Dépôt numérique
RECHERCHER

Oncostatin M and STAT3 Signaling Pathways Support Human Trophoblast Differentiation by Inhibiting Inflammatory Stress in Response to IFNγ and GM-CSF

Téléchargements

Téléchargements par mois depuis la dernière année

Plus de statistiques...

Ravelojaona, Marion; Girouard, Julie; Kana Tsapi, Emmanuelle Stella; Chambers, Megan; Vaillancourt, Cathy ORCID logoORCID: https://orcid.org/0000-0003-0543-6244; Van Themsche, Céline; Thornton, Catherine A et Reyes-Moreno, Carlos (2024). Oncostatin M and STAT3 Signaling Pathways Support Human Trophoblast Differentiation by Inhibiting Inflammatory Stress in Response to IFNγ and GM-CSF Cells , vol. 13 , nº 3:229. pp. 1-24. DOI: 10.3390/cells13030229.

[thumbnail of cells-13-00229-v2.pdf]
Prévisualisation
PDF - Version publiée
Disponible sous licence Creative Commons Attribution.

Télécharger (5MB) | Prévisualisation

Résumé

Interleukin-6 (IL-6) superfamily cytokines play critical roles during human pregnancy by promoting trophoblast differentiation, invasion, and endocrine function, and maintaining embryo immunotolerance and protection. In contrast, the unbalanced activity of pro-inflammatory factors such as interferon gamma (IFNγ) and granulocyte-macrophage colony-stimulating factor (GM-CSF) at the maternal-fetal interface have detrimental effects on trophoblast function and differentiation. This study demonstrates how the IL-6 cytokine family member oncostatin M (OSM) and STAT3 activation regulate trophoblast fusion and endocrine function in response to pro-inflammatory stress induced by IFNγ and GM-CSF. Using human cytotrophoblast-like BeWo (CT/BW) cells, differentiated in villous syncytiotrophoblast (VST/BW) cells, we show that beta-human chorionic gonadotrophin (βhCG) production and cell fusion process are affected in response to IFNγ or GM-CSF. However, those effects are abrogated with OSM by modulating the activation of IFNγ-STAT1 and GM-CSF-STAT5 signaling pathways. OSM stimulation enhances the expression of STAT3, the phosphorylation of STAT3 and SMAD2, and the induction of negative regulators of inflammation (e.g., IL-10 and TGFβ1) and cytokine signaling (e.g., SOCS1 and SOCS3). Using STAT3-deficient VST/BW cells, we show that STAT3 expression is required for OSM to regulate the effects of IFNγ in βhCG and E-cadherin expression. In contrast, OSM retains its modulatory effect on GM-CSF-STAT5 pathway activation even in STAT3-deficient VST/BW cells, suggesting that OSM uses STAT3-dependent and -independent mechanisms to modulate the activation of pro-inflammatory pathways IFNγ-STAT1 and GM-CSF-STAT5. Moreover, STAT3 deficiency in VST/BW cells leads to the production of both a large amount of βhCG and an enhanced expression of activated STAT5 induced by GM-CSF, independently of OSM, suggesting a key role for STAT3 in βhCG production and trophoblast differentiation through STAT5 modulation. In conclusion, our study describes for the first time the critical role played by OSM and STAT3 signaling pathways to preserve and regulate trophoblast biological functions during inflammatory stress.

Type de document: Article
Mots-clés libres: : beta-human chorionic gonadotrophin; granulocyte–macrophage colony-stimulating factor; inflammatory stress; interferon gamma; oncostatin M; pregnancy; placenta; syncytiotrophoblast
Centre: Centre INRS-Institut Armand Frappier
Date de dépôt: 12 févr. 2024 06:12
Dernière modification: 12 févr. 2024 06:12
URI: https://espace.inrs.ca/id/eprint/14199

Gestion Actions (Identification requise)

Modifier la notice Modifier la notice