首页 | 本学科首页   官方微博 | 高级检索  
     


Complement C5a exacerbates acute lung injury induced through autophagy-mediated alveolar macrophage apoptosis
Authors:R Hu  Z-F Chen  J Yan  Q-F Li  Y Huang  H Xu  X Zhang  H Jiang
Affiliation:1.Department of Anesthesiology, Shanghai Ninth People''s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China;2.Institute of Medical Intervention Engineering, Tongji University, No.727 North Zhongshan Road, Shanghai, China;3.Department of Interventional Radiology, Shanghai Tenth People''s Hospital, Tongji University, No. 301, Yanchang Road, Shanghai, China
Abstract:Intestinal ischemia has a high mortality and often causes acute lung injury (ALI), which is a serious complication, and is accompanied by high mortality up to 40%. An intense local and systemic inflammation occurs during intestinal ischemia/reperfusion (IR)-induced lung injury resulting from activation of immune responses. It has been reported that one component of complement, C5a, is indispensable for the full development of IR-induced lung injury, whereas the detailed molecular mechanism remains to be elucidated. In this study, we found that intestinal IR induced ALI-like symptoms, and C5a receptor (C5aR) expression was upregulated in alveolar macrophages, which are resident macrophages in lung tissue and are important in pulmonary homeostasis. C5a produced during lung injury binds to C5aR in alveolar macrophages, initiates downstream signaling that promotes autophagy, leading to apoptosis of alveolar macrophages. Using Mφ-ATG5−/− mice, in which the atg5 is deficient specifically in macrophages and autophagy is inhibited, we confirmed that in vivo C5a interacting with C5aR induced autophagy in alveolar macrophages, which promoted alveolar macrophage apoptosis. Further study indicated that autophagy was induced through C5aR-mediated degradation of bcl-2. Taken together, our results demonstrated that C5aR-mediated autophagy induced apoptosis in alveolar macrophages, disrupting pulmonary homeostasis and contributing to the development of ALI. This novel mechanism suggests new therapeutic potential of autophagy regulation in ALI.During diverse clinical procedures, transient ischemia and reperfusion, known as ischemia/reperfusion (IR) clinically, are found in organs or tissues, and cause intense inflammation, both locally and systemically,1, 2 which in turn leads to various types of injury, even multiple organ failure, contributing to high mortality. Acute lung injury (ALI) is a common outcome of IR, and usually occurs in patients with intestinal ischemia, leading to high mortality of 60–80%.3 In addition, ALI is a life-threatening complication associated with sepsis, pneumonia, trauma, and many other clinical conditions. Despite improvements in the management of critically ill patients, ALI mortality is approximately 40%, and survivors often do not return to a normal life.4 During the IR process, ischemia initiates a local inflammatory response, by releasing pro-inflammatory factors and activating/attracting inflammatory cells, such as neutrophils, macrophages, and lymphocytes.5 Oxidative stress resulting from ischemia also contributes to IR injury. Owing to the unique anatomic and physiological features, the lung is susceptible to IR injury through pro-inflammatory cytokines storm.6 Only a few pharmacologic treatment options are available for IR-induced ALI, which work by inhibiting inflammation or anti-oxidative effects.7 Obviously, more effort is needed to clarify the underlying pathophysiological mechanisms of ALI and find more efficient therapeutic methods.Macrophages are believed to derive from hematopoietic stem cells and are distributed all over the body. Macrophages are of vital importance in immune homeostasis, tissue remodeling, and biological events. Alveolar macrophages are resident lung macrophages, and present the first line of encountering inhaled substances.8 Alveolar macrophages have essential roles in maintaining pulmonary homeostasis, without pro-inflammatory effects.9 More importantly, alveolar macrophages suppress excessive inflammation, putatively through the strong inhibition of local immune cells, such as T lymphocytes and DCs. For example, rodent alveolar macrophages render inhibition on T-cell activation in the presence of DCs in vitro, through multiple mechanisms, such as releasing the suppressive cytokines, transforming growth factor-β and interleukin-10 (IL-10).8, 9, 10, 11, 12 If alveolar macrophages are depleted, the animals display stronger inflammatory responses to otherwise innocuous inhaled antigens.13 During ALI, cytokines and chemokines produced by tissue macrophages recruit neutrophils to the injury sites,14 but the neutrophil recruitment also affects alveolar macrophage activity.15,16 IL-10 production is induced by macrophages after phagocytosis of apoptotic neutrophils, which in turn suppresses additional cytokine production and inflammation, affecting both pro-inflammatory and anti-inflammatory cellular components of ALI.12 For these reasons, alveolar macrophages have attracted interest in studies on the mechanisms of ALI.8, 9, 10, 11Complements are key mediators of the first line in protecting hosts from pathogen invasions and have been shown to be involved in IR-induced inflammation. During the ignition and amplification stages, complement activation contributes to inflammation-mediated tissue injury,1, 2, 17 which would be significantly diminished if complement factors were depleted.18, 19 The complement activation product, C5a, is essential for the full development of injury. C5a has the ability of chemotaxis20 and it can also directly activate neutrophils and macrophages for chemokine production.21 C5a receptor (C5aR) signaling is required for C5a to render its effects on the process, as blockade of C5aR signaling will have similar effects to depletion of C5a in the survival of animals with cecal ligation and puncture,22 suggesting that intercepting C5a or C5aR signaling may provide a potential target for therapeutic treatment in inflammatory diseases.23Although significant effort has been aimed at determining the mechanism of macrophages in ALI, the activity of C5aR on macrophages is unclear. This study aimed to clarify the role of C5aR in macrophage biology during ALI development, and found that elevated C5a induced C5aR signaling in alveolar macrophages, and contributed to autophagy-mediated apoptosis, thus exacerbating the ALI symptoms. This novel mechanism provides a potential role for autophagy regulation in ALI therapeutic applications.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号