Share this post on:

Ive regulatory CD4+CD25+ cells following A.C.NPs-legumain and empty A.C.NPs treatment is significantly lower than that for the other three groups (Fig. S2A, B). This implies that suppression of T cells immunity by A.C.NPs might contribute to the protective effect of oral legumain DNA vaccine against tumors.DiscussionDevelopment of non-toxic and efficient delivery systems has always been a challenge with the application of DNA vaccines [32,33]. As a result, the biocompatible and biodegradable features of chitosan nanoparticles have gained more attention as a carrier for oral DNA vaccination [34]. Although many efforts have been put into the design and modification of chitosan nanoparticles, a viable oral delivery system for DNA vaccines is still lacking [33]. One major obstacle to achieving satisfactory oral MedChemExpress Rubusoside vaccinations in the DNA degradation results when exposed to the acidic gastric environment and the subsequent decreased uptake via lymphoid tissue in the gastrointestinal tract [35,36,37]. We demonstrated here, that compared with plain chitosan nanoparticles, A.C.NPs protect DNA against degradation in stimulated gastric fluid (pH 1.5). At pH 7.0, alginic acid forms amide bonds with DNA-loaded chitosan nanoparticles by reaction of alginic acid carboxyl groups with chitosan amino groups. Interestingly, after modification with alginic acid, DNAloaded chitosan nanoparticles demonstrate a distinct trend as pH values increase. A.C.NPs tend to aggregate into micrometerscale complexes at pH 1.5, whereas they disperse into smaller particles as pH increases. This difference might be attributed to the hydrolysis of the amide bond in the acidic condition and the subsequent decomposition of the K162 site cross-linked structure between the carboxyl groups of alginic acid and amino groups of chitosan. Excess alginic acid is insoluble at pH 1.5 and potentially forms an amorphous shell on the nanoparticle surface; protecting DNA against enzymatic and acidic degradation (Fig. 9A). Uptake and expression by antigen-presenting cells in Peyer’s patches is a critical step toward successful oral DNA vaccination [18,20]. We visualized the expression of DNA plasmids when loaded in diverse delivery carriers using enhanced green fluorescence protein DNA plasmids. Our results suggest that after leaving the acidic gastric environment and entering the intestines, DNA loaded in A.C.NPs are effectively taken up and expressed by the Peyer’s patches. Expression of EGFP was predominately located 15755315 in macrophages and dendritic cells. It appears as though modification with alginic acid largely increased the amount of EGFP expression in Peyer’s patches after oral administration (illustrated schematically, Fig. 9B). The absorption promoting effect of C.NPs has been extensively studied. Chitosan has been shown to exhibit increased mucoadhesion due to electrostatic interaction, as well as influence the transient opening of tight junctions; both of which may promote the transportation of a loaded DNA vaccine across the mucosal barrier [38]. In addition, studies also indicated that chitosan uptake increases the accumulation and activation of macrophages, providing an extra advantage in improving vaccination efficiency. A noninvasive, controllable, stable, and effective DNA vaccine for cancer therapy would offer a new therapeutic approach. We utilize a full-length legumain DNA vaccine and the murineEffect of A.C.NPs Loaded with Legumain DNA on Tumor GrowthTo further evaluate the protective.Ive regulatory CD4+CD25+ cells following A.C.NPs-legumain and empty A.C.NPs treatment is significantly lower than that for the other three groups (Fig. S2A, B). This implies that suppression of T cells immunity by A.C.NPs might contribute to the protective effect of oral legumain DNA vaccine against tumors.DiscussionDevelopment of non-toxic and efficient delivery systems has always been a challenge with the application of DNA vaccines [32,33]. As a result, the biocompatible and biodegradable features of chitosan nanoparticles have gained more attention as a carrier for oral DNA vaccination [34]. Although many efforts have been put into the design and modification of chitosan nanoparticles, a viable oral delivery system for DNA vaccines is still lacking [33]. One major obstacle to achieving satisfactory oral vaccinations in the DNA degradation results when exposed to the acidic gastric environment and the subsequent decreased uptake via lymphoid tissue in the gastrointestinal tract [35,36,37]. We demonstrated here, that compared with plain chitosan nanoparticles, A.C.NPs protect DNA against degradation in stimulated gastric fluid (pH 1.5). At pH 7.0, alginic acid forms amide bonds with DNA-loaded chitosan nanoparticles by reaction of alginic acid carboxyl groups with chitosan amino groups. Interestingly, after modification with alginic acid, DNAloaded chitosan nanoparticles demonstrate a distinct trend as pH values increase. A.C.NPs tend to aggregate into micrometerscale complexes at pH 1.5, whereas they disperse into smaller particles as pH increases. This difference might be attributed to the hydrolysis of the amide bond in the acidic condition and the subsequent decomposition of the cross-linked structure between the carboxyl groups of alginic acid and amino groups of chitosan. Excess alginic acid is insoluble at pH 1.5 and potentially forms an amorphous shell on the nanoparticle surface; protecting DNA against enzymatic and acidic degradation (Fig. 9A). Uptake and expression by antigen-presenting cells in Peyer’s patches is a critical step toward successful oral DNA vaccination [18,20]. We visualized the expression of DNA plasmids when loaded in diverse delivery carriers using enhanced green fluorescence protein DNA plasmids. Our results suggest that after leaving the acidic gastric environment and entering the intestines, DNA loaded in A.C.NPs are effectively taken up and expressed by the Peyer’s patches. Expression of EGFP was predominately located 15755315 in macrophages and dendritic cells. It appears as though modification with alginic acid largely increased the amount of EGFP expression in Peyer’s patches after oral administration (illustrated schematically, Fig. 9B). The absorption promoting effect of C.NPs has been extensively studied. Chitosan has been shown to exhibit increased mucoadhesion due to electrostatic interaction, as well as influence the transient opening of tight junctions; both of which may promote the transportation of a loaded DNA vaccine across the mucosal barrier [38]. In addition, studies also indicated that chitosan uptake increases the accumulation and activation of macrophages, providing an extra advantage in improving vaccination efficiency. A noninvasive, controllable, stable, and effective DNA vaccine for cancer therapy would offer a new therapeutic approach. We utilize a full-length legumain DNA vaccine and the murineEffect of A.C.NPs Loaded with Legumain DNA on Tumor GrowthTo further evaluate the protective.

Share this post on: