首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 0 毫秒
1.
Interferon-inducible transmembrane protein 3 (IFITM3) is essential for innate defense against influenza virus in mice and humans. IFITM3 localizes to endolysosomes where it prevents virus fusion, although mechanisms controlling its trafficking to this cellular compartment are not fully understood. We determined that both mouse and human IFITM3 are phosphorylated by the protein-tyrosine kinase FYN on tyrosine 20 (Tyr20) and that mouse IFITM3 is also phosphorylated on the non-conserved Tyr27. Phosphorylation led to a cellular redistribution of IFITM3, including plasma membrane accumulation. Mutation of Tyr20 caused a similar redistribution of IFITM3 and resulted in decreased antiviral activity against influenza virus, whereas Tyr27 mutation of mouse IFITM3 showed minimal effects on localization or activity. Using FYN knockout cells, we also found that IFITM3 phosphorylation is not a requirement for its antiviral activity. Together, these results indicate that Tyr20 is part of an endocytosis signal that can be blocked by phosphorylation or by mutation of this residue. Further mutagenesis narrowed this endocytosis-controlling region to four residues conforming to a YXXΦ (where X is any amino acid and Φ is Val, Leu, or Ile) endocytic motif that, when transferred to CD4, resulted in its internalization from the cell surface. Additionally, we found that phosphorylation of IFITM3 by FYN and mutagenesis of Tyr20 both resulted in decreased IFITM3 ubiquitination. Overall, these results suggest that modification of Tyr20 may serve in a cellular checkpoint controlling IFITM3 trafficking and degradation and demonstrate the complexity of posttranslational regulation of IFITM3.  相似文献   

2.
The interferon-induced transmembrane (IFITM) family of proteins have recently been identified as important host effector molecules of the type I interferon response against viruses. IFITM1 has been identified as a potent antiviral effector against hepatitis C virus (HCV), whereas the related family members IFITM2 and IFITM3 have been described to have antiviral effects against a broad range of RNA viruses. Here, we demonstrate that IFITM2 and IFITM3 play an integral role in the interferon response against HCV and act at the level of late entry stages of HCV infection. We have established that in hepatocytes, IFITM2 and IFITM3 localize to the late and early endosomes, respectively, as well as the lysosome. Furthermore, we have demonstrated that S-palmitoylation of all three IFITM proteins is essential for anti-HCV activity, whereas the conserved tyrosine residue in the N-terminal domain of IFITM2 and IFITM3 plays a significant role in protein localization. However, this tyrosine was found to be dispensable for anti-HCV activity, with mutation of the tyrosine resulting in an IFITM1-like phenotype with the retention of anti-HCV activity and co-localization of IFITM2 and IFITM3 with CD81. In conclusion, we propose that the IFITM proteins act in a coordinated manner to restrict HCV infection by targeting the endocytosed HCV virion for lysosomal degradation and demonstrate that the actions of the IFITM proteins are indeed virus and cell-type specific.  相似文献   

3.
Previous studies revealed an essential role for the lipid-binding Sec14 domain of kalirin (KalSec14), but its mechanism of action is not well understood. Because alternative promoter usage appends unique N-terminal peptides to the KalSec14 domain, we used biophysical, biochemical, and cell biological approaches to examine the two major products, bKalSec14 and cKalSec14. Promoter B encodes a charged, unstructured peptide, whereas promoter C encodes an amphipathic helix (Kal-C-helix). Both bKalSec14 and cKalSec14 interacted with lipids in PIP strip and liposome flotation assays, with significantly greater binding by cKalSec14 in both assays. Disruption of the hydrophobic face of the Kal-C-helix in cKalSec14KKED eliminated its increased liposome binding. Although cKalSec14 showed significantly reduced binding to liposomes lacking phosphatidylinositol phosphates or cholesterol, liposome binding by bKalSec14 and cKalSec14KKED was not affected. When expressed in AtT-20 cells, bKalSec14-GFP was diffusely localized, whereas cKalSec14-GFP localized to the trans-Golgi network and secretory granules. The amphipathic C-helix was sufficient for this localization. When AtT-20 cells were treated with a cell-permeant derivative of the Kal-C-helix (Kal-C-helix-Arg9), we observed increased secretion of a product stored in mature secretory granules, with no effect on basal secretion; a cell-permeant control peptide (Kal-C-helixKKED-Arg9) did not have this effect. Through its ability to control expression of a novel, phosphoinositide-binding amphipathic helix, Kalrn promoter usage is expected to affect function.  相似文献   

4.
Interferon inducible transmembrane proteins (IFITMs) are broad‐spectrum antiviral factors. In cell culture the entry of many enveloped viruses, including orthomyxo‐, flavi‐, and filoviruses, is inhibited by IFITMs, though the mechanism(s) involved remain unclear and may vary between viruses. We demonstrate that Sindbis and Semliki Forest virus (SFV), which both use endocytosis and acid‐induced membrane fusion in early endosomes to infect cells, are restricted by the early endosomal IFITM3. The late endosomal IFITM2 is less restrictive and the plasma membrane IFITM1 does not inhibit normal infection by either virus. IFITM3 inhibits release of the SFV capsid into the cytosol, without inhibiting binding, internalization, trafficking to endosomes or low pH‐induced conformational changes in the envelope glycoprotein. Infection by SFV fusion at the cell surface was inhibited by IFITM1, but was equally inhibited by IFITM3. Furthermore, an IFITM3 mutant (Y20A) that is localized to the plasma membrane inhibited infection by cell surface fusion more potently than IFITM1. Together, these results indicate that IFITMs, in particular IFITM3, can restrict alphavirus infection by inhibiting viral fusion with cellular membranes. That IFITM3 can restrict SFV infection by fusion at the cell surface equivalently to IFITM1 suggests that IFITM3 has greater antiviral potency against SFV.   相似文献   

5.
The interferon‐induced transmembrane (IFITM) proteins protect host cells from diverse virus infections. IFITM proteins also incorporate into HIV‐1 virions and inhibit virus fusion and cell‐to‐cell spread, with IFITM3 showing the greatest potency. Here, we report that amino‐terminal mutants of IFITM3 preventing ubiquitination and endocytosis are more abundantly incorporated into virions and exhibit enhanced inhibition of HIV‐1 fusion. An analysis of primate genomes revealed that IFITM3 is the most ancient antiviral family member of the IFITM locus and has undergone a repeated duplication in independent host lineages. Some IFITM3 genes in nonhuman primates, including those that arose following gene duplication, carry amino‐terminal mutations that modify protein localization and function. This suggests that “runaway” IFITM3 variants could be selected for altered antiviral activity. Furthermore, we show that adaptations in IFITM3 result in a trade‐off in antiviral specificity, as variants exhibiting enhanced activity against HIV‐1 poorly restrict influenza A virus. Overall, we provide the first experimental evidence that diversification of IFITM3 genes may boost the antiviral coverage of host cells and provide selective functional advantages.  相似文献   

6.
Interferon-inducible transmembrane (IFITM) protein family members IFITM1, -2, and -3 restrict the infection of multiple enveloped viruses. Significant enrichment of a minor IFITM3 allele was recently reported for patients who were hospitalized for seasonal and 2009 H1N1 pandemic flu. This IFITM3 allele lacks the region corresponding to the first amino-terminal 21 amino acids and is unable to inhibit influenza A virus. In this study, we found that deleting this 21-amino-acid region relocates IFITM3 from the endosomal compartments to the cell periphery. This finding likely underlies the lost inhibition of influenza A virus that completes its entry exclusively within endosomes at low pH. Yet, wild-type IFITM3 and the mutant with the 21-amino-acid deletion inhibit HIV-1 replication equally well. Given the pH-independent nature of HIV-1 entry, our results suggest that IFITM3 can inhibit viruses that enter cells via different routes and that its N-terminal region is specifically required for controlling pH-dependent viruses.  相似文献   

7.
GroEL is an essential Escherichia coli molecular chaperon that uses ATP to facilitate correct folding of a range of proteins in a cell. Central to the GroEL substrate diversity is how GroEL recognizes the substrates. The interaction between GroEL and substrate has been proposed to be largely hydrophobic because GroEL interacts with proteins in non-native conformations but not in native forms. Analysis of GroEL substrate proteins reveals that one of its main substrates are proteins with αβ folding domains, suggesting that GroEL may stabilize the collapsed αβ core by binding to hydrophobic surfaces that are usually buried between the α and β elements. In this study, we characterize the interaction between GroEL and a peptide derived from our previous selection via a phage display method. NMR studies map the peptide-binding site to the region containing Helices H and I, which is consistent with evidence that this region comprises the primary substrate-binding site. The peptide is largely unstructured in solution but adopts a helical conformation when bound to the GroEL apical domain with a moderate affinity (Kd = 17.1 ± 2.5 μm). The helical conformation aligns residues to form an amphipathic structure, and the hydrophobic side of this amphipathic helix interacts with GroEL as suggested by fluorescence quenching studies. Together with previous structural studies on the GroEL-peptide complexes, our work supports the notion that the amphipathic secondary elements in the substrate proteins may be the structural motif recognized by GroEL.The bacterial chaperonin GroEL and its co-chaperonin GroES are essential for cell viability by assisting folding of a wide range of proteins via an ATP-dependent mechanism (13). Structurally, fourteen 57-kDa GroEL subunits assemble into two back-to-back stacking heptameric rings, giving rise to two functionally independent central cavities (4). Each GroEL subunit folds into three distinctive domains: equatorial domain, intermediate domain, and apical domain. The equatorial domains contain the ATP-binding sites and provide most of the intra-ring interactions and all the inter-ring interactions. The apical domains form the rims of the central cavities and contain the binding sites for the substrate proteins and GroES. The intermediate domains link the apical domains and the equatorial domains. For the co-chaperonin GroES, seven GroES subunits, of 10 kDa each, assemble into a heptamer ring (5, 6). In forming the GroEL-GroES complex, GroES caps one end of GroEL, and large structural changes are observed in both GroEL and GroES (7). In GroEL, the apical domain is rotated 90° along its axis and 60° upwards, and the intermediate domain is closed down ∼25° to the equatorial domain. A loop in GroES (residues 17–33) that is unstructured in the isolated GroES adopts a β-turn structure and forms contact with the GroEL apical domain. Compared with the unliganded GroEL, the volume of the enclosed GroEL-GroES cavity is doubled, and the surface lining the wall of the GroEL cavity changes from hydrophobic to hydrophilic.A wealth of information derived from both intensive biochemical and structural characterizations has revealed a general role of GroEL-GroES in assisting protein folding (see reviews in Refs. 3, 8, and 9). Briefly, GroEL binds the substrate proteins in their aggregation-prone non-native states, preventing them from aggregating. Binding of ATP to the substrate occupied GroEL ring (cis-ring) presumably induces large conformational change in GroEL that promotes binding of GroES to the cis-ring. As a result of ATP and GroES binding, the substrate protein is displaced into the GroEL central cavity, initiating the folding process. Both hydrolysis of ATP in the cis-ring and binding of ATP to the substrate unoccupied ring (trans ring) weaken the GroES-GroEL interaction, and ATP binding to the trans ring results in the dissociation of GroES from GroEL, releasing substrate from the central cavity of GroEL. The released substrate may continue folding into the native state if in a folding competent state or may rebind to GroEL if it is still misfolded.One of the most intriguing aspects of the GroE-assisted folding is the substrate promiscuity. It has been shown that about 300 Escherichia coli proteins can interact with GroEL, and these proteins are diverse in terms of both structures and functions (10). A range of techniques have been applied to investigate this important yet complex aspect, and salient features regarding GroEL-substrate interactions have emerged. The apical domains, on the rim of the GroEL central cavity, contain the main substrate-binding site (1113). Structural flexibility, reflected by both high temperature factors of the apical domain in the crystal structure of tetradecameric GroEL (14) and conformational multiplicity around Helix H and I (15), is proposed to account for the diverse spectrum of GroEL substrates. Mutational studies on GroEL suggest that the GroEL-substrate interactions are largely hydrophobic (16). Structural study on GroEL-substrate interaction, however, is hindered mainly because of the multiple conformations of the bound substrate protein. Very recently, NMR techniques have been used to directly investigate the bound conformations of the substrate (17, 18); yet the nature of GroEL-substrate interaction is not revealed. Peptides may mimic segments of substrate proteins, and studies of GroEL-peptide interactions have uncovered detailed intermolecular interactions and provided insights into principles of substrate recognition by GroEL. The bound peptides may adopt α-helix (1923), β-hairpin (15), or extended conformations (24), and despite different conformations, they all appear to bind to Helix H and I of GroEL. Hydrophobic interaction dominates the interface between GroEL and peptides in either β-hairpin or extended structures and is proposed so between GroEL and α-helical peptides. These detailed structural characterizations on GroEL-peptide interactions have contributed to dissecting the complex nature of the substrate recognition by GroEL (25).We previously identified a high affinity peptide (strong binding peptide (SBP))2 for GroEL using a phage display method and found that SBP adopts a β-hairpin structure bound to GroEL (15, 26). To investigate the contribution of the β-turn in SBP to the GroEL-SBP interaction, we have created various SBP variants with the intension to disrupt the β-turn structure and have studied their binding to GroEL. One of the peptides (termed SBP-W2DP6V), however, adopts a helical conformation when bound to GroEL by NMR analysis. NMR results also map the peptide-binding site on GroEL to be a region formed by Helix H and I. The helical peptide has an amphipathic feature, and fluorescence studies provide direct evidence that the hydrophobic face is involved in the interaction with GroEL. Our structural analysis, combined with previous studies, suggests that GroEL recognizes the amphipathic property in the secondary structures of the substrate protein and binds preferably to the hydrophobic side of these structural elements to stabilize and preserve their structures.  相似文献   

8.
目的:探讨干扰素诱导的跨膜转运蛋白3(IFITM3)在LPS刺激的RAW264.7细胞系的脓毒症模型中的表达以及胆碱能抗炎模型中的表达。方法:用1μg/mL LPS刺激RAW264.7细胞24、48、72 h后,用Western-Blot法检测各组细胞IFITM3蛋白表达水平。用1μg/mL LPS刺激RAW264.7细胞后,给予50μM胆碱能受体激动剂GTS-21以及同时给予100 n M胆碱能受体拮抗剂α-BGT刺激细胞24 h后,用Western-Blot法检测各组细胞IFITM3蛋白表达水平。用ELISA法检测IL-1β的方法验证脓毒症模型和胆碱能抗炎模型的建立。结果:(1)1μg/mL LPS刺激RAW264.7细胞后,IFITM3蛋白表达明显降低(P0.01)。(2)1μg/mL LPS刺激RAW264.7细胞后再给予50μM GTS-21,IFITM3蛋白表达明显升高(P0.001);而给予100 nMα-BGT后,IFITM3蛋白表达明显降低(P0.001)。结论:LPS刺激的RAW264.7细胞IFITM3蛋白表达降低。给予胆碱能激动剂GTS-21后能够逆转LPS诱导的IFITM3表达的降低,给予胆碱能受体拮抗剂α-BGT则能阻断这种现象。IFITM3有可能在脓毒症中发挥保护作用,并且参与了胆碱能抗炎通路抗炎过程的调节。  相似文献   

9.
Interferon‐induced transmembrane proteins (IFITMs) restrict infections by many viruses, but a subset of IFITMs enhance infections by specific coronaviruses through currently unknown mechanisms. We show that SARS‐CoV‐2 Spike‐pseudotyped virus and genuine SARS‐CoV‐2 infections are generally restricted by human and mouse IFITM1, IFITM2, and IFITM3, using gain‐ and loss‐of‐function approaches. Mechanistically, SARS‐CoV‐2 restriction occurred independently of IFITM3 S‐palmitoylation, indicating a restrictive capacity distinct from reported inhibition of other viruses. In contrast, the IFITM3 amphipathic helix and its amphipathic properties were required for virus restriction. Mutation of residues within the IFITM3 endocytosis‐promoting YxxФ motif converted human IFITM3 into an enhancer of SARS‐CoV‐2 infection, and cell‐to‐cell fusion assays confirmed the ability of endocytic mutants to enhance Spike‐mediated fusion with the plasma membrane. Overexpression of TMPRSS2, which increases plasma membrane fusion versus endosome fusion of SARS‐CoV‐2, attenuated IFITM3 restriction and converted amphipathic helix mutants into infection enhancers. In sum, we uncover new pro‐ and anti‐viral mechanisms of IFITM3, with clear distinctions drawn between enhancement of viral infection at the plasma membrane and amphipathicity‐based mechanisms used for endosomal SARS‐CoV‐2 restriction.  相似文献   

10.
Interferon-inducible transmembrane protein 3 (IFITM3) inhibits influenza virus infection by blocking viral membrane fusion, but the exact mechanism remains elusive. Here, we investigated the function and key region of IFITM3 in blocking influenza virus entry mediated by hemagglutinin (HA). The restriction of IFITM3 on HA-mediated viral entry was confirmed by pseudovirus harboring HA protein from H5 and H7 influenza viruses. Subcellular co-localization and immunocoprecipitation analyses revealed that IFITM3 partially co-located with the full-length HA protein and could directly interact with HA2 subunit but not HA1 subunit of H5 and H7 virus. Truncated analyses showed that the transmembrane domain of the IFITM3 and HA2 subunit might play an important role in their interaction. Finally, this interaction of IFITM3 was also verified with HA2 subunits from other subtypes of influenza A virus and influenza B virus. Overall, our data demonstrate for the first time a direct interaction between IFITM3 and influenza HA protein via the transmembrane domain, providing a new perspective for further exploring the biological significance of IFITM3 restriction on influenza virus infection or HA-mediated antagonism or escape.  相似文献   

11.
Interferon‐induced transmembrane protein 3 (IFITM3) is a cellular factor that blocks virus fusion with cell membranes. IFITM3 has been suggested to alter membrane curvature and fluidity, though its exact mechanism of action is unclear. Using a bioinformatic approach, we predict IFITM3 secondary structures and identify a highly conserved, short amphipathic helix within a hydrophobic region of IFITM3 previously thought to be a transmembrane domain. Consistent with the known ability of amphipathic helices to alter membrane properties, we show that this helix and its amphipathicity are required for the IFITM3‐dependent inhibition of influenza virus, Zika virus, vesicular stomatitis virus, Ebola virus, and human immunodeficiency virus infections. The homologous amphipathic helix within IFITM1 is also required for the inhibition of infection, indicating that IFITM proteins possess a conserved mechanism of antiviral action. We further demonstrate that the amphipathic helix of IFITM3 is required to block influenza virus hemagglutinin‐mediated membrane fusion. Overall, our results provide evidence that IFITM proteins utilize an amphipathic helix for inhibiting virus fusion.  相似文献   

12.
Ferroxidase II (Fox II) was developed in serum by acid incubation for 24h. The resulting activity showed a strong positive correlation with the serum cholesterol concentration in normal subjects and patients with hyperlipidaemia. The potentiating effect of cholesterol on developed Fox II has been confirmed by the in-vitro addition of cholesterol to serum. There was no significant correlation between developed Fox II and caeruloplasmin (ferroxidase I) or between cholesterol and caeruloplasmin.  相似文献   

13.
目的:获得IFITM1基因片段并构建真核表达质粒.方法:采用RT-PCR技术扩增IFITM1,将扩增产物连接至pcDNA3.1载体,对重组质粒进行测序验证,结果:构建了真核表达质粒pcDNA3.1-IFITM1,通过酶切、测序等方法验证完全正确.结论:成功构建了IFITM1基因的真核表达质粒,为下一步探讨IFITM1基因在子宫颈癌HeLa细胞中的作用提供了实验基础.  相似文献   

14.
《Free radical research》2013,47(1-5):79-83
Ferroxidase II (Fox II) was developed in serum by acid incubation for 24h. The resulting activity showed a strong positive correlation with the serum cholesterol concentration in normal subjects and patients with hyperlipidaemia. The potentiating effect of cholesterol on developed Fox II has been confirmed by the in-vitro addition of cholesterol to serum. There was no significant correlation between developed Fox II and caeruloplasmin (ferroxidase I) or between cholesterol and caeruloplasmin.  相似文献   

15.
目的:探讨干扰素介导的跨膜蛋白1(Interferon-induced transmembrane protein 1,IFITM1)基因在卵巢上皮性癌中表达的相关性及其意义。方法:应用Western blotting检测正常卵巢、卵巢良性肿瘤、卵巢交界性肿瘤和卵巢上皮性癌组织中IFITM1蛋白表达。免疫组织化学检测12例正常卵巢、21例卵巢良性肿瘤、18例卵巢交界性肿瘤和85例卵巢上皮性癌组织中IFITM1的蛋白表达,同时分析IFITM1表达状况与临床病理因素之间的相关性。结果:Western blotting显示卵巢上皮性癌和卵巢交界性肿瘤中IFITM1表达水平明显高于正常卵巢组织和卵巢良性肿瘤。免疫组化显示在正常卵巢组织中IFITM1阳性表达率为41.7%(5/12),在卵巢良性肿瘤组织中71.4%(15/21),在卵巢交界性肿瘤组织中为72.2%(13/18),在卵巢上皮性癌中为77.6%(66/85),IFITM1蛋白表达强度在正常卵巢、良性卵巢肿瘤、交界性卵巢肿瘤、上皮性卵巢癌间的比较有统计学意义(P0.05)。IFITM1蛋白表达与病理类型、肿瘤分化程度、肿瘤FIGO分期有关(P0.05),与淋巴结转移、腹水无明显相关性。化疗敏感组和耐药组的IFITM1表达强度间差异有统计学意义(P0.05)。结论:IFITM1在正常卵巢、卵巢良性肿瘤、卵巢交界性肿瘤和卵巢上皮性癌组织中的表达依次升高,并与卵巢癌以铂类为基础的化疗耐药性产生有相关性,为进一步研究IFITM1在卵巢癌诊治及化疗中的应用前景提供依据。  相似文献   

16.
C-type lectin receptors (CLRs) are an emerging family of pattern recognition receptors that recognizes pathogens or damaged tissue to trigger innate immune responses. However, endogenous ligands for CLRs are not fully understood. In this study, we sought to identify an endogenous ligand(s) for human macrophage-inducible C-type lectin (hMincle). A particular fraction of lipid extracts from liver selectively activated reporter cells expressing hMincle. MS analysis determined the chemical structure of the active component as cholesterol. Purified cholesterol in plate-coated and crystalized forms activates reporter cells expressing hMincle but not murine Mincle (mMincle). Cholesterol crystals are known to activate immune cells and induce inflammatory responses through lysosomal damage. However, direct innate immune receptors for cholesterol crystals have not been identified. Murine macrophages transfected with hMincle responded to cholesterol crystals by producing pro-inflammatory cytokines. Human dendritic cells expressed a set of inflammatory genes in response to cholesterol crystals, and this was inhibited by anti-human Mincle. Importantly, other related CLRs did not bind cholesterol crystals, whereas other steroids were not recognized by hMincle. These results suggest that cholesterol crystals are an endogenous ligand for hMincle and that they activate innate immune responses.  相似文献   

17.
Immunoadhesins are engineered proteins combining the constant domain (Fc) of an antibody with a ligand‐binding (adhesion) domain. They have significant potential as therapeutic agents, because they maintain the favourable pharmacokinetics of antibodies with an expanded repertoire of ligand‐binding domains: proteins, peptides, or small molecules. We have recently reported that the addition of a cholesterol group to two HIV antibodies can dramatically improve their antiviral potency. Cholesterol, which can be conjugated at various positions in the antibody, including the constant (Fc) domain, endows the conjugate with affinity for the membrane lipid rafts, thus increasing its concentration at the site where viral entry occurs. Here, we extend this strategy to an HIV immunoadhesin, combining a cholesterol‐conjugated Fc domain with the peptide fusion inhibitor C41. The immunoadhesin C41‐Fc‐chol displayed high affinity for Human Embryonic Kidney (HEK) 293 cells, and when tested on a panel of HIV‐1 strains, it was considerably more potent than the unconjugated C41‐Fc construct. Potentiation of antiviral activity was comparable to what was previously observed for the cholesterol‐conjugated HIV antibodies. Given the key role of cholesterol in lipid raft formation and viral fusion, we expect that the same strategy should be broadly applicable to enveloped viruses, for many of which it is already known the sequence of a peptide fusion inhibitor similar to C41. Moreover, the sequence of heptad repeat‐derived fusion inhibitors can often be predicted from genomic information alone, opening a path to immunoadhesins against emerging viruses. Copyright © 2015 European Peptide Society and John Wiley & Sons, Ltd.  相似文献   

18.
The interferon (IFN)-induced transmembrane protein 3 (IFITM3) is a cellular restriction factor that inhibits infection by influenza virus and many other pathogenic viruses. IFITM3 prevents endocytosed virus particles from accessing the host cytoplasm although little is known regarding its regulatory mechanisms. Here we demonstrate that IFITM3 localization to and antiviral remodeling of endolysosomes is differentially regulated by S-palmitoylation and lysine ubiquitination. Although S-palmitoylation enhances IFITM3 membrane affinity and antiviral activity, ubiquitination decreases localization with endolysosomes and decreases antiviral activity. Interestingly, autophagy reportedly induced by IFITM3 expression is also negatively regulated by ubiquitination. However, the canonical ATG5-dependent autophagy pathway is not required for IFITM3 activity, indicating that virus trafficking from endolysosomes to autophagosomes is not a prerequisite for influenza virus restriction. Our characterization of IFITM3 ubiquitination sites also challenges the dual-pass membrane topology predicted for this protein family. We thus evaluated topology by N-linked glycosylation site insertion and protein lipidation mapping in conjunction with cellular fractionation and fluorescence imaging. Based on these studies, we propose that IFITM3 is predominantly an intramembrane protein where both the N and C termini face the cytoplasm. In sum, by characterizing S-palmitoylation and ubiquitination of IFITM3, we have gained a better understanding of the trafficking, activity, and intramembrane topology of this important IFN-induced effector protein.  相似文献   

19.
Interferon-induced transmembrane protein 3 (IFITM3; FRAGILIS; MIL-1) is part of a larger family of important small interferon-induced transmembrane genes and proteins involved in early development, cell adhesion, and cell proliferation, and which also play a major role in response to bacterial and viral infections and, more recently, in pronounced malignancies. IFITM3, together with tissue-nonspecific alkaline phosphatase (TNAP), PRDM1, and STELLA, has been claimed to be a hallmark of segregated primordial germ cells (PGCs) (Saitou et al., 2002). However, whether IFITM3, like STELLA, is part of a broader stem/progenitor pool that builds the posterior region of the mouse conceptus (Mikedis and Downs, 2012) is obscure. To discover the whereabouts of IFITM3 during mouse gastrulation (~E6.5-9.0), systematic immunohistochemical analysis was carried out at closely spaced 2-4-h intervals. Results revealed diverse, yet consistent, profiles of IFITM3 localization throughout the gastrula. Within the putative PGC trajectory and surrounding posterior tissues, IFITM3 localized as a large cytoplasmic spot with or without staining in the plasma membrane. IFITM3, like STELLA, was also found in the ventral ectodermal ridge (VER), a posterior progenitor pool that builds the tailbud. The large cytoplasmic spot with plasma membrane staining was exclusive to the posterior region; the visceral yolk sac, non-posterior tissues, and epithelial tissues exhibited spots of IFITM3 without cell surface staining. Colocalization of the intracellular IFITM3 spot with the endoplasmic reticulum, Golgi apparatus, or endolysosomes was not observed. That relatively high levels of IFITM3 were found throughout the posterior primitive streak and its derivatives is consistent with evidence that IFITM3, like STELLA, is part of a larger stem/progenitor cell pool at the posterior end of the primitive streak that forms the base of the allantois and builds the fetal-umbilical connection, thus further obfuscating practical phenotypic distinctions between so-called PGCs and surrounding soma.  相似文献   

20.
The broadly neutralizing antibodies HIV 2F5 and 4E10, which bind to overlapping epitopes in the membrane-proximal external region of the fusion protein gp41, have been proposed to use a two-step mechanism for neutralization; first, they bind and preconcentrate at the viral membrane through their long, hydrophobic CDRH3 loops, and second, they form a high affinity complex with the protein epitope. Accordingly, mutagenesis of the CDRH3 can abolish their neutralizing activity, with no change in the affinity for the peptide epitope. We show here that we can mimic this mechanism by conjugating a cholesterol group outside of the paratope of an antibody. Cholesterol-conjugated antibodies bind to lipid raft domains on the membrane, and because of this enrichment, they show increased antiviral potency. In particular, we find that cholesterol conjugation (i) rescues the antiviral activity of CDRH3-mutated 2F5, (ii) increases the antiviral activity of WT 2F5, (iii) potentiates the non-membrane-binding HIV antibody D5 10–100-fold (depending on the virus strain), and (iv) increases synergy between 2F5 and D5. Conjugation can be made at several positions, including variable and constant domains. Cholesterol conjugation therefore appears to be a general strategy to boost the potency of antiviral antibodies, and, because membrane affinity is engineered outside of the antibody paratope, it can complement affinity maturation strategies.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号