cytokine secretion profile
Recently Published Documents


TOTAL DOCUMENTS

36
(FIVE YEARS 1)

H-INDEX

16
(FIVE YEARS 0)

2021 ◽  
Vol 12 ◽  
Author(s):  
Rui-Jún Eveline Li ◽  
Aram de Haas ◽  
Ernesto Rodríguez ◽  
Hakan Kalay ◽  
Anouk Zaal ◽  
...  

Dendritic cells (DCs) are key initiators of the adaptive immunity, and upon recognition of pathogens are able to skew T cell differentiation to elicit appropriate responses. DCs possess this extraordinary capacity to discern external signals using receptors that recognize pathogen-associated molecular patterns. These can be glycan-binding receptors that recognize carbohydrate structures on pathogens or pathogen-associated patterns that additionally bind receptors, such as Toll-like receptors (TLRs). This study explores the early signaling events in DCs upon binding of α2-3 sialic acid (α2-3sia) that are recognized by Immune inhibitory Sialic acid binding immunoglobulin type lectins. α2-3sias are commonly found on bacteria, e.g. Group B Streptococcus, but can also be expressed by tumor cells. We investigated whether α2-3sia conjugated to a dendrimeric core alters DC signaling properties. Through phosphoproteomic analysis, we found differential signaling profiles in DCs after α2-3sia binding alone or in combination with LPS/TLR4 co-stimulation. α2-3sia was able to modulate the TLR4 signaling cascade, resulting in 109 altered phosphoproteins. These phosphoproteins were annotated to seven biological processes, including the regulation of the IL-12 cytokine pathway. Secretion of IL-10, the inhibitory regulator of IL-12 production, by DCs was found upregulated after overnight stimulation with the α2-3sia dendrimer. Analysis of kinase activity revealed altered signatures in the JAK-STAT signaling pathway. PhosphoSTAT3 (Ser727) and phosphoSTAT5A (Ser780), involved in the regulation of the IL-12 pathway, were both downregulated. Flow cytometric quantification indeed revealed de- phosphorylation over time upon stimulation with α2-3sia, but no α2-6sia. Inhibition of both STAT3 and -5A in moDCs resulted in a similar cytokine secretion profile as α-3sia triggered DCs. Conclusively, this study revealed a specific alteration of the JAK-STAT pathway in DCs upon simultaneous α2-3sia and LPS stimulation, altering the IL10:IL-12 cytokine secretion profile associated with reduction of inflammation. Targeted control of the STAT phosphorylation status is therefore an interesting lead for the abrogation of immune escape that bacteria or tumors impose on the host.


2018 ◽  
Vol 8 (1) ◽  
Author(s):  
Daniëlle G. Leuning ◽  
Nick R. M. Beijer ◽  
Nadia A. du Fossé ◽  
Steven Vermeulen ◽  
Ellen Lievers ◽  
...  

2014 ◽  
Vol 1 (1) ◽  
Author(s):  
Kevin Anton ◽  
John Glod

AbstractThe tumor microenvironment plays a critical role in the survival, growth, invasion, and metastasis of solid tumors. However, the mechanisms by which it influences these aspects of tumor progression remain incompletely characterized. In this study, we show that human glioblastoma cells secrete soluble factors that alter the phenotype and cytokine secretion profile of both macrophages and mesenchymal stem cells (MSCs). Macrophages and MSCs respond to tumor-secreted factors by increasing the release of interleukin-6 (IL-6) and this response is potentiated when macrophages and MSCs are combined in co-culture. In glioblastoma, IL-6 has been associated with tumor cell invasion, angiogenesis, tumor cell proliferation, immune suppression, and poor prognosis. Our results suggest that the orchestrated response of macrophages and stromal elements to neoplastic cells enhances tumor progression through the release of soluble factors.


Sign in / Sign up

Export Citation Format

Share Document