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 48 毫秒
1.
Min Zhang  Yu Wang 《Autophagy》2018,14(5):918-920
Autophagosomal membrane sources generate autophagic membrane precursors, which later assemble into the double-membrane autophagosome. The key events happening on the membrane sources during autophagic membrane generation remain poorly characterized. Our previous work found the ER-Golgi intermediate compartment (ERGIC) as a membrane source for the phagophore, the precursor to the autophagosome. A relocation of the COPII machinery from the ER-exit sites (ERES) to the ERGIC generates vesicles for LC3 lipidation. In recent work, we made a further step by showing that a starvation-induced remodeling of ERES facilitates the relocation of COPII to the ERGIC and the generation of the autophagic membrane.  相似文献   

2.
Liang Ge  Livia Wilz  Randy Schekman 《Autophagy》2015,11(12):2372-2374
Autophagosome biogenesis requires efficient mobilization and delivery of membranes from intracellular sources. How these membranes are mobilized remains poorly understood. Our recent work reported an autophagic signal-induced membrane mobilization event from the ER-Golgi intermediate compartment (ERGIC) to generate an early autophagosomal membrane precursor. We found that starvation activates the autophagic phosphatidylinositol 3-kinase, which promotes a relocation of COPII proteins from the ER-exit sites to the ERGIC. The relocation of COPII generates ERGIC-derived COPII vesicles as a membrane template for LC3 lipidation, a key step for autophagosome biogenesis.  相似文献   

3.
Background information. Syntaxin 17 is a SNARE (soluble N‐ethylmaleimide‐sensitive‐factor‐attachment protein receptor) protein that predominantly localizes to the ER (endoplasmic reticulum) and to some extent in the ERGIC (ER—Golgi intermediate compartment). Syntaxin 17 has been suggested to function as a receptor at the ER membrane that mediates trafficking between the ER and post‐ER compartments. It has a unique 33 amino acid luminal tail whose function is not known. Here we have investigated the structural requirements for localization of syntaxin 17 to the ERGIC and its role in trafficking. Results. Deletion analysis showed that syntaxin 17 required its cytoplasmic domain to exit the ER and localize to the ERGIC. Mutation of a conserved tyrosine residue in the cytoplasmic domain resulted in reduced localization of syntaxin 17 in the ERGIC and ER‐exit sites, suggesting the presence of a tyrosine‐based ER export motif. Syntaxin 17 also required its C‐terminal tail to localize to the ERES (ER exit sites) and ERGIC. Knockdown of syntaxin 17 destabilized the ERGIC organization and also caused fragmentation of the Golgi complex. Syntaxin 17 showed direct interaction with transmembrane proteins p23 and p25 (cargo receptors that cycle between the ER and Golgi) with the help of its C‐terminal tail. Overexpression of syntaxin 17 redistributed β‐COP (β‐coatomer protein) which required its C‐terminal tail. Overexpression of syntaxin 17 also blocked the anterograde transport of VSVG (vesicular stomatitis virus G‐protein) in the ERGIC. Conclusions. We show that syntaxin 17 has a tyrosine‐based motif which is required for its incorporation into COPII (coatomer protein II) vesicles, exit from the ER and localization to the ERGIC. Our results suggest that syntaxin 17 cycles between the ER and ERGIC through classical trafficking pathways involving COPII and COPI (coatomer protein I) vesicles, which requires its unique C‐terminal tail. We also show that syntaxin 17 is essential for maintaining the architecture of ERGIC and Golgi.  相似文献   

4.
In mammalian cells, cargo‐laden secretory vesicles leave the endoplasmic reticulum (ER) en route to ER‐Golgi intermediate compartments (ERGIC) in a manner dependent on the COPII coat complex. We report here that COPII‐coated transport carriers traverse a submicron, TFG (Trk‐fused gene)‐enriched zone at the ER/ERGIC interface. The architecture of TFG complexes as determined by three‐dimensional electron microscopy reveals the formation of flexible, octameric cup‐like structures, which are able to self‐associate to generate larger polymers in vitro. In cells, loss of TFG function dramatically slows protein export from the ER and results in the accumulation of COPII‐coated carriers throughout the cytoplasm. Additionally, the tight association between ER and ERGIC membranes is lost in the absence of TFG. We propose that TFG functions at the ER/ERGIC interface to locally concentrate COPII‐coated transport carriers and link exit sites on the ER to ERGIC membranes. Our findings provide a new mechanism by which COPII‐coated carriers are retained near their site of formation to facilitate rapid fusion with neighboring ERGIC membranes upon uncoating, thereby promoting interorganellar cargo transport.  相似文献   

5.
Wei T  Wang A 《Journal of virology》2008,82(24):12252-12264
Single-stranded positive-sense RNA viruses induce the biogenesis of cytoplasmic membranous vesicles, where viral replication takes place. However, the mechanism underlying this characteristic vesicular proliferation remains poorly understood. Previously, a 6-kDa potyvirus membrane protein (6K) was shown to interact with the endoplasmic reticulum (ER) and to induce the formation of the membranous vesicles. In this study, the involvement of the early secretory pathway in the formation of the 6K-induced vesicles was investigated in planta. By means of live-cell imaging, it was found that the 6K protein was predominantly colocalized with Sar1, Sec23, and Sec24, which are known markers of ER exit sites (ERES). The localization of 6K at ERES was prevented by the coexpression of a dominant-negative mutant of Sar1 that disables the COPII activity or by the coexpression of a mutant of Arf1 that disrupts the COPI complex. The secretion of a soluble secretory marker targeting the apoplast was arrested at the level of the ER in cells overexpressing 6K or infected by a potyvirus. This blockage of protein trafficking out of the ER by 6K and the distribution of 6K toward the ERES may account for the aggregation of the 6K-bound vesicles. Finally, virus infection was reduced when the accumulation of 6K at ERES was inhibited by impairing either the COPI or COPII complex. Taken together, these results imply that the cellular COPI and COPII coating machineries are involved in the biogenesis of the potyvirus 6K vesicles at the ERES for viral-genome replication.  相似文献   

6.
COPII and COPI mediate the formation of membrane vesicles translocating in opposite directions within the secretory pathway. Live-cell and electron microscopy revealed a novel mode of function for COPII during cargo export from the ER. COPII is recruited to membranes defining the boundary between the ER and ER exit sites, facilitating selective cargo concentration. Using direct observation of living cells, we monitored cargo selection processes, accumulation, and fission of COPII-free ERES membranes. CRISPR/Cas12a tagging, the RUSH system, and pharmaceutical and genetic perturbations of ER-Golgi transport demonstrated that the COPII coat remains bound to the ER–ERES boundary during protein export. Manipulation of the cargo-binding domain in COPII Sec24B prohibits cargo accumulation in ERES. These findings suggest a role for COPII in selecting and concentrating exported cargo rather than coating Golgi-bound carriers. These findings transform our understanding of coat proteins’ role in ER-to-Golgi transport.  相似文献   

7.
Runz H  Miura K  Weiss M  Pepperkok R 《The EMBO journal》2006,25(13):2953-2965
Alterations in endoplasmic reticulum (ER) cholesterol are fundamental for a variety of cellular processes such as the regulation of lipid homeostasis or efficient protein degradation. We show that reduced levels of cellular sterols cause a delayed ER-to-Golgi transport of the secretory cargo membrane protein ts-O45-G and a relocation to the ER of an endogenous protein cycling between the ER and the Golgi complex. Transport inhibition is characterized by a delay in the accumulation of ts-O45-G in ER-exit sites (ERES) and correlates with a reduced mobility of ts-O45-G within ER membranes. A simple mathematical model describing the kinetics of ER-exit predicts that reduced cargo loading to ERES and not the reduced mobility of ts-O45-G accounts for the delayed ER-exit and arrival at the Golgi. Consistent with this, membrane turnover of the COPII component Sec23p is delayed in sterol-depleted cells. Altogether, our results demonstrate the importance of sterol levels in COPII mediated ER-export.  相似文献   

8.
The autophagosome, a double‐membrane structure mediating degradation of cytoplasmic materials by macroautophagy, is formed in close proximity to the endoplasmic reticulum (ER). However, how the ER membrane is involved in autophagy initiation and to which membrane structures the autophagy‐initiation complex is localized have not been fully characterized. Here, we were able to biochemically analyze autophagic intermediate membranes and show that the autophagy‐initiation complex containing ULK and FIP200 first associates with the ER membrane. To further characterize the ER subdomain, we screened phospholipid biosynthetic enzymes and found that the autophagy‐initiation complex localizes to phosphatidylinositol synthase (PIS)‐enriched ER subdomains. Then, the initiation complex translocates to the ATG9A‐positive autophagosome precursors in a PI3P‐dependent manner. Depletion of phosphatidylinositol (PI) by targeting bacterial PI‐specific phospholipase C to the PIS domain impairs recruitment of downstream autophagy factors and autophagosome formation. These findings suggest that the autophagy‐initiation complex, the PIS‐enriched ER subdomain, and ATG9A vesicles together initiate autophagosome formation.  相似文献   

9.
In contrast with animals, plant cells contain multiple mobile Golgi stacks distributed over the entire cytoplasm. However, the distribution and dynamics of protein export sites on the plant endoplasmic reticulum (ER) surface have yet to be characterized. A widely accepted model for ER-to-Golgi transport is based on the sequential action of COPII and COPI coat complexes. The COPII complex assembles by the ordered recruitment of cytosolic components on the ER membrane. Here, we have visualized two early components of the COPII machinery, the small GTPase Sar1p and its GTP exchanging factor Sec12p in live tobacco (Nicotiana tabacum) leaf epidermal cells. By in vivo confocal laser scanning microscopy and fluorescence recovery after photobleaching experiments, we show that Sar1p cycles on mobile punctate structures that track with the Golgi bodies in close proximity but contain regions that are physically separated from the Golgi bodies. By contrast, Sec12p is uniformly distributed along the ER network and does not accumulate in these structures, consistent with the fact that Sec12p does not become part of a COPII vesicle. We propose that punctate accumulation of Sar1p represents ER export sites (ERES). The sites may represent a combination of Sar1p-coated ER membranes, nascent COPII membranes, and COPII vectors in transit, which have yet to lose their coats. ERES can be induced by overproducing Golgi membrane proteins but not soluble bulk-flow cargos. Few punctate Sar1p loci were observed that are independent of Golgi bodies, and these may be nascent ERES. The vast majority of ERES form secretory units that move along the surface of the ER together with the Golgi bodies, but movement does not influence the rate of cargo transport between these two organelles. Moreover, we could demonstrate using the drug brefeldin A that formation of ERES is strictly dependent on a functional retrograde transport route from the Golgi apparatus.  相似文献   

10.
The selective export of proteins and lipids from the endoplasmic reticulum (ER) is mediated by the coat protein complex II (COPII) that assembles onto the ER membrane. In higher eukaryotes, COPII proteins assemble at discrete sites on the membrane known as ER exit sites (ERES). Here, we identify Sec16 as the protein that defines ERES in mammalian cells. Sec16 localizes to ERES independent of Sec23/24 and Sec13/31. Overexpression, and to a lesser extent, small interfering RNA depletion of Sec16, both inhibit ER-to-Golgi transport suggesting that Sec16 is required in stoichiometric amounts. Sar1 activity is required to maintain the localization of Sec16 at discrete locations on the ER membrane, probably through preventing its dissociation. Our data suggest that Sar1-GTP-dependent assembly of Sec16 on the ER membrane forms an organized scaffold defining an ERES.  相似文献   

11.
Autophagy is an intracellular degradation process, through which cytosolic materials are delivered to the lysosome. Despite recent identification of many autophagy-related genes, how autophagosomes are generated remains unclear. Here, we examined the hierarchical relationships among mammalian Atg proteins. Under starvation conditions, ULK1, Atg14, WIPI-1, LC3 and Atg16L1 target to the same compartment, whereas DFCP1 localizes adjacently to these Atg proteins. In terms of puncta formation, the protein complex including ULK1 and FIP200 is the most upstream unit and is required for puncta formation of the Atg14-containing PI3-kinase complex. Puncta formation of both DFCP1 and WIPI-1 requires FIP200 and Atg14. The Atg12-Atg5-Atg16L1 complex and LC3 are downstream units among these factors. The punctate structures containing upstream Atg proteins such as ULK1 and Atg14 tightly associate with the ER, where the ER protein vacuole membrane protein 1 (VMP1) also transiently localizes. These structures are formed even when cells are treated with wortmannin to suppress autophagosome formation. These hierarchical analyses suggest that ULK1, Atg14 and VMP1 localize to the ER-associated autophagosome formation sites in a PI3-kinase activity-independent manner.Key words: autophagosome, PI3-kinase, isolation membrane, endoplasmic reticulum, ULK  相似文献   

12.
细胞自噬是一种重要且保守的细胞内降解过程,通过形成双层膜的自噬体包裹细胞内容物进行降解。内质网来源的COPII囊泡被认为是饥饿诱导的应激过程中自噬体的膜源。探究了COPII囊泡衣被蛋白SEC24A在巨自噬通路中的作用。利用siRNA干扰技术敲低SEC24A的表达,EBSS饥饿处理对照组和SEC24A敲低组HeLa细胞2 h诱导自噬发生,经Western blot和免疫荧光实验检测自噬底物蛋白p62和自噬标志蛋白LC3-II的蛋白水平变化,以确定SEC24A是否参与自噬。通过RFP-GFP-LC3串联荧光检测自噬体和自噬溶酶体的数目,利用蛋白酶K保护实验验证自噬缺陷发生在自噬体闭合之前或者之后,利用免疫荧光实验检测敲低SEC24A对自噬通路上ATG复合物的影响,以确定SEC24A调控自噬通路的位点。通过免疫共沉淀实验验证SEC24A与自噬相关蛋白ATG9A是否存在相互作用。蛋白检测实验发现,饥饿条件下与对照细胞相比,敲低SEC24A细胞内自噬底物蛋白p62积累,而标志蛋白LC3-II减少。RFP-GFP-LC3串联荧光实验显示,敲低SEC24A后自噬体及自噬溶酶体的数目均减少。蛋白酶K保护实验显示,SEC24A敲低细胞中受膜结构保护的p62和GFP-LC3均减少,提示SEC24A作用位点在自噬体闭合之前。免疫荧光实验显示,敲低SEC24A的表达后ATG14L、ATG16L1点状结构减少,而ATG9A点状结构的数量没有明显变化,提示SEC24A作用于ATG14L、ATG16L1上游。免疫共沉淀实验显示SEC24A与ATG9A存在相互作用。研究结果不仅有助于深化对自噬体形成过程和分子机制的了解,也为全面解读COPII囊泡及其衣被蛋白在自噬中的重要作用提供了信息。  相似文献   

13.
Double membrane structure, autophagosome, is formed de novo in the process of autophagy in the yeast Saccharomyces cerevisiae, and many Apg proteins participate in this process. To further understand autophagy, we analyzed the involvement of factors engaged in the secretory pathway. First, we showed that Sec18p (N-ethylmaleimide-sensitive fusion protein, NSF) and Vti1p (soluble N-ethylmaleimide-sensitive fusion protein attachment protein, SNARE), and soluble N-ethylmaleimide-sensitive fusion protein receptor are required for fusion of the autophagosome to the vacuole but are not involved in autophagosome formation. Second, Sec12p was shown to be essential for autophagy but not for the cytoplasm to vacuole-targeting (Cvt) (pathway, which shares mostly the same machinery with autophagy. Subcellular fractionation and electron microscopic analyses showed that Cvt vesicles, but not autophagosomes, can be formed in sec12 cells. Three other coatmer protein (COPII) mutants, sec16, sec23, and sec24, were also defective in autophagy. The blockage of autophagy in these mutants was not dependent on transport from endoplasmic reticulum-to-Golgi, because mutations in two other COPII genes, SEC13 and SEC31, did not affect autophagy. These results demonstrate the requirement for subgroup of COPII proteins in autophagy. This evidence demonstrating the involvement of Sec proteins in the mechanism of autophagosome formation is crucial for understanding membrane flow during the process.  相似文献   

14.
《Autophagy》2013,9(6):764-776
Autophagy is an intracellular degradation process, through which cytosolic materials are delivered to the lysosome. Despite recent identification of many autophagy-related genes, how autophagosomes are generated remains unclear. Here, we examined the hierarchical relationships among mammalian Atg proteins. Under starvation conditions, ULK1, Atg14, WIPI-1, LC3 and Atg16L1 target to the same compartment, whereas DFCP1 localizes adjacently to these Atg proteins. In terms of puncta formation, the protein complex including ULK1 and FIP200 is the most upstream unit and is required for puncta formation of the Atg14-containing PI3-kinase complex. Puncta formation of both DFCP1 and WIPI-1 requires FIP200 and Atg14. The Atg12-Atg5-Atg16L1 complex and LC3 are downstream units among these factors. The punctate structures containing upstream Atg proteins such as ULK1 and Atg14 tightly associate with the ER, where the ER protein Vacuole membrane protein 1 (VMP1) also transiently localizes. These structures are formed even when cells are treated with wortmannin to suppress autophagosome formation. These hierarchical analyses suggest that ULK1, Atg14 and VMP1 localize to the ER-associated autophagosome formation sites in a PI3-kinase activity-independent manner.  相似文献   

15.
Giardia lamblia parasitism accounts for the majority of cases of parasitic diarrheal disease, making this flagellated eukaryote the most successful intestinal parasite worldwide. This organism has undergone secondary reduction/elimination of entire organelle systems such as mitochondria and Golgi. However, trophozoite to cyst differentiation (encystation) requires neogenesis of Golgi‐like secretory organelles named encystation‐specific vesicles (ESVs), which traffic, modify and partition cyst wall proteins produced exclusively during encystation. In this work we ask whether neogenesis of Golgi‐related ESVs during G. lamblia differentiation, similarly to Golgi biogenesis in more complex eukaryotes, requires the maintenance of distinct COPII‐associated endoplasmic reticulum (ER) subdomains in the form of ER exit sites (ERES) and whether ERES are also present in non‐differentiating trophozoites. To address this question, we identified conserved COPII components in G. lamblia cells and determined their localization, quantity and dynamics at distinct ERES domains in vegetative and differentiating trophozoites. Analogous to ERES and Golgi biogenesis, these domains were closely associated to early stages ofnewly generated ESV. Ectopic expression of non‐functional Sar1 GTPase variants caused ERES collapse and, consequently, ESV ablation, leading to impaired parasite differentiation. Thus, our data show how ERES domains remain conserved in G. lamblia despite elimination of steady‐state Golgi. Furthermore, the fundamental eukaryotic principle of ERES to Golgi/Golgi‐like compartment correspondence holds true in differentiating Giardia presenting streamlined machinery for secretory organelle biogenesis and protein trafficking. However, in the Golgi‐less trophozoites ERES exist as stable ER subdomains, likely as the sole sorting centres for secretory traffic.  相似文献   

16.
The plant endoplasmic reticulum (ER) contains functionally distinct subdomains at which cargo molecules are packed into transport carriers. To study these ER export sites (ERES), we used tobacco (Nicotiana tabacum) leaf epidermis as a model system and tested whether increased cargo dosage leads to their de novo formation. We have followed the subcellular distribution of the known ERES marker based on a yellow fluorescent protein (YFP) fusion of the Sec24 COPII coat component (YFP-Sec24), which, differently from the previously described ERES marker, tobacco Sar1-YFP, is visibly recruited at ERES in both the presence and absence of overexpressed membrane cargo. This allowed us to quantify variation in the ERES number and in the recruitment of Sec24 to ERES upon expression of cargo. We show that increased synthesis of membrane cargo leads to an increase in the number of ERES and induces the recruitment of Sec24 to these ER subdomains. Soluble proteins that are passively secreted were found to leave the ER with no apparent up-regulation of either the ERES number or the COPII marker, showing that bulk flow transport has spare capacity in vivo. However, de novo ERES formation, as well as increased recruitment of Sec24 to ERES, was found to be dependent on the presence of the diacidic ER export motif in the cytosolic domain of the membrane cargo. Our data suggest that the plant ER can adapt to a sudden increase in membrane cargo-stimulated secretory activity by signal-mediated recruitment of COPII machinery onto existing ERES, accompanied by de novo generation of new ERES.  相似文献   

17.
Proteins trafficking through the secretory pathway must first exit the endoplasmic reticulum (ER) through membrane vesicles created and regulated by the COPII coat protein complex. Cranio-lenticulo-sutural dysplasia (CLSD) was recently shown to be caused by a missense mutation in SEC23A, a gene encoding one of two paralogous COPII coat proteins. We now elucidate the molecular mechanism underlying this disease. In vitro assays reveal that the mutant form of SEC23A poorly recruits the Sec13-Sec31 complex, inhibiting vesicle formation. Surprisingly, this effect is modulated by the Sar1 GTPase paralog used in the reaction, indicating distinct affinities of the two human Sar1 paralogs for the Sec13-Sec31 complex. Patient cells accumulate numerous tubular cargo-containing ER exit sites devoid of observable membrane coat, likely representing an intermediate step in COPII vesicle formation. Our results indicate that the Sar1-Sec23-Sec24 prebudding complex is sufficient to form cargo-containing tubules in vivo, whereas the Sec13-Sec31 complex is required for membrane fission.  相似文献   

18.
The variant surface glycoprotein (VSG) of bloodstream form Trypanosoma brucei (Tb) is a critical virulence factor. The VSG glycosylphosphatidylinositol (GPI)-anchor strongly influences passage through the early secretory pathway. Using a dominant-negative mutation of TbSar1, we show that endoplasmic reticulum (ER) exit of secretory cargo in trypanosomes is dependent on the coat protein complex II (COPII) machinery. Trypanosomes have two orthologues each of the Sec23 and Sec24 COPII subunits, which form specific heterodimeric pairs: TbSec23.1/TbSec24.2 and TbSec23.2/TbSec24.1. RNA interference silencing of each subunit is lethal but has minimal effects on trafficking of soluble and transmembrane proteins. However, silencing of the TbSec23.2/TbSec24.1 pair selectively impairs ER exit of GPI-anchored cargo. All four subunits colocalize to one or two ER exit sites (ERES), in close alignment with the postnuclear flagellar adherence zone (FAZ), and closely juxtaposed to corresponding Golgi clusters. These ERES are nucleated on the FAZ-associated ER. The Golgi matrix protein Tb Golgi reassembly stacking protein defines a region between the ERES and Golgi, suggesting a possible structural role in the ERES:Golgi junction. Our results confirm a selective mechanism for GPI-anchored cargo loading into COPII vesicles and a remarkable degree of streamlining in the early secretory pathway. This unusual architecture probably maximizes efficiency of VSG transport and fidelity in organellar segregation during cytokinesis.  相似文献   

19.
Autophagy is an intracellular degradation system, by which cytoplasmic contents are degraded in lysosomes. Autophagy is dynamically induced by nutrient depletion to provide necessary amino acids within cells, thus helping them adapt to starvation. Although it has been suggested that mTOR is a major negative regulator of autophagy, how it controls autophagy has not yet been determined. Here, we report a novel mammalian autophagy factor, Atg13, which forms a stable ~3-MDa protein complex with ULK1 and FIP200. Atg13 localizes on the autophagic isolation membrane and is essential for autophagosome formation. In contrast to yeast counterparts, formation of the ULK1–Atg13–FIP200 complex is not altered by nutrient conditions. Importantly, mTORC1 is incorporated into the ULK1–Atg13–FIP200 complex through ULK1 in a nutrient-dependent manner and mTOR phosphorylates ULK1 and Atg13. ULK1 is dephosphorylated by rapamycin treatment or starvation. These data suggest that mTORC1 suppresses autophagy through direct regulation of the ~3-MDa ULK1–Atg13–FIP200 complex.  相似文献   

20.
Export from the ER is COPII-dependent. However, there is disagreement on the nature of the cargo-containing carriers that exit the ER. Two new studies from Shomron et al. (2021. J. Cell Biol. https://doi.org/10.1083/jcb.201907224) and Weigel et al. (2021. Cell. https://doi.org/10.1016/j.cell.2021.03.035) present a new model, where COPII helps to select secretory cargo but does not coat the carriers leaving the ER.

An analogy for ER-to-Golgi transport is day-to-day logistics, whereby we have developed different types of carriers to accommodate cargo of varying shapes, quantities, and sizes. Following the same logic, we could assume that our cells are equipped with carriers of different shapes and sizes to shuttle small or bulky cargo or different cargo quantities out of the ER. Although this appears straightforward, the molecular details of ER-to-Golgi transport has been the subject of intensive debate.Early work on secretory trafficking from the 1960s noted that secretory proteins leave the ER at ribosome free regions, which were termed transitional elements (or transitional ER; 1). These transitional elements were postulated to give rise to “transport vesicles with fuzzy coats,” which mediate transport from the ER (1). Sec23 was found to be enriched at these transitional elements (2) and was later shown to be part of the COPII complex that mediates export from the ER in small vesicles (3). Since then, the standard model for export from the ER was that small and round COPII vesicles (60–80 nm in diameter) leave the ER from transitional elements, now referred to as ER exit sites (ERES; Fig. 1 A). This model was later expanded to include pre-Golgi intermediates such as the ER-Golgi-intermediate compartment (ERGIC) generated from the homotypic fusion of COPII vesicles (4).Open in a separate windowFigure 1.Depiction of different possible modes of ER export. (A) The classical (vesicular) transport model that includes budding of a coated vesicle followed by homotypic fusion of COPII vesicles. (B) ERES can give rise to tubular structures, which either depart as tubules followed by COPI recruitment or may form tunnels with distal compartments.Despite its wide acceptance, experimental evidence for the presence of COPII vesicles at ERES in intact cells was scarce. The main evidence for COPII vesicles came from in vitro assays from which such vesicles were isolated. Furthermore, the relatively small size of COPII vesicles (60–80 nm) could not explain transport of bulky cargo such as type I collagen (with a length of 300 nm). A major challenge to the vesicular transport model came from a paper by Mironov et al. (5), who presented evidence that ER-derived carriers are large uncoated saccules that mature toward the Golgi. Notably, round and small COPII vesicles as carriers mediating the ER export were absent during synchronized transport of type I collagen and VSVG-ts045 (a temperature-sensitive mutant of the vesicular stomatitis virus glycoprotein). Both types of cargo rely on retention in the ER at 40°C followed by export upon lowering the temperature (and addition of ascorbic acid in the case of type I collagen). Supporters of the conventional vesicular transport model argued that the size of type I collagen as well as the large quantities of molecules queuing to leave the ER simultaneously might have led the ER export machinery to adapt to this situation, thereby leading to formation of the observed carriers. Further support for the vesicle transport model came from work using 3D electron tomography showing that ERES are domains that are continuous with the ER, which are surrounded by COPII vesicles (6). The electron tomography was performed in both chemically fixed as well as high-pressure-frozen cells in the absence of secretory cargo overexpression or synchronized trafficking waves. Because ERES exhibited budding profiles coated with COPII, it was concluded that the COPII vesicles bud as coated carriers from ERES. Despite this support for the vesicular transport model, it was becoming increasingly clear that ER-to-Golgi transport cannot be explained solely by small COPII vesicles. Thus, the idea that different types of carriers operate in the ER-Golgi route began to ripen in the community.In yeast, cis-Golgi compartments were shown to touch ERES to “pick up” secretory cargo (7). Whether this “hug and kiss” model involves fusion of the cis-Golgi with budded COPII vesicles or whether it forms a “tunnel” between the ERES and the cis-Golgi remains unclear. In support of the existence of tunnels is work from the Malhotra group showing that collagen transport might occur via recruitment of ERGIC membranes to ERES enriched in pro-collagen (8). The observation that a tunnel is formed between the ERES and ERGIC necessitates the preexistence of an ERGIC membrane container. As discussed, the ERGIC might form by homotypic fusion of COPII vesicles, and thus the tunnel model proposed by Raote and Malhotra (9) can easily be reconciled with the vesicle transport model.In this issue, Shomron et al. (10) present a major challenge to the vesicle transport model. They suggest that COPII complexes only decorate the neck of an ERES, where they solely serve to concentrate cargo into transport containers. This confirms earlier papers showing that COPII mediates concentrative ER export (11, 12). Strikingly, Shomron et al. observe with live imaging that secretory cargo enters a tubule that segregates from COPII at the level of ERES, indicating that the departing transport carrier is not coated. Furthermore, COPII was confined to the neck of the tubular carrier. This finding agrees with previously observed (noncoated) saccules that leave the ER (5). A concurrent study from Weigel et al. (13) reached a similar conclusion. To overcome prior difficulties associated with fixation, low sampling, and thick sections, they aimed at imaging ERES in living cells by combining focused ion beam scanning electron microscopy with cryo-structured illumination microscopy. Furthermore, they used the retention using selective hook technology (14) to perform synchronized cargo release experiments, thus avoiding problems associated with temperature shifts. In agreement with Shomron et al., they show that ERES give rise to a network of tubules that contain secretory cargo devoid of COPII components. Again, COPII components were only found to localize to the neck of these tubules, implicating that the main role of COPII is to concentrate cargo into carriers. They also showed that ERES are structures continuous with the ER (confirming the earlier data from 3D electron-tomography; 6) that adapt in size to accommodate the load of secretory cargo (again confirming earlier work by others; 15, 16).Another interesting finding by both groups was that the tubule acquired COPI as it moved toward the Golgi (10, 13). Therefore, they independently conclude that this presents evidence for a role of COPI in anterograde ER-to-Golgi transport, which challenges the classical model whereby COPI is thought exclusively to mediate retrograde transport from the Golgi back to the ER. It remains unclear what role COPI would precisely play in anterograde transport. Simply because the tubular membrane container is positive for COPI does not necessarily mean that COPI regulates anterograde transport. Carriers need tethering factors such as p115/Uso1, which are recruited by Rab1 to deliver their content to the next compartment. No role for COPI is known in this process. An alternative explanation for the recruitment of COPI to the ER-derived carriers is that this marks the beginning of retrograde transport back to the ER. This is supported by the observation that a mutant of ERGIC-53 (LMAN1) that does not bind COPI is capable of leaving the ER and without exhibiting any defect in anterograde transport (17). Strikingly, this mutant ERGIC-53 mislocalizes to the plasma membrane because it cannot use COPI for retrograde transport (16). Thus, recruitment of COPI might contribute to the maturation of the forward moving membrane carrier by retrieving back ER proteins.Altogether, it appears that several types of carriers (tubules, saccules, tunnels, and coated vesicles) may coexist and operate along the ER-to-Golgi route. The papers by Shomron et al. and Weigel el al. do not cancel or revoke the other models of trafficking. Rather, they add a new model and show us how diverse and flexible this trafficking route is. It is possible that our cells are equipped with all types of carriers, which cells use depending on the size, quantity, or type of cargo, as well as on the cellular and the environmental context. This diversity might confer robustness of the ER-to-Golgi transport pathway. This might explain why different groups reached sometimes opposing conclusions. For instance, papers that relied on waves of synchronized trafficking or on bulky cargo might have shifted the balance toward a certain type of carrier. Most cells contain several hundred ERES, with some of them at several microns’ distance to the Golgi. It is therefore possible that different types of carriers might operate in a manner depending on the type of ERES. Future work will clarify and reconcile all these open questions.  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