Open Access

Crucial role of OX40/OX40L signaling in a murine model of asthma

  • Authors:
    • Wei Lei
    • Daxiong Zeng
    • Gaoqin Liu
    • Yehan Zhu
    • Jiajia Wang
    • Hongya Wu
    • Junhong Jiang
    • Jianan Huang
  • View Affiliations

  • Published online on: January 18, 2018     https://doi.org/10.3892/mmr.2018.8453
  • Pages: 4213-4220
  • Copyright: © Lei et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to explore the roles of OX40/OX40 ligand (OX40L) signaling and OX40+ T cells in ovalbumin (OVA)‑induced mouse asthma model. Asthma was induced by OVA exposure and subsequent co‑treatment with OX40L protein, neutralizing anti‑OX40L blocking antibody, OX40+ T cells or PBS. The protein expression levels of interleukin (IL)‑4, IL‑6, IL‑13, IL‑17, tumor necrosis factor (TNF)‑α and interferon (IFN)‑γ in bronchoalveolar lavage fluid (BALF) were examined using murine cytokine‑specific ELISA. Eosinophil accumulation as well as proliferation and apoptosis of T cells in BALF were detected by Cell Counting kit‑8 and flow cytometric assays. Expression of the apoptosis‑related protein cleaved caspase‑3 was examined in OX40+ T cells using western blot assay. Flow cytometric analysis revealed that OVA‑treated mice that were co‑treated with OX40L or OX40+ T cells exhibited higher eosinophil infiltration compared with control mice treated only with OVA, whereas neutralizing anti‑OX40L blocking antibody inhibited eosinophil infiltration. ELISA assays demonstrated that the expression of IL‑4, IL‑6, IL‑13, IL‑17, TNF‑α and IFN‑γ in BALF in OX40L‑treated and OX40+ T cell‑treated mice was increased compared with expression levels in control mice. Treatment with OX40L protein effectively reduced apoptosis of T cells and the expression of cleaved caspase‑3 in T cells. OX40L‑treated and OX40+ T cell‑treated mice exhibited increased asthma through OX40/OX40L signaling, which probably promoted inflammatory factor expression, eosinophil infiltration and T cell proliferation.

Introduction

Asthma is a complex disease that involves a number of genetic and environmental influences (1). The chronic inflammation in asthma is characterized by eosinophilic recruitment, airway hyperresponsiveness (AHR), goblet cell hyperplasia/metaplasia, epithelial hypertrophy/hyperplasia, mucus hypersecretion, collagen deposition, smooth muscle cell hypertrophy/hyperplasia and subepithelial fibrosis (2,3). Worldwide ~300 million people suffer from asthma, and this number is predicted to rise over the next 10–15 years to >400 million (4). It has been reported that chronic airway inflammatory processes lead to the recruitment of activated eosinophils and T helper 2 (Th2) lymphocytes to the site of injury and an improper immune response to common allergens (5). Recurrent inflammation and subsequent abnormalities in the tissue repair mechanisms may lead to structural alterations in the airway wall that may develop the clinically detectable features of epithelial injury, goblet cell hyperplasia, subepithelial thickening, airway hyperplasia and angiogenesis (6). Inflammatory infiltrates in the airways that are characteristic of asthma may affect the structural cells and lead to AHR (7). There are a number of different immune cells in the infiltrates, including T lymphocytes that produce cytokines, such as interleukin (IL)-4, IL-5 and IL-13, which serve important roles in the pathogenesis of asthma (810). Therefore, targeting these T lymphocytes may have the potential to effectively treat asthma.

OX40 (also known as CD134) and its binding partner OX40 ligand (OX40L; also known as CD252) are members of the tumor necrosis factor (TNF)/TNF receptor superfamily and are expressed on activated CD4 and CD8 T cells, and on a number of lymphoid and non-lymphoid cells (11). OX40L is mainly expressed by antigen-presenting cells (APCs), such as dendritic cells, but is also expressed by B cells, macrophages and Langerhans cells (12). Dendritic cells in the airway express OX40L in response to epithelial cell-derived thymic stromal lymphopoietin stimulation (TSLP) (13). In vivo studies using murine and nonhuman primate models of asthma have reported that the inhibition of OX40L suppressed TSLP-mediated Th2 inflammation and reduced the number of OX40L+ dendritic cells in the lungs (14). OX40/OX40L interactions have been demonstrated to serve a central role in numerous inflammatory and autoimmune disease development, which suggested that they may be suitable candidates for clinical intervention (15); however, the effects and precise mechanisms of OX40/OX40L signaling in the development of asthma remains unclear. Clarification of the underlying mechanisms of the OX40/OX40L signaling in mediating inflammation, immunoreactions or other cell functions in asthma may lead to improved clinical treatment on asthma.

The present study examined the effects of OX40/OX40L signaling on inflammation and T cell functions in a mouse asthma model and investigated the possible underlying mechanisms. The aim was to provide a new perspective and deeper understanding of the etiology of asthma and to provide additional evidence for the potential involvement of OX40/OX40L signaling in the development of asthma.

Materials and methods

Reagents and antibodies

Murine interleukin (IL-) 4 (catalog no. BMS613), IL-6 (catalog no. BMS603-2), IL-13 (catalog no. KMC2221), IL-17 (catalog no. BMS6001), tumor necrosis factor (TNF-)α (catalog no. BMS607-3) and interferon (IFN-)γ (catalog no. 88-8314-77) ELISA kits were purchased from Invitrogen (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Ovalbumin (OVA) was purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Neutralizing rat anti-OX40L monoclonal antibody was purchased from Bio X Cell (West Lebanon, NH, USA; catalog no. BE0033-1-25MG). Mouse recombinant OX40L protein was purchased from R&D Systems, Inc. (Minneapolis, MN, USA; catalog no. 1236-OX-025). Rabbit anti-cleaved caspase 3 (Asp175), polyclonal antibody was purchased from Abbexa, Ltd. (Cambridge, UK; catalog no. abx015533). Rabbit anti-NF-κB polyclonal antibody (Aviva Systems Biology, San Diego, CA, USA; catalog no. OAAI00072; phosphorylated (p-)Ser337). Anti-GAPDH antibody was purchased from Beyotime Institute of Biotechnology (Shanghai, China; catalog no. AF0006). Cell Counting Kit-8 (CCK-8) was purchased from Dojindo Molecular Technologies, Inc. (Kumamoto, Japan). Dead Cell Apoptosis kit with Annexin V Alexa Fluor 488 & propidium iodide (PI) was purchased from Thermo Fisher Scientific, Inc. (catalog no. V13241). Fluorescein isothiocyanate-conjugated rat anti-CD4 monoclonal antibody was purchased from LifeSpan BioSciences, Inc. (Seattle, WA, USA; catalog no. LS-C62734-300). Phycoerythrin (PE-)conjugated goat anti-OX40 polyclonal antibody was purchased from R&D Systems, Inc. (catalog no. FAB1256P).

Experimental animals

Specific-pathogen-free female BALB/c mice (n=156; age, 6–8 weeks; weight, 20–25 g) were obtained from Shanghai SLAC Laboratory Animal Co. Ltd. (Shanghai, China), and were kept at 19–22°C and 40–75% relative humidity at all times in the animal facility under specific-pathogen-free conditions. A 12-h light/dark cycle was maintained during the course of the present study. Animals were kept in groups of five and fed regular lab chow and water ad libitum. All animal experiments performed in this study conformed to the Guide for the Care and Use of Laboratory Animals (16) and were approved by the Institutional Animal Care and Use Committee of Soochow University (Suzhou, China).

OX40+ T cell sorting protocol

Murine CD4+ T cells were obtained from mononuclear cells prepared from the bronchoalveolar lavage fluid (BALF) of 18 OVA-challenged mice or the spleen of 6 BALB/c mice and T cells were isolated using a Pan T Cell Isolation Kit II (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany; catalog no. 130-095-130) according to the manufacturer's protocol (17) and collected in sterile PBS containing 50% fetal calf serum (FCS). The purified T cells (106 cells/ml) were then cultured in DMEM containing 10% FCS in a 6-well plate with plate-bound anti-CD3 antibody (3 µg/ml; R&D Systems, Inc.; catalog no. MAB4841-SP) and soluble anti-CD28 antibody (10 µg/ml; R&D Systems, Inc.; catalog no. MAB4832-SP), as well as IL-2 (5 ng/ml; R&D Systems, Inc.; catalog no. P04351), IL-4 (20 ng/ml; R&D Systems, Inc.; catalog no. P07750), anti-IFN-γ antibody (10 µg/ml; R&D Systems, Inc.; catalog no. MAB485-SP) and anti-IL-12 antibody (10 µg/ml; R&D Systems, Inc.; catalog no. AF-419-SP) for 3 days at 37°C. Cells were subsequently removed, washed and cultured with DMEM containing 10% FCS for a further 3-6 days at 37°C without further stimulation (18). T cells were subsequently processed for the sorting assay with phycoethryin-conjugated OX40 antibodies (10 µl/106 cells; R&D Systems, Inc.; catalog no. FAB1256P) using flow cytometry to isolate OX40+ T cells for further in vivo experiments. Cells were analyzed with FlowJo software (version 7.6; FlowJo LLC, Ashland, OR, USA).

Immunization and intervention

Protocols for immunization and intervention were as previously described (19). Briefly, 120 mice were immunized with an intraperitoneal injection of OVA (100 µg; Sigma-Aldrich; Merck KGaA) and aluminum hydroxide (2 mg; Pierce; Thermo Fisher Scientific, Inc.) in sterile saline on days 1 and 8. On days 9–14 following the initial sensitization, mice were challenged intranasally with 20 µg of 2% OVA in sterile saline. In other experiments for intervention, 100 µg/kg of recombinant murine OX40L protein, 12 mg/kg of neutralizing anti-OX40L antibody or 5×106 isolated OX40+ T cells or PBS (control) were injected intravenously through the tail vein following anesthesia (20,21).

Cytokine and protein measurements in bronchoalveolar lavage fluid (BALF)

BALF was collected by flushing 1 ml ice-cold PBS back and forth three times through a tracheal cannula followed by centrifugation at 2,000 × g at 4°C for 10 min, as previously described (22). Protein concentrations of IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ in the supernatant were measured using murine cytokine-specific ELISA kits (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The concentration of IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ in the BALF were measured as markers of inflammatory reaction using a microplate reader at 450 nm (Thermo Fisher Scientific, Inc.; catalog no. 51119000).

Determination of eosinophils

Eosinophils were detected and counted as previously described (23). Briefly, BALF was performed by instilling 0.9% NaCl containing 0.6 mmol/l EDTA in two separate 0.5 ml aliquots. The fluid was recovered by gentle suction and placed on ice for immediate processing. An aliquot of ~0.5 ml BALF was processed immediately for eosinophil count. BALF cell suspensions were stained with PE-conjugated rat anti-mouse CCR3 at 4°C for 30 min (1:500; R&D Systems, Inc.; catalog no. FAB1551T-100UG), followed by staining with fluorescein isothiocyanate-conjugated anti-mouse CD16 (1:500; Antigenix America Inc., Huntington Station, NY, USA; catalog no. RM160323) for 30 min at 4°C in the dark. Samples stained with non-immunized rat IgG mAb (1:100; R&D Systems, Inc.; catalog no. IC005P) for 30 min at 4°C were used as an isotype control. The percentage of eosinophils (CCR3+CD16) was calculated as the number of eosinophils divided by the total number of cells in the BALF sample, as determined by flow cytometry and were analyzed with FlowJo software (version 7.6). All analyses were performed in a blinded fashion. The remainder of the lavage fluid was centrifuged at 1,000 × g for 10 min at 4°C and the supernatant was removed aseptically and stored in individual aliquots at −80°C.

Western blot analysis

Cell pellets (1×107) from fresh BALF collected from lung tissues was homogenized in 600 ml radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology; catalog no. P0013B) in the presence of protease inhibitors for 30 min at 4°C. Protein concentration was determined with the Bradford assay. Lysates were centrifuged at 9,000 × g for 15 min at 4°C. Samples (20 µg/lane) were boiled for 5 min at 100°C and separated by 12% SDS-PAGE under denaturing conditions and electroblotted to a polyvinylidene difluoride membrane (Bio-Rad Laboratories, Inc., Hercules, CA). Membranes were blocked with PBS and 5% non-fat dry milk for 12 h at 4°C and incubated with the following antibodies for 1 h at room temperature: Anti-NF-κB (1:500; Aviva Systems Biology; catalog no. OAAI00072; p-Ser337) and anti-cleaved caspase 3 (1:500; Abbexa Ltd., Cambridge, UK; catalog no. abx015533). Immunoblot assays were then washed and incubated with a horseradish peroxidase-labeled secondary antibody (1:5,000 dilution; GE Healthcare Life Sciences, Shanghai, China; catalog nos. RPN4301 and RPN4201) for 1 h at room temperature. Protein bands were visualized using ECL Plus Enhanced Chemiluminescence Reagent (GE Healthcare Life Sciences), according to the manufacturer's protocol. Protein band intensity was determined relative to GAPDH using ImageJ software (version 2.1.4.7; National Institutes of Health, Bethesda, MD, USA).

Cell proliferation assay in vitro

The CCK-8 assay was used to evaluate proliferation of isolated T cells. T cells were exposed to increasing concentrations of OX40L protein (10, 50, 100 or 200 ng/ml) or 200 ng/ml of OX40L protein combined with 200 ng/ml of neutralizing anti-OX40L antibody. Briefly, T cells (3×103 cells/well) were seeded in each well of a 96-well plate and stimulated with 0.10 mg/ml phytohemagglutinin (Thermo Fisher Scientific, Inc.; catalog no. 00-4977-93) for 24 h. Subsequently, the cells were cultured in serum-free DMEM for 24 h starvation at 37°C. Cells were treated with different concentrations of OX40L protein (10–200 ng/ml) or 200 ng/ml OX40L protein and 200 ng/ml neutralizing anti-OX40L antibody (200 ng/ml) for 24 h at 37°C, and the proliferative activity was determined by CCK-8 assay, according to the manufacturer's protocol. CCK-8 (10 µl) was added to each well followed by incubation for an additional 2–4 h at 37°C. When the media changed from red to yellow, the absorbance value was detected at a wavelength of 450 nm using a microplate reader (Thermo Fisher Scientific, Inc.; catalog no. 51119000). The experiment was performed at least three times.

Flow cytometric analysis of cell apoptosis

Mononuclear T cells were isolated from BALF according to a previously described procedure with some modifications (24). Briefly, T cells (1×105 cells/test) were washed twice with PBS and centrifuged at 1,000 × g for 5 min at room temperature. Each pellet (~1×105 cells) of the T cells was then re-suspended in 400 µl PBS followed by incubation with 5 µl Annexin V and 1 µl propidium iodide (PI; 1 mg/ml) for 15 min at room temperature. Cells were subsequently analyzed using a flow cytometer without washing the cells. Cells that were PI and Annexin V negative were considered healthy cells, PI negative and Annexin V positive were considered apoptotic and cells that were PI and Annexin V positive were considered necrotic.

Statistical analysis

Data are presented as the mean ± standard error of the mean. Statistical analyses were performed using SPSS 18.0 software package (SPSS Inc., Chicago, IL, USA). Comparisons among multiple groups were performed using one-way analysis of variance followed by the Bonferroni post-hoc test if the data were normally distributed. P<0.05 was considered to indicate a statistically significant difference.

Results

OX40/OX40L signaling effects on eosinophil recruitment in lungs following sensitization and challenge with OVA

The presence of eosinophils in the BALF and lungs is an important indicator of airway inflammation (25). OVA challenged mice had a significantly higher eosinophil count (56±6.8%) compared with PBS-treated control mice (4.4±3.6%; P<0.01); OX40L protein-treated mice and OX40+ T cell-treated mice also had a high eosinophil count (65±5.5 and 67±7.2%, respectively) in the BALF following sensitization and challenge with OVA (Fig. 1), compared with those treated with OVA alone. By contrast, eosinophils accounted for only 41±4.5% of all cells in the BALF of mice treated with neutralizing anti-OX40L antibody (Fig. 1). These results suggested that OX40/OX40L signaling may serve an important role in eosinophil recruitment into the lungs.

Effects of OX40/OX40L signaling on cytokine expression levels in vivo

Mice challenged with OVA exhibited a significant increase in IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ expression levels in BALF, compared with the levels in untreated mice (P<0.05; Fig. 2). In mice sensitized and challenged with OVA that were co-treated with neutralizing anti-mouse OX40L antibody, this increase in IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ cytokine levels was significantly suppressed (P<0.05 or P<0.01 vs. OVA; Fig. 2); whereas mice co-treated with OX40L protein or OX40+ T cells exhibited a significant increase in these cytokine levels in the BALF (P<0.05 or P<0.01; Fig. 2), compared with the levels the OVA group. These results suggested that OX40/OX40L signaling may significantly increase the IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ cytokine levels in the BALF.

NF-κB activation promotes inflammatory cell migration and proliferation, and mediates inflammatory factor secretion (26). To determine whether OX40/OX40L signaling is able to effect NF-κB activation, the expression of activated NF-κB was detected in inflammatory cells from the BALF. Activated NF-κB expression was significantly increased in the OVA-challenged group compared with the PBS group (P<0.01). Furthermore, pNF-κB expression was significantly increased in OX40L protein or OX40+ T cell-treated mice and significantly reduced in anti-OX40L antibody-treated mice compared with mice in the OVA group (Fig. 3). These results indicated that OX40/OX40L signaling may induce NF-κB activation and suggests that OX40/OX40L signaling had a pro-inflammatory effect by regulating the expression of IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ through NF-κB activation.

Effects of OX40/OX40L signaling on proliferation and polarization of CD4+ T cells in vitro

It has been previously reported that OX40L promoted the number of OX40+ CD4 T cells in an asthma model (27), which indicated that OX40/OX40L signaling may serve an important role in the process of CD4+ T cell polarization, which may eventually affect the progression of asthma. The present study examined the effects of OX40/OX40L signaling on CD4 T cell proliferation and polarization in vitro. CD4+ T cells isolated from mouse spleen cultured with 100 or 200 ng/ml OX40L protein exhibited a significant increase in proliferation, whereas no change in proliferation was detected between the OX40L protein and neutralizing anti-OX40L antibody co-culture group and the PBS-treated control group (Fig. 4A). Furthermore, OX40L treatment significantly promoted the number of OX40+ T cells, whereas co-treatment with neutralizing anti-OX40L antibody suppresses OX40+ T cell number (Fig. 4B-D). These data indicated that OX40/OX40L signaling may be involved in CD4+ T cell proliferation and polarization, and thus influences T cell bio-function in asthma pathology.

OX40/OX40L signaling effects on T cell apoptosis in lungs following sensitization and challenge with OVA

To further explore the mechanism underlying OX40/OX40L signaling involved in asthma process, the expression levels of apoptosis-related protein cleaved caspase-3 were examined in isolated T cells from PBS-treated, OX40L protein-treated or neutralizing anti-OX40L antibody-treated mice. Western blot analysis demonstrated that the relative protein expression level of cleaved-caspase-3 was significantly increased in anti-OX40L antibody-treated mice (0.87±0.11) and decreased in OX40L protein-treated mice (0.36±0.18) compared with PBS-treated mice (0.51±0.17; P<0.05 Fig. 5A and B). Similar results were obtained by flow cytometric analyses for apoptosis (Fig. 5C and D).

Discussion

OX40/OX40L signaling serves a key role in the development, differentiation and physiological functions of T cells and other immunological cells (28). The expression of OX40 is upregulated on activated T cells (29), whereas it is constitutively expressed on T regulatory cells (30,31). This difference in expression is consistent with the previously hypothesized role of OX40/OX40L interactions in the propagation of the immune response and in initial T cell priming (32). OX40L is predominantly expressed in APCs, such as B cells, macrophages, microglia, dendritic cells and endothelial cells (3339). Signaling through OX40/OX40L interactions during effector T cell responses has been previously reported to enhance T cell survival (40,41), cytokine production (42) and increase the number of memory CD4+ T cells (43). Experimental models of autoimmunity and inflammation have indicated the potential role for OX40/OX40L, as inhibiting the interaction between OX40 and OX40L attenuates disease progression or severity (44,45). Owing to this important role in the pathological process of numerous diseases, the present study examined the effects and the mechanisms of OX40/OX40L signaling in experimentally induced asthma.

A number of previous in vivo studies (15,4648) have demonstrated a pathogenic role for OX40/OX40L signaling in autoimmune diseases, and the disruption of this axis was reported to be beneficial for prevention and treatment (49). However, whether OX40/OX40L serves pathogenic roles in human asthma requires further exploration. Our previous study demonstrated that OX40L was overexpressed by myeloid APCs in peripheral blood and BALF in a mouse asthma model (27). Another previous study reported that the number of OX40L-expressing myeloid APCs was positively correlated with disease activity, as assessed by intrafluid expression of inflammatory factors (50). In the present study, the results indicated that OX40/OX40L signaling promoted CD4+ T cell proliferation and polarization into CD4+OX40+ T cells, as evidenced by the significantly increased proportion of CD4+OX40+ T cells present when OX40L was administered to isolated T cells from experimental mice in vitro. Conversely, CD4+ T cell proliferation was suppressed in cells co-treated with the neutralizing anti-mouse OX40L antibody. These observations suggest that OX40/OX40L interactions may have activated CD4+OX40+ T cells and this event may represent a crucial component for affecting the progression of experimental asthma (51).

Although OX40/OX40L signaling inhibition was demonstrated to reduce inflammation, as determined by its effect on IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ expression in peripheral blood and bronchoalveolar lavage fluid, there are still many signal transduction processes that may be involved in the pathogenesis of asthma progression, which makes these types of investigations very complex. Previous studies have focused on NF-κB signaling, which is necessary for the proliferation and migration of cells, the expression of multiple cytokines and the inflammatory response (52). The present study examined activated NF-κB expression in asthma lung tissues by western blot analysis and demonstrated that activated NF-κB expression was elevated in OX40L protein- and CD4+OX40+ T cell-treated groups, and it was decreased in anti-OX40L antibody treated group, suggesting that OX40/OX40L interactions may affect asthma progression through the NF-κB pathway. Song et al (53) reported that the activation of NF-κB1 by OX40 contributes to antigen-driven T cell expansion and survival. Burrows et al (54) demonstrated that OX40 blockade inhibits house dust mite-driven allergic lung inflammation in mice and allergic responses in humans in vitro. The effects of OX40/OX40L on T cell function and the inflammatory response through the NF-κB pathway demonstrated in these previous reports is consistent with the results of the present study.

Inflammatory responses are important factors for asthma-induced lung injury (5557). During the acute pathological process, inflammatory cells (including neutrophils and macrophages) are recruited into lung lesions, where they are induced to secret pro-inflammatory cytokines to further accelerate inflammatory responses and promote asthma (58). Therefore, effective blocking or inhibiting of the inflammatory responses may be a fundamental treatment strategy for asthma. OX40/OX40L interaction is a promoting factor in pathological processes of asthma (59). Consistently, OX40/OX40L interactions in the present study were also demonstrated to promote inflammatory cell infiltration and facilitated pro-inflammatory cytokine expression. A clear association has been demonstrated between an experimental model of asthma and the regulation of the inflammatory response by OX40/OX40L (60,61).

In conclusion, although it requires further exploration, OX40/OX40L signaling may be a prospective intervention target for inhibiting CD4+OX40+ T cell activation, inflammatory factor secretion and inflammatory cell infiltration into BALF in clinical settings of asthma therapy. Results from the present study revealed that the neutralizing anti-mouse OX40L antibody was an efficient inhibitor of CD4+OX40+ T cell production in OVA-induced asthma, and this inhibiting effect may further impact IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ expression, reduce inflammatory responses and thus alleviate asthma progression. Targeting of the OX40/OX40L axis may provide therapeutic effects in the treatment of asthma.

Acknowledgements

This study was supported by The National Natural Science Foundation of China (grant nos. 81300026 and 31600736), The Science and Education of Public Health Project for Young Medical Talents of Jiangsu Province (grant nos. QNRC2016747 and QNRC2016718), The Societal and Developmental Project of Suzhou (grant no. SS201630), The Suzhou Key Laboratory for Respiratory Medicine (grant no. SZS201617) and The Clinical Medical Center of Suzhou (grant no. Szzx201502).

References

1 

Blume C and Davies DE: In vitro and ex vivo models of human asthma. Eur J Pharm Biopharm. 84:394–400. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Kumar RK and Foster PS: Modeling allergic asthma in mice: Pitfalls and opportunities. Am J Respir Cell Mol Biol. 27:267–272. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Saw S, Kale SL and Arora N: Serine protease inhibitor attenuates ovalbumin induced inflammation in mouse model of allergic airway disease. PLoS One. 7:e411072012. View Article : Google Scholar : PubMed/NCBI

4 

Hamid Q and Tulic M: Immunobiology of asthma. Annu Rev Physiol. 71:489–507. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Paul WE and Zhu J: How are T(H)2-type immune responses initiated and amplified? Nat Rev Immunol. 10:225–235. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Busse WW and Lemanske RF Jr: Asthma. N Engl J Med. 344:350–362. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Holgate ST: Pathogenesis of asthma. Clin Exp Allergy. 38:872–897. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Medoff BD, Thomas SY and Luster AD: T cell trafficking in allergic asthma: The ins and outs. Annu Rev Immunol. 26:205–232. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Schuijs MJ, Willart MA, Hammad H and Lambrecht BN: Cytokine targets in airway inflammation. Curr Opin Pharmacol. 13:351–361. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Koshy S, Huq R, Tanner MR, Atik MA, Porter PC, Khan FS, Pennington MW, Hanania NA, Corry DB and Beeton C: Blocking KV1.3 channels inhibits Th2 lymphocyte function and treats a rat model of asthma. J Biol Chem. 289:12623–12632. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Webb GJ, Hirschfield GM and Lane PJ: OX40, OX40L and autoimmunity: A comprehensive review. Clin Rev Allergy Immunol. 50:312–332. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Wang YH and Liu YJ: Thymic stromal lymphopoietin, OX40-ligand, and interleukin-25 in allergic responses. Clin Exp Allergy. 39:798–806. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Liu YJ: Thymic stromal lymphopoietin and OX40 ligand pathway in the initiation of dendritic cell-mediated allergic inflammation. J Allergy Clin Immunol. 120:238–244. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Seshasayee D, Lee WP, Zhou M, Shu J, Suto E, Zhang J, Diehl L, Austin CD, Meng YG, Tan M, et al: In vivo blockade of OX40 ligand inhibits thymic stromal lymphopoietin driven atopic inflammation. J Clin Invest. 117:3868–3878. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Croft M, So T, Duan W and Soroosh P: The significance of OX40 and OX40L to T-cell biology and immune disease. Immunol Rev. 229:173–191. 2009. View Article : Google Scholar : PubMed/NCBI

16 

National Research and Council (US), . Committee for the update of the guide for the care and use of laboratory animals: Guide for the care and use of laboratory animals. 8th. Washington (DC): National Academies Press (US); 2011

17 

Wu Q, Tang Y, Hu X, Wang Q, Lei W, Zhou L and Huang J: Regulation of Th1/Th2 balance through OX40/OX40L signalling by glycyrrhizic acid in a murine model of asthma. Respirology. 21:102–111. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Salek-Ardakani S, Song J, Halteman BS, Jember AG, Akiba H, Yagita H and Croft M: OX40 (CD134) controls memory T helper 2 cells that drive lung inflammation. J Exp Med. 198:315–324. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Lee MY, Lee JA, Seo CS, Ha H, Lee NH and Shin HK: Protective effects of Mentha haplocalyx ethanol extract (MH) in a mouse model of allergic asthma. Phytother Res. 25:863–869. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Malmström V, Shipton D, Singh B, Al-Shamkhani A, Puklavec MJ, Barclay AN and Powrie F: CD134L expression on dendritic cells in the mesenteric lymph nodes drives colitis in T cell-restored SCID mice. J Immunol. 166:6972–6981. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Haley KJ, Ciota A, Contreras JP, Boothby MR, Perkins DL and Finn PW: Alterations in lung collectins in an adaptive allergic immune response. Am J Physiol Lung Cell Mol Physiol. 282:L573–L584. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Zhang M, Fei X, Zhang GQ, Zhang PY, Li F, Bao WP, Zhang YY and Zhou X: Role of neutralizing anti-murine interleukin-17A monoclonal antibody on chronic ozone-induced airway inflammation in mice. Biomed Pharmacother. 83:247–256. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Arestides RS, He H, Westlake RM, Chen AI, Sharpe AH, Perkins DL and Finn PW: Costimulatory molecule OX40L is critical for both Th1 and Th2 responses in allergic inflammation. Eur J Immunol. 32:2874–2880. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Li YY, Chang JW, Chou WC, Liaw CC, Wang HM, Huang JS, Wang CH and Yeh KY: Zoledronic acid is unable to induce apoptosis, but slows tumor growth and prolongs survival for non-small-cell lung cancers. Lung Cancer. 59:180–191. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Lucas CD, Dorward DA, Sharma S, Rennie J, Felton JM, Alessandri AL, Duffin R, Schwarze J, Haslett C and Rossi AG: Wogonin induces eosinophil apoptosis and attenuates allergic airway inflammation. Am J Respir Crit Care Med. 191:626–636. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Carrero R, Cerrada I, Lledó E, Dopazo J, García-García F, Rubio MP, Trigueros C, Dorronsoro A, Ruiz-Sauri A, Montero JA and Sepúlveda P: IL1β induces mesenchymal stem cells migration and leucocyte chemotaxis through NF-κB. Stem Cell Rev. 8:905–916. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Lei W, Zeng DX, Zhu CH, Liu GQ, Zhang XQ, Wang CG, Wang Q and Huang JA: The upregulated expression of OX40/OX40L and their promotion of T cells proliferation in the murine model of asthma. J Thorac Dis. 6:979–987. 2014.PubMed/NCBI

28 

Kow NY and Mak A: Costimulatory pathways: Physiology and potential therapeutic manipulation in systemic lupus erythematosus. Clin Dev Immunol. 2013:2459282013. View Article : Google Scholar : PubMed/NCBI

29 

Gramaglia I, Weinberg AD, Lemon M and Croft M: OX40 ligand: A potent costimulatory molecule for sustaining primary CD4 T cell responses. J Immunol. 161:6510–6517. 1998.PubMed/NCBI

30 

Takeda I, Ine S, Killeen N, Ndhlovu LC, Murata K, Satomi S, Sugamura K and Ishii N: Distinct roles for the OX40-OX40 ligand interaction in regulatory and nonregulatory T cells. J Immunol. 172:3580–3589. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Valzasina B, Guiducci C, Dislich H, Killeen N, Weinberg AD and Colombo MP: Triggering of OX40 (CD134) on CD4(+)CD25+ T cells blocks their inhibitory activity: A novel regulatory role for OX40 and its comparison with GITR. Blood. 105:2845–2851. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Ekkens MJ, Liu Z, Liu Q, Whitmire J, Xiao S, Foster A, Pesce J, VanNoy J, Sharpe AH, Urban JF and Gause WC: The role of OX40 ligand interactions in the development of the Th2 response to the gastrointestinal nematode parasite Heligmosomoides polygyrus. J Immunol. 170:384–393. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Miura S, Ohtani K, Numata N, Niki M, Ohbo K, Ina Y, Gojobori T, Tanaka Y, Tozawa H, Nakamura M, et al: Molecular cloning and characterization of a novel glycoprotein, gp34, that is specifically induced by the human T-cell leukemia virus type I transactivator p40tax. Mol Cell Biol. 11:1313–1325. 1991. View Article : Google Scholar : PubMed/NCBI

34 

Baum PR, Gayle RB III, Ramsdell F, Srinivasan S, Sorensen RA, Watson ML, Seldin MF, Baker E, Sutherland GR, Clifford KN, et al: Molecular characterization of murine and human OX40/OX40 ligand systems: Identification of a human OX40 ligand as the HTLV-1-regulated protein gp34. EMBO J. 13:3992–4001. 1994.PubMed/NCBI

35 

Stüber E, Neurath M, Calderhead D, Fell HP and Strober W: Cross-linking of OX40 ligand, a member of the TNF/NGF cytokine family, induces proliferation and differentiation in murine splenic B cells. Immunity. 2:507–521. 1995. View Article : Google Scholar : PubMed/NCBI

36 

Stüber E and Strober W: The T cell-B cell interaction via OX40-OX40L is necessary for the T cell-dependent humoral immune response. J Exp Med. 183:979–989. 1996. View Article : Google Scholar : PubMed/NCBI

37 

Ohshima Y, Tanaka Y, Tozawa H, Takahashi Y, Maliszewski C and Delespesse G: Expression and function of OX40 ligand on human dendritic cells. J Immunol. 159:3838–3848. 1997.PubMed/NCBI

38 

Brocker T, Gulbranson-Judge A, Flynn S, Riedinger M, Raykundalia C and Lane P: CD4 T cell traffic control: In vivo evidence that ligation of OX40 on CD4 T cells by OX40-ligand expressed on dendritic cells leads to the accumulation of CD4 T cells in B follicles. Eur J Immunol. 29:1610–1616. 1999. View Article : Google Scholar : PubMed/NCBI

39 

Imura A, Hori T, Imada K, Ishikawa T, Tanaka Y, Maeda M, Imamura S and Uchiyama T: The human OX40/gp34 system directly mediates adhesion of activated T cells to vascular endothelial cells. J Exp Med. 183:2185–2195. 1996. View Article : Google Scholar : PubMed/NCBI

40 

Rogers PR, Song J, Gramaglia I, Killeen N and Croft M: OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity. 15:445–455. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Kinnear G, Wood KJ, Marshall D and Jones ND: Anti-OX40 prevents effector T-cell accumulation and CD8+ T-cell mediated skin allograft rejection. Transplantation. 90:1265–1271. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Flynn S, Toellner KM, Raykundalia C, Goodall M and Lane P: CD4 T cell cytokine differentiation: The B cell activation molecule, OX40 ligand, instructs CD4 T cells to express interleukin 4 and upregulates expression of the chemokine receptor, Blr-1. J Exp Med. 188:297–304. 1998. View Article : Google Scholar : PubMed/NCBI

43 

Vu MD, Clarkson MR, Yagita H, Turka LA, Sayegh MH and Li XC: Critical, but conditional, role of OX40 in memory T cell-mediated rejection. J Immunol. 176:1394–1401. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Murata K, Nose M, Ndhlovu LC, Sato T, Sugamura K and Ishii N: Constitutive OX40/OX40 ligand interaction induces autoimmune-like diseases. J Immunol. 169:4628–4236. 2002. View Article : Google Scholar : PubMed/NCBI

45 

Ueno H and Blanco P: OX40/OX40L axis: Not a friend in autoimmunity. Oncotarget. 6:21779–21780. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Gough MJ and Weinberg AD: OX40 (CD134) and OX40L. Adv Exp Med Biol. 647:94–107. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Hori T: Roles of OX40 in the pathogenesis and the control of diseases. Int J Hematol. 83:17–22. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Redmond WL and Weinberg AD: Targeting OX40 and OX40L for the treatment of autoimmunity and cancer. Crit Rev Immunol. 27:415–436. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Park BS, Hong GU and Ro JY: Foxp3(+)-Treg cells enhanced by repeated low-dose gamma-irradiation attenuate ovalbumin-induced allergic asthma in mice. Radiat Res. 179:570–583. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Obermeier F, Schwarz H, Dunger N, Strauch UG, Grunwald N, Schölmerich J and Falk W: OX40/OX40L interaction induces the expression of CXCR5 and contributes to chronic colitis induced by dextran sulfate sodium in mice. Eur J Immunol. 33:3265–3274. 2003. View Article : Google Scholar : PubMed/NCBI

51 

Kay AB: The role of T lymphocytes in asthma. Chem Immunol Allergy. 91:59–75. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Yang T, Zhang X, Wang M, Zhang J, Huang F, Cai J, Zhang Q, Mao F, Zhu W, Qian H and Xu W: Activation of mesenchymal stem cells by macrophages prompts human gastric cancer growth through NF-κB pathway. PLoS One. 9:e975692014. View Article : Google Scholar : PubMed/NCBI

53 

Song J, So T and Croft M: Activation of NF-kappaB1 by OX40 contributes to antigen-driven T cell expansion and survival. J Immunol. 180:7240–7248. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Burrows KE, Dumont C, Thompson CL, Catley MC, Dixon KL and Marshall D: OX40 blockade inhibits house dust mite driven allergic lung inflammation in mice and in vitro allergic responses in humans. Eur J Immunol. 45:1116–1128. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Robinson DP, Hall OJ, Nilles TL, Bream JH and Klein SL: 17β-estradiol protects females against influenza by recruiting neutrophils and increasing virus-specific CD8 T cell responses in the lungs. J Virol. 88:4711–4720. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Jandl K, Stacher E, Bálint Z, Sturm EM, Maric J, Peinhaupt M, Luschnig P, Aringer I, Fauland A, Konya V, et al: Activated prostaglandin D2 receptors on macrophages enhance neutrophil recruitment into the lung. J Allergy Clin Immunol. 137:833–843. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Jeon J, Kim Y, Kim H, Kang JS and Lee WJ: Anti-inflammatory effect of alloferon on ovalbumin-induced asthma. Immune Netw. 15:304–312. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Bouchard JC, Beal DR, Kim J, Vaickus LJ and Remick DG: Chemokines mediate ethanol-induced exacerbations of murine cockroach allergen asthma. Clin Exp Immunol. 172:203–216. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Farres MN, Sabry MK, Ahmed EE, Elkady HM and Mohamed NA: OX40 ligand: A potential costimulatory molecule in atopic asthma. J Asthma. 51:573–577. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Kaur D and Brightling C: OX40/OX40 ligand interactions in T-cell regulation and asthma. Chest. 141:494–499. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Lei W, Zhu CH, Zeng da X, Wang Q, Zhang XQ, Chen YB, Mu CY and Huang JA: SOX40L: An important inflammatory mediator in adult bronchial asthma. Ann Acad Med Singapore. 41:200–204. 2012.PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 17 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lei W, Zeng D, Liu G, Zhu Y, Wang J, Wu H, Jiang J and Huang J: Crucial role of OX40/OX40L signaling in a murine model of asthma. Mol Med Rep 17: 4213-4220, 2018
APA
Lei, W., Zeng, D., Liu, G., Zhu, Y., Wang, J., Wu, H. ... Huang, J. (2018). Crucial role of OX40/OX40L signaling in a murine model of asthma. Molecular Medicine Reports, 17, 4213-4220. https://doi.org/10.3892/mmr.2018.8453
MLA
Lei, W., Zeng, D., Liu, G., Zhu, Y., Wang, J., Wu, H., Jiang, J., Huang, J."Crucial role of OX40/OX40L signaling in a murine model of asthma". Molecular Medicine Reports 17.3 (2018): 4213-4220.
Chicago
Lei, W., Zeng, D., Liu, G., Zhu, Y., Wang, J., Wu, H., Jiang, J., Huang, J."Crucial role of OX40/OX40L signaling in a murine model of asthma". Molecular Medicine Reports 17, no. 3 (2018): 4213-4220. https://doi.org/10.3892/mmr.2018.8453