Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 25(7): 1193-1206, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38834865

ABSTRACT

Immune cells experience large cell shape changes during environmental patrolling because of the physical constraints that they encounter while migrating through tissues. These cells can adapt to such deformation events using dedicated shape-sensing pathways. However, how shape sensing affects immune cell function is mostly unknown. Here, we identify a shape-sensing mechanism that increases the expression of the chemokine receptor CCR7 and guides dendritic cell migration from peripheral tissues to lymph nodes at steady state. This mechanism relies on the lipid metabolism enzyme cPLA2, requires nuclear envelope tensioning and is finely tuned by the ARP2/3 actin nucleation complex. We also show that this shape-sensing axis reprograms dendritic cell transcription by activating an IKKß-NF-κB-dependent pathway known to control their tolerogenic potential. These results indicate that cell shape changes experienced by immune cells can define their migratory behavior and immunoregulatory properties and reveal a contribution of the physical properties of tissues to adaptive immunity.


Subject(s)
Cell Movement , Dendritic Cells , Homeostasis , Lymph Nodes , Mice, Inbred C57BL , Receptors, CCR7 , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymph Nodes/immunology , Lymph Nodes/cytology , Receptors, CCR7/metabolism , Mice , Cell Movement/immunology , Cell Shape , NF-kappa B/metabolism , Mice, Knockout , Signal Transduction/immunology , I-kappa B Kinase/metabolism , Actin-Related Protein 2-3 Complex/metabolism
2.
Immunity ; 55(11): 2103-2117.e10, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36323311

ABSTRACT

The surface of the central nervous system (CNS) is protected by the meninges, which contain a dense network of meningeal macrophages (MMs). Here, we examined the role of tissue-resident MM in viral infection. MHC-II- MM were abundant neonatally, whereas MHC-II+ MM appeared over time. These barrier macrophages differentially responded to in vivo peripheral challenges such as LPS, SARS-CoV-2, and lymphocytic choriomeningitis virus (LCMV). Peripheral LCMV infection, which was asymptomatic, led to a transient infection and activation of the meninges. Mice lacking macrophages but conserving brain microglia, or mice bearing macrophage-specific deletion of Stat1 or Ifnar, exhibited extensive viral spread into the CNS. Transcranial pharmacological depletion strategies targeting MM locally resulted in several areas of the meninges becoming infected and fatal meningitis. Low numbers of MHC-II+ MM, which is seen upon LPS challenge or in neonates, corelated with higher viral load upon infection. Thus, MMs protect against viral infection and may present targets for therapeutic manipulation.


Subject(s)
COVID-19 , Lymphocytic Choriomeningitis , Animals , Mice , Lipopolysaccharides , Mice, Inbred C57BL , SARS-CoV-2 , Lymphocytic choriomeningitis virus/physiology , Macrophages , Meninges
3.
Immunity ; 42(4): 627-39, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25862089

ABSTRACT

Migratory non-lymphoid tissue dendritic cells (NLT-DCs) transport antigens to lymph nodes (LNs) and are required for protective immune responses in the context of inflammation and to promote tolerance to self-antigens in steady-state. However, the molecular mechanisms that elicit steady-state NLT-DC maturation and migration are unknown. By comparing the transcriptome of NLT-DCs in the skin with their migratory counterparts in draining LNs, we have identified a novel NF-κB-regulated gene network specific to migratory DCs. We show that targeted deletion of IKKß in DCs, a major activator of NF-κB, prevents NLT-DC accumulation in LNs and compromises regulatory T cell conversion in vivo. This was associated with impaired tolerance and autoimmunity. NF-κB is generally considered the prototypical pro-inflammatory transcription factor, but this study describes a role for NF-κB signaling in DCs for immune homeostasis and tolerance that could have implications in autoimmune diseases and immunity.


Subject(s)
Dendritic Cells/immunology , Gene Regulatory Networks/immunology , Homeostasis/immunology , Immune Tolerance , NF-kappa B/immunology , Signal Transduction/immunology , Animals , Autoantigens/genetics , Autoantigens/immunology , Autoimmunity , Cell Movement , Dendritic Cells/cytology , Gene Expression Profiling , Gene Expression Regulation , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Knockout , Microarray Analysis , NF-kappa B/genetics , Skin/cytology , Skin/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
4.
Immunity ; 41(1): 14-20, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25035950

ABSTRACT

Description of macrophage activation is currently contentious and confusing. Like the biblical Tower of Babel, macrophage activation encompasses a panoply of descriptors used in different ways. The lack of consensus on how to define macrophage activation in experiments in vitro and in vivo impedes progress in multiple ways, including the fact that many researchers still consider there to be only two types of activated macrophages, often termed M1 and M2. Here, we describe a set of standards encompassing three principles-the source of macrophages, definition of the activators, and a consensus collection of markers to describe macrophage activation-with the goal of unifying experimental standards for diverse experimental scenarios. Collectively, we propose a common framework for macrophage-activation nomenclature.


Subject(s)
Macrophage Activation/immunology , Macrophages/immunology , Terminology as Topic , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Guidelines as Topic , Humans , Macrophage Colony-Stimulating Factor/immunology , Mice , Research
5.
EMBO J ; 37(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30131424

ABSTRACT

Plasmacytoid dendritic cells (pDC) are the major source of type I interferons (IFN-I) during viral infections, in response to triggering of endosomal Toll-like receptors (TLRs) 7 or 9 by viral single-stranded RNA or unmethylated CpG DNA, respectively. Synthetic ligands have been used to disentangle the underlying signaling pathways. The adaptor protein AP3 is necessary to transport molecular complexes of TLRs, synthetic CpG DNA, and MyD88 into endosomal compartments allowing interferon regulatory factor 7 (IRF7) recruitment whose phosphorylation then initiates IFN-I production. High basal expression of IRF7 by pDC and its further enhancement by positive IFN-I feedback signaling appear to be necessary for robust cytokine production. In contrast, we show here that in vivo during mouse cytomegalovirus (MCMV) infection pDC produce high amounts of IFN-I downstream of the TLR9-to-MyD88-to-IRF7 signaling pathway without requiring IFN-I positive feedback, high IRF7 expression, or AP3-driven endosomal routing of TLRs. Hence, the current model of the molecular requirements for professional IFN-I production by pDC, established by using synthetic TLR ligands, does not strictly apply to a physiological viral infection.


Subject(s)
Dendritic Cells/immunology , Herpesviridae Infections/immunology , Interferon Type I/immunology , Muromegalovirus/immunology , Signal Transduction/immunology , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex 3/immunology , Animals , Dendritic Cells/pathology , Endosomes/genetics , Endosomes/immunology , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
6.
Immunol Cell Biol ; 100(9): 691-704, 2022 10.
Article in English | MEDLINE | ID: mdl-35849045

ABSTRACT

Clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) is an efficient tool for establishing genetic models including cellular models, and has facilitated unprecedented advancements in biomedical research. In both patients and cancer animal models, immune cells infiltrate the tumor microenvironment and some of them migrate to draining lymph nodes to exert antitumor effects. Among these immune cells, phagocytes such as macrophages and dendritic cells engulf tumor antigens prior to their crosstalk with T cells and elicit adaptive immune response against tumors. Melanoma cells are frequently used as a tumor model because of their relatively high level of somatic mutations and antigenicity. However, few genetic models have been developed using melanoma cell lines to track tumor cell phagocytosis, which is essential for understanding protective immune response in vivo. In this study, we used CRISPR/Cas9-mediated DNA cleavage and homologous recombination to develop a novel knock-in tool which expresses the ultra-bright fluorescent probe ZsGreen in YUMM1.7 melanoma cells. Using this novel tool, we measured the macrophagic engulfment of melanoma cells inside the tumor microenvironment. We also found that in tumor-grafted mice, a subset of dendritic cells efficiently engulfed YUMM1.7 cells and was preferentially trafficking tumor antigens to draining lymph nodes. In addition, we used this knock-in tool to assess the impact of a point mutation of CD11b on phagocytosis in the tumor microenvironment. Our results demonstrate that the ZsGreen-expressing YUMM1.7 melanoma model provides a valuable tool for the study of phagocytosis in vivo.


Subject(s)
CD11b Antigen , Melanoma , Phagocytosis , Animals , Antigens, Neoplasm , CD11b Antigen/genetics , Cell Line , Cell Line, Tumor , Fluorescent Dyes , Melanoma/genetics , Mice , Point Mutation , Tumor Microenvironment
7.
EMBO J ; 36(22): 3336-3355, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29038174

ABSTRACT

Accumulation of tumor-associated macrophages (TAMs) associates with malignant progression in cancer. However, the mechanisms that drive the pro-tumor functions of TAMs are not fully understood. ZEB1 is best known for driving an epithelial-to-mesenchymal transition (EMT) in cancer cells to promote tumor progression. However, a role for ZEB1 in macrophages and TAMs has not been studied. Here we describe that TAMs require ZEB1 for their tumor-promoting and chemotherapy resistance functions in a mouse model of ovarian cancer. Only TAMs that expressed full levels of Zeb1 accelerated tumor growth. Mechanistically, ZEB1 expression in TAMs induced their polarization toward an F4/80low pro-tumor phenotype, including direct activation of Ccr2 In turn, expression of ZEB1 by TAMs induced Ccl2, Cd74, and a mesenchymal/stem-like phenotype in cancer cells. In human ovarian carcinomas, TAM infiltration and CCR2 expression correlated with ZEB1 in tumor cells, where along with CCL2 and CD74 determined poorer prognosis. Importantly, ZEB1 in TAMs was a factor of poorer survival in human ovarian carcinomas. These data establish ZEB1 as a key factor in the tumor microenvironment and for maintaining TAMs' tumor-promoting functions.


Subject(s)
Carcinogenesis/pathology , Macrophages/metabolism , Macrophages/pathology , Neoplasms/metabolism , Neoplasms/pathology , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Count , Cell Differentiation/drug effects , Chemokine CCL2/pharmacology , Colony-Stimulating Factors/pharmacology , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class II/metabolism , Humans , Macrophage Activation/drug effects , Macrophages/drug effects , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Models, Biological , Monocytes/drug effects , Monocytes/pathology , Neoplasms/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phenotype , Receptors, CCR2/metabolism , Survival Analysis , Up-Regulation/drug effects
8.
Nat Immunol ; 9(9): 1019-27, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18677317

ABSTRACT

The mitogen-activated protein kinase p38 mediates cellular responses to injurious stress and immune signaling. Among the many p38 isoforms, p38 alpha is the most widely expressed in adult tissues and can be targeted by various pharmacological inhibitors. Here we investigated how p38 alpha activation is linked to cell type-specific outputs in mouse models of cutaneous inflammation. We found that both myeloid and epithelial p38 elicit inflammatory responses, yet p38 alpha signaling in each cell type served distinct inflammatory functions and varied depending on the mode of skin irritation. In addition, myeloid p38 alpha limited acute inflammation via activation of anti-inflammatory gene expression dependent on mitogen- and stress-activated kinases. Our results suggest a dual function for p38 alpha in the regulation of inflammation and show mixed potential for its inhibition as a therapeutic strategy.


Subject(s)
Inflammation Mediators/metabolism , Inflammation/immunology , MAP Kinase Signaling System/drug effects , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cells, Cultured/metabolism , Disease Models, Animal , Epithelial Cells , Gene Expression/drug effects , Mice , Myeloid Cells , Protein Kinase Inhibitors/pharmacology , Skin Diseases/genetics , Skin Diseases/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
J Lipid Res ; 60(12): 2006-2019, 2019 12.
Article in English | MEDLINE | ID: mdl-31570505

ABSTRACT

During foam cell formation and atherosclerosis development, the scavenger receptor CD36 plays critical roles in lipid uptake and triggering of atherogenicity via the activation of Vav molecules. The Vav family includes three highly conserved members known as Vav1, Vav2, and Vav3. As Vav1 and Vav3 were found to exert function in atherosclerosis development, it remains thus to decipher whether Vav2 also plays a role in the development of atherosclerosis. In this study we found that Vav2 deficiency in RAW264.7 macrophages significantly diminished oxidized LDL uptake and CD36 signaling, demonstrating that each Vav protein family member was required for foam cell formation. Genetic disruption of Vav2 in ApoE-deficient C57BL/6 mice significantly inhibited the severity of atherosclerosis. Strikingly, we further found that the genetic deletion of each member of the Vav protein family by CRISPR/Cas9 resulted in a similar alteration of transcriptomic profiles of macrophages. The three members of the Vav proteins were found to form complexes, and genetic ablation of each single Vav molecule was sufficient to prevent endocytosis of CD36. The functional interdependence of the three Vav family members in foam cell formation was due to their indispensable roles in transcriptomic programing, lipid uptake, and activation of the JNK kinase in macrophages.


Subject(s)
Atherosclerosis/metabolism , Foam Cells/cytology , Protein Multimerization , Proto-Oncogene Proteins c-vav/chemistry , Proto-Oncogene Proteins c-vav/metabolism , Animals , Apolipoproteins E/deficiency , Atherosclerosis/genetics , Base Sequence , CD36 Antigens/metabolism , Cell Differentiation , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Phenotype , Protein Structure, Quaternary , Protein Transport , Proto-Oncogene Proteins c-vav/deficiency , Proto-Oncogene Proteins c-vav/genetics , RAW 264.7 Cells
10.
EMBO J ; 33(10): 1104-16, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24737868

ABSTRACT

Dendritic cells (DC) are key regulators of both protective immune responses and tolerance to self-antigens. Soon after their discovery in lymphoid tissues by Steinman and Cohn, as cells with the unique ability to prime naïve antigen-specific T cells, it was realized that DC can exist in at least two distinctive states characterized by morphological, phenotypic and functional changes-this led to the description of DC maturation. It is now well appreciated that there are several subsets of DC in both lymphoid and non-lymphoid tissues of mammals, and these cells show remarkable functional specialization and specificity in their roles in tolerance and immunity. This review will focus on the specific characteristics of DC subsets and how their functional specialization may be regulated by distinctive gene expression programs and signaling responses in both steady-state and in the context of inflammation. In particular, we will highlight the common and distinctive genes and signaling pathways that are associated with the functional maturation of DC subsets.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Animals , Humans , Immune Tolerance/immunology , Immune Tolerance/physiology , Signal Transduction/immunology , Signal Transduction/physiology
11.
EMBO J ; 32(6): 816-28, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23422957

ABSTRACT

Dendritic cells (DC) are required for priming antigen-specific T cells and acquired immunity to many important human pathogens, including Mycobacteriuim tuberculosis (TB) and influenza. However, inappropriate priming of auto-reactive T cells is linked with autoimmune disease. Understanding the molecular mechanisms that regulate the priming and activation of naïve T cells is critical for development of new improved vaccines and understanding the pathogenesis of autoimmune diseases. The serine/threonine kinase IKKα (CHUK) has previously been shown to have anti-inflammatory activity and inhibit innate immunity. Here, we show that IKKα is required in DC for priming antigen-specific T cells and acquired immunity to the human pathogen Listeria monocytogenes. We describe a new role for IKKα in regulation of IRF3 activity and the functional maturation of DC. This presents a unique role for IKKα in dampening inflammation while simultaneously promoting adaptive immunity that could have important implications for the development of new vaccine adjuvants and treatment of autoimmune diseases.


Subject(s)
Adaptive Immunity/genetics , Cell Differentiation/genetics , Dendritic Cells/physiology , I-kappa B Kinase/physiology , Infections/immunology , Adoptive Transfer/methods , Animals , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Infections/genetics , Inflammation/genetics , Inflammation/immunology , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
Arterioscler Thromb Vasc Biol ; 35(6): 1374-81, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25908768

ABSTRACT

OBJECTIVE: To investigate the roles and signaling pathways of CD40L and CD40 in platelet-platelet interactions and thrombus formation under conditions relevant for atherothrombosis. APPROACH AND RESULTS: Platelets from mice prone to atherosclerosis lacking CD40L (Cd40lg(-/-)Apoe(-/-)) showed diminished αIIbß3 activation and α-granule secretion in response to glycoprotein VI stimulation, whereas these responses of CD40-deficient platelets (Cd40(-/-)Apoe(-/-)) were not decreased. Using blood from Cd40lg(-/-)Apoe(-/-) and Cd40(-/-)Apoe(-/-) mice, the glycoprotein VI-dependent formation of dense thrombi was impaired on atherosclerotic plaque material or on collagen, in comparison with Apoe(-/-) blood. In all genotypes, addition of CD40L to the blood enhanced the growth of dense thrombi on plaques and collagen. Similarly, CD40L enhanced glycoprotein VI-induced platelet aggregation, even with platelets deficient in CD40. This potentiation was antagonized in Pik3cb(R/R) platelets or by inhibiting phosphatidylinositol 3-kinase ß (PI3Kß). Addition of CD40L also enhanced collagen-induced Akt phosphorylation, which was again antagonized by absence or inhibition of PI3Kß. Finally, platelets from Chuk1(A/A)Apoe(-/-) mice deficient in IκB kinase α (IKKα), implicated in CD40 signaling to nuclear factor (NF) κB, showed unchanged responses to CD40L in aggregation or thrombus formation. CONCLUSIONS: Under atherogenic conditions, CD40L enhances collagen-induced platelet-platelet interactions by supporting integrin αIIbß3 activation, secretion and thrombus growth via PI3Kß, but not via CD40 and IKKα/NFκB. This role of CD40L exceeds the no more than modest role of CD40 in thrombus formation.


Subject(s)
Atherosclerosis/metabolism , Blood Platelets/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , I-kappa B Kinase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Thrombosis/metabolism , Animals , Atherosclerosis/pathology , Collagen/metabolism , Mice , Platelet Activation , Signal Transduction , Thrombosis/pathology
13.
Cancer Cell ; 12(4): 300-1, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17936555

ABSTRACT

Recent literature has highlighted an important role of inflammation in promoting cancer. However, the immune system can also play a central role in protecting the body against cancer as well as infection, although its role in cancer is not well understood. A study published in the September issue of Nature Medicine adds a new twist to the role of inflammation in cancer. Apetoh et al. describe how activation of innate immunity after conventional radiation or chemotherapy can trigger protective antitumor immunity.


Subject(s)
Antineoplastic Agents/pharmacology , Immunity, Cellular , Immunity, Innate , Inflammation/immunology , Neoplasms/drug therapy , Neoplasms/radiotherapy , Toll-Like Receptor 4/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Death , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/radiation effects , HMGB1 Protein/metabolism , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/radiation effects , Immunity, Innate/drug effects , Immunity, Innate/radiation effects , Inflammation/pathology , Mice , Myeloid Differentiation Factor 88/metabolism , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/radiation effects
14.
Biochem J ; 461(3): 531-7, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24911653

ABSTRACT

IKKß {IκB [inhibitor of NF-κB (nuclear factor κB)] kinase ß} is required to activate the transcription factor NF-κB, but how IKKß itself is activated in vivo is still unclear. It was found to require phosphorylation by one or more 'upstream' protein kinases in some reports, but by autophosphorylation in others. In the present study, we resolve this contro-versy by demonstrating that the activation of IKKß induced by IL-1 (interleukin-1) or TNF (tumour necrosis factor) in embryonic fibroblasts, or by ligands that activate Toll-like receptors in macrophages, requires two distinct phosphorylation events: first, the TAK1 [TGFß (transforming growth factor ß)-activated kinase-1]-catalysed phosphorylation of Ser¹77 and, secondly, the IKKß-catalysed autophosphorylation of Ser¹8¹. The phosphorylation of Ser¹77 by TAK1 is a priming event required for the subsequent autophosphorylation of Ser¹8¹, which enables IKKß to phosphorylate exogenous substrates. We also provide genetic evidence which indicates that the IL-1-stimulated, LUBAC (linear ubiquitin chain assembly complex)-catalysed formation of linear ubiquitin chains and their interaction with the NEMO (NF-κB essential modulator) component of the canonical IKK complex permits the TAK1-catalysed priming phosphorylation of IKKß at Ser¹77 and IKKα at Ser¹76. These findings may be of general significance for the activation of other protein kinases.


Subject(s)
I-kappa B Kinase/metabolism , MAP Kinase Kinase Kinases/metabolism , Protein Processing, Post-Translational , Amino Acid Substitution , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Enzyme Activation/drug effects , Gene Knock-In Techniques , HEK293 Cells , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/chemistry , I-kappa B Kinase/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice , Mice, Transgenic , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphorylation/drug effects , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine/metabolism , Ubiquitination
15.
Nat Rev Immunol ; 2(10): 787-95, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12360216

ABSTRACT

The pro-inflammatory signalling pathways and cellular mechanisms that initiate the inflammatory response have become increasingly well characterized. However, little is known about the mediators and mechanisms that switch off inflammation. Recent data indicate that the resolution of inflammation is an active process controlled by endogenous mediators that suppress pro-inflammatory gene expression and cell trafficking, as well as induce inflammatory-cell apoptosis and phagocytosis, which are crucial determinants of successful resolution. This review focuses on this emerging area of inflammation research and describes the mediators and mechanisms that are currently stealing the headlines.


Subject(s)
Inflammation/etiology , Lipids/immunology , Animals , Anti-Inflammatory Agents/immunology , Apoptosis , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/physiopathology , Inflammation Mediators/immunology , Models, Immunological , NF-kappa B/immunology , Phagocytosis , Signal Transduction
16.
PLoS Pathog ; 8(7): e1002812, 2012.
Article in English | MEDLINE | ID: mdl-22829768

ABSTRACT

Group B Streptococcus (GBS) is a leading cause of invasive bacterial infections in human newborns and immune-compromised adults. The pore-forming toxin (PFT) ß hemolysin/cytolysin (ßh/c) is a major virulence factor for GBS, which is generally attributed to its cytolytic functions. Here we show ßh/c has immunomodulatory properties on macrophages at sub-lytic concentrations. ßh/c-mediated activation of p38 MAPK drives expression of the anti-inflammatory and immunosuppressive cytokine IL-10, and inhibits both IL-12 and NOS2 expression in GBS-infected macrophages, which are critical factors in host defense. Isogenic mutant bacteria lacking ßh/c fail to activate p38-mediated IL-10 production in macrophages and promote increased IL-12 and NOS2 expression. Furthermore, targeted deletion of p38 in macrophages increases resistance to invasive GBS infection in mice, associated with impaired IL-10 induction and increased IL-12 production in vivo. These data suggest p38 MAPK activation by ßh/c contributes to evasion of host defense through induction of IL-10 expression and inhibition of macrophage activation, a new mechanism of action for a PFT and a novel anti-inflammatory role for p38 in the pathogenesis of invasive bacterial infection. Our studies suggest p38 MAPK may represent a new therapeutic target to blunt virulence and improve clinical outcome of invasive GBS infection.


Subject(s)
Bacterial Proteins/metabolism , Hemolysin Proteins/metabolism , Interleukin-10/biosynthesis , Macrophages/immunology , Macrophages/microbiology , Streptococcal Infections/immunology , Streptococcus agalactiae/pathogenicity , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Immunity, Innate , Interleukin-12/biosynthesis , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophage Activation , Macrophages/metabolism , Mice , Nitric Oxide Synthase/biosynthesis , Streptococcal Infections/microbiology , Streptococcus agalactiae/immunology , Streptococcus agalactiae/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
17.
Drug Alcohol Depend ; 246: 109846, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37004463

ABSTRACT

BACKGROUND: Deficits in executive function are common in methamphetamine use disorder (MUD), likely contributing to difficulties in sustained treatment success. Cognitive remediation interventions are designed to treat such deficits but have not been adapted to the needs of people with MUD. This study presents a proof-of-concept trial to evaluate a new cognitive remediation program for MUD, Goal Management Training+ (GMT+). METHODS: This was a cluster-randomised crossover trial comparing GMT+ with a psychoeducation-based control (Brain Health Workshop; BHW). GMT+ is a therapist-administered group-based cognitive remediation for executive dysfunction comprising four 90-minute weekly sessions and daily journal activities. BHW is a lifestyle psychoeducation program matched to GMT+ for therapist involvement, format, and duration. Participants (n = 36; GMT n = 17; BHW n = 19) were recruited from therapeutic communities in Victoria, Australia. Primary outcomes included intervention acceptability, feasibility, and improvements in self-reported executive function. Secondary outcomes included cognitive tests of executive function, severity of methamphetamine dependence, craving, and quality of life. We performed mixed linear modelling and calculated Hedges' g effect sizes. RESULTS: GMT+ participant ratings and program retention indicated high acceptability. There was no difference between GMT+ and BHW on self-reported executive function (g = 0.06). Cognitive tasks suggested benefits of GMT+ on information gathering (g = 0.88) and delay-discounting (g = 0.80). Severity of methamphetamine dependence decreased more in GMT+ (g = 1.47). CONCLUSIONS: GMT+ was well-accepted but did not improve self-reported executive functioning. Secondary outcomes suggested GMT+ was beneficial for objective cognitive performance and severity of dependence.


Subject(s)
Brain Injuries , Executive Function , Humans , Goals , Quality of Life , Brain Injuries/complications , Brain Injuries/psychology , Treatment Outcome , Victoria
18.
Dev Cell ; 58(17): 1548-1561.e10, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37442140

ABSTRACT

Tumor-associated macrophages (TAMs) are a heterogeneous population of cells that facilitate cancer progression. However, our knowledge of the niches of individual TAM subsets and their development and function remain incomplete. Here, we describe a population of lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1)-expressing TAMs, which form coordinated multi-cellular "nest" structures that are heterogeneously distributed proximal to vasculature in tumors of a spontaneous murine model of breast cancer. We demonstrate that LYVE-1+ TAMs develop in response to IL-6, which induces their expression of the immune-suppressive enzyme heme oxygenase-1 and promotes a CCR5-dependent signaling axis, which guides their nest formation. Blocking the development of LYVE-1+ TAMs or their nest structures, using gene-targeted mice, results in an increase in CD8+ T cell recruitment to the tumor and enhanced response to chemotherapy. This study highlights an unappreciated collaboration of a TAM subset to form a coordinated niche linked to immune exclusion and resistance to anti-cancer therapy.


Subject(s)
Neoplasms , Mice , Animals , Neoplasms/pathology , Macrophages/metabolism
19.
J Exp Med ; 203(8): 1883-9, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16880258

ABSTRACT

Glucocorticoids (GCs), which are used in the treatment of immune-mediated inflammatory diseases, inhibit the expression of many inflammatory mediators. They can also induce the expression of dual specificity phosphatase 1 (DUSP1; otherwise known as mitogen-activated protein kinase [MAPK] phosphatase 1), which dephosphorylates and inactivates MAPKs. We investigated the role of DUSP1 in the antiinflammatory action of the GC dexamethasone (Dex). Dex-mediated inhibition of c-Jun N-terminal kinase and p38 MAPK was abrogated in DUSP1-/- mouse macrophages. Dex-mediated suppression of several proinflammatory genes (including tumor necrosis factor, cyclooxygenase 2, and interleukin 1alpha and 1beta) was impaired in DUSP1-/- mouse macrophages, whereas other proinflammatory genes were inhibited by Dex in a DUSP1-independent manner. In vivo antiinflammatory effects of Dex on zymosan-induced inflammation were impaired in DUSP1-/- mice. Therefore, the expression of DUSP1 is required for the inhibition of proinflammatory signaling pathways by Dex in mouse macrophages. Furthermore, DUSP1 contributes to the antiinflammatory effects of Dex in vitro and in vivo.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Cycle Proteins/biosynthesis , Dexamethasone/pharmacology , Immediate-Early Proteins/biosynthesis , Phosphoprotein Phosphatases/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , Animals , Bone Marrow/drug effects , Dose-Response Relationship, Drug , Dual Specificity Phosphatase 1 , Enzyme Induction/drug effects , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Immediate-Early Proteins/deficiency , Inflammation , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/drug effects , Mice , Phosphoprotein Phosphatases/deficiency , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/deficiency , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Curr Top Microbiol Immunol ; 349: 171-84, 2011.
Article in English | MEDLINE | ID: mdl-21080151

ABSTRACT

Macrophages are tissue resident phagocytes with important roles in development, wound healing, and inflammation. There is enormous heterogeneity in macrophage phenotype, from 'classically' activated macrophages that have important roles in inflammation and innate immunity, to 'alternative' macrophage activation that is associated with wound healing, angiogenesis, and immune-suppression. Most, if not all, solid tumors have a significant macrophage population, clinical and experimental evidence suggests tumor-associated macrophages (TAM) are linked with tumor progression. The trophic functions of TAM are associated with increased angiogenesis, malignant cell invasion, and metastasis. NF-κB is s central regulator of inflammation and NF-κB activation particularly in TAM is linked with promotion of carcinogenesis in various experimental models of inflammation-associated cancer. NF-κB activation in TAM has, therefore, been suggested to represent a molecular link between inflammation and cancer. However, TAM frequently display an anti-inflammatory phenotype linked with immune-suppression that is not easily reconciled with a pro-inflammatory function for NF-κB in TAM. Here, I review the form and function of TAM and discuss the role of NF-κB activation in TAM in carcinogenesis.


Subject(s)
Macrophages/physiology , NF-kappa B/physiology , Neoplasms/etiology , Animals , Humans , Neoplasms/immunology , Neovascularization, Pathologic/etiology , Tumor Escape
SELECTION OF CITATIONS
SEARCH DETAIL