Share this post on:

(F) The proportion of DQ-BSA-labeled p62-good mycobacteria in DC2.4 cells. Data symbolize the imply and SD of three impartial experiments.Determine 5. Localization of MHC class II to mycobacterial autophagosomes in DC. (A, C) Localization of MHC class II to p62positive or ubiquitin-constructive mycobacteria in JAWSII cells.1354825-62-9 chemical information JAWSII cells had been contaminated with Alexa Fluor 405-labeled M. tuberculosis (blue) for 24 h and immunostained with anti-MHC class II (eco-friendly) and anti-p62 antibodies (pink) (A) or anti-MHC class II (green) and anti-ubiquitin antibodies (red) (C). (B, D) The proportion of MHCII-localized p62-optimistic (B) orubiquitin-good (D) mycobacteria in JAWSII cells. Information represent the suggest and SD of 3 unbiased experiments that autophagosomes had been fashioned in reaction to mycobacteria an infection in macrophages [36]. Recently, it was demonstrated that M. tuberculosis infection induced autophagy but impaired the autophagic flux in human principal macrophages and DC [twenty,40]. In this study, we demonstrated that autophagosome markers localized to M. tuberculosis in BMDC but not in BMM (Determine 1). In DC cell strains, we also showed that autophagosomes are formed in reaction to M. tuberculosis infection (Determine two), and identified that lysosomal vesicles fuse with mycobacterial autophagosomes in DC (Determine 4). Since autophagy is believed to be associated in antigen presentation via MHC II [22], it is assumed that autophagy promotes the presentation of mycobacterial antigens in DC. In this examine, we shown that M. tuberculosis an infection induced selective autophagy in DC and that mycobacterial autophagosomes fuse with lysosomes followed by the recruitment of MHC II. These benefits suggest that the autophagosome formation in DC encourages degradation of infected mycobacteria and the resulting degradative peptides are then loaded on to MHC II, which are recruited to mycobacterial autophagosomes, top the antigen presentation of mycobacterial peptides to CD4+ T lymphocytes through MHC II. What is the original celebration that triggers the induction of autophagosome formation to mycobacteria in phagocytic cells The ubiquitination of microorganisms or bacterial phagosomes is essential for selective autophagy because ubiquitinated substrates are connected with the autophagy adaptor proteins which recruit LC3 and autophagosome membranes [41]. In Mycobacterium marinum, the ubiquitination of this bacterium is dependent upon the ESAT-six and ESX-1 secretion program [42]. ESAT-6 of M. marinum disrupts the phagosomal membranes to support bacilli escape from phagosomes to the cytosol [43-forty six].Determine six. p62-dependent ubiquitination of mycobacteria in DC. (A) The proportion of LC3, p62 or ubiquitin recruitment to mycobacteria in DC dealt with with 3-MA. DC2.four were contaminated with DsRed-expressing M. tuberculosis for 24 h with or with no 3-MA and immunostained with anti-LC3, anti-p62 or anti-ubiquitin antibodies. Information symbolize the mean and SD of a few or four impartial experiments. p < 0.05 (unpaired Student's t-test). (B) Immunoblot analysis on the silencing effects of p62 and Atg5. DC2.4 cells were transfected with siRNA for p62 or Atg5 for 48 h and subjected to immunoblot analysis using indicated antibodies. (C, D) The proportion of p62 or ubiquitin recruitment to mycobacteria in DC. DC2.4 cells transfected with siRNA for p62 or Atg5 were infected with DsRed-expressing M. tuberculosis for 12 h and immunostained with anti-p62 (C) or anti-ubiquitin (D) antibody. Data represent the mean and SD of three independent experiments. p < 0.05 (unpaired Student's t-test).M. tuberculosis can also damage the phagosome membrane and translocate from phagosomes to the cytosol in a mechanism that is dependent upon the ESAT-6 and ESX-1 secretion system [47,48]. In macrophages, the permeabilization of the phagosomal membrane signals the ubiquitination of M. tuberculosis [36,49,50]. Our results suggest that the initial step for ubiquitination of mycobacteria in DC is also triggered by the permeabilization of phagosomal membrane by ESAT-6 because BCG bacilli were not ubiquitinated in DC (data not shown). Recently, an E3 ligase was identified that interacts with NDP52 and is responsible for ubiquitination of Salmonella [51]. Manzanillo et al. demonstrated that a ubiquitin ligase, Parkin mediates the ubiquitination of intracellular bacteria in macrophages [50]. Results from the current study suggest the possibility that p62 mediates the recruitment of an E3 ligase for ubiquitination of M. tuberculosis bacilli or bacilli-containing phagosomes in DC.Figure 7. Atg5-dependent localization of LAMP1 and MHC class II to mycobacterial autophagosomes. (A-C) Thin-section electron micrograph of M. tuberculosis bacilli in p62- or Atg5-knockdown DC. DC2.4 cells transfected with siRNA for control (A), p62 (B) or Atg5 (C) were infected with M. tuberculosis for 24 h and observed by thin-section electron microscopy. Autophagosomes are indicated by arrowheads. (D) Proportion of mycobacteria in multi-membrane structures in DC2.4 transfected with siRNA for p62 or Atg5. (E, F) The proportion of LAMP1 (E) or MHC class II (F) localization to ubiquitin-positive mycobacteria is shown. JAWSII cells transfected with control or Atg5 siRNA for 24 h were infected with Alexa Fluor 405-labeled M. tuberculosis and immunostained with anti-LAMP1 and anti-ubiquitin antibodies or anti-MHC class II and anti-ubiquitin antibodies. Data represent the mean and SD of three independent experiments. p < 0.05 (unpaired Student's t-test).Atg5 is involved in the elongation of isolation membrane in autophagosome formation [52]. We did not observe autophagosome membrane structures around infecting mycobacteria in DC by thin-section electron microscopy, however p62 and ubiquitin were recruited to mycobacteria in Atg5-knockdown DC (Figures 6 and 7). These results suggest that p62 and ubiquitin are recruited to infecting mycobacteria followed by the formation of autophagosome membrane depending on the function of Atg5. This hypothesis is consistent with the results that lysosomal markers do not localize to ubiqutinated mycobacteria in Atg5-knockdown DC (Figure 7). Atg16L is another autophagic-related protein involved in elongation of autophagic isolation membrane by forming an Atg5-Atg12-Atg16L complex [53]. We found that ubiquitin is recruited to M. tuberculosis but that LAMP1 does not localize to ubiquitinated mycobacteria in Atg16Lknockdown DC (Figure S5). These results suggest that ubiquitination of infected mycobacteria in DC is followed by formation of the autophagosome membrane and the autolysosome. We found that autophagosomal markers did not localize to infected mycobacteria in macrophages, but did so in DC (Figure 1). These results imply that phagosomal membranes of DC are more susceptible to ESAT-6 and/or other secreted proteins from M. tuberculosis than those of macrophages. We have previously demonstrated that depletion of Coronin-1a leads the autophagosome formation to infecting M. tuberculosis in macrophages [26]. Coronin-1a associates with F-actin and localizes to mycobacterial phagosomes [54,55], suggesting that Coronin-1a supports the phagosomal membranes in macrophages but not in DC. However, proteomic analysis revealed that Coronin-1a localizes to mycobacterial phagosomes in both macrophages and DC [56], suggesting that other proteins localizing to mycobacterial phagosomes in macrophages but not in DC would contribute the autophagosome formation to infecting mycobacteria. In conclusion, M. tuberculosis infection in DC induced autophagosome formation followed by the fusion with lysosomes and MHC II recruitment. p62 and Atg5 function in the initiation and progression of autophagosome formation to M. tuberculosis, respectively. Thus, p62 mediates the ubiquitination of M. tuberculosis and Atg5 is involved in the fusion of lysosomes with mycobacterial autophagosomes. These results imply that autophagosome formation in M. tuberculosis-infected DC contributes to activate CD4+ T lymphocytes via MHC II antigen presentation.siRNA for autophagy-related genes. JAWSII cells transfected with siRNA for p62 or Atg5 genes for 48 h were subjected to immunoblot analysis using the indicated antibodies. (B) The proportion of ubiquitinated mycobacteria in JAWSII cells. JAWSII cells transfected with siRNA for p62 or Atg5 were infected with DsRed-expressing M. tuberculosis for 24 h and immunostained with anti-ubiquitin antibody. Data represent the mean and SD of three independent experiments. p < 0.05 (unpaired Student's t-test). (TIF) Figure S5. Ubiquitination of mycobacteria in Atg16Lknockdown DC. (A) Immunoblot analysis of DC2.4 cells transfected with siRNA for Atg16L. DC2.4 cells were transfected with Atg16L siRNA for 48 h and subjected to immunoblot analysis using anti-Atg16L antibody. (B) The proportion of ubiquitinated mycobacteria in Atg16L-knockdown DC. DC2.4 cells transfected with siRNA for Atg16 were infected with DsRed-expressing M. tuberculosis for 24 h and immunostained with anti-ubiquitin antibody. (C) The proportion of LAMP1 localization to ubiqutinated mycobacteria in Atg16Lknockdown DC. DC2.4 cells transfected with control or Atg16 siRNA for 24 h were infected with Alexa Fluor 405-labeled M. tuberculosis and immunostained with anti-LAMP1 and antiubiquitin antibodies. Data represent the mean and SD of three independent experiments. p < 0.05 (unpaired Student's t-test).The envelope glycoprotein (Env) of human immunodeficiency virus 1 (HIV-1) binds to receptors and triggers conformational changes to initiate viral infection. The entry of HIV-1 into target cells requires fusion between the viral and cellular membranes, which is mediated by the Env [1,2]. Env is expressed as a heavily glycosylated precursor (gp160) that is cleaved intracellularly into two non-covalently-associated functional subunits: an extracellular subunit (gp120), responsible for CD4 and coreceptor (primarily CCR5 and/or CXCR4) binding, and a transmembrane subunit (gp41) that mediates the fusion between the viral and host cell membranes. HIV-1 Env is highly variable in five sequence regions (variable regions, V1 to V5) and highly conserved in other regions (conserved regions, C1 to C5). The HIV-1 Env polymorphism reflects the selective pressures of the immune system on viruses [3] and assists in virus escape. In contrast, Env conservation reflects the stability of virus evolution and it is necessary for maintaining virus function. Notably, the gp120 V4 region is highly polymorphic in length, amino acid composition, and glycosylation [4]. However, the biological significance of the extensive evolution of the V4 region and its association with viral infectivity have not been well established. Some studies have revealed that the V4 region is associated with disease progression in SIV and HIV infection [7]. The sequences within the V4-V5 regions in subjects with slow CD4 cell loss were more variable than in subjects with rapid CD4 cell loss [7]. The envelopes isolated from individuals infected with subtype C HIV-1 demonstrated that slow progressors had a significantly longer C3-V4 region compared with progressors [8]. In addition, rapid progressors among SIV-infected macaques exhibited high diversity in the V4 region [9]. Furthermore, neutralization escape has been associated with amino acid substitutions and sequence insertion/deletion in the V4 region in SIV [10,11] andaqqa HIV infection [12]. During HIV-1 infection, the disease progression is primarily associated with massive damage of target cells caused by the successful entry of viruses and the infection of target cells. It was found that the V45 regions may play an important role in affecting fusion efficiency [13], and the V4 mutations at positions 407D and 386N can modulate resistance to CCR5 antagonists [14]. Moreover, V4 variation correlates with coreceptor preference because a CCR5-tropic virus strain with a V4-V5 region replaced by the V4-V5 region from a CXCR4-tropic virus can utilize CXCR4 [15], and rapid and extensive changes in the amino acid sequences in the V4-V5 regions have also been demonstrated to accompany coreceptor switching from CCR5 to R5X4 [16]. It is still unclear how the highly variable V4 region affects disease progression and whether it is a result of affecting virus entry capacity. The identification of concrete amino acid residues or subregions in the V4 region corresponding to the Env functions to find a potentially novel target for HIV-1 entry inhibitors is worthwhile. In this study, we focused on exploring the correlation between the polymorphism of V4 region and its influence on virus entry, and the possible mechanisms for how the HIV-1 gp120 V4 region contributes to virus infectivity fetal calf serum (FCS), 2 mM L-glutamine, 0.1 mM non-essential amino acids (GibcoBRL, Grand Island, NY, USA), 110 mg/l sodium pyruvate (GibcoBRL, Grand Island, NY, USA), 100 mg/ ml penicillin, and 100 mg/ml streptomycin (GibcoBRL, Grand Island, NY, USA). Human astroglia cell lines expressing CD4 and the coreceptors CXCR4 (U87.CD4.CXCR4) or CCR5 (U87.CD4.CCR5) were obtained from the AIDS Research and Reference Reagent Program and maintained in DMEM 6502210and 10% FCS [15]. We added 300 mg/ml G418 (GibcoBRL, Grand Island, NY, USA) and 1 mg/ml puromycin (Sigma, St. Louis, MO, USA) as the selection reagents for CD4 and the coreceptors.Two well-characterized R5-tropic and X4-tropic HIV-1 strains, ADA and HXB2, were used in this study. We constructed a series of gp160 mutants by either deleting a portion or substituting an individual residue of the V4 region and maintaining the two terminal cysteines to retain the loop conformation. To generate luciferase-reporter Env-pseudoviruses, 293T cells were co-transfected with the expressing plasmid pNL4-3-Luc-E-R- (provided by Dr D.R. Littman) and each of the gp160-expressing plasmids [18]. The pseudoviruses were filtered with 0.45 mM membrane filters and quantified using the P24 antigen ELISA assay (ZeptoMetrix Corporation, Buffalo, NY, USA). The coreceptor usage and the infectivity of the pseudoviruses with wild-type (wt) or V4-mutated Env glycoproteins were examined in a single-round infection assay [18]. Pseudoviruses, at 20 ng or 40 ng of P24, were used to infect 26104 U87.CD4.CCR5 or U87.CD4.CXCR4 cells grown in 48well plates in triplicate. The luciferase activity (relative light units, RLU) was examined 48 h post-infection using a luminometer (Turner 20/20n, Turner Designs, Mountain View, CA, USA) and the Luciferase Assay System (Promega, Madison, WI, USA). The experiments were performed three to five times.

Share this post on:

Author: M2 ion channel