Interleukin 9 promotes influx and local maturation of eosinophils

Blood ◽  
2001 ◽  
Vol 97 (4) ◽  
pp. 1035-1042 ◽  
Author(s):  
Jamila Louahed ◽  
Yuhong Zhou ◽  
W. Lee Maloy ◽  
Pyapalli U. Rani ◽  
Christine Weiss ◽  
...  

Abstract The interleukin 9 (IL-9) pathway has recently been associated with the asthmatic phenotype including an eosinophilic tissue inflammation. The mechanism by which IL-9 affects eosinophils (eos) is not known. To investigate whether this cytokine has a direct activity on the development of eos and eosinophilic inflammation, a model of thioglycolate-induced peritoneal inflammation was used in IL-9 transgenic (TG5) and background strain (FVB) mice. In this model, a transient eosinophilic infiltration in the peritoneal cavity was observed in FVB mice 12 to 24 hours after thioglycolate injection that coincided with peak IL-5 and IL-9 release. In contrast, TG5 mice developed a massive eosinophilia that persisted at high levels (81% of total cells) even 72 hours after thioglycolate injection. Release of eosinophilic major basic protein (MBP), IL-4, and IL-5 to the peritoneal cavity of these mice was significantly increased when compared with the control FVB strain. To study the mechanism by which IL-9 exerts its effect on eos, bone marrow or peritoneal cells were cultured in the presence of IL-5, IL-9, or their combination in vitro. IL-5 alone was able to generate significant numbers of eos in TG5 but not FVB mice, whereas a combination of IL-5 and IL-9 induced marked eosinophilia in both strains indicating a synergism between these 2 cytokines. These data suggest that IL-9 may promote and sustain eosinophilic inflammation via IL-5–driven eos maturation of precursors.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 18-18
Author(s):  
Muithi Mwanthi ◽  
Gracie Michels ◽  
Karl Staser ◽  
Sarita Sehra ◽  
Michal Jander ◽  
...  

Abstract Abstract 18 Eosinophils are increasingly recognized as important myeloid effector cells in the inflammatory environment of many human diseases. Although eosinophils critically contribute to chronic asthmatic inflammation, few therapies directly target these cells. Eosinophils rapidly migrate to eotaxin elicited by allergic sensitization and challenge, a chemokine that ligates the CCR3 receptor. Eotaxin:CCR3 signaling critically regulates allergen-induced eosinophil infiltration in murine models by activating the Rho-family proteins. In several cell systems, the Rho proteins Rac and CDC42 activate p21-activated kinase 1 (PAK1), which we have previously shown to regulate F-actin dynamics and histamine release in the degranulating mast cell. In these studies, we examined eotaxin-induced eosinophil migration using genetic and hematopoietic ablation of Pak1 (Pak1−/−) in a murine asthma model. Using an in vitro transwell migration assay system, we evaluated the migration of bone marrow derived eosinophils of both genotypes to eotaxin (N=10). Pak1−/− eosinophils exhibited profoundly diminished eotaxin-induced chemotaxis in vitro relative to wild-type (Pak1+/+) eosinophils (p < 0.0001) with a 30% overall decrease in migrating Pak1−/− compared to Pak1+/+ eosinophils. Furthermore, we compared the eotaxin-induced localization and arrangement of F-actin in eosinophils of both genotypes by fluorescence cytometry and deconvolution confocal microscopy of fluorescently-tagged phalloidin in seeking to explain this migration defect. Preliminary findings suggest decreased F-actin polymerization in eotaxin-treated Pak1−/− eosinophils. In an independent line of experiments designed to compare eotaxin-mediated eosinophil recruitment in vivo we injected mice of both genotypes with an intraperitoneal dose of eotaxin or saline. Pak1+/+ mice showed an 8 fold eotaxin-mediated increase in eosinophil recruitment over control whereas Pak1−/− mice demonstrated only a modest 3–4 fold increase (p< 0.05). Finally we pursued PAK1's function in an experimental disease model in which the eosinophil's key role in pathogenesis is well documented. In 3 cohorts of 7 age, gender and strain matched Pak1+/+ and Pak1−/− ova albumin (OVA)-sensitized and challenged mice, we scored lung eosinophilic inflammation by histology and compared eosinophil counts and eotaxin concentrations in broncho-alveolar lavage fluid (BALF) by fluorescence cytometry and ELISA respectively. We also assessed OVA-specific T-cell subset cytokine secretion in our asthma mice by ELISA. Lung-parenchymal eosinophilic inflammation was diminished in Pak1−/− ova-sensitized mice versus Pak1+/+'s (p<0.01) with neither differences in BALF eotaxin content nor OVA-specific in vitro T-helper cell secretion of asthma-induced cytokines between the 2 genotypes. Based on our findings in this model, we assessed PAK1's hematopoietic role using two complementary chimeric mouse models. In a cohort of matched recipient Pak1+/+ mice we transplanted Pak1+/+ and Pak1−/− bone marrow and after hematopoietic reconstitution we incited asthmatic inflammation in these mice. Significantly, hosts transplanted with Pak1−/− bone marrow developed decreased eosinophilic inflammation scores compared to Pak1+/+ bone marrow recipients (p<0.05). To complement the bone marrow experiments, we transplanted left-lung grafts from Pak1+/+ and Pak1−/− mice into matched Pak1+/+ and Pak1−/− recipient mice and after surgical recuperation we elicited asthmatic inflammation as above. Similar to our bone marrow transplant experiments, irrespective of the genotype of the lung graft, hosts with Pak1−/− bone marrow developed decreased lung eosinophil infiltrate. Our data suggest that genetic PAK1 disruption hinders the in vitro and in vivo eotaxin-mediated migration of eosinophils by altering polymerization of F-actin. In an OVA murine model of asthma, we show that the genetic ablation of PAK1 attenuates the eosinophilic inflammation without affecting T-cell function. We similarly demonstrate that hematopoietic expression of PAK1 is critical to the development of eosinophil inflammation in two complementary transplant murine models. Pharmacologically targeting PAK1 may thus provide a specific way to impede eosinophil tissue infiltration, alleviate chronic eosinophil inflammation, and hamper long-term tissue remodeling in diseases like asthma. Disclosures: No relevant conflicts of interest to declare.


Allergy ◽  
2004 ◽  
Vol 59 (10) ◽  
pp. 1080-1086 ◽  
Author(s):  
A.-K. Johansson ◽  
M. Sjostrand ◽  
M. Tomaki ◽  
A.-M. Samulesson ◽  
J. Lotvall

Author(s):  
Margaret J. Hukee ◽  
Randa I. Abu-Ghazaleh ◽  
Franklyn G. Prendergast

The eosinophil major basic protein (MBP) has been localized in the crystalloid core of the specific eosinophil granule. High levels of MBP are present in sputa and at sites of epithelial damage in patients with brochial asthma suggesting that MBP is released from the eosinophil. In vitro, the toxicity of MBP to mammalian cells, helminths, protozoa, and bacteria, has been demonstrated. MBP also induced the degranulation of platelets, basophils, and mast cells. Since these events involve extracellular MBP and rupture of plasma membranes, the mechanism of toxicity may be due to a direct interaction between MBP and biological membranes. Fluorescence spectroscopy studies using synthetic lipid bilayers (liposomes) showed the ability of MBP to induce disorder, fusion and lysis of those membranes. In this report, structural evidence is presented for liposome aggregation and possible lysis in the presence of MBP.


1999 ◽  
Vol 6 (5) ◽  
pp. 453-457 ◽  
Author(s):  
Redwan Moqbel

Recent advances in the treatment and management of asthma have suggested that leukotriene (LT) receptor antagonists may be very beneficial as a second generation therapy with steroid-sparing properties and negligible side effects. These agents have shown interesting effects on peripheral blood and sputum eosinophils. A major contributor to the damage in the airway of asthmatic patients is the eosinophil, which, upon activation, releases a battery of granule-associated cytotoxic, cationic proteins, including the major basic protein and eosinophil peroxidase, and membrane-derived de novo-synthesized bioactive lipid mediators, including LTC4, LTD4and LTE4, as well as platelet activating factor. These products have deleterious effects on the airway tissue including mucosal and smooth muscle layers. Accumulating evidence suggests that these agents may also influence the accumulation and maintenance of eosinophilic responses at the site of inflammation. This article reviews the possible anti-inflammatory mode of action of these therapies. It also discusses where there may be a gap in the knowledge regarding the potential direct and indirect effects of LT modifiers on eosinophil function and recruitment.


2005 ◽  
Vol 289 (3) ◽  
pp. L413-L418 ◽  
Author(s):  
Toshiaki Homma ◽  
Jason H. T. Bates ◽  
Charles G. Irvin

Major basic protein and other native cationic proteins increase airway hyperresponsiveness when administered to the luminal surface of the airways in vitro. To determine whether the same applies in vivo, we assessed airway responsiveness in rats challenged with both aerosolized and intravenously infused methacholine. We partitioned total lung resistance into its airway and tissue components using the alveolar capsule technique. Neither poly-l-lysine nor major basic protein altered baseline mechanics or its dependence on positive end-expiratory pressures ranging from 1 to 13 cmH2O. When methacholine was administered to the lungs as an aerosol, both cationic proteins increased responsiveness as measured by airway resistance, tissue resistance, and tissue elastance. However, responsiveness of all three parameters was unchanged when the methacholine was infused. Together, these findings suggest that cationic proteins alter airway responsiveness in vivo by an effect that is apparently limited to the bronchial epithelium.


1998 ◽  
Vol 274 (6) ◽  
pp. L997-L1005 ◽  
Author(s):  
Mark E. Wylam ◽  
Nesli Gungor ◽  
Richard W. Mitchell ◽  
Jason G. Umans

Previous studies in vivo or in isolated airway preparations have suggested that eosinophil-derived polycationic proteins enhance airway smooth muscle tone in an epithelium-dependent manner. We assessed the direct effects of activated human eosinophil supernatant, major basic protein (MBP), and polycationic polypeptides on basal and agonist-stimulated intracellular Ca2+concentrations ([Ca2+]i) in cultured bovine tracheal smooth muscle (TSM) cells. A 1-h incubation of myocytes with activated eosinophil buffer resulted in a doubling of basal [Ca2+]iand increased responsivity to histamine compared with myocytes that were exposed to sham-activated eosinophil buffer. In addition, concentration-dependent acute transient increases and subsequent 1-h sustained elevations of basal [Ca2+]iwere observed immediately after addition of MBP and model polycationic proteins. Finally, both peak and plateau [Ca2+]iresponses to bradykinin addition were augmented significantly in cultured myocytes that had been exposed to low concentrations of MBP or model polycationic proteins but were inhibited at greater concentrations. This elevated [Ca2+]ito polycationic proteins was manifest in epithelium-denuded bovine TSM strips as concentration-dependent increased basal tone. We conclude that activated eosinophil supernatant, MBP, and other polycationic proteins have a direct effect on both basal and subsequent agonist-elicited Ca2+mobilization in cultured TSM cells; TSM strips in vitro demonstrated, respectively, augmented and diminished responses to the contractile agonist acetylcholine. It is possible that alteration in myocyte Ca2+mobilization induced by these substances may influence clinical states of altered airway tone, such as asthma.


1990 ◽  
Vol 86 (1) ◽  
pp. 52-63 ◽  
Author(s):  
Ken-ichi Hisamatsu ◽  
Tetsuya Ganbo ◽  
Tsutomu Nakazawa ◽  
Yoshihiko Murakami ◽  
Gerald J. Gleich ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document