Moreover,

in vitro binding studies demonstrated that NRP-

Moreover,

in vitro binding studies demonstrated that NRP-1 increases PDGF binding affinity for PDGFR-expressing cells. HSCs from NRP-1–deleted mice exhibited decreased migration in response to PDGF, whereas overexpression of NRP-1 promoted selective activation of Rac1 in the presence of PDGF without affecting Akt and ERK activity. Interestingly, Rac activity was diminished in c-Abl–deficient mouse embryonic fibroblasts overexpressing NRP-1, suggesting that NRP-1 directs the PDGFR signals to Rac1 through its ability to bind and activate c-Abl (Fig. 1). Furthermore, Cao et al. investigate the role of NRP-1 in the regulation of collagen deposition induced by the PDGF and TGF-β LEE011 mw pathways. Surprisingly, both cytokines induce collagen deposition after overexpression of NRP-1. Collagen deposition is inhibited in NRP-1– and c-Abl–deficient mouse embryonic fibroblasts after treatment with PDGF or TGF-β, suggesting the presence of an NRP-1/c-Abl pathway in enhancing collagen deposition. In a recently published parallel study, the same group demonstrated that

NRP-1 mediates R-SMAD signaling via TGFβ11 and suggested that NRP-1 amplifies TGFβ-induced myofibroblast activation by increasing the profibrogenic Smad2/3 pathway and suppressing the antifibrogenic Smad 1/5 pathway. In summary, these studies convincingly establish NRP-1 as an amplifier for profibrogenic signaling pathways such as PDGF and TGF-β, leading to increased HSC activation 上海皓元医药股份有限公司 and fibrosis in the liver. A few questions have yet to be answered, Dorsomorphin concentration however. Culture-activated HSCs and HSC cell lines employed for mechanistic experiments in this study may differ significantly from in vivo–activated HSCs.12, 13 In this regard, additional in vivo studies may be helpful to further delineate whether NRP-1 promotes HSC activation and liver fibrosis acting through its role as a VEGF and semaphorin coreceptor. Notably, the two antibodies employed in the present studies differ in their epitope binding,

with NRP-1a blocking semaphoring binding and NRP-1b blocking VEGF binding. Because NRP-1b antibody reduced CCl4-induced liver fibrosis, one needs to consider whether the VEGF blocking abilities of this antibody played a role in the improved fibrosis observed in vivo. Importantly, angiogenesis has been suggested to contribute to hepatic fibrosis.14 Although Cao et al. investigated the role of NRP-1 in regulating the ability of HSCs to promote the formation of vascular tubes, they did not investigate angiogenesis in vivo. Moreover, the current study did not employ genetic methods to inhibit NRP-1 expression in vivo. The floxed NRP-1 mice employed for in vitro experiments in this study should ideally be used to delete NRP-1 in HSCs during liver fibrosis. Finally, it is intriguing that NRP-1 was strongly up-regulated in HSCs from CCl4-treated livers but only very moderately in HSCs isolated form bile duct–ligated livers.

Comments are closed.