TGF-β3 Induces Autophagic Activity by Increasing ROS Generation in a NOX4-Dependent Pathway

المؤلفون المشاركون

Wang, Xiaoyun
Zhang, Yun
Tang, Hong-Mei
Liu, Chun-Feng
Yuan, Xie-Fang
Ma, Ning
Xu, Guo-Feng
Wang, Song-Ping
Deng, Jun
Wang, Xing

المصدر

Mediators of Inflammation

العدد

المجلد 2019، العدد 2019 (31 ديسمبر/كانون الأول 2019)، ص ص. 1-17، 17ص.

الناشر

Hindawi Publishing Corporation

تاريخ النشر

2019-12-31

دولة النشر

مصر

عدد الصفحات

17

التخصصات الرئيسية

الأمراض

الملخص EN

Higher concentrations of reactive oxygen species (ROS) have been associated with epithelial cell damage, cell shedding, and airway hyperresponsiveness.

Previous studies have indicated that transforming growth factor-beta (TGF-β) mediates ROS production and NADPH oxidase (NOX) activity.

In our previous study, we also observed that TGF-β3 increases mucus secretion in airway epithelial cells in an autophagy-dependent fashion.

Although it is well known that the relationship between ROS and autophagy is cell context-dependent, the exact mechanism of action remains unclear.

The following study examined whether ROS act as upstream of autophagy activation in response to TGF-β3 induction.

Using an allergic inflammation mouse model induced by house dust mite (HDM), we observed elevated lung amounts of TGF-β3 accompanied by increased ROS levels.

And we found that ROS levels were elevated and NOX4 expression was increased in TGF-β3-induced epithelial cells, while the lack of NOX4 in the epithelial cells could reduce ROS generation and autophagy-dependent MUC5AC expression treated with TGF-β3.

Furthermore, our studies demonstrated that the Smad2/3 pathway was involved in TGF-β3-induced ROS generation by promoting NOX4 expression.

The inhibition of ROS generation by N-Acetyl-L-cysteine (NAC) resulted in a decrease in mucus expression and autophagy activity in vivo as well as in vitro.

Finally, TGF-β3-neutralizing antibody significantly reduced the ROS generation, mucus expression, and autophagy activity and also decreased the phosphorylation of Smad2 and Smad3.

Taken together, the obtained results revealed that persistent TGF-β3 activation increased ROS levels in a NOX4-dependent pathway and subsequently induced autophagy as well as MUC5AC expression in the epithelial cells.

نمط استشهاد جمعية علماء النفس الأمريكية (APA)

Zhang, Yun& Tang, Hong-Mei& Liu, Chun-Feng& Yuan, Xie-Fang& Wang, Xiaoyun& Ma, Ning…[et al.]. 2019. TGF-β3 Induces Autophagic Activity by Increasing ROS Generation in a NOX4-Dependent Pathway. Mediators of Inflammation،Vol. 2019, no. 2019, pp.1-17.
https://search.emarefa.net/detail/BIM-1192777

نمط استشهاد الجمعية الأمريكية للغات الحديثة (MLA)

Zhang, Yun…[et al.]. TGF-β3 Induces Autophagic Activity by Increasing ROS Generation in a NOX4-Dependent Pathway. Mediators of Inflammation No. 2019 (2019), pp.1-17.
https://search.emarefa.net/detail/BIM-1192777

نمط استشهاد الجمعية الطبية الأمريكية (AMA)

Zhang, Yun& Tang, Hong-Mei& Liu, Chun-Feng& Yuan, Xie-Fang& Wang, Xiaoyun& Ma, Ning…[et al.]. TGF-β3 Induces Autophagic Activity by Increasing ROS Generation in a NOX4-Dependent Pathway. Mediators of Inflammation. 2019. Vol. 2019, no. 2019, pp.1-17.
https://search.emarefa.net/detail/BIM-1192777

نوع البيانات

مقالات

لغة النص

الإنجليزية

الملاحظات

Includes bibliographical references

رقم السجل

BIM-1192777