Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 441
Filter
1.
Biochem Biophys Res Commun ; 533(3): 493-500, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32977946

ABSTRACT

Silica crystals (silica), which are a major mineral component of volcanic ash and desert dust, contribute to the pathogenesis of pulmonary disorders such as asthma and fibrosis. Although administration of silica or sand dust to rodents exacerbates development of ovalbumin-induced or house dust mite-induced asthma-like airway inflammation, the detailed mechanisms remain unclear. Here, using murine models, we found that silica can induce IL-33 expression in pulmonary epithelial cells. IL-33, but not IL-25 or TSLP, and type 2 cytokines such as IL-5 and IL-13 were critically involved in silica's exacerbation of OVA-induced airway eosinophilia in mice. Innate lymphoid cells (ILCs), but not T, B or NKT cells, were also involved in the setting. Moreover, a scavenger receptor that recognized silica was important for silica's exacerbating effect. These observations suggest that IL-33 induced in epithelial cells by silica activates ILCs to produce IL-5 and/or IL-13, contributing to silica's exacerbation of OVA-induced airway eosinophilia in mice. Our findings provide new insight into the underlying mechanisms of exacerbation of pulmonary disorders such as asthma following inhalation of silica-containing materials such as volcanic ash and desert dust.


Subject(s)
Interleukin-33/physiology , Pulmonary Eosinophilia/immunology , Silicon Dioxide/toxicity , Animals , Asthma/immunology , Cytokines/physiology , Interleukin-13/physiology , Interleukin-33/biosynthesis , Interleukin-5/physiology , Interleukins/physiology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/immunology , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Eosinophilia/chemically induced , Receptors, Scavenger/physiology , Thymic Stromal Lymphopoietin
2.
PLoS One ; 15(5): e0233728, 2020.
Article in English | MEDLINE | ID: mdl-32469969

ABSTRACT

BACKGROUND: Carotid artery plaque is an established marker of subclinical atherosclerosis with pronounced sex-dimorphism. Here, we aimed to identify genetic variants associated with carotid plaque burden (CPB) and to examine potential sex-specific genetic effects on plaque sizes. METHODS AND RESULTS: We defined six operationalizations of CPB considering plaques in common carotid arteries, carotid bulb, and internal carotid arteries. We performed sex-specific genome-wide association analyses for all traits in the LIFE-Adult cohort (n = 727 men and n = 550 women) and tested significantly associated loci for sex-specific effects. In order to identify causal genes, we analyzed candidate gene expression data for correlation with CPB traits and corresponding sex-specific effects. Further, we tested if previously reported SNP associations with CAD and plaque prevalence are also associated with CBP. We found seven loci with suggestive significance for CPB (p<3.33x10-7), explaining together between 6 and 13% of the CPB variance. Sex-specific analysis showed a genome-wide significant hit for men at 5q31.1 (rs201629990, ß = -0.401, p = 5.22x10-9), which was not associated in women (ß = -0.127, p = 0.093) with a significant difference in effect size (p = 0.008). Analyses of gene expression data suggested IL5 as the most plausible candidate, as it reflected the same sex-specific association with CPBs (p = 0.037). Known plaque prevalence or CAD loci showed no enrichment in the association with CPB. CONCLUSIONS: We showed that CPB is a complementary trait in analyzing genetics of subclinical atherosclerosis. We detected a novel locus for plaque size in men only suggesting a role of IL5. Several estrogen response elements in this locus point towards a functional explanation of the observed sex-specific effect.


Subject(s)
Arteriosclerosis/genetics , Carotid Artery Diseases/genetics , Genetic Predisposition to Disease/genetics , Interleukin-5 , Plaque, Atherosclerotic/genetics , Sex Characteristics , Aged , Carotid Artery, External/diagnostic imaging , Carotid Artery, External/pathology , Carotid Artery, Internal/diagnostic imaging , Carotid Artery, Internal/pathology , Cohort Studies , Female , Genome-Wide Association Study , Humans , Interleukin-5/genetics , Interleukin-5/physiology , Male , Middle Aged , Sex Factors
3.
Front Immunol ; 9: 2673, 2018.
Article in English | MEDLINE | ID: mdl-30505309

ABSTRACT

Even though more than 30 years have passed since the eradication of smallpox, high titers of smallpox-specific antibodies are still detected in the blood of subjects vaccinated in childhood. In fact, smallpox-specific antibody levels are maintained in serum for more than 70 years. The generation of life-long immunity against infectious diseases such as smallpox and measles has been thoroughly documented. Although the mechanisms behind high persisting antibody titers in the absence of the causative agent are still unclear, long lived plasma cells (LLPCs) play an important role. Most of the current knowledge on LLPCs is based on experiments performed in mouse models, although the amount of data derived from human studies is increasing. As the results from mouse models are often directly extrapolated to humans, it is important to keep in mind that there are differences. These are not only the obvious such as the life span but there are also anatomical differences, for instance the adiposity of the bone marrow (BM) where LLPCs reside. Whether these differences have an effect on the function of the immune system, and in particular on LLPCs, are still unknown. In this review, we will briefly discuss current knowledge of LLPCs, comparing mice and humans.


Subject(s)
Plasma Cells/cytology , Plasma Cells/immunology , Adiposity/physiology , Animals , Antibodies, Monoclonal/therapeutic use , Bone Marrow/immunology , Germinal Center/immunology , Humans , Interleukin-5/physiology , Interleukin-6/physiology , Longevity/physiology , Mice , Receptors, Cell Surface , Transcription Factors
4.
Int J Parasitol ; 47(14): 951-960, 2017 12.
Article in English | MEDLINE | ID: mdl-28859850

ABSTRACT

Approximately 100 million people suffer from filarial diseases including lymphatic filariasis (elephantiasis), onchocerciasis (river blindness) and loiasis. These diseases are amongst the most devastating of the neglected tropical diseases in terms of social and economic impact. Moreover, many infection-induced immune mechanisms in the host, their relationship to disease-related symptoms and the development of pathology within the site of infection remain unclear. To improve on current drug therapies or vaccines, further studies are necessary to decipher the mechanisms behind filaria-driven immune responses and pathology development, and thus the rodent model of Litomosoides sigmodontis can be used to unravel host-filaria interactions. Interestingly, BALB/c mice develop a patent state (release of microfilariae, the transmission life-stage, into the periphery) when exposed to L. sigmodontis. Thus, using this model, we determined levels of host inflammation and pathology development during a L. sigmodontis infection in vivo for the first known time. Our study reveals that after 30days p.i., inflammation and pathology began to develop in infected wild type BALB/c mice between the lung and diaphragm, close to the site of infection - the thoracic cavity. Interestingly, infected IL-4Rα/IL-5-/- BALB/c mice had accentuated inflammation of the pleural lung and pleural diaphragm, and higher parasite burdens. Corresponding to the pleural inflammation, levels of IP-10, MIP-1α, MIP-1ß, MIP-2 and RANTES were significantly elevated in the thoracic cavity fluid of infected IL-4Rα/IL-5-/- mice compared with wild type controls. Moreover, upon L. sigmodontis antigen stimulation, IFN-γ and IL-17A secretions by cells isolated from draining lymph nodes of IL-4Rα/IL-5-/- mice were significantly elevated, whereas secretion of IL-5, IL-13 and IL-10 was reduced. Elevated filaria-specific IFN-γ secretion was also observed in spleen-derived CD4+ T cell co-cultures from IL-4Rα/IL-5-/- mice. In summary, this study unravels the essential role of IL-4/IL-5 signalling in controlling immunity against filarial infections and demonstrates the requirement of this pathway for the host to control ensuing pathology and inflammation.


Subject(s)
Filariasis/immunology , Filarioidea/immunology , Interleukin-4/physiology , Interleukin-5/physiology , Animals , Chemokines/metabolism , Diaphragm/parasitology , Diaphragm/pathology , Female , Filariasis/pathology , Filarioidea/pathogenicity , Interferon-gamma/metabolism , Lung/parasitology , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Pleural Cavity/parasitology , Pleural Cavity/pathology , Signal Transduction , Specific Pathogen-Free Organisms , Spleen/cytology , Spleen/immunology
5.
J Exp Med ; 214(4): 943-957, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28302646

ABSTRACT

Inflammatory dilated cardiomyopathy (DCMi) is a major cause of heart failure in children and young adults. DCMi develops in up to 30% of myocarditis patients, but the mechanisms involved in disease progression are poorly understood. Patients with eosinophilia frequently develop cardiomyopathies. In this study, we used the experimental autoimmune myocarditis (EAM) model to determine the role of eosinophils in myocarditis and DCMi. Eosinophils were dispensable for myocarditis induction but were required for progression to DCMi. Eosinophil-deficient ΔdblGATA1 mice, in contrast to WT mice, showed no signs of heart failure by echocardiography. Induction of EAM in hypereosinophilic IL-5Tg mice resulted in eosinophilic myocarditis with severe ventricular and atrial inflammation, which progressed to severe DCMi. This was not a direct effect of IL-5, as IL-5TgΔdblGATA1 mice were protected from DCMi, whereas IL-5-/- mice exhibited DCMi comparable with WT mice. Eosinophils drove progression to DCMi through their production of IL-4. Our experiments showed eosinophils were the major IL-4-expressing cell type in the heart during EAM, IL-4-/- mice were protected from DCMi like ΔdblGATA1 mice, and eosinophil-specific IL-4 deletion resulted in improved heart function. In conclusion, eosinophils drive progression of myocarditis to DCMi, cause severe DCMi when present in large numbers, and mediate this process through IL-4.


Subject(s)
Cardiomyopathy, Dilated/etiology , Eosinophils/physiology , Interleukin-4/physiology , Myocarditis/complications , Animals , Autoimmune Diseases/complications , Disease Progression , Fibrosis , Humans , Interferon-gamma/physiology , Interleukin-13/physiology , Interleukin-17/physiology , Interleukin-5/physiology , Mice , Mice, Inbred BALB C
6.
Am J Respir Crit Care Med ; 193(1): 31-42, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26378386

ABSTRACT

RATIONALE: Group 2 innate lymphoid cells (ILC2s) robustly produce IL-5 and IL-13, cytokines central to the asthma phenotype; however, the effect of prostaglandin (PG) I2 on ILC2 function is unknown. OBJECTIVES: To determine the effect of PGI2 on mouse and human ILC2 cytokine expression in vitro and the effect of endogenous PGI2 and the PGI2 analog cicaprost on lung ILC2s in vivo. METHODS: Flow-sorted bone marrow ILC2s of wild-type (WT) and PGI2 receptor-deficient (IP(-/-)) mice were cultured with IL-33 and treated with the PGI2 analog cicaprost. WT and IP(-/-) mice were challenged intranasally with Alternaria alternata extract for 4 consecutive days to induce ILC2 responses, and these were quantified. Prior to A. alternata extract, challenged WT mice were treated with cicaprost. Human flow-sorted peripheral blood ILC2s were cultured with IL-33 and IL-2 and treated with the PGI2 analog cicaprost. MEASUREMENT AND MAIN RESULTS: We demonstrate that PGI2 inhibits IL-5 and IL-13 protein expression by IL-33-stimulated ILC2s purified from mouse bone marrow in a manner that was dependent on signaling through the PGI2 receptor IP. In a mouse model of 4 consecutive days of airway challenge with an extract of A. alternata, a fungal aeroallergen associated with severe asthma exacerbations, endogenous PGI2 signaling significantly inhibited lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s, as well as the mean fluorescence intensity of IL-5 and IL-13 staining. In addition, exogenous administration of a PGI2 analog inhibited Alternaria extract-induced lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s and the mean fluorescence intensity of IL-5 and IL-13 staining. Finally, a PGI2 analog inhibited IL-5 and IL-13 expression by human ILC2s that were stimulated with IL-2 and IL-33. CONCLUSIONS: These results suggest that PGI2 may be a potential therapy to reduce the ILC2 response to protease-containing aeroallergens, such as Alternaria.


Subject(s)
Epoprostenol/physiology , Lymphocytes/physiology , Signal Transduction/physiology , Alternaria/immunology , Animals , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Humans , In Vitro Techniques , Interleukin-13/physiology , Interleukin-33/pharmacology , Interleukin-5/physiology , Lung/cytology , Lung/immunology , Lymphocytes/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction/drug effects
7.
Cancer Res ; 75(8): 1624-1634, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25691457

ABSTRACT

Although the lung is the most common metastatic site for cancer cells, biologic mechanisms regulating lung metastasis are not fully understood. Using heterotopic and intravenous injection models of lung metastasis in mice, we found that IL5, a cytokine involved in allergic and infectious diseases, facilitates metastatic colonization through recruitment of sentinel eosinophils and regulation of other inflammatory/immune cells in the microenvironment of the distal lung. Genetic IL5 deficiency offered marked protection of the lungs from metastasis of different types of tumor cells, including lung cancer, melanoma, and colon cancer. IL5 neutralization protected subjects from metastasis, whereas IL5 reconstitution or adoptive transfer of eosinophils into IL5-deficient mice exerted prometastatic effects. However, IL5 deficiency did not affect the growth of the primary tumor or the size of metastatic lesions. Mechanistic investigations revealed that eosinophils produce CCL22, which recruits regulatory T cells to the lungs. During early stages of metastasis, Treg created a protumorigenic microenvironment, potentially by suppressing IFNγ-producing natural killer cells and M1-polarized macrophages. Together, our results establish a network of allergic inflammatory circuitry that can be co-opted by metastatic cancer cells to facilitate lung colonization, suggesting interventions to target this pathway may offer therapeutic benefits to prevent or treat lung metastasis.


Subject(s)
Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Interleukin-5/physiology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Tumor Microenvironment/immunology , Animals , Carcinoma, Lewis Lung/genetics , Cell Line, Tumor , Eosinophils/pathology , Female , Lung/immunology , Lung/pathology , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , Tumor Escape/genetics , Tumor Microenvironment/genetics
8.
Am J Respir Crit Care Med ; 190(12): 1383-94, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25390970

ABSTRACT

RATIONALE: Cytokine receptors can be markers defining different T-cell subsets and considered as therapeutic targets. The association of IL-6 and IL-6 receptor α (IL-6Rα) with asthma was reported, suggesting their involvement in asthma. OBJECTIVES: To determine whether and how IL-6Rα defines a distinct effector memory (EM) CD8+ T-cell population in health and disease. METHODS: EM CD8+ T cells expressing IL-6Rα (IL-6Rα(high)) were identified in human peripheral blood and analyzed for function, gene, and transcription factor expression. The relationship of these cells with asthma was determined using blood and sputum. MEASUREMENTS AND MAIN RESULTS: A unique population of IL-6Rα(high) EM CD8+ T cells was found in peripheral blood. These cells that potently proliferated, survived, and produced high levels of the Th2-type cytokines IL-5 and IL-13 had increased levels of GATA3 and decreased levels of T-bet and Blimp-1 in comparison with other EM CD8+ T cells. In fact, GATA3 was required for IL-6Rα expression. Patients with asthma had an increased frequency of IL-6Rα(high) EM CD8+ T cells in peripheral blood compared with healthy control subjects. Also, IL-6Rα(high) EM CD8+ T cells exclusively produced IL-5 and IL-13 in response to asthma-associated respiratory syncytial virus and bacterial superantigens. CONCLUSIONS: Human IL-6Rα(high) EM CD8+ T cells is a unique cell subset that may serve as a reservoir for effector CD8+ T cells, particularly the ones producing Th2-type cytokines, and expand in asthma.


Subject(s)
Asthma/physiopathology , CD8-Positive T-Lymphocytes/physiology , Interleukin-13/physiology , Interleukin-5/physiology , Interleukin-6 Receptor alpha Subunit/physiology , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
9.
Am J Respir Crit Care Med ; 190(12): 1373-82, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25350863

ABSTRACT

RATIONALE: Rhinoviruses are the major cause of asthma exacerbations; however, its underlying mechanisms are poorly understood. We hypothesized that the epithelial cell-derived cytokine IL-33 plays a central role in exacerbation pathogenesis through augmentation of type 2 inflammation. OBJECTIVES: To assess whether rhinovirus induces a type 2 inflammatory response in asthma in vivo and to define a role for IL-33 in this pathway. METHODS: We used a human experimental model of rhinovirus infection and novel airway sampling techniques to measure IL-4, IL-5, IL-13, and IL-33 levels in the asthmatic and healthy airways during a rhinovirus infection. Additionally, we cultured human T cells and type 2 innate lymphoid cells (ILC2s) with the supernatants of rhinovirus-infected bronchial epithelial cells (BECs) to assess type 2 cytokine production in the presence or absence of IL-33 receptor blockade. MEASUREMENTS AND MAIN RESULTS: IL-4, IL-5, IL-13, and IL-33 are all induced by rhinovirus in the asthmatic airway in vivo and relate to exacerbation severity. Further, induction of IL-33 correlates with viral load and IL-5 and IL-13 levels. Rhinovirus infection of human primary BECs induced IL-33, and culture of human T cells and ILC2s with supernatants of rhinovirus-infected BECs strongly induced type 2 cytokines. This induction was entirely dependent on IL-33. CONCLUSIONS: IL-33 and type 2 cytokines are induced during a rhinovirus-induced asthma exacerbation in vivo. Virus-induced IL-33 and IL-33-responsive T cells and ILC2s are key mechanistic links between viral infection and exacerbation of asthma. IL-33 inhibition is a novel therapeutic approach for asthma exacerbations.


Subject(s)
Asthma/etiology , Inflammation/etiology , Interleukins/physiology , Picornaviridae Infections/complications , Adult , Asthma/physiopathology , Asthma/virology , Cells, Cultured , Female , Humans , Inflammation/physiopathology , Interleukin-13/physiology , Interleukin-33 , Interleukin-4/physiology , Interleukin-5/physiology , Lymphocyte Subsets/physiology , Male , Picornaviridae Infections/physiopathology , Rhinovirus , Severity of Illness Index , T-Lymphocytes/physiology , Th2 Cells/physiology , Viral Load
10.
Am J Rhinol Allergy ; 28(5): 378-82, 2014.
Article in English | MEDLINE | ID: mdl-25198022

ABSTRACT

BACKGROUND: Sinus disease is commonly seen in patients with asthma, and several studies have been published describing the relationship between sinus disease and the inflammation seen in the sputum of asthmatic subjects. In this article, we expand on this knowledge by studying patients with eosinophilic bronchitis with and without asthma. METHODS: We describe the relationship between the severity of sinus disease determined by the Lund-Mackay score and sputum eosinophilia. Comparisons with blood eosinophil and total immunoglobulin (IgE) measurements are made. RESULTS: We have shown that the severity of sinus disease is positively correlated with sputum eosinophil counts, and the site of sinus disease affected the level of eosinophilia. There was a positive correlation between sputum eosinophils and blood eosinophils, but there was no relationship with blood total IgE levels. CONCLUSIONS: We have confirmed that there is a link between upper and lower airway inflammation and that this is not limited to patients with asthma. The process is associated with systemic inflammation as evidenced by increased blood eosinophils but appears to be independent of IgE.


Subject(s)
Bronchitis/immunology , Eosinophilia/immunology , Paranasal Sinus Diseases/immunology , Sputum/immunology , Asthma/immunology , Humans , Immunoglobulin E/blood , Interleukin-5/physiology , Retrospective Studies , Sputum/cytology , Tomography, X-Ray Computed
11.
Immunotherapy ; 6(3): 321-31, 2014.
Article in English | MEDLINE | ID: mdl-24762076

ABSTRACT

Eosinophilic esophagitis (EoE) is a clinical pathologic disease characterized by symptoms of esophageal dysfunction and eosinophilia of the esophagus. When the diagnosis is confirmed, it is important to treat the eosinophilic inflammation not only to control the presenting symptoms, but also to prevent acute and chronic complications. The pathogenesis of EoE is most likely a mixed IgE and non-IgE food-mediated reaction, where Th2 cytokines drive esophageal eosinophilia as in other atopic diseases. Hence, it is not surprising that therapy is based on inflammation control, with steroids (oral or topical) and/or food antigen avoidance. However, these treatment options are not specific, reduce the quality of life of patients and have significant side effects, therefore, there is an ongoing effort to design more specific immunotherapies. In this review, we review standard and immunotherapeutic options for EoE treatment, such as anti-IL-5, anti-TNFα, anti-IgE, anti-CRTH, oral allergy desensitization and environmental immunotherapy.


Subject(s)
Eosinophilic Esophagitis/therapy , Immunotherapy/methods , Adrenal Cortex Hormones/therapeutic use , Allergens/adverse effects , Allergens/therapeutic use , Animals , Antibodies, Anti-Idiotypic/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Combined Modality Therapy , Cytokines/genetics , Cytokines/immunology , Cytokines/physiology , Desensitization, Immunologic , Diagnosis, Differential , Eosinophilic Esophagitis/diagnosis , Eosinophilic Esophagitis/diet therapy , Eosinophilic Esophagitis/drug therapy , Eosinophilic Esophagitis/immunology , Food Hypersensitivity/immunology , Gastroesophageal Reflux/complications , Gastroesophageal Reflux/diagnosis , Humans , Immunoglobulin E/immunology , Immunosuppressive Agents/therapeutic use , Interleukin-5/antagonists & inhibitors , Interleukin-5/physiology , Mice , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/immunology , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Thymic Stromal Lymphopoietin
12.
Cytokine Growth Factor Rev ; 24(3): 189-201, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23535386

ABSTRACT

The GM-CSF, IL-3 and IL-5 family of cytokines, also known as the ßc family due to their receptors sharing the signalling subunit ßc, regulates multiple biological processes such as native and adaptive immunity, inflammation, normal and malignant hemopoieis, and autoimmunity. Australian scientists played a major role in the discovery and biological characterisation of the ßc cytokines and their recent work is revealing unique features of cytokine receptor assembly and signalling. Furthermore, specific antibodies have been generated to modulate their function. Characterisation of the structural and dynamic requirements for the activation of the ßc receptor family and the molecular definition of downstream signalling pathways are providing new insights into cytokine receptor signalling as well as new therapeutic opportunities.


Subject(s)
Cytokines/physiology , Signal Transduction/physiology , Animals , Autoimmunity/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Humans , Inflammation/physiopathology , Interleukin-3/physiology , Interleukin-5/physiology , Janus Kinases/metabolism , NF-kappa B/physiology , Receptors, Cytokine/physiology
13.
J Exp Med ; 210(3): 535-49, 2013 Mar 11.
Article in English | MEDLINE | ID: mdl-23420878

ABSTRACT

Eosinophils in visceral adipose tissue (VAT) have been implicated in metabolic homeostasis and the maintenance of alternatively activated macrophages (AAMs). The absence of eosinophils can lead to adiposity and systemic insulin resistance in experimental animals, but what maintains eosinophils in adipose tissue is unknown. We show that interleukin-5 (IL-5) deficiency profoundly impairs VAT eosinophil accumulation and results in increased adiposity and insulin resistance when animals are placed on a high-fat diet. Innate lymphoid type 2 cells (ILC2s) are resident in VAT and are the major source of IL-5 and IL-13, which promote the accumulation of eosinophils and AAM. Deletion of ILC2s causes significant reductions in VAT eosinophils and AAMs, and also impairs the expansion of VAT eosinophils after infection with Nippostrongylus brasiliensis, an intestinal parasite associated with increased adipose ILC2 cytokine production and enhanced insulin sensitivity. Further, IL-33, a cytokine previously shown to promote cytokine production by ILC2s, leads to rapid ILC2-dependent increases in VAT eosinophils and AAMs. Thus, ILC2s are resident in VAT and promote eosinophils and AAM implicated in metabolic homeostasis, and this axis is enhanced during Th2-associated immune stimulation.


Subject(s)
Eosinophils/physiology , Intra-Abdominal Fat/cytology , Macrophages/physiology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Helminthiasis/immunology , Immunity, Innate , Insulin Resistance , Interleukin-13/physiology , Interleukin-33 , Interleukin-5/physiology , Interleukins/pharmacology , Intestinal Diseases, Parasitic/immunology , Lymphocyte Activation , Macrophage Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
14.
Oncol Rep ; 28(3): 1084-90, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22710862

ABSTRACT

Interleukin-5 (IL-5) plays an important role in the growth and differentiation of human B cells and eosinophils. However, little is known about the effect of IL-5 on cancer cells. In this study, we investigated the molecular mechanisms involved in the IL-5-induced migration of HT1376 bladder cancer cells. Our results indicated that IL-5 significantly enhanced migration and MMP-9 expression in HT1376 cells. We also found that IL-5 induces transcriptional activation of the binding of NF-κB and AP-1, which are two important nuclear transcription factors that are linked to MMP-9 expression in HT1376 cells. In subsequent experiments, we found activation of ERK1/2 in IL-5-treated HT1376 cells. To examine the involvement of the ERK1/2 signaling pathway on IL-5-induced cell responses, we pretreated HT1376 cells with the ERK1/2 inhibitor U0126 followed by IL-5 treatment. The results showed that U0126 treatment inhibited migration of IL-5-treated HT1376 cells. Moreover, IL-5-stimulated MMP-9 expression was suppressed by the addition of U0126. Inhibition of ERK1/2 function consistently rescued transcriptional activity of NF-κB, without altering AP-1 activation, in IL-5-treated cells. Finally, inhibition of the IL-5-specific receptor IL-5Rα by small interfering RNA (siRNA) suppressed migration, ERK1/2 activation, MMP-9 expression and binding activation of NF-κB in IL-5-treated HT1376 cells. The results of the present study indicate that the IL-28A/IL-28AR1 dyad induces cell migration through ERK1/2-mediated expression of MMP-9 by binding activation of NF-κB in bladder cancer cells. In conclusion, these novel findings indicate that binding of IL-5 to IL-5Rα plays a critical role in MMP-9 expression, which may be involved in the migration of bladder cancer.


Subject(s)
Cell Movement , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-5/physiology , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , 5' Flanking Region , Butadienes/pharmacology , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Gene Expression , Gene Knockdown Techniques , Humans , Interleukin-5/genetics , Interleukin-5/metabolism , Interleukin-5 Receptor alpha Subunit/genetics , Interleukin-5 Receptor alpha Subunit/metabolism , MAP Kinase Signaling System , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Nitriles/pharmacology , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , RNA Interference , Transcription Factor AP-1/metabolism
15.
PLoS One ; 7(4): e34912, 2012.
Article in English | MEDLINE | ID: mdl-22523564

ABSTRACT

The Fip1-like1 (FIP1L1)-platelet-derived growth factor receptor alpha fusion gene (F/P) arising in the pluripotent hematopoietic stem cell (HSC),causes 14% to 60% of patients with hypereosinophilia syndrome (HES). These patients, classified as having F/P (+) chronic eosinophilic leukemia (CEL), present with clonal eosinophilia and display a more aggressive disease phenotype than patients with F/P (-) HES patients. The mechanisms underlying predominant eosinophil lineage targeting and the cytotoxicity of eosinophils in this leukemia remain unclear. Given that the Janus tyrosine kinase (JAK)/signal transducers and activators of transcription (Stat) signaling pathway is key to cytokine receptor-mediated eosinophil development and activated Stat3 and Stat5 regulate the expression of genes involved in F/P malignant transformation, we investigated whether and how JAK proteins were involved in the pathogenesis of F/P-induced CEL. F/P activation of JAK2, Stat3 and Stat5, were confirmed in all the 11 F/P (+) CEL patients examined. In vitro inhibition of JAK2 in EOL-1, primary F/P(+) CEL cells (PC) and T674I F/P Imatinib resistant cells(IR) by either JAK2-specific short interfering RNA (siRNA) or the tryphostin derivative AG490(AG490), significantly reduced cellular proliferation and induced cellular apoptosis. The F/P can enhance the IL-5-induced JAK2 activation, and further results indicated that JAK2 inhibition blocked IL-5-induced cellular migration and activation of the EOL-1 and PC cells in vitro. F/P-stimulation of the JAK2 suppressed cells led to a significantly reduction in Stat3 activation, but relatively normal induction of Stat5 activation. Interestingly, JAK2 inhibition also reduced PI3K, Akt and NF-κB activity in a dose-dependent manner, and suppressed expression levels of c-Myc and Survivin. These results strongly suggest that JAK2 is activated by F/P and is required for F/P stimulation of cellular proliferation and infiltration, possibly through induction of c-Myc and Survivin expression via activation of multiple signaling pathways, including NF-κB, Stat3, and PI3K/Akt.


Subject(s)
Eosinophils/physiology , Hypereosinophilic Syndrome/genetics , Janus Kinase 2/metabolism , Oncogene Proteins, Fusion/physiology , Receptor, Platelet-Derived Growth Factor alpha/physiology , mRNA Cleavage and Polyadenylation Factors/physiology , Benzamides , Down-Regulation , Enzyme Activation , Hematopoietic Stem Cells/metabolism , Humans , Hypereosinophilic Syndrome/enzymology , Hypereosinophilic Syndrome/physiopathology , Imatinib Mesylate , Interleukin-5/physiology , Janus Kinase 2/antagonists & inhibitors , NF-kappa B/metabolism , Piperazines/therapeutic use , Pyrimidines/therapeutic use , RNA, Small Interfering/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Tumor Cells, Cultured , Tyrphostins/pharmacology
16.
J Immunol ; 188(2): 703-13, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22174445

ABSTRACT

IL-5 is involved in a number of immune responses such as helminth infection and allergy. IL-5 also plays roles in innate immunity by maintaining B-1 B cells and mucosal IgA production. However, the identity of IL-5-producing cells has not been unambiguously characterized. In this report, we describe the generation of an IL-5 reporter mouse and identify IL-5-producing non-T lymphoid cells that reside in the intestine, peritoneal cavity, and lungs in naive mice. They share many characteristics with natural helper cells, nuocytes, and Ih2 cells, including surface Ags and responsiveness to cytokines. However, these phenotypes do not completely overlap with any particular one of these cell types. Innate non-T IL-5-producing cells localized most abundantly in the lung and proliferated and upregulated IL-5 production in response to IL-25 and IL-33. IL-33 was more effective than IL-25. These cells contribute to maintaining sufficient numbers of lung eosinophils and are important for eosinophil recruitment mediated by IL-25 and IL-33. Given that eosinophils are shown to possess antitumor activity, we studied lung tumor metastasis and showed that innate IL-5-producing cells were increased in response to tumor invasion, and their regulation of eosinophils is critical to suppress tumor metastasis. Genetic blockade or neutralization of IL-5 impaired eosinophil recruitment into the lung and resulted in increased tumor metastasis. Conversely, exogenous IL-5 treatment resulted in suppressed tumor metastasis and augmented eosinophil infiltration. These newly identified innate IL-5-producing cells thus play a role in tumor surveillance through lung eosinophils and may contribute to development of novel immunotherapies for cancer.


Subject(s)
Cell Movement/immunology , Eosinophils/immunology , Immunity, Innate , Interleukin-5/biosynthesis , Lung Neoplasms/immunology , Melanoma, Experimental/immunology , Tumor Escape/immunology , Animals , Cell Line, Tumor , Cells, Cultured , Eosinophils/pathology , Female , Gene Knock-In Techniques , Interleukin-5/physiology , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Male , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic
17.
Am J Respir Crit Care Med ; 184(10): 1125-32, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21852542

ABSTRACT

RATIONALE: Eosinophilic asthma is a phenotype of asthma characterized by the persistence of eosinophils in the airways. IL-5 is involved in the activation and survival of eosinophils. OBJECTIVES: To evaluate the effect of the antibody to IL-5, reslizumab, in patients with eosinophilic asthma that is poorly controlled with high-dose inhaled corticosteroid. METHODS: Patients were randomly assigned to receive infusions of reslizumab at 3.0 mg/kg (n = 53) or placebo (n = 53) at baseline and at Weeks 4, 8, and 12, with stratification by baseline Asthma Control Questionnaire (ACQ) score less than or equal to 2 or greater than 2. The primary efficacy measure was the difference between the reslizumab and placebo groups in the change in ACQ score from baseline to end of therapy (Week 15 or early withdrawal). MEASUREMENTS AND MAIN RESULTS: Mean changes from baseline to end of therapy in ACQ score were -0.7 in the reslizumab group and -0.3 in the placebo group (P = 0.054) and in FEV(1) were 0.18 and -0.08 L, respectively (P = 0.002). In those patients with nasal polyps, the changes in ACQ score were -1.0 and -0.1, respectively (P = 0.012). Median percentage reductions from baseline in sputum eosinophils were 95.4 and 38.7%, respectively (P = 0.007). Eight percent of patients in the reslizumab group and 19% of patients in the placebo group had an asthma exacerbation (P = 0.083). The most common adverse events with reslizumab were nasopharyngitis, fatigue, and pharyngolaryngeal pain. CONCLUSIONS: Patients receiving reslizumab showed significantly greater reductions in sputum eosinophils, improvements in airway function, and a trend toward greater asthma control than those receiving placebo. Reslizumab was generally well tolerated.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/drug therapy , Interleukin-5/antagonists & inhibitors , Pulmonary Eosinophilia/drug therapy , Adult , Asthma/physiopathology , Double-Blind Method , Eosinophils/drug effects , Female , Forced Expiratory Volume/drug effects , Humans , Interleukin-5/physiology , Leukocyte Count , Male , Middle Aged , Sputum/cytology , Surveys and Questionnaires , Treatment Outcome
18.
J Immunol ; 186(1): 516-26, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21106848

ABSTRACT

Human blood eosinophils exhibit a hyperactive phenotype in response to chemotactic factors after cell "priming" with IL-5 family cytokines. Earlier work has identified ERK1/2 as molecular markers for IL-5 priming, and in this article, we show that IL-3, a member of the IL-5 family, also augments fMLP-stimulated ERK1/2 phosphorylation in primary eosinophils. Besides ERK1/2, we also observed an enhancement of chemotactic factor-induced Akt phosphorylation after IL-5 priming of human blood eosinophils. Administration of a peptide antagonist that targets the Src family member Lyn before cytokine (IL-5/IL-3) priming of blood eosinophils inhibited the synergistic increase of fMLP-induced activation of Ras, ERK1/2 and Akt, as well as the release of the proinflammatory factor leukotriene C(4). In this study, we also examined a human eosinophil-like cell line HL-60 clone-15 and observed that these cells exhibited significant surface expression of IL-3Rs and GM-CSFRs, as well as ERK1/2 phosphorylation in response to the addition of IL-5 family cytokines or the chemotactic factors fMLP, CCL5, and CCL11. Consistent with the surface profile of IL-5 family receptors, HL-60 clone-15 recapitulated the enhanced fMLP-induced ERK1/2 phosphorylation observed in primary blood eosinophils after priming with IL-3/GM-CSF, and small interfering RNA-mediated knockdown of Lyn expression completely abolished the synergistic effects of IL-3 priming on fMLP-induced ERK1/2 phosphorylation. Altogether, our data demonstrate a central role for Lyn in the mechanisms of IL-5 family priming and suggest that Lyn contributes to the upregulation of the Ras-ERK1/2 and PI3K-Akt cascades, as well as the increased leukotriene C(4) release observed in response to fMLP in "primed" eosinophils.


Subject(s)
Eosinophils/immunology , Interleukin-3/physiology , Interleukin-5/physiology , Leukotriene C4/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , ras Proteins/physiology , src-Family Kinases/physiology , Adolescent , Adult , Amino Acid Sequence , Asthma/enzymology , Asthma/immunology , Asthma/metabolism , Eosinophils/metabolism , HL-60 Cells , Humans , Middle Aged , Molecular Sequence Data , Signal Transduction/immunology
19.
Neurobiol Dis ; 41(3): 717-24, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21168500

ABSTRACT

Mesenchymal stem cells (MSCs) are reported to possess immunomodulatory properties. Previous reports have demonstrated the beneficial effects of MSC-transplantation in focal cerebral ischemia animal models. In this study, we have investigated the neuroimmunomodulatory functions of human MSCs, transplanted in a rat focal ischemia model of transient middle cerebral artery occlusion (MCAO). Our results revealed that in a human mesenchymal stem cell line, B10 cell transplantation decreased the accumulation of Iba-1(+) microglia and GFAP(+) astrocytes, and inhibited proinflammatory gene expression in the core and ischemic border zone (IBZ). Among the proinflammatory genes iNOS, which was expressed in microglia/macrophage, was persistently inhibited up to 7days after MCAO. In vivo laser capture microdissection and double immunofluorescence staining, and in vitro B10 cell culture experiments showed that, in inflammatory conditions, B10 cells expressed cytokines and growth factors including IL-5, fractalkine, IGF-1, GDNF and VEGF. Fractalkine and IL-5 inhibited cytokine-induced proinflammatory gene expression including iNOS in a human microglia cell line. Thus, our results demonstrate that MSC transplantation suppresses MCAO focal ischemia-induced inflammation, possibly through expression of fractalkine and IL-5.


Subject(s)
Brain Ischemia/pathology , Brain Ischemia/prevention & control , Chemokine CX3CL1/physiology , Interleukin-5/physiology , Mesenchymal Stem Cell Transplantation/methods , Neurons/pathology , Animals , Brain Ischemia/metabolism , Cell Line , Cells, Cultured , Humans , Inflammation/pathology , Inflammation/prevention & control , Male , Neurons/metabolism , Neurons/physiology , Rats , Rats, Wistar
20.
Am J Pathol ; 177(6): 2850-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21037078

ABSTRACT

Substantial data show that infection with helminth parasites ameliorates colitis; however, oxazolone-induced colitis is exaggerated in mice infected with the tapeworm, Hymenolepis diminuta. We tested the hypothesis that the IL-5 response to helminth infection enhances the severity of oxazolone-induced colitis. Mice were infected with H. diminuta and 8 days later were treated with oxazolone ± anti-IL-5 antibodies. Colitis was assessed 72 hours postoxazolone treatment by disease activity scores, myeloperoxidase activity, and histopathology. Other mice received injections of a replication-deficient adenovirus that carried the IL-5 (Ad.IL-5) gene or a control adenovirus (Ad.delete) ± oxazolone. The effect of H. diminuta+oxazolone in CCL11/CCL22 (eotaxin-1 and 2) knockout (KO) mice was determined. Helminth infection and Ad.IL-5 treatment increased IL-5 and eosinophil numbers. In vivo neutralization of IL-5 significantly reduced the severity of colitis in H. diminuta+oxazolone-treated mice, and H. diminuta did not exaggerate oxazolone-induced colitis in CCL11/CCL22 KO mice. Mice receiving Ad.IL-5 only had no colitis, while oxazolone-induced colitis was more severe in animals cotreated with Ad.IL-5 (Ad.delete + oxazolone was not significantly different from oxazolone only). Thus, while there is much to be gleaned about antiinflammatory mechanisms from rodent-helminth model systems, these data illustrate the caveat that infection with helminth parasites as a therapy could be contraindicated in patients with eosinophilia or elevated IL-5 unless coupled to appropriate measures to block IL-5 and/or eosinophil activity.


Subject(s)
Colitis/complications , Disease Progression , Eosinophils/physiology , Hymenolepiasis/complications , Hymenolepis diminuta/physiology , Interleukin-5/physiology , Animals , Antibodies/therapeutic use , Chemokine CCL11/genetics , Chemokine CCL22/genetics , Colitis/chemically induced , Colitis/pathology , Colitis/therapy , Eosinophils/immunology , Helminths/physiology , Hymenolepiasis/immunology , Hymenolepiasis/pathology , Hymenolepiasis/therapy , Hymenolepis diminuta/immunology , Immunotherapy, Adoptive , Interleukin-5/genetics , Interleukin-5/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Oxazolone , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...