...
首页> 外文期刊>The journal of clinical investigation >Placenta growth factor augments airway hyperresponsiveness via leukotrienes and IL-13
【24h】

Placenta growth factor augments airway hyperresponsiveness via leukotrienes and IL-13

机译:胎盘生长因子通过白三烯和IL-13增强气道高反应性

获取原文

摘要

Airway hyperresponsiveness (AHR) affects 55%–77% of children with sickle cell disease (SCD) and occurs even in the absence of asthma. While asthma increases SCD morbidity and mortality, the mechanisms underlying the high AHR prevalence in a hemoglobinopathy remain unknown. We hypothesized that placenta growth factor (PlGF), an erythroblast-secreted factor that is elevated in SCD, mediates AHR. In allergen-exposed mice, loss of Plgf dampened AHR, reduced inflammation and eosinophilia, and decreased expression of the Th2 cytokine IL-13 and the leukotriene-synthesizing enzymes 5-lipoxygenase and leukotriene-C4-synthase. Plgf~(–/–) mice treated with leukotrienes phenocopied the WT response to allergen exposure; conversely, anti-PlGF Ab administration in WT animals blunted the AHR. Notably, Th2-mediated STAT6 activation further increased PlGF expression from lung epithelium, eosinophils, and macrophages, creating a PlGF/leukotriene/Th2-response positive feedback loop. Similarly, we found that the Th2 response in asthma patients is associated with increased expression of PlGF and its downstream genes in respiratory epithelial cells. In an SCD mouse model, we observed increased AHR and higher leukotriene levels that were abrogated by anti-PlGF Ab or the 5-lipoxygenase inhibitor zileuton. Overall, our findings indicate that PlGF exacerbates AHR and uniquely links the leukotriene and Th2 pathways in asthma. These data also suggest that zileuton and anti-PlGF Ab could be promising therapies to reduce pulmonary morbidity in SCD. Keywords: Genetics, Hematology, Immunology, Inflammation, PulmonologyIntroductionSickle cell disease (SCD) is one of the most prevalent genetic disorders worldwide. It is caused by a mutation in the β-globin gene that results in sickle-shaped rbc during deoxygenation (1, 2). Adhesion of rbc and leukocytes to the endothelium, rbc sickling, and adhesion of sickle rbc to wbc result in vascular occlusions and multiple end-organ complications. However, the oxygen tension in lungs is too high for sickle hemoglobin polymerization. Indeed, the lung is the only organ where sickled rbc returning from the venous vasculature can unsickle. Despite this, acute and chronic lung diseases are major complications of SCD, and independent risk factors for morbidity and mortality in these patients (3, 4) and mechanisms behind them are largely unknown. Airway hyperresponsiveness (AHR), a dynamic narrowing of the lower airways, is highly prevalent in patients with SCD, affecting 55%–77% of children (5–7) and 33% of adults with SCD (8, 9). In contrast, the reported prevalence of AHR is 6%–20% in the African-American and black Caribbean pediatric population (6, 10).Clinically, AHR is a key feature of asthma and is assessed using a provocative challenge (e.g., methacholine challenge) to induce a measurable bronchoconstriction. In SCD, the incidence of AHR is much higher than that of clinically symptomatic asthma (5, 6, 10–12) and is present without clinical features of asthma (wheezing, exhaled NO, and positive skin test to aeroallergens). But unlike asthma, which is a known risk factor for morbidity in SCD (13–16), the mechanism behind high AHR and its clinical significance is unknown. In SCD, biomarkers of hemolysis rather than those of asthma show a strong correlation with AHR (7). This led us to hypothesize that the etiology of the high AHR prevalence in SCD may be related to erythroid cells.Following allergen stimulation, AHR is driven by a potent Th2 immune response that primes B cells to release allergen-specific IgE and drives lung inflammation. Th2 cytokines, such as IL-13 and IL-4 (17), stimulate airway smooth muscle contraction, recruit inflammatory cells, and increase mucus production; congestion and constriction of the airway lumen contributes to its remodeling (18, 19). In SCD, AHR has been presumed to occur secondarily to the leukocytosis and inflammation present at steady state. Notably, SCD patients also have elevated levels of LTE_(4) and LTB_(4) at steady state (20, 21). LTB_(4), a potent neutrophil inflammatory molecule (22), and cysteinyl LTs (CysLT) have been implicated in different aspects of allergen-induced asthma (18, 23). CysLT receptor inhibitors of 5-lipoxygenase (5-LO, encoded by Alox5 ) inhibitors are currently licensed for use for the treatment of asthma in humans (24, 25).Our group previously reported that placenta growth factor (PlGF), an erythroid cell–secreted growth factor that belongs to the vascular endothelial growth factor family, is increased in plasma of patients with SCD and contributes to the baseline inflammation and features of pulmonary hypertension (26, 27). Herein, we hypothesized that PLGF mediates the high prevalence of AHR in SCD. We utilized genetic and pharmacological ablation of Plgf in WT mice and Berkeley sickle mice (hereafter termed HbS mice) (28). We have previously reported that crosses between Plgf KO and HbS mice were born extremely rarely, and the few bo
机译:气道高反应性(AHR)会影响55%–77%的镰状细胞病(SCD)儿童,甚至在没有哮喘的情况下也会发生。尽管哮喘增加了SCD的发病率和死亡率,但血红蛋白病中高AHR患病率的潜在机制仍然未知。我们假设胎盘生长因子(PlGF)(一种在SCD中升高的成红细胞分泌因子)介导了AHR。在暴露于变应原的小鼠中,Plgf的丢失会抑制AHR,减轻炎症和嗜酸性粒细胞,并降低Th2细胞因子IL-13和白三烯合成酶5-脂氧合酶和白三烯C4-合成酶的表达。用白三烯处理的Plgf〜(– / –)小鼠表型化了WT对过敏原暴露的反应;相反,在野生动物中抗PlGF Ab的给药使AHR变钝。值得注意的是,Th2介导的STAT6激活进一步增加了肺上皮,嗜酸性粒细胞和巨噬细胞中PlGF的表达,从而形成了PlGF /白三烯/ Th2反应阳性反馈环。同样,我们发现哮喘患者的Th2反应与呼吸道上皮细胞中PlGF及其下游基因的表达增加有关。在SCD小鼠模型中,我们观察到被抗PlGF Ab或5-脂氧合酶抑制剂zileuton消除的AHR升高和白三烯水平升高。总体而言,我们的发现表明,PlGF会加重AHR,并唯一关联哮喘中的白三烯和Th2途径。这些数据还表明,齐留通和抗PlGF Ab有望成为减少SCD肺部疾病的有前途的疗法。关键词:遗传学,血液学,免疫学,炎症,肺病简介镰状细胞病(SCD)是世界上最流行的遗传性疾病之一。它是由β-珠蛋白基因的突变引起的,该突变导致脱氧过程中出现镰刀状的rbc(1、2)。 rbc和白细胞对内皮的粘附,rbc镰状化以及镰刀rbc对wbc的粘附导致血管闭塞和多种终末器官并发症。但是,对于镰刀型血红蛋白聚合而言,肺中的氧气张力过高。确实,肺是唯一可以使镰状的rbc从静脉血管返回的器官。尽管如此,急性和慢性肺部疾病是SCD的主要并发症,这些患者发病和死亡的独立危险因素(3、4)及其背后的机制在很大程度上尚不清楚。气道高反应性(AHR)是下呼吸道的动态变窄,在SCD患者中非常普遍,影响55%至77%的儿童(5-7)和33%的SCD成人(8、9)。相比之下,据报道,非洲裔美国人和加勒比海黑人儿童的AHR患病率为6%–20%(6,10)。临床上,AHR是哮喘的关键特征,并通过挑衅性的挑战(例如,乙酰甲胆碱)进行评估挑战)以诱导可测量的支气管收缩。在SCD中,AHR的发生率远高于临床上有症状的哮喘(5、6、10-12),并且没有哮喘的临床特征(喘息,呼气NO和气敏性皮肤试验阳性)。但是与哮喘不同,哮喘是SCD发病的已知危险因素(13-16),而高AHR背后的机制及其临床意义尚不清楚。在SCD中,溶血的生物标志物而不是哮喘的生物标志物与AHR密切相关(7)。这导致我们假设SCD中高AHR患病率的病因可能与类红细胞有关。在过敏原刺激后,AHR受有效的Th2免疫反应驱动,引发B细胞释放过敏原特异性IgE并驱动肺部炎症。 Th2细胞因子,例如IL-13和IL-4(17),刺激气道平滑肌收缩,募集炎性细胞,并增加粘液产生;气管腔的充血和收缩有助于其重塑(18、19)。在SCD中,AHR已被认为次于白细胞增多和稳定状态的炎症。值得注意的是,SCD患者在稳定状态下的LTE_(4)和LTB_(4)水平也升高(20,21)。 LTB_(4),一种有效的嗜中性粒细胞炎性分子(22)和半胱氨酰LTs(CysLT)已牵涉到变应原诱发哮喘的不同方面(18,23)。 5-脂氧合酶的CysLT受体抑制剂(5-LO,由Alox5编码)目前已获准用于治疗人类哮喘(24,25)。我们的研究小组先前曾报道胎盘生长因子(PlGF)是一种类红细胞–分泌的生长因子,属于血管内皮生长因子家族,在SCD患者的血浆中升高,并且有助于基线炎症和肺动脉高压的特征(26、27)。在这里,我们假设PLGF介导SCD中AHR的高患病率。我们在WT小鼠和伯克利镰刀小鼠(以下称为HbS小鼠)中利用了Plgf的遗传和药理学消融(28)。我们之前曾报道过,Plgf KO和HbS小鼠之间的杂交非常少,而

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号