The suppressed immune responses mediated by liver-infiltrating im

The suppressed immune responses mediated by liver-infiltrating immune cells may also play a key role. In this study, decreased infiltration of F4/80+

macrophages was observed in DEN-treated TLR2-mutant livers. This finding is consistent with the suppression of immune signaling pathways and cytokine production in the TLR2-mutant livers. However, these results do not agree with a recent report13 in which TLR2 deficiency was shown to enhance tumor development in a mouse model of colitis-induced cancer by increasing the number of colon-infiltrating inflammatory cells and the production of inflammatory cytokines in local tissues. Obviously, the difference between the two studies can be attributed to the use of different animal models. In the DEN-induced HCC model, sterile inflammation Dabrafenib order can be induced by the unfolding protein response to oxidative/ER stress or by PRRs, such as TLR2, interacting

with DAMPs released from the damaged liver cells. In the FK228 colitis-induced cancer model, the microbial infection recruits a large number of inflammatory cells to the tissue; the tissue-infiltrating inflammatory cells produce inflammatory cytokines, such as IL-6 and IL-17, to promote cancer development.13, 34 Interestingly, TLR2 mutations promote tumorigenesis in two different cancer models. These studies indicate the complexity of the role of TLR2 activity learn more in the regulation of tumor development. In future studies, it will be worthwhile to determine how TLR2 mutations can affect communication between immune and liver parenchymal cells, particularly as it relates to dissecting the significance of TLR2 in the regulation of HCC development for individual hepatic cell populations

using bone marrow chimeras and other molecular approaches.35 Our study demonstrates a critical protective role of TLR2-mediated p62-dependent activation of autophagy in DEN-induced tumorigenesis through the clearing of intracellularly accumulated ROS and p62 aggregates. Recent studies indicate that the accumulated ROS and p62 aggregates can form a positive feedback loop, and each of these factors is toxic to the liver and acts as a trigger for HCC development.23, 36 By clearing p62 aggregates from the cell, autophagy protects the liver from ROS-related ER stress, DNA damage, and carcinogenesis.23 Moreover, a link has been recently established between autophagy and cellular senescence: autophagy is a consequence of cellular senescence, and it can also trigger cellular senescence.19 Indeed, TLR2 deficiency-attenuated senescence and suppressed autophagy flux can be reversed by the administration of IFN-γ, a positive modulator of senescence and autophagy.

Comments are closed.