GM-CSF-, TNF-, and CCL17-driven monoarticular arthritis) and inflammatory pain models (intraplantar zymosan, GM-CSF, TNF, and CCL17). Additionally, IL-23-induced inflammatory pain was measured in GM-CSF-/-, Tnf-/-, and Ccl17E/E mice and in the presence of indomethacin. Pain-like behaviour and arthritis were assessed by relative weight distribution in hindlimbs and histology, respectively. Cytokine mRNA expression in knees and paw skin was analysed by quantitative PCR. Blood and synovial cell populations were analysed by flow cytometry. RESULTS: We report, using Il23p19-/- mice, that innate immune (zymosan)-driven arthritic pain-like behaviour (herein referred to as pain) was completely dependent upon IL-23; optimal arthritic disease development required IL-23 (P < 0.05). Zymosan-induced inflammatory pain was also completely dependent on IL-23. In addition, we found that exogenous TNF-, GM-CSF-, and CCL17-driven arthritic pain, as well as inflammatory pain driven by each of these cytokines, were absent in Il23p19-/- mice; optimal disease in these mBSA-primed models was dependent on IL-23 (P < 0.05). Supporting this cytokine connection, it was found conversely that IL-23 (200 ng) can induce inflammatory pain at 4 h (P < 0.0001) with a requirement for each of the other cytokines as well as cyclooxygenase activity. CONCLUSIONS: These findings indicate a role for IL-23 in innate immune-mediated arthritic and inflammatory pain with potential links to TNF, GM-CSF, CCL17, and eicosanoid function.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , Interleukin-23 , Animals , Cytokines , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Mice , Pain , Tumor Necrosis Factor-alpha
9.
J Clin Invest ; 129(10): 4239-4244, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31483287

ABSTRACT

Macrophage activation in response to LPS is coupled to profound metabolic changes, typified by accumulation of the TCA cycle intermediates citrate, itaconate, and succinate. We have identified that endogenous type I IFN controls the cellular citrate/α-ketoglutarate ratio and inhibits expression and activity of isocitrate dehydrogenase (IDH); and, via 13C-labeling studies, demonstrated that autocrine type I IFN controls carbon flow through IDH in LPS-activated macrophages. We also found that type I IFN-driven IL-10 contributes to inhibition of IDH activity and itaconate synthesis in LPS-stimulated macrophages. Our findings have identified the autocrine type I IFN pathway as being responsible for the inhibition of IDH in LPS-stimulated macrophages.


Subject(s)
Interferon Type I/metabolism , Isocitrate Dehydrogenase/antagonists & inhibitors , Macrophages/immunology , Macrophages/metabolism , Animals , Autocrine Communication , Citric Acid Cycle , Humans , Interleukin-10/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Succinates/metabolism
10.
Front Immunol ; 10: 2054, 2019.
Article in English | MEDLINE | ID: mdl-31555280

ABSTRACT

Monocytes in humans consist of 3 subsets; CD14+CD16- (classical), CD14+CD16+ (intermediate) and CD14dimCD16+ (non-classical), which exhibit distinct and heterogeneous responses to activation. During acute inflammation CD14+CD16- monocytes are significantly elevated and migrate to the sites of injury via the adhesion cascade. The field of immunometabolism has begun to elucidate the importance of the engagement of specific metabolic pathways in immune cell function. Yet, little is known about monocyte metabolism and the role of metabolism in mediating monocyte activation and adherence to vessels. Accordingly, we aimed to determine whether manipulating the metabolism of CD14+CD16- monocytes alters their ability to become activated and adhere. We discovered that LPS stimulation increased the rate of glycolysis in human CD14+CD16- monocytes. Inhibition of glycolysis with 2-deoxy-D-glucose blunted LPS-induced activation and adhesion of monocytes. Mechanistically, we found that increased glycolysis was regulated by mTOR-induced glucose transporter (GLUT)-1. Furthermore, enhanced glycolysis increased accumulation of reactive oxygen species (ROS) and activation of p38 MAPK, which lead to activation and adhesion of monocytes. These findings reveal that glycolytic metabolism is critical for the activation of CD14+CD16- monocytes and contributes to our understanding of the interplay between metabolic substrate preference and immune cell function.


Subject(s)
Inflammation/immunology , Monocytes/metabolism , Reactive Oxygen Species/metabolism , Cell Adhesion , Cells, Cultured , Deoxyglucose/metabolism , Glucose Transporter Type 1/metabolism , Glycolysis , Humans , Immunophenotyping , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/metabolism , MAP Kinase Signaling System , Monocytes/immunology , Receptors, IgG/metabolism , TOR Serine-Threonine Kinases/metabolism
11.
J Immunol ; 202(10): 3033-3040, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30988114

ABSTRACT

Studies have demonstrated the importance of a GM-CSF→IFN regulatory factor 4 (IRF4)→CCL17 pathway, first identified in monocytes/macrophages, for arthritic pain and disease development. In this study, we further investigated the involvement of this new pathway in shaping the inflammatory response using the zymosan-induced peritonitis (ZIP) model. ZIP (8 mg of zymosan, i.p., day 0) was induced in C57BL/6 wild-type (WT), GM-CSF-/- , Irf4-/- , and Ccl17E/E mice. In comparison with WT mice, GM-CSF-/- and Irf4-/- mice had a reduced ZIP response, as judged by a reduced number of neutrophils and macrophages in the peritoneal cavity. Moreover, the phenotype of the ZIP macrophages was altered by a lack of GM-CSF or IRF4 (increased IL-10 secretion and Arg1 mRNA expression), with IRF4 levels being lower in GM-CSF-/- ZIP macrophages than in the WT cells. In addition, GM-CSF ̶IRF4 signaling upregulated MHC class II expression in ZIP macrophages and bone marrow-derived macrophages. Although Ccl17 mRNA expression was reduced in ZIP macrophages in the absence of either GM-CSF or IRF4, thus supporting the presence of the new pathway in inflammatory macrophages, CCL17 did not modulate the inflammatory response, both in terms of number of myeloid cells or the macrophage phenotype. Thus, during an inflammatory response, both macrophage numbers and their phenotype can depend on GM-CSF- and IRF4-dependent signaling independently of CCL17.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interferon Regulatory Factors/immunology , Macrophages/immunology , Signal Transduction/immunology , Animals , Chemokine CCL17/genetics , Chemokine CCL17/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon Regulatory Factors/genetics , Macrophages/pathology , Mice , Mice, Knockout , Signal Transduction/genetics , Up-Regulation/immunology
12.
J Immunol ; 201(7): 2042-2053, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30120124

ABSTRACT

Pain is one of the most debilitating symptoms in many diseases for which there is inadequate management and understanding. CSF-1, also known as M-CSF, acts via its receptor (CSF-1R, c-Fms) to regulate the development of the monocyte/macrophage lineage and to act locally in tissues to control macrophage numbers and function. It has been implicated in the control of neuropathic pain via a central action on microglia. We report in this study that systemic administration of a neutralizing anti-CSF-1R or CSF-1 mAb inhibits the development of inflammatory pain induced by zymosan, GM-CSF, and TNF in mice. This approach also prevented but did not ameliorate the development of arthritic pain and optimal disease driven by the three stimuli in mice, suggesting that CSF-1 may only be relevant when the driving inflammatory insults in tissues are acute and/or periodic. Systemic CSF-1 administration rapidly induced pain and enhanced the arthritis in an inflamed mouse joint, albeit via a different pathway(s) from that used by systemic GM-CSF and TNF. It is concluded that CSF-1 can function peripherally during the generation of inflammatory pain and hence may be a target for such pain and associated disease, including when the clinically important cytokines, TNF and GM-CSF, are involved. Our findings have ramifications for the selection and design of anti-CSF-1R/CSF-1 trials.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Inflammation/immunology , Joints/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/physiology , Monocytes/physiology , Animals , Antibodies, Neutralizing/administration & dosage , Cell Differentiation , Cell Lineage , Humans , Macrophage Colony-Stimulating Factor/immunology , Mice , Mice, Inbred C57BL , Pain , Receptor, Macrophage Colony-Stimulating Factor/immunology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction
13.
J Biol Chem ; 293(29): 11415-11423, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29871928

ABSTRACT

Interleukin 4 (IL4) is generally viewed as a Th2 cytokine capable of polarizing macrophages into an anti-inflammatory phenotype, whereas granulocyte macrophage-colony-stimulating factor (GM-CSF) is often viewed as a proinflammatory cytokine with part of this function due to its action on monocytes/macrophages. Paradoxically, these two cytokines act additively to enhance the in vitro differentiation of dendritic cells from precursors such as monocytes. One up-regulated marker of an IL4-polarized M2 macrophage is the chemokine (C-C motif) ligand 17 (CCL17), which we have recently reported to be induced by GM-CSF in monocytes/macrophages in an interferon regulatory factor 4 (IRF4)-dependent manner. In this study, we report that IL4 also induces CCL17 production by acting through IRF4 in human monocytes and murine macrophages. Furthermore, evidence is presented that IL4 up-regulates IRF4 expression at the epigenetic level by enhancing the expression and activity of jumonji domain-containing protein 3 (JMJD3) demethylase. Intriguingly, silencing the signal transducer and activator of transcription 6 (STAT6) gene led to a decrease in not only CCL17 formation, but also in that of its upstream regulators, JMJD3 and IRF4. Moreover, IL4 treatment of human monocytes resulted in an increased association of STAT6 to the promoter regions of the CCL17, IRF4, and JMJD3 genes. Thus, despite their vastly different functions, IL4 and GM-CSF appear to share elements of a common signaling pathway in regulating CCL17 production in human monocytes and murine macrophages.


Subject(s)
Chemokine CCL17/genetics , Epigenesis, Genetic , Interleukin-4/genetics , Macrophages/metabolism , Monocytes/metabolism , Transcriptional Activation , Animals , Cells, Cultured , Humans , Interferon Regulatory Factors/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, Inbred C57BL , Up-Regulation
14.
Methods Mol Biol ; 1784: 215-223, 2018.
Article in English | MEDLINE | ID: mdl-29761402

ABSTRACT

Animal models of arthritis enable us to investigate the pathogenesis of the disease and also to evaluate new therapies. Here we describe two different acute inflammatory monoarticular arthritis models (mBSA/IL1ß and mBSA/GM-CSF) providing a more rapid and potentially simplified approach to investigate the pathogenesis.


Subject(s)
Arthritis, Experimental/genetics , Cytokines/toxicity , Inflammation/genetics , Rheumatic Fever/genetics , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Inflammation/chemically induced , Inflammation/pathology , Mice , Rheumatic Fever/chemically induced , Rheumatic Fever/pathology
15.
Arthritis Res Ther ; 20(1): 62, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29622035

ABSTRACT

BACKGROUND: Granulocyte macrophage-colony stimulating factor (GM-CSF) has been implicated in the pathogenesis of a number of inflammatory diseases and in osteoarthritis (OA). We identified previously a new GM-CSF→Jmjd3→interferon regulatory factor 4 (IRF4)→chemokine (c-c motif) ligand 17 (CCL17) pathway, which is important for the development of inflammatory arthritis pain and disease. Tumour necrosis factor (TNF) can also be linked with this pathway. Here we investigated the involvement of the pathway in OA pain and disease development using the GM-CSF-dependent collagenase-induced OA (CiOA) model. METHODS: CiOA was induced in C57BL/6 wild-type (WT), Irf4 -/- , Ccl17 E/E , Ccr4 -/- , Tnf -/- and GM-CSF -/- mice. Additionally, therapeutic targeting of CCL17, Jmjd3 and cyclooxygenase 2 (COX-2) was evaluated. Development of pain (assessment of weight distribution) and OA disease (histologic scoring of synovitis, cartilage destruction and osteophyte size) were assessed. Synovial joint cells, including neutrophils, macrophages, fibroblasts and endothelial cells, were isolated (cell sorting) and gene expression analyzed (quantitative PCR). RESULTS: Studies in the gene-deficient mice indicated that IRF4, CCL17 and the CCL17 receptor, CCR4, but not TNF, were required for CiOA pain and optimal cartilage destruction and osteophyte size. Therapeutic neutralization of CCL17 and Jmjd3 ameliorated both pain and disease, whereas the COX-2 inhibitor only ameliorated pain. In the synovium Ccl17 mRNA was expressed only in the macrophages in a GM-CSF-dependent and IRF4-dependent manner. CONCLUSIONS: The GM-CSF→Jmjd3→IRF4→CCL17 pathway is important for the development of CiOA, with CCL17 thus being a potential therapeutic target for the treatment of both OA pain and disease.


Subject(s)
Chemokine CCL17/antagonists & inhibitors , Chemokine CCL17/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Pain/drug therapy , Pain/metabolism , Animals , Disease Progression , Knee Joint/drug effects , Knee Joint/metabolism , Knee Joint/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis/pathology , Pain/pathology
16.
JCI Insight ; 3(6)2018 03 22.
Article in English | MEDLINE | ID: mdl-29563337

ABSTRACT

TNF and granulocyte macrophage-colony stimulating factor (GM-CSF) have proinflammatory activity and both contribute, for example, to rheumatoid arthritis pathogenesis. We previously identified a new GM-CSF→JMJD3 demethylase→interferon regulatory factor 4 (IRF4)→CCL17 pathway that is active in monocytes/macrophages in vitro and important for inflammatory pain, as well as for arthritic pain and disease. Here we provide evidence for a nexus between TNF and this pathway, and for TNF and GM-CSF interdependency. We report that the initiation of zymosan-induced inflammatory pain and zymosan-induced arthritic pain and disease are TNF dependent. Once arthritic pain and disease are established, blockade of GM-CSF or CCL17, but not of TNF, is still able to ameliorate them. TNF is required for GM-CSF-driven inflammatory pain and for initiation of GM-CSF-driven arthritic pain and disease, but not once they are established. TNF-driven inflammatory pain and TNF-driven arthritic pain and disease are dependent on GM-CSF and mechanistically require the same downstream pathway involving GM-CSF→CCL17 formation via JMJD3-regulated IRF4 production, indicating that GM-CSF and CCL17 can mediate some of the proinflammatory and algesic actions of TNF. Given we found that TNF appears important only early in arthritic pain and disease progression, targeting a downstream mediator, such as CCL17, which appears to act throughout the course of disease, could be effective at ameliorating chronic inflammatory conditions where TNF is implicated.


Subject(s)
Arthritis, Rheumatoid/metabolism , Chemokine CCL17/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/physiopathology , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocytes , Inflammation/immunology , Inflammation/pathology , Inflammation/physiopathology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Jumonji Domain-Containing Histone Demethylases , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain/chemically induced , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Zymosan/pharmacology
17.
Cell Death Dis ; 9(3): 267, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449600

ABSTRACT

Glucocorticoids (GCs) are potent anti-inflammatory drugs whose mode of action is complex and still debatable. One likely cellular target of GCs are monocytes/macrophages. The role of GCs in monocyte survival is also debated. Although both granulocyte macrophage-colony stimulating factor (GM-CSF) and macrophage-CSF (M-CSF) are important regulators of macrophage lineage functions including their survival, the former is often associated with proinflammatory functions while the latter is important in lineage homeostasis. We report here that the GC, dexamethasone, induces apoptosis in GM-CSF-treated human monocytes while having no impact on M-CSF-induced monocyte survival. To understand how GCs, GM-CSF, and M-CSF are regulating monocyte survival and other functions during inflammation, we firstly examined the transcriptomic changes elicited by these three agents in human monocytes, either acting alone or in combination. Transcriptomic and Ingenuity pathway analyses found that dexamethasone differentially modulated dendritic cell maturation and TREM1 signaling pathways in GM-CSF-treated and M-CSF-treated monocytes, two pathways known to be regulated by ERK1/2 activity. These analyses led us to provide evidence that the GC inhibits ERK1/2 activity selectively in GM-CSF-treated monocytes to induce apoptosis. It is proposed that this inhibition of ERK1/2 activity leads to inactivation of p90 ribosomal-S6 kinase and Bad dephosphorylation leading in turn to enhanced caspase-3 activity and subsequent apoptosis. Furthermore, pharmacological inhibition of GC receptor activity restored the ERK1/2 signaling and prevented the GC-induced apoptosis in GM-CSF-treated monocytes. Increased tissue macrophage numbers, possibly from enhanced survival due to mediators such as GM-CSF, can correlate with inflammatory disease severity; also reduction in these numbers can correlate with the therapeutic benefit of a number of agents, including GCs. We propose that the ERK1/2 signaling pathway promotes survival of GM-CSF-treated proinflammatory monocytes, which can be selectively targeted by GCs as a novel mechanism to reduce local monocyte/macrophage numbers and hence inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Inflammation/prevention & control , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Monocytes/drug effects , Protein Kinase Inhibitors/pharmacology , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Inflammation/enzymology , Inflammation/pathology , Macrophage Colony-Stimulating Factor/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/enzymology , Monocytes/pathology , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Triggering Receptor Expressed on Myeloid Cells-1/metabolism
18.
Article in English | MEDLINE | ID: mdl-28824884

ABSTRACT

Porphyromonas gingivalis is one of the bacterial species most closely associated with periodontitis and can shed large numbers of outer membrane vesicles (OMVs), which are increasingly thought to play a significant role in bacterial virulence and pathogenicity. Macrophages are amongst the first immune cells to respond to bacteria and their products, so we sought to directly compare the response of macrophages to P. gingivalis or its purified OMVs. Macrophages stimulated with OMVs produced large amounts of TNFα, IL-12p70, IL-6, IL-10, IFNß, and nitric oxide compared to cells infected with P. gingivalis, which produced very low levels of these mediators. Both P. gingivalis and OMVs induced a shift in macrophage metabolism from oxidative phosphorylation (OXPHOS) to glycolysis, which was supported by enhanced lactate release, decreased mitochondrial oxygen consumption with reduced spare respiratory capacity, as well as increased mitochondrial reactive oxygen species (ROS) production. Corresponding to this metabolic shift, gene expression analysis of macrophages infected with P. gingivalis or stimulated with OMVs revealed a broad transcriptional upregulation of genes critical to glycolysis and a downregulation of genes associated with the TCA cycle. Upon examination of inflammasome signaling and pyroptosis it was found that P. gingivalis did not activate the inflammasome in macrophages as the mature forms of caspase-1, IL-1ß, and IL-18 were not detected and there was no extracellular release of lactate dehydrogenase (LDH) or 7-AAD staining. In comparison, macrophages stimulated with OMVs potently activated caspase-1, produced large amounts of IL-1ß, IL-18, released LDH, and were positive for 7-AAD indicative of pyroptotic cell death. These data directly quantitate the distinct effects of P. gingivalis and its OMVs on macrophage inflammatory phenotype, mitochondrial function, inflammasome activation, and pyroptotic cell death that may have potential implications for their roles in chronic periodontitis.


Subject(s)
Extracellular Vesicles/metabolism , Inflammasomes/immunology , Macrophages/metabolism , Macrophages/microbiology , Porphyromonas gingivalis/immunology , Pyroptosis , Animals , Caspase 1/metabolism , Cytokines/metabolism , Gene Expression , Glycolysis , Humans , Inflammation , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-18 , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Nitric Oxide/metabolism , Oxidative Phosphorylation , Oxygen Consumption , Porphyromonas gingivalis/pathogenicity , Tumor Necrosis Factor-alpha/metabolism
19.
Regul Toxicol Pharmacol ; 87: 54-63, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28461231

ABSTRACT

In response to increasing concerns around the potential environmental impact of industrial chemicals, the pharmaceutical industry is seeking alternatives for traditional solvents used during the manufacturing process. Taking into consideration the principles of green chemistry, 2-methyltetrahydrofuran (2-MeTHF) is proposed as a suitable replacement for the structurally similar solvent tetrahydrofuran (THF). 2-MeTHF is derived from renewable sources and is more easily recovered thereby facilitating its reuse. However, 2-MeTHF is currently not included in the International Conference on Harmonisation (ICH) Q3C residual solvent guidelines and there is no Permitted Daily Exposure (PDE) limit proposed below which there would be negligible safety concerns for patients exposed to it as a residual impurity in a drug product. To enable the calculation of a PDE, a GLP compliant 3-month repeat-dose oral toxicity study in rats with a 1-month recovery period was conducted with doses of 2-MeTHF of 0, 80, 250, 500 and 1000 mg/kg/day. Administration of doses of up to 1000 mg/kg/day was tolerated. Based upon minimal observed effects on the liver at ≥500 mg/kg/day, the NOAEL in this study was considered to be 250 mg/kg/day. Inclusion of this NOAEL, and a safety factor of 250 a PDE of 50 mg/day was derived to support the safe use of 2-MeTHF in the pharmaceutical industry.


Subject(s)
Furans , Solvents , Animals , Drug Industry , Humans , Maximum Allowable Concentration , No-Observed-Adverse-Effect Level , Pharmaceutical Preparations/chemistry , Rats
20.
J Immunol ; 198(9): 3565-3575, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28320832

ABSTRACT

G-CSF or CSF-3, originally defined as a regulator of granulocyte lineage development via its cell surface receptor (G-CSFR), can play a role in inflammation, and hence in many pathologies, due to its effects on mature lineage populations. Given this, and because pain is an extremely important arthritis symptom, the efficacy of an anti-G-CSFR mAb for arthritic pain and disease was compared with that of a neutrophil-depleting mAb, anti-Ly6G, in both adaptive and innate immune-mediated murine models. Pain and disease were ameliorated in Ag-induced arthritis, zymosan-induced arthritis, and methylated BSA/IL-1 arthritis by both prophylactic and therapeutic anti-G-CSFR mAb treatment, whereas only prophylactic anti-Ly6G mAb treatment was effective. Efficacy for pain and disease correlated with reduced joint neutrophil numbers and, importantly, benefits were noted without necessarily the concomitant reduction in circulating neutrophils. Anti-G-CSFR mAb also suppressed zymosan-induced inflammatory pain. A new G-CSF-driven (methylated BSA/G-CSF) arthritis model was established enabling us to demonstrate that pain was blocked by a cyclooxygenase-2 inhibitor, suggesting an indirect effect on neurons. Correspondingly, dorsal root ganglion neurons cultured in G-CSF failed to respond to G-CSF in vitro, and Csf3r gene expression could not be detected in dorsal root ganglion neurons by single-cell RT-PCR. These data suggest that G-CSFR/G-CSF targeting may be a safe therapeutic strategy for arthritis and other inflammatory conditions, particularly those in which pain is important, as well as for inflammatory pain per se.


Subject(s)
Antibodies, Blocking/therapeutic use , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/therapy , Immunotherapy/methods , Neurons/drug effects , Neutrophils/immunology , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Animals , Antigens, Ly/immunology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Cells, Cultured , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Leukocyte Reduction Procedures , Mice , Mice, Inbred C57BL , Neurons/physiology , Neutrophils/drug effects , Neutrophils/pathology , Pain Management , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...