English
 
Help Privacy Policy Disclaimer
  Advanced SearchBrowse

Item

ITEM ACTIONSEXPORT
  Maternal and perinatal obesity induce bronchial obstruction and pulmonary hypertension via IL-6-FoxO1-axis in later life

Selle, J., Dinger, K., Jentgen, V., Zanetti, D., Will, J., Georgomanolis, T., et al. (2022). Maternal and perinatal obesity induce bronchial obstruction and pulmonary hypertension via IL-6-FoxO1-axis in later life. NATURE COMMUNICATIONS, 13(1): 4352. doi:10.1038/s41467-022-31655-z.

Item is

Files

show Files

Locators

show

Creators

show
hide
 Creators:
Selle, Jaco, Author
Dinger, Katharina, Author
Jentgen, Vanessa, Author
Zanetti, Daniela, Author
Will, Johannes, Author
Georgomanolis, Theodoros, Author
Vohlen, Christina, Author
Wilke, Rebecca, Author
Kojonazarov, Baktybek, Author
Klymenko, Oleksiy, Author
Mohr, Jasmine, Author
Koningsbruggen-Rietschel, Silke v, Author
Rhodes, Christopher J., Author
Ulrich, Anna, Author
Hirani, Dharmesh, Author
Nestler, Tim, Author
Odenthal, Margarete, Author
Mahabir, Esther, Author
Nayakanti, Sreenath1, Author           
Dabral, Swati1, Author           
Wunderlich, Thomas, AuthorPriest, James, AuthorSeeger, Werner1, Author           Doetsch, Joerg, AuthorPullamsetti, Soni S.1, Author           Alcazar, Miguel A. Alejandre, Author more..
Affiliations:
1Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Max Planck Society, ou_2591698              

Content

show
hide
Free keywords: -
 Abstract: This study shows that maternal and perinatal obesity cause bronchial and vascular smooth muscle cell proliferation through an IL-6-FoxO1 axis, and favor thereby the emergence of bronchial obstruction and pulmonary hypertension later in life.
Obesity is a pre-disposing condition for chronic obstructive pulmonary disease, asthma, and pulmonary arterial hypertension. Accumulating evidence suggests that metabolic influences during development can determine chronic lung diseases (CLD). We demonstrate that maternal obesity causes early metabolic disorder in the offspring. Here, interleukin-6 induced bronchial and microvascular smooth muscle cell (SMC) hyperproliferation and increased airway and pulmonary vascular resistance. The key anti-proliferative transcription factor FoxO1 was inactivated via nuclear exclusion. These findings were confirmed using primary SMC treated with interleukin-6 and pharmacological FoxO1 inhibition as well as genetic FoxO1 ablation and constitutive activation. In vivo, we reproduced the structural and functional alterations in offspring of obese dams via the SMC-specific ablation of FoxO1. The reconstitution of FoxO1 using IL-6-deficient mice and pharmacological treatment did not protect against metabolic disorder but prevented SMC hyperproliferation. In human observational studies, childhood obesity was associated with reduced forced expiratory volume in 1 s/forced vital capacity ratio Z-score (used as proxy for lung function) and asthma. We conclude that the interleukin-6-FoxO1 pathway in SMC is a molecular mechanism by which perinatal obesity programs the bronchial and vascular structure and function, thereby driving CLD development. Thus, FoxO1 reconstitution provides a potential therapeutic option for preventing this metabolic programming of CLD.

Details

show
hide
Language(s):
 Dates: 2022-07-27
 Publication Status: Published online
 Pages: -
 Publishing info: -
 Table of Contents: -
 Rev. Type: -
 Identifiers: ISI: 000831732000004
DOI: 10.1038/s41467-022-31655-z
PMID: 35896539
 Degree: -

Event

show

Legal Case

show

Project information

show

Source 1

show
hide
Title: NATURE COMMUNICATIONS
Source Genre: Journal
 Creator(s):
Affiliations:
Publ. Info: -
Pages: - Volume / Issue: 13 (1) Sequence Number: 4352 Start / End Page: - Identifier: -