Hypoxia is a key component of the tumor microenvironment (TME) and promotes not only tumor growth and metastasis, but also negatively affects infiltrating immune cells by impairing host immunity. Dendritic cells (DCs) are the most potent antigen-presenting cells and their biology is weakened in the TME in many ways, including the modulation of their viability. RNASET2 belongs to the T2 family of extracellular ribonucleases and, besides its nuclease activity, it exerts many additional functions. Indeed, RNASET2 is involved in several human pathologies, including cancer, and it is functionally relevant in the TME. RNASET2 functions are not restricted to cancer cells and its expression could be relevant also in other cell types which are important players in the TME, including DCs. Therefore, this study aimed to unravel the effect of hypoxia (2% O2) on the expression of RNASET2 in DCs. Here, we showed that hypoxia enhanced the expression and secretion of RNASET2 in human monocyte-derived DCs. This paralleled the HIF-1α accumulation and HIF-dependent and-independent signaling, which are associated with DCs’ survival/autophagy/apoptosis. RNASET2 expression, under hypoxia, was regulated by the PI3K/AKT pathway and was almost completely abolished by TLR4 ligand, LPS. Taken together, these results highlight how hypoxia-dependent and-independent pathways shape RNASET2 expression in DCs, with new perspectives on its implication for TME and, therefore, in anti-tumor immunity.

Monaci, S., Coppola, F., Giuntini, G., Roncoroni, R., Acquati, F., Sozzani, S., et al. (2021). Hypoxia enhances the expression of RNASET2 in human monocyte-derived dendritic cells: Role of PI3K/AKT pathway. INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, 22(14) [10.3390/ijms22147564].

Hypoxia enhances the expression of RNASET2 in human monocyte-derived dendritic cells: Role of PI3K/AKT pathway

Monaci, S.;Coppola, F.;Giuntini, G.;Carraro, F.;Naldini, A.
2021-01-01

Abstract

Hypoxia is a key component of the tumor microenvironment (TME) and promotes not only tumor growth and metastasis, but also negatively affects infiltrating immune cells by impairing host immunity. Dendritic cells (DCs) are the most potent antigen-presenting cells and their biology is weakened in the TME in many ways, including the modulation of their viability. RNASET2 belongs to the T2 family of extracellular ribonucleases and, besides its nuclease activity, it exerts many additional functions. Indeed, RNASET2 is involved in several human pathologies, including cancer, and it is functionally relevant in the TME. RNASET2 functions are not restricted to cancer cells and its expression could be relevant also in other cell types which are important players in the TME, including DCs. Therefore, this study aimed to unravel the effect of hypoxia (2% O2) on the expression of RNASET2 in DCs. Here, we showed that hypoxia enhanced the expression and secretion of RNASET2 in human monocyte-derived DCs. This paralleled the HIF-1α accumulation and HIF-dependent and-independent signaling, which are associated with DCs’ survival/autophagy/apoptosis. RNASET2 expression, under hypoxia, was regulated by the PI3K/AKT pathway and was almost completely abolished by TLR4 ligand, LPS. Taken together, these results highlight how hypoxia-dependent and-independent pathways shape RNASET2 expression in DCs, with new perspectives on its implication for TME and, therefore, in anti-tumor immunity.
2021
Monaci, S., Coppola, F., Giuntini, G., Roncoroni, R., Acquati, F., Sozzani, S., et al. (2021). Hypoxia enhances the expression of RNASET2 in human monocyte-derived dendritic cells: Role of PI3K/AKT pathway. INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, 22(14) [10.3390/ijms22147564].
File in questo prodotto:
File Dimensione Formato  
ijms-22-07564.pdf

accesso aperto

Tipologia: PDF editoriale
Licenza: Creative commons
Dimensione 1.98 MB
Formato Adobe PDF
1.98 MB Adobe PDF Visualizza/Apri

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11365/1185861