Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 14(5): 1160-1171, 2021 09.
Article in English | MEDLINE | ID: mdl-34045680

ABSTRACT

Regulation of epithelial cell death has emerged as a key mechanism controlling immune homeostasis in barrier surfaces. Necroptosis is a type of regulated necrotic cell death induced by receptor interacting protein kinase 3 (RIPK3) that has been shown to cause inflammatory pathologies in different tissues. The role of regulated cell death and particularly necroptosis in lung homeostasis and disease remains poorly understood. Here we show that mice with Airway Epithelial Cell (AEC)-specific deficiency of Fas-associated with death domain (FADD), an adapter essential for caspase-8 activation, developed exacerbated allergic airway inflammation in a mouse model of asthma induced by sensitization and challenge with house dust mite (HDM) extracts. Genetic inhibition of RIPK1 kinase activity by crossing to mice expressing kinase inactive RIPK1 as well as RIPK3 or MLKL deficiency prevented the development of exaggerated HDM-induced asthma pathology in FADDAEC-KO mice, suggesting that necroptosis of FADD-deficient AECs augmented the allergic immune response. These results reveal a role of AEC necroptosis in amplifying airway allergic inflammation and suggest that necroptosis could contribute to asthma exacerbations caused by respiratory virus infections inducing AEC death.


Subject(s)
Allergens/immunology , Asthma/etiology , Asthma/metabolism , Necroptosis/immunology , Pyroglyphidae/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Airway Remodeling , Animals , Asthma/pathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers , Disease Models, Animal , Disease Progression , Disease Susceptibility , Enzyme Activation , Fas-Associated Death Domain Protein/deficiency , Immunoglobulin E/immunology , Immunohistochemistry , Mice , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Respiratory Mucosa/pathology
2.
Nat Commun ; 12(1): 2048, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33824345

ABSTRACT

Loss of TP53 and RB1 in treatment-naïve small cell lung cancer (SCLC) suggests selective pressure to inactivate cell death pathways prior to therapy. Yet, which of these pathways remain available in treatment-naïve SCLC is unknown. Here, through systemic analysis of cell death pathway availability in treatment-naïve SCLC, we identify non-neuroendocrine (NE) SCLC to be vulnerable to ferroptosis through subtype-specific lipidome remodeling. While NE SCLC is ferroptosis resistant, it acquires selective addiction to the TRX anti-oxidant pathway. In experimental settings of non-NE/NE intratumoral heterogeneity, non-NE or NE populations are selectively depleted by ferroptosis or TRX pathway inhibition, respectively. Preventing subtype plasticity observed under single pathway targeting, combined treatment kills established non-NE and NE tumors in xenografts, genetically engineered mouse models of SCLC and patient-derived cells, and identifies a patient subset with drastically improved overall survival. These findings reveal cell death pathway mining as a means to identify rational combination therapies for SCLC.


Subject(s)
Ferroptosis , Neuroendocrine Tumors/pathology , Small Cell Lung Carcinoma/pathology , Animals , Antioxidants/metabolism , Apoptosis , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Survival , Humans , Lipid Metabolism , Male , Mice, Nude , Models, Biological , Necroptosis , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipids/metabolism , Prognosis , Thioredoxins/metabolism
3.
PLoS One ; 13(3): e0194048, 2018.
Article in English | MEDLINE | ID: mdl-29522531

ABSTRACT

Type 1 diabetes (T1D) is caused by the autoimmune destruction of the insulin-producing pancreatic beta cells. While the role of adaptive immunity has been extensively studied, the role of innate immune responses and particularly of Toll- like Receptor (TLR) signaling in T1D remains poorly understood. Here we show that myeloid cell-specific MyD88 deficiency considerably protected mice from the development of streptozotocin (STZ)-induced diabetes. The protective effect of MyD88 deficiency correlated with increased expression of the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO) in pancreatic lymph nodes from STZ-treated mice and in bone marrow-derived dendritic cells (BMDC) stimulated with apoptotic cells. Mice with myeloid cell specific TIR-domain-containing adapter-inducing interferon-ß (TRIF) knockout showed a trend towards accelerated onset of STZ-induced diabetes, while TRIF deficiency resulted in reduced IDO expression in vivo and in vitro. Moreover, myeloid cell specific MyD88 deficiency delayed the onset of diabetes in Non-Obese Diabetic (NOD) mice, whereas TRIF deficiency had no effect. Taken together, these results identify MyD88 signaling in myeloid cells as a critical pathogenic factor in autoimmune diabetes, which is antagonized by TRIF-dependent responses. This differential function of MyD88 and TRIF depends at least in part on their opposite effects in regulating IDO expression in phagocytes exposed to apoptotic cells.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 1/etiology , Myeloid Cells/immunology , Myeloid Differentiation Factor 88/physiology , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Animals , Apoptosis , Dendritic Cells/physiology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Enzyme Induction , Female , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Macrophages, Peritoneal/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Phagocytosis , Specific Pathogen-Free Organisms , Streptozocin , T-Lymphocyte Subsets/pathology
4.
Cell Rep ; 8(5): 1308-17, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25159141

ABSTRACT

Interferon Regulatory Factor 5 (IRF5) plays a major role in setting up an inflammatory macrophage phenotype, but the molecular basis of its transcriptional activity is not fully understood. In this study, we conduct a comprehensive genome-wide analysis of IRF5 recruitment in macrophages stimulated with bacterial lipopolysaccharide and discover that IRF5 binds to regulatory elements of highly transcribed genes. Analysis of protein:DNA microarrays demonstrates that IRF5 recognizes the canonical IRF-binding (interferon-stimulated response element [ISRE]) motif in vitro. However, IRF5 binding in vivo appears to rely on its interactions with other proteins. IRF5 binds to a noncanonical composite PU.1:ISRE motif, and its recruitment is aided by RelA. Global gene expression analysis in macrophages deficient in IRF5 and RelA highlights the direct role of the RelA:IRF5 cistrome in regulation of a subset of key inflammatory genes. We map the RelA:IRF5 interaction domain and suggest that interfering with it would offer selective targeting of macrophage inflammatory activities.


Subject(s)
Interferon Regulatory Factors/metabolism , Macrophages/metabolism , Transcription Factor RelA/metabolism , Animals , Cells, Cultured , Genome , Interferon Regulatory Factors/genetics , Macrophage Activation/genetics , Macrophages/immunology , Mice , Mice, Inbred C57BL , Protein Binding , Response Elements , Transcription Factor RelA/genetics , Transcriptional Activation
5.
PLoS One ; 9(5): e97060, 2014.
Article in English | MEDLINE | ID: mdl-24835211

ABSTRACT

Chronic activation of innate immunity takes place in obesity and initiated by the hypertrophic adipocytes which obtain a pro-inflammatory phenotype. The corticotrophin-releasing factor (CRF) family of neuropeptides and their receptors (CRF1 and CRF2) affect stress response and innate immunity. Adipose tissue expresses a complete CRF system. The aim of this study was to examine the role of CRF neuropeptides in the immune phenotype of adipocytes assessed by their expression of the toll-like receptor-4 (TLR4), the production of inflammatory cytokines IL-6, TNF-α and IL-1ß, chemokines IL-8, monocyte attractant protein-1 (MCP-1) and of the adipokines adiponectin, resistin and leptin. Our data are as follows: (a) CRF, UCN2 and UCN3 are expressed in human white adipocytes as well as CRFR1a, CRFR2a and CRFR2b but not CRFR2c. 3T3L1 pre-adipocytes and differentiated adipocytes expressed both CRF1 and CRF2 receptors and UCN3, while UCN2 was detected only in differentiated adipocytes. CRF2 was up-regulated in mouse mature adipocytes. (b) CRF1 agonists suppressed media- and LPS-induced pre-adipocyte differentiation while CRF2 receptor agonists had no effect. (c) In mouse pre-adipocytes, CRF2 agonists suppressed TLR4 expression and the production of IL-6, CXCL1 and adiponectin while CRF1 agonists had no effect. (d) In mature mouse adipocytes LPS induced IL-6 and CXCL1 production and suppressed leptin. (e) In human visceral adipocytes LPS induced IL-6, TNF-α, IL-8, MCP-1 and leptin production and suppressed adiponectin and resistin. (f) In mouse mature adipocytes CRF1 and CRF2 agonists suppressed basal and LPS-induced production of inflammatory cytokines, TLR4 expression and adiponectin production, while in human visceral adipocytes CRF and UCN1 suppressed basal and LPS-induced IL-6, TNF-α, IL-8 and MCP-1 production. In conclusion, the effects of the activation of CRF1 and CRF2 may be significant in ameliorating the pro-inflammatory activity of adipocytes in obesity.


Subject(s)
3T3-L1 Cells/metabolism , Adipocytes, White/metabolism , Cell Differentiation/physiology , Corticotropin-Releasing Hormone/metabolism , Immunity, Innate/immunology , Inflammation/metabolism , Urocortins/metabolism , 3T3-L1 Cells/physiology , Adipokines/metabolism , Analysis of Variance , Animals , Cytokines/metabolism , DNA Primers/genetics , Flow Cytometry , Humans , Inflammation/immunology , Mice , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
6.
Proc Natl Acad Sci U S A ; 109(24): 9517-22, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22647600

ABSTRACT

Activated macrophages are described as classically activated or M1 type and alternatively activated or M2 type, depending on their response to proinflammatory stimuli and the expression of genetic markers including iNOS, arginase1, Ym1, and Fizz1. Here we report that Akt kinases differentially contribute to macrophage polarization, with Akt1 ablation giving rise to an M1 and Akt2 ablation resulting in an M2 phenotype. Accordingly, Akt2(-/-) mice were more resistant to LPS-induced endotoxin shock and to dextran sulfate sodium (DSS)-induced colitis than wild-type mice, whereas Akt1(-/-) mice were more sensitive. Cell depletion and reconstitution experiments in a DSS-induced colitis model confirmed that the effect was macrophage-dependent. Gene-silencing studies showed that the M2 phenotype of Akt2(-/-) macrophages was cell autonomous. The microRNA miR-155, whose expression was repressed in naive and in LPS-stimulated Akt2(-/-) macrophages, and its target C/EBPß appear to play a key role in this process. C/EBPß, a hallmark of M2 macrophages that regulates Arg1, was up-regulated upon Akt2 ablation or silencing. Overexpression or silencing of miR-155 confirmed its central role in Akt isoform-dependent M1/M2 polarization of macrophages.


Subject(s)
Cell Polarity , Macrophages/immunology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Macrophages/enzymology , Mice , Proto-Oncogene Proteins c-akt/genetics
7.
PLoS One ; 6(11): e27435, 2011.
Article in English | MEDLINE | ID: mdl-22114673

ABSTRACT

Listeria monocytogenes is a gram-positive facultative intracellular pathogen, causing serious illness in immunocompromised individuals and pregnant women. Upon detection by macrophages, which are key players of the innate immune response against infection, L. monocytogenes induces specific host cell responses which need to be tightly controlled at transcriptional and post-transcriptional levels. Here, we ask whether and how host miRNAs, which represent an important mechanism of post-transcriptional regulation in a wide array of biological processes, are altered by a model pathogen upon live infection of murine bone marrow derived macrophages. We first report that L. monocytogenes subverts the host genome-wide miRNA profile of macrophages in vitro. Specifically, we show that miR-155, miR-146a, miR-125a-3p/5p and miR-149 were amongst the most significantly regulated miRNAs in infected macrophages. Strikingly, these miRNAs were highly upregulated upon infection with the Listeriolysin-deficient L. monocytogenes mutant Δhly, that cannot escape from the phagosome thus representing a vacuolar-contained infection. The vacuolar miRNA response was significantly reduced in macrophages deficient for MyD88. In addition, miR-146a and miR-125a-3p/5p were regulated at transcriptional levels upon infection, and miR-125a-3p/5p were found to be TLR2 responsive. Furthermore, miR-155 transactivation in infection was regulated by NF-κB p65, while miR-146a and miR-125a-3p/5p expression was unaffected in p65-deficient primary macrophages upon L. monocytogenes infection. Our results demonstrate that L. monocytogenes promotes significant changes in the miRNA expression profile in macrophages, and reveal a vacuolar-dependent miRNA signature, listeriolysin-independent and MyD88-dependent. These miRNAs are predicted to target immune genes and are therefore most likely involved in regulation of the macrophage innate immune response against infection at post-transcriptional levels.


Subject(s)
Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Macrophages/microbiology , Macrophages/pathology , MicroRNAs/physiology , Vacuoles/immunology , Animals , Bacterial Toxins/pharmacology , Cells, Cultured , DNA, Bacterial/genetics , Gene Expression Profiling , Gene Expression Regulation , Heat-Shock Proteins/pharmacology , Hemolysin Proteins/pharmacology , Immunity, Innate , Listeria monocytogenes/genetics , Listeriosis/genetics , Listeriosis/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Toll-Like Receptor 2 , Transcriptional Activation , Vacuoles/genetics , Vacuoles/microbiology
8.
Nature ; 471(7340): 637-41, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21455181

ABSTRACT

SHARPIN is a ubiquitin-binding and ubiquitin-like-domain-containing protein which, when mutated in mice, results in immune system disorders and multi-organ inflammation. Here we report that SHARPIN functions as a novel component of the linear ubiquitin chain assembly complex (LUBAC) and that the absence of SHARPIN causes dysregulation of NF-κB and apoptotic signalling pathways, explaining the severe phenotypes displayed by chronic proliferative dermatitis (cpdm) in SHARPIN-deficient mice. Upon binding to the LUBAC subunit HOIP (also known as RNF31), SHARPIN stimulates the formation of linear ubiquitin chains in vitro and in vivo. Coexpression of SHARPIN and HOIP promotes linear ubiquitination of NEMO (also known as IKBKG), an adaptor of the IκB kinases (IKKs) and subsequent activation of NF-κB signalling, whereas SHARPIN deficiency in mice causes an impaired activation of the IKK complex and NF-κB in B cells, macrophages and mouse embryonic fibroblasts (MEFs). This effect is further enhanced upon concurrent downregulation of HOIL-1L (also known as RBCK1), another HOIP-binding component of LUBAC. In addition, SHARPIN deficiency leads to rapid cell death upon tumour-necrosis factor α (TNF-α) stimulation via FADD- and caspase-8-dependent pathways. SHARPIN thus activates NF-κB and inhibits apoptosis via distinct pathways in vivo.


Subject(s)
Apoptosis , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin/metabolism , Animals , Apoptosis/drug effects , B-Lymphocytes/metabolism , Carrier Proteins/metabolism , Caspase 8/metabolism , Cells, Cultured , Dermatitis/genetics , Dermatitis/metabolism , Dermatitis/pathology , Fas-Associated Death Domain Protein/metabolism , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Mice , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
J Angiogenes Res ; 2: 22, 2010 Oct 25.
Article in English | MEDLINE | ID: mdl-20974005

ABSTRACT

BACKGROUND: Quantitative determination of the development of new blood vessels is crucial for our understanding of the progression of several diseases, including cancer. However, in most cases a high throughput technique that is simple, accurate, user-independent and cost-effective for small animal imaging is not available. METHODS: In this work we present a simple approach based on spectral imaging to increase the contrast between vessels and surrounding tissue, enabling accurate determination of the blood vessel area. This approach is put to test with a 4T1 breast cancer murine in vivo model and validated with histological and microvessel density analysis. RESULTS: We found that one can accurately measure the vascularization area by using excitation/emission filter pairs which enhance the surrounding tissue's autofluorescence, significantly increasing the contrast between surrounding tissue and blood vessels. Additionally, we found excellent correlation between this technique and histological and microvessel density analysis. CONCLUSIONS: Making use of spectral imaging techniques we have shown that it is possible to accurately determine blood vessel volume intra-vitally. We believe that due to the low cost, accuracy, user-independence and simplicity of this technique, it will be of great value in those cases where in vivo quantitative information is necessary.

10.
Mol Cancer ; 9: 261, 2010 Sep 27.
Article in English | MEDLINE | ID: mdl-20875132

ABSTRACT

INTRODUCTION: Stress has been shown to be a tumor promoting factor. Both clinical and laboratory studies have shown that chronic stress is associated with tumor growth in several types of cancer. Corticotropin Releasing Factor (CRF) is the major hypothalamic mediator of stress, but is also expressed in peripheral tissues. Earlier studies have shown that peripheral CRF affects breast cancer cell proliferation and motility. The aim of the present study was to assess the significance of peripheral CRF on tumor growth as a mediator of the response to stress in vivo. METHODS: For this purpose we used the 4T1 breast cancer cell line in cell culture and in vivo. Cells were treated with CRF in culture and gene specific arrays were performed to identify genes directly affected by CRF and involved in breast cancer cell growth. To assess the impact of peripheral CRF as a stress mediator in tumor growth, Balb/c mice were orthotopically injected with 4T1 cells in the mammary fat pad to induce breast tumors. Mice were subjected to repetitive immobilization stress as a model of chronic stress. To inhibit the action of CRF, the CRF antagonist antalarmin was injected intraperitoneally. Breast tissue samples were histologically analyzed and assessed for neoangiogenesis. RESULTS: Array analysis revealed among other genes that CRF induced the expression of SMAD2 and ß-catenin, genes involved in breast cancer cell proliferation and cytoskeletal changes associated with metastasis. Cell transfection and luciferase assays confirmed the role of CRF in WNT- ß-catenin signaling. CRF induced 4T1 cell proliferation and augmented the TGF-ß action on proliferation confirming its impact on TGFß/SMAD2 signaling. In addition, CRF promoted actin reorganization and cell migration, suggesting a direct tumor-promoting action. Chronic stress augmented tumor growth in 4T1 breast tumor bearing mice and peripheral administration of the CRF antagonist antalarmin suppressed this effect. Moreover, antalarmin suppressed neoangiogenesis in 4T1 tumors in vivo. CONCLUSION: This is the first report demonstrating that peripheral CRF, at least in part, mediates the tumor-promoting effects of stress and implicates CRF in SMAD2 and ß-catenin expression.


Subject(s)
Breast Neoplasms/metabolism , Corticotropin-Releasing Hormone/metabolism , Stress, Physiological/physiology , Animals , Blotting, Western , Breast Neoplasms/etiology , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Mice , Mice, Inbred BALB C , Pyrimidines/pharmacology , Pyrroles/pharmacology , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological/genetics
11.
Immunity ; 31(2): 220-31, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19699171

ABSTRACT

MicroRNAs regulated by lipopolysaccharide (LPS) target genes that contribute to the inflammatory phenotype. Here, we showed that the protein kinase Akt1, which is activated by LPS, positively regulated miRNAs let-7e and miR-181c but negatively regulated miR-155 and miR-125b. In silico analyses and transfection studies revealed that let-7e repressed Toll-like receptor 4 (TLR4), whereas miR-155 repressed SOCS1, two proteins critical for LPS-driven TLR signaling, which regulate endotoxin sensitivity and tolerance. As a result, Akt1(-/-) macrophages exhibited increased responsiveness to LPS in culture and Akt1(-/-) mice did not develop endotoxin tolerance in vivo. Overexpression of let-7e and suppression of miR-155 in Akt1(-/-) macrophages restored sensitivity and tolerance to LPS in culture and in animals. These results indicate that Akt1 regulates the response of macrophages to LPS by controlling miRNA expression.


Subject(s)
Lipopolysaccharides/immunology , Macrophages/immunology , MicroRNAs/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Immune Tolerance/immunology , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/genetics , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/immunology , Toll-Like Receptor 4/immunology
12.
Mol Cancer ; 8: 30, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19490624

ABSTRACT

INTRODUCTION: Cancer cells secrete bioactive peptides that act in an autocrine or paracrine fashion affecting tumor growth and metastasis. Corticotropin-releasing factor (CRF), a hypothalamic neuropeptide that controls the response to stress, has been detected in breast cancer tissues and cell lines. CRF can affect breast cancer cells in an autocrine or paracrine manner via its production from innervating sympathetic neurons or immune cells. METHODS: In the present study we report our findings regarding the impact of CRF on breast cancer cell motility and invasiveness. For this purpose we used the MCF7 breast cancer cell line and evaluated the effect of CRF on motility and invasiveness using the wound-healing and boyden-chamber assays. In addition, we measured the effect of CRF on molecules that mediate motility by western blot, immunofluorescence, ELISA and RT-PCR. RESULTS: Our findings show that: 1. CRF transiently inhibited the apoptosis of MCF7 cells. 2. CRF enhanced MCF7 cell motility in a wound healing assay and their invasiveness through extracellular matrix. 3. CRF increased actin polymerization, phosphorylation of Focal Adhesion Kinase (FAK), providing a potential mechanism for the observed induction of MCF7 motility. 4. CRF induced the expression of Cox-1 but not Cox-2 in MCF7 cells as well as the production of prostaglandins, factors known to promote invasiveness and metastasis. CONCLUSION: Overall, our data suggest that CRF stimulates cell motility and invasiveness of MCF7 cells most probably via induction of FAK phosphorylation and actin filament reorganization and production of prostaglandins via Cox1. Based on these findings we postulate that the stress neuropeptide CRF present in the vicinity of tumors (either produced locally by the tumor cells themselves or by nearby normal cells or secreted from the innervations of surrounding tissues) may play an important role on breast tumor growth and metastatic capacity, providing a potential link between stress and tumor progression.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Corticotropin-Releasing Hormone/physiology , Actins/metabolism , Analysis of Variance , Apoptosis , Blotting, Western , Cell Line, Tumor , Cell Movement , Corticotropin-Releasing Hormone/pharmacology , Cyclooxygenase 2/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Focal Adhesion Kinase 1/metabolism , Humans , Neoplasm Invasiveness , Phosphorylation , Prostaglandins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
J Immunol ; 182(10): 6444-51, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414798

ABSTRACT

High levels of plasma adiponectin are associated with low levels of inflammatory markers and cardioprotection. The mechanism via which adiponectin exerts its anti-inflammatory effect is yet unknown. In the present study, we demonstrate that globular adiponectin (gAd) induces the expression of the inactive isoform of IL-1R-associated kinases (IRAK), IRAK-M. Homologous deletion of IRAK-M in IRAK-M(-/-) mice abolished the tolerogenic properties of gAd because pretreatment of IRAK-M(-/-) macrophages with gAd did not suppress LPS-induced proinflammatory cytokine production. GAd activated the MAPKs MEK1/2 and ERK1/2 in macrophages via their upstream regulator Tpl2. Activation of ERK1/2 via Tpl2 appeared necessary for the induction of IRAK-M because gAd did not induce IRAK-M in Tpl2(-/-) macrophages or in macrophages pretreated with the MEK1/2 inhibitor UO126. In addition, activation of PI3K and Akt1 also appeared necessary for the induction of IRAK-M by gAd, because treatment of Akt1(-/-) macrophages or pretreatment of macrophages with the PI3K inhibitor wortmannin abolished gAd-induced IRAK-M expression. Analysis of IRAK-M expression in human peripheral blood cells confirmed that serum adiponectin was negatively associated with IRAK-M and responsiveness to LPS. In conclusion, our data demonstrate that IRAK-M is a major mediator of gAd-induced endotoxin tolerance in primary macrophages, expression of which depends on the activation of Tpl2/ERK and PI3K/Akt1 signaling pathways.


Subject(s)
Adiponectin/immunology , Endotoxins/immunology , Immune Tolerance , Interleukin-1 Receptor-Associated Kinases/biosynthesis , Macrophages/immunology , Adiponectin/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/immunology , Humans , Interleukin-1 Receptor-Associated Kinases/immunology , MAP Kinase Kinase Kinases/immunology , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/immunology , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology
14.
Mol Immunol ; 45(10): 2970-80, 2008 May.
Article in English | MEDLINE | ID: mdl-18336909

ABSTRACT

Toll-like receptor 4 (TLR4) recognizes and initiates signals from Gram-negative bacterial lipopolysaccharide (LPS) triggering the inflammatory response. Expression levels of TLR4 on macrophages partly regulate the magnitude of the response to LPS. Vasoactive Intestinal Peptide (VIP) is known to block inflammatory responses by inhibiting pro-inflammatory cytokine production from activated macrophages. In the present report we demonstrate that VIP directly suppressed TLR4 expression on naïve primary mouse macrophages utilizing signalling cascades that control TLR4 transcription. VIP-induced suppression of TLR4 occurred at the transcriptional level by decreasing PU.1 DNA binding. Mutation of the proximal PU.1 but not the AP-1-binding site on the TLR4 promoter abrogated VIP-induced suppression of TLR4 transcription. Moreover, inhibition of PI3K by wortmannin or homologous deletion of the Akt1 isoform, a pathway known to act as a negative regulator of macrophage activation, alleviated the suppressive action of VIP on TLR4 expression. To evaluate the biological significance of VIP effect on TLR4 expression, Raw264.7 macrophages were pre-treated with VIP for 24h and then exposed to LPS. Pre-treatment with VIP rendered macrophages hypo-responsive to LPS resulting in reduced pro-inflammatory cytokine production. Moreover, in vivo administration of VIP in C57BL/6 mice resulted in lower IL-6 production upon treatment with LPS. Overall, the results indicate that VIP promotes endotoxin tolerance by downregulating TLR4 expression via Akt1.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/enzymology , Macrophages/immunology , Proto-Oncogene Proteins c-akt/metabolism , Toll-Like Receptor 4/immunology , Vasoactive Intestinal Peptide/pharmacology , Animals , Cell Line , Cytokines/biosynthesis , DNA/metabolism , Gene Expression Regulation/drug effects , Immune Tolerance/drug effects , Inflammation Mediators , Interleukin-1 Receptor-Associated Kinases/immunology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptor 4/genetics , Trans-Activators/metabolism , Transcription, Genetic/drug effects
15.
Proc Natl Acad Sci U S A ; 105(8): 2987-92, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18287049

ABSTRACT

The protein kinase encoded by the Tpl2 protooncogene plays an obligatory role in the transduction of Toll-like receptor and death receptor signals in macrophages, B cells, mouse embryo fibroblasts, and epithelial cells in culture and promotes inflammatory responses in animals. To address its role in T cell activation, we crossed the T cell receptor (TCR) transgene 2C, which recognizes class I MHC presented peptides, into the Tpl2(-/-) genetic background. Surprisingly, the TCR2C(tg/tg)/Tpl2(-/-) mice developed T cell lymphomas with a latency of 4-6 months. The tumor cells were consistently TCR2C(+)CD8(+)CD4(-), suggesting that they were derived either from chronically stimulated mature T cells or from immature single positive (ISP) cells. Further studies showed that the population of CD8(+) ISP cells was not expanded in the thymus of TCR2C(tg/tg)/Tpl2(-/-) mice, making the latter hypothesis unlikely. Mature peripheral T cells of Tpl2(-/-) mice were defective in ERK activation and exhibited enhanced proliferation after TCR stimulation. The same cells were defective in the induction of CTLA4, a negative regulator of the T cell response, which is induced by TCR signals via ERK. These findings suggest that Tpl2 functions normally in a feedback loop that switches off the T cell response to TCR stimulation. As a result, Tpl2, a potent oncogene, functions as a tumor suppressor gene in chronically stimulated T cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/metabolism , Antigens, Differentiation/metabolism , Blotting, Western , CTLA-4 Antigen , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Enzyme Activation/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Flow Cytometry , Immunohistochemistry , MAP Kinase Kinase Kinases/immunology , Mice , Mice, Knockout , Proto-Oncogene Proteins/immunology , T-Lymphocytes/metabolism , Transgenes/genetics
16.
J Cell Physiol ; 210(3): 774-83, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17117478

ABSTRACT

Corticotropin-releasing factor (CRF), the principal regulator of the hypothalamus-pituitary-adrenal (HPA) axis, also modulates the inflammatory response directly, via its effect on mast cells and macrophages. On macrophages, it augments production of lipopolysaccharide (LPS)-induced pro-inflammatory cytokines. CRF and its related peptides may also act as anti-inflammatory agents. Aim of the present work was to examine the role of macrophages on the anti-inflammatory effects of CRF-peptides and the mechanism involved. Thus, we examined if CRF receptor 1 (CRF1) and CRF2 agonists exert any anti-inflammatory effect on primary mouse macrophages. We have found that: (a) CRF, Urocortin (UCN)1 and UCN2 transiently suppressed the release of Tumor Necrosis Factor-alpha (TNF-alpha) in LPS-activated macrophages, an effect peaking at 4 h. This effect did not involve changes on TNF-alpha transcription. (b) CRF peptide-induced suppression of TNF-alpha release depended on induction of COX-2 and PGE2 synthesis. (c) Use of specific CRF1 and CRF2 antagonists suggested that this effect involved both CRF receptor types. (d) The effect of CRF-peptides on COX-2 was mediated via PI3K and p38MAPK. (e) Longer exposure of macrophages to CRF-peptides resulted in induction of TNF-alpha production via enhancement of its transcription. In conclusion, this is the first report suggesting that CRF1 and CRF2 agonists exert a biphasic effect on macrophages. During the early stages of the inflammatory response, they suppress TNF-alpha release via induction of COX-2/PGE2 while later on they induce TNF-alpha transcription. Hence, the reported anti-inflammatory effect of CRF-peptides appears to involve macrophages and is confined at the early stage of inflammation.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Inflammation/prevention & control , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Inflammation/physiopathology , Macrophages/cytology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/drug effects , Receptors, Corticotropin-Releasing Hormone/physiology , Urocortins , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Ann N Y Acad Sci ; 1083: 185-95, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17148740

ABSTRACT

Several recently published reports, including ours, suggest that adiponectin is a strong proinflammatory agent. Indeed, exposure of human placenta and adipose tissue to adiponectin induces the production of interleukin-1beta (IL-1beta), IL-6, tumor necrosis factor alpha (TNF-alpha), and prostaglandin E2 (PGE2). We have previously shown that adiponectin is a powerful inducer of proinflammatory cytokines production by macrophages. The reported anti-inflammatory effect of adiponectin may be due to the induction of macrophage tolerance to further adiponectin exposure or to other proinflammatory stimuli including the Toll-like receptor (TLR) 3 ligand polyI:C and the TLR4 ligand lipopolysaccharide (LPS). We now present additional data supporting the hypothesis that adiponectin is a strong proinflammatory adipokine. More specifically, we demonstrate that adiponectin induces IL-1beta and IL-8 from THP-1 macrophage cell line. The effect of adiponectin is not restricted to differentiated THP-1 macrophages but it is evident at lower levels in undifferentiated THP-1 monocytes promoting TNF-alpha, IL-6, and IL-8 production. Thus, its high levels in the circulation of lean subjects render their macrophages resistant to several proinflammatory stimuli including its own thus acting in effect as an anti-inflammatory agent. Lowering of its high levels, as a consequence of increased body mass index (BMI), renders macrophages sensitive to any proinflammatory insult.


Subject(s)
Adiponectin/physiology , Cytokines/biosynthesis , Metabolic Syndrome/immunology , Adiponectin/pharmacology , Cell Line , Dose-Response Relationship, Drug , Humans , Inflammation/immunology , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Macrophages/metabolism , Monocytes/metabolism , Obesity/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
18.
Neuroendocrinology ; 84(1): 49-57, 2006.
Article in English | MEDLINE | ID: mdl-17090973

ABSTRACT

The stress neuropeptides, corticotropin-releasing hormone (CRH) and urocortin (UCN), modulate the inflammatory response via the hypothalamus-pituitary-adrenal axis and locally, in a paracrine manner, act on mast and macrophage cells. Kupffer cells (KCs) are the resident macrophages of the liver. They represent the bulk of tissue macrophages in the body and they are the first to face invading noxious agents reaching the body via the portal circulation. The aim of the present report was to study the expression of the CRH system in rat KC and test its functionality. Our findings are as follows: (1) In highly purified KCs the transcripts of UCN, of its receptors CRHR1, CRHR2 and that of the pseudoreceptor CRH-binding protein (CRHBP) were present while that of CRH was not detectable. (2) Similarly, immunoreactive UCN, CRHR1, CRHR2 and CRHBP were easily detectable by immunohistochemistry and immunofluorescence in sections of whole rat liver (localized in KC) as well as in purified KC while CRH was again not detectable. (3) Exposure of purified KC to CRH or UCN suppressed lipopolysaccharide-induced tumor necrosis factor alpha production, an effect completely prevented by the CRHR1 and CRHR2 receptor antagonist astressin. Our data demonstrate the presence of UCN and its receptors in rat KC, the absence of CRH, and the functionality of these receptors. We propose that a UCN-based system may affect local inflammatory phenomena in the liver acting in a paracrine manner.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Kupffer Cells/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/pharmacology , Corticotropin-Releasing Hormone/physiology , Gene Expression Regulation , Male , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Urocortins
19.
Int J Biochem Cell Biol ; 38(10): 1654-61, 2006.
Article in English | MEDLINE | ID: mdl-16713323

ABSTRACT

Cyclooxygenease-2 (COX-2) is the key enzyme regulating the production of prostaglandins, central mediators of inflammation. The expression of cyclooxygenease-2 is induced by several extra cellular signals including pro-inflammatory and growth-promoting stimuli. All signals converge to the activation of mitogen-activated protein kinases (MAPK) that regulate cyclooxygenease-2 mRNA levels both at the transcriptional and post-transcriptional level. The machinery appears to be highly specialized involving activation of distinct signalling molecules depending on the type of extracellular stimulus. Expression of cyclooxygenease-2 mRNA is regulated by several transcription factors including the cyclic-AMP response element binding protein (CREB), nuclear factor kappa B (NFkB) and the CCAAT-enhancer binding protein (C/EBP). Cyclooxygenease-2 is also affected post-transcriptionaly, at the level of mRNA stability. Finally, cyclooxygenease-2 can be affected directly at its enzymatic activity by nitric oxide and nitric oxide synthase (iNOS). Each step of cyclooxygenease-2 regulation can be used as potential therapeutic target.


Subject(s)
Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase 2/genetics , Cyclooxygenase 2/physiology , Gene Expression Regulation, Enzymologic , Animals , Cyclooxygenase 2/drug effects , Cyclooxygenase 2 Inhibitors/adverse effects , Female , Humans , Mice , Prostaglandins/metabolism , Signal Transduction
20.
J Immunol ; 176(3): 1869-77, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16424218

ABSTRACT

Corticotropin-releasing factor (CRF) augments LPS-induced proinflammatory cytokine production from macrophages. The aim of the present study was to determine the mechanism by which CRF and its related peptides urocortins (UCN) 1 and 2 affect LPS-induced cytokine production. We examined their role on TLR4 expression, the signal-transducing receptor of LPS. For this purpose, the murine macrophage cell line RAW 264.7 and primary murine peritoneal macrophages were used. Exposure of peritoneal macrophages and RAW 264.7 cells to CRF, UCN1, or UCN2 up-regulated TLR4 mRNA and protein levels. To study whether that effect occurred at the transcriptional level, RAW 264.7 cells were transfected with a construct containing the proximal region of the TLR4 promoter linked to the luciferase gene. CRF peptides induced activation of the TLR4 promoter, an effect abolished upon mutation of a proximal PU.1-binding consensus or upon mutation of an AP-1-binding element. Indeed, all three peptides promoted PU.1 binding to the proximal PU.1 site and increased DNA-binding activity to the AP-1 site. The effects of CRF peptides were inhibited by the CRF2 antagonist anti-sauvagine-30, but not by the CRF1 antagonist antalarmin, suggesting that CRF peptides mediated the up-regulation of TLR4 via the CRF2 receptor. Finally, CRF peptides blocked the inhibitory effect of LPS on TLR4 expression. In conclusion, our data suggest that CRF peptides play an important role on macrophage function. They augment the effect of LPS by inducing Tlr4 gene expression, through CRF2, via activation of the transcription factors PU.1 and AP-1.


Subject(s)
Corticotropin-Releasing Hormone/physiology , Macrophages/metabolism , Proto-Oncogene Proteins/metabolism , Toll-Like Receptor 4/biosynthesis , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line , Gene Expression Regulation/immunology , Genes, Reporter , Lipopolysaccharides/metabolism , Mice , Peptide Fragments/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins/physiology , Receptors, Corticotropin-Releasing Hormone/metabolism , Toll-Like Receptor 4/genetics , Trans-Activators/physiology , Transcription Factor AP-1/physiology , Transfection , Urocortins
SELECTION OF CITATIONS
SEARCH DETAIL
...