Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(51): e2303075120, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38100414

ABSTRACT

Adipose tissue macrophages (ATM) are key players in the development of obesity and associated metabolic inflammation which contributes to systemic metabolic dysfunction. We here found that fibroblast activation protein α (FAP), a well-known marker of cancer-associated fibroblast, is selectively expressed in murine and human ATM among adipose tissue-infiltrating leukocytes. Macrophage FAP deficiency protects mice against diet-induced obesity and proinflammatory macrophage infiltration in obese adipose tissues, thereby alleviating hepatic steatosis and insulin resistance. Mechanistically, FAP specifically mediates monocyte chemokine protein CCL8 expression by ATM, which is further upregulated upon high-fat-diet (HFD) feeding, contributing to the recruitment of monocyte-derived proinflammatory macrophages with no effect on their classical inflammatory activation. CCL8 overexpression restores HFD-induced metabolic phenotypes in the absence of FAP. Moreover, macrophage FAP deficiency enhances energy expenditure and oxygen consumption preceding differential body weight after HFD feeding. Such enhanced energy expenditure is associated with increased levels of norepinephrine (NE) and lipolysis in white adipose tissues, likely due to decreased expression of monoamine oxidase, a NE degradation enzyme, by Fap-/- ATM. Collectively, our study identifies FAP as a previously unrecognized regulator of ATM function contributing to diet-induced obesity and metabolic inflammation and suggests FAP as a potential immunotherapeutic target against metabolic disorders.


Subject(s)
Adipose Tissue , Insulin Resistance , Animals , Humans , Mice , Adipose Tissue/metabolism , Diet, High-Fat , Inflammation/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Obesity/metabolism
2.
Allergy ; 74(3): 495-506, 2019 03.
Article in English | MEDLINE | ID: mdl-30390302

ABSTRACT

BACKGROUND: Epithelial cells (ECs) play a crucial role in allergic sensitization to inhaled protease allergens by instructing type 2 innate lymphoid cells (ILC2) and dendritic cells (DCs) via release of pro-type 2 cytokines, particularly interleukin-33 (IL-33). Leukotriene B4 (LTB4) is a well-known leukocyte chemoattractant via engagement of its receptor 1 (BLT1). However, the role of LTB4-BLT1 axis in allergic sensitization via activation of ECs is still unknown. METHODS: We evaluated the effect of LTB4-BLT1 axis on IL-33 expression and ILC2 activation in vivo and in vitro. Chimeric mice were established to evaluate the contribution of BLT1 expression in nonimmune cell to allergic sensitization. RESULTS: Genetical or pharmacological interruption of LTB4-BLT1 axis during sensitization phase markedly reduced papain-induced IL-33 expression, decreased ILC2 activation and DC migration, thereby impairing the priming of allergic Th2 responses. Furthermore, papain inhalation induced a rapid release of LTB4 preceding IL-33, and intranasal administration of LTB4 to naïve WT mice significantly increased IL-33 expression and ILC2 activation in lung, which was absent in Il33-/- or Ltb4r1-/- mice. Furthermore, BLT1 was expressed in primary mouse ECs or normal human bronchial ECs (NHBE), and papain induced LTB4 release by NHBE, which in turn amplified IL-33 production dependent on Akt activation via BLT1. Consequently, bone marrow chimeric mice lacking BLT1 in radio-resistant structural cells failed to develop allergic lung inflammation to papain. CONCLUSION: Our study reveals a functional role of LTB4-BLT1 axis in nonimmune cells, most likely lung ECs, in controlling allergic sensitization as an upstream regulator of IL-33.


Subject(s)
Epithelial Cells/metabolism , Hypersensitivity/immunology , Hypersensitivity/metabolism , Interleukin-33/biosynthesis , Receptors, Leukotriene B4/metabolism , Signal Transduction , Allergens/immunology , Animals , Cytokines/metabolism , Hypersensitivity/genetics , Immunization , Interleukin-33/genetics , Leukotriene B4/metabolism , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Papain/immunology , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
3.
Cytokine ; 111: 530-540, 2018 11.
Article in English | MEDLINE | ID: mdl-29884308

ABSTRACT

BLT1, the primary functional receptor of Leukotriene B4 (LTB4), is involved in tissue inflammation by mediating leukocyte recruitment, and recently LTB4-dependent inflammation was reported to promote lung tumor growth. Exposure to diesel exhaust particle (DEP), the major component of particulate matter 2.5 (PM2.5), can elicit lung inflammation, which may increase the risk of lung cancer. However, it remains unknown about the critical factors mediating DEP-induced lung inflammation and the subsequent effect on tumor metastasis. In this study, we found that DEP exposure led to acute lung inflammation, characterized by abundant infiltration of neutrophils and elevated lung levels in LTB4, as well as several pro-inflammatory cytokines and chemokines, including IL-1ß, IL-6, TNF-α, CXCL1/2. Furthermore, DEP exposure promoted lung metastasis of 3LL and 4T1 cells. BLT1 blockade by its specific antagonist U75302 significantly inhibited neutrophilic lung inflammation following DEP exposure. Importantly, BLT1 blockade before the onset of inflammation significantly reduced DEP-enhanced lung metastasis, which was associated with greatly decreased infiltrating neutrophils in lungs. Interestingly, BLT1 blockade after the occurrence of lung metastases had no effect on the magnitude of lung metastasis, suggesting that inhibition of BLT1-mediated lung inflammation was insufficient to suppress established metastatic tumor. Administration of BLT2 inhibitor LY255283 fails to inhibit DEP-induced lung inflammation and tumor metastasis. Collectively, our results demonstrate that DEP exposure causes BLT1-mediated lung neutrophilic inflammation, which is critical for tumor lung metastasis, and suggest that interruption of the LTB4-BLT1 axis could be useful for preventing PM2.5-induced inflammation and subsequent susceptible to lung metastasis.


Subject(s)
Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Lung/pathology , Neutrophils/metabolism , Pneumonia/metabolism , Receptors, Leukotriene B4/metabolism , Vehicle Emissions/toxicity , Animals , Cell Line, Tumor , Chemokines/metabolism , Female , Interleukin-1beta/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , Leukocytes/pathology , Lung/drug effects , Lung/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/pathology , Pneumonia/chemically induced , Pneumonia/pathology , Signal Transduction/drug effects
4.
J Immunol ; 198(4): 1673-1684, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28077599

ABSTRACT

Leukotriene B4 (LTB4) and its functional receptor BLT1 are closely involved in tissue inflammation by primarily mediating leukocyte recruitment and activation. Elevated LTB4 was reported in patients with lung fibrosis; however, the role of the LTB4/BLT1 axis in lung fibrosis remains unknown. In this study, we demonstrated that BLT1-/- mice exhibited significantly attenuated bleomycin (BLM)-induced lung fibrosis. Interestingly, BLT1 blockade with its specific antagonist U75302 in the acute injury phase (days 0-10 after BLM treatment) significantly attenuated lung fibrosis, which was accompanied by significant decreases in early infiltrating neutrophils and later infiltrating CD4+ T cells and the production of TGF-ß, IL-13, and IL-17A. In contrast, BLT1 blockade in the fibrotic phase (days 10-21 after BLM treatment) had no effect on lung fibrosis and TGF-ß production, although it significantly decreased CD4+ T cell infiltration. Furthermore, depletion of neutrophils or CD4+ T cells had no effect on BLM-induced lung fibrosis, suggesting the independence of profibrotic activity of the LTB4/BLT1 axis on BLT1-dependent lung recruitment of these two leukocytes. Finally, although BLT1 blockade had no effect on the recruitment and phenotype of macrophages in BLM-induced lung fibrosis, the LTB4/BLT1 axis could promote TGF-ß production by macrophages stimulated with BLM or supernatants from BLM-exposed airway epithelial cells in an autocrine manner, which further induced collagen secretion by lung fibroblasts. Collectively, our study demonstrates that the LTB4/BLT1 axis plays a critical role in acute injury phase to promote BLM-induced lung fibrosis, and it suggests that early interruption of the LTB4/BLT1 axis in some inflammatory diseases could prevent the later development of tissue fibrosis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Neutrophils/immunology , Pulmonary Fibrosis/immunology , Receptors, Leukotriene B4/metabolism , Animals , Bleomycin , CD4-Positive T-Lymphocytes/physiology , Fatty Alcohols/administration & dosage , Glycols/administration & dosage , Inflammation , Interleukin-13/biosynthesis , Interleukin-13/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Lung/immunology , Lung/pathology , Mice , Neutrophil Infiltration , Neutrophils/physiology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/prevention & control , Receptors, Leukotriene B4/deficiency , Receptors, Leukotriene B4/genetics , Signal Transduction , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/immunology
5.
Nat Commun ; 7: 10680, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26893169

ABSTRACT

Stimulator of interferon genes (STING) is known be involved in control of DNA viruses but has an unexplored role in control of RNA viruses. During infection with DNA viruses STING is activated downstream of cGAMP synthase (cGAS) to induce type I interferon. Here we identify a STING-dependent, cGAS-independent pathway important for full interferon production and antiviral control of enveloped RNA viruses, including influenza A virus (IAV). Further, IAV interacts with STING through its conserved hemagglutinin fusion peptide (FP). Interestingly, FP antagonizes interferon production induced by membrane fusion or IAV but not by cGAMP or DNA. Similar to the enveloped RNA viruses, membrane fusion stimulates interferon production in a STING-dependent but cGAS-independent manner. Abolishment of this pathway led to reduced interferon production and impaired control of enveloped RNA viruses. Thus, enveloped RNA viruses stimulate a cGAS-independent STING pathway, which is targeted by IAV.


Subject(s)
Influenza A virus/physiology , Influenza, Human/enzymology , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , RNA Virus Infections/enzymology , Animals , Humans , Influenza A virus/genetics , Influenza, Human/genetics , Influenza, Human/metabolism , Influenza, Human/virology , Interferon Type I/genetics , Interferon Type I/metabolism , Membrane Proteins/genetics , Mice , Nucleotidyltransferases/genetics , RNA Virus Infections/genetics , RNA Virus Infections/metabolism , RNA Virus Infections/virology , RNA Viruses/genetics , RNA Viruses/physiology
6.
Immunology ; 146(1): 50-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25959240

ABSTRACT

Leukotriene B4 (LTB4 ) is a lipid mediator that is rapidly generated in inflammatory sites, and its functional receptor, BLT1, is mostly expressed on immune cells. Contact dermatitis is a common inflammatory skin disease characterized by skin oedema and abundant inflammatory infiltrates, primarily including neutrophils and CD8(+) T cells. The role of the LTB4 -BLT1 axis in contact dermatitis remains largely unknown. In this study, we found up-regulated gene expression of 5-lipoxygenase and leukotriene A4 hydrolase, two critical enzymes for LTB4 synthesis, BLT1 and elevated LTB4 levels in skin lesions of oxazolone (OXA)-induced contact dermatitis. BLT1 deficiency or blockade of LTB4 and BLT1 by the antagonists, bestatin and U-75302, respectively, in the elicitation phase caused significant decreases in ear swelling and skin-infiltrating neutrophils and CD8(+) T cells, which was accompanied by significantly reduced skin expression of CXCL1, CXCL2, interferon-γ and interleukin-1ß. Furthermore, neutrophil depletion during the elicitation phase of OXA-induced contact dermatitis also caused significant decreases in ear swelling and CD8(+) T-cell infiltration accompanied by significantly decreased LTB4 synthesis and gene expression of CXCL2, interferon-γ and interleukin-1ß. Importantly, subcutaneous injection of exogenous LTB4 restored the skin infiltration of CD8(+) T cells in neutrophil-depleted mice following OXA challenge. Collectively, our results demonstrate that the LTB4 -BLT1 axis contributes to OXA-induced contact dermatitis by mediating skin recruitment of neutrophils, which are a major source of LTB4 that sequentially direct CD8(+) T-cell homing to OXA-challenged skin. Hence, LTB4 and BLT1 could be potential therapeutic targets for the treatment of contact dermatitis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dermatitis, Contact/immunology , Leukotriene B4/immunology , Neutrophils/immunology , Receptors, Leukotriene B4/immunology , Animals , Arachidonate 5-Lipoxygenase/biosynthesis , Chemokine CXCL1/biosynthesis , Chemokine CXCL2/biosynthesis , Dermatitis, Contact/drug therapy , Epoxide Hydrolases/biosynthesis , Fatty Alcohols/pharmacology , Female , Glycols/pharmacology , Inflammation/drug therapy , Inflammation/immunology , Interferon-gamma/biosynthesis , Interleukin-1beta/biosynthesis , Leucine/analogs & derivatives , Leucine/pharmacology , Leukotriene B4/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxazolone , Receptors, Leukotriene B4/antagonists & inhibitors , Receptors, Leukotriene B4/biosynthesis , Skin/cytology , Skin/immunology
7.
Lab Invest ; 95(3): 342-50, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25502178

ABSTRACT

Daily subcutaneous (sc) injection of bleomycin (BLM) causes dermal fibrosis but rarely causes lung changes in mice. There are also significant disadvantages to this traditional model for systemic sclerosis, including a variable distribution of lesions and a requirement for repetitive procedures. The present study was undertaken to develop a convenient method of BLM administration that yields stable dermal inflammation and fibrosis with extensive and reproducible interstitial lung disease (ILD) in mice. Osmotic minipumps containing BLM (150 mg/kg) or saline were implanted sc in C57BL/6 mice and the drug was delivered as a continuous infusion over 1∼4 weeks. The time course of morphological features, collagen content, and pro-inflammatory cytokine expression in the skin and the lungs were analyzed. Pathological examination demonstrated dominant inflammatory infiltrates at week 1 and significant fibrosis at week 4. Decreased microvessel density and increased myofibroblast counts were observed in the skin of BLM-treated mice at week 4. In addition, there were obvious increases in dermal infiltration of CD45(+) leukocytes, including F4/80(+) macrophages, Gr-1(+) neutrophils, and CD3(+) T lymphocytes in BLM-treated mice. IL-1ß, IL-4, and CXCL2 transcripts were continually upregulated by BLM in the skin and lung tissues. In addition, lungs from BLM-treated mice showed significant inflammatory infiltrates and confluent subpleural fibrosis at week 4. In conclusion, this modified murine model for drug-induced systemic inflammation and fibrosis uses a single procedure and provides reproducible skin and lung lesions, mimicking human systemic sclerosis (SSc) with ILD-like manifestation.


Subject(s)
Bleomycin/administration & dosage , Disease Models, Animal , Pneumonia/pathology , Pulmonary Fibrosis/pathology , Scleroderma, Systemic/pathology , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Chemokine CXCL2/genetics , Gene Expression , Humans , Infusion Pumps, Implantable , Interleukin-1beta/genetics , Interleukin-4/genetics , Leukocytes/metabolism , Leukocytes/pathology , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Pneumonia/chemically induced , Pneumonia/genetics , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Reverse Transcriptase Polymerase Chain Reaction , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...