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 31 毫秒
1.
We have identified a 55 kDa protein, named GRASP55 (Golgi reassembly stacking protein of 55 kDa), as a component of the Golgi stacking machinery. GRASP55 is homologous to GRASP65, an N-ethylmaleimide-sensitive membrane protein required for the stacking of Golgi cisternae in a cell-free system. GRASP65 exists in a complex with the vesicle docking protein receptor GM130 to which it binds directly, and the membrane tethering protein p115, which also functions in the stacking of Golgi cisternae. GRASP55 binding to GM130, could not be detected using biochemical methods, although a weak interaction was detected with the yeast two-hybrid system. Cryo-electron microscopy revealed that GRASP65, like GM130, is present on the cis-Golgi, while GRASP55 is on the medial-Golgi. Recombinant GRASP55 and antibodies to the protein block the stacking of Golgi cisternae, which is similar to the observations made for GRASP65. These results demonstrate that GRASP55 and GRASP65 function in the stacking of Golgi cisternae.  相似文献   

2.
In vitro assays identified the Golgi peripheral protein GRASP65 as a Golgi stacking factor that links adjacent Golgi cisternae by forming mitotically regulated trans‐oligomers. These conclusions, however, require further confirmation in the cell. In this study, we showed that the first 112 amino acids at the N‐terminus (including the first PDZ domain, PDZ1) of the protein are sufficient for oligomerization. Systematic electron microscopic analysis showed that the expression of non‐regulatable GRASP65 mutants in HeLa cells enhanced Golgi stacking in interphase and inhibited Golgi fragmentation during mitosis. Depletion of GRASP65 by small interference RNA (siRNA) reduced the number of cisternae in the Golgi stacks; this reduction was rescued by expressing exogenous GRASP65. These results provided evidence and a molecular mechanism by which GRASP65 stacks Golgi cisternal membranes. Further experiments revealed that inhibition of mitotic Golgi disassembly by expressing non‐regulatable GRASP65 mutants did not affect equal partitioning of the Golgi membranes into the daughter cells. However, it delayed mitotic entry and suppressed cell growth; this effect was diminished by dispersing the Golgi apparatus with Brefeldin A treatment prior to mitosis, suggesting that Golgi disassembly at the onset of mitosis plays a role in cell cycle progression.  相似文献   

3.
Wang Y  Wei JH  Bisel B  Tang D  Seemann J 《PloS one》2008,3(2):e1647
The Golgi apparatus in mammalian cells is composed of flattened cisternae that are densely packed to form stacks. We have used the Golgi stacking protein GRASP65 as a tool to modify the stacking state of Golgi cisternae. We established an assay to measure protein transport to the cell surface in post-mitotic cells in which the Golgi was unstacked. Cells with an unstacked Golgi showed a higher transport rate compared to cells with stacked Golgi membranes. Vesicle budding from unstacked cisternae in vitro was significantly increased compared to stacked membranes. These results suggest that Golgi cisternal stacking can directly regulate vesicle formation and thus the rate of protein transport through the Golgi. The results further suggest that at the onset of mitosis, unstacking of cisternae allows extensive and rapid vesiculation of the Golgi in preparation for its subsequent partitioning.  相似文献   

4.
In mammalian cells, flat Golgi cisternae closely arrange together to form stacks. During mitosis, the stacked structure undergoes a continuous fragmentation process. The generated mitotic Golgi fragments are distributed into the daughter cells, where they are reassembled into new Golgi stacks. In this study, an in vitro assay has been developed using purified proteins and Golgi membranes to reconstitute the Golgi disassembly and reassembly processes. This technique provides a useful tool to delineate the mechanisms underlying the morphological change. There are two processes during Golgi disassembly: unstacking and vesiculation. Unstacking is mediated by two mitotic kinases, cdc2 and plk, which phosphorylate the Golgi stacking protein GRASP65 and thus disrupt the oligomer of this protein. Vesiculation is mediated by the COPI budding machinery ARF1 and the coatomer complex. When treated with a combination of purified kinases, ARF1 and coatomer, the Golgi membranes were completely fragmented into vesicles. After mitosis, there are also two processes in Golgi reassembly: formation of single cisternae by membrane fusion, and restacking. Cisternal membrane fusion requires two AAA ATPases, p97 and NSF (N-ethylmaleimide-sensitive fusion protein), each of which functions together with specific adaptor proteins. Restacking of the newly formed Golgi cisternae requires dephosphorylation of Golgi stacking proteins by the protein phosphatase PP2A. This systematic study revealed the minimal machinery that controls the mitotic Golgi disassembly and reassembly processes.  相似文献   

5.
GRASP55 and GRASP65 have been implicated in stacking of Golgi cisternae and lateral linking of stacks within the Golgi ribbon. However, RNAi or gene knockout approaches to dissect their respective roles have often resulted in conflicting conclusions. Here, we gene-edited GRASP55 and/or GRASP65 with a degron tag in human fibroblasts, allowing for induced rapid degradation by the proteasome. We show that acute depletion of either GRASP55 or GRASP65 does not affect the Golgi ribbon, while chronic degradation of GRASP55 disrupts lateral connectivity of the ribbon. Acute double depletion of both GRASPs coincides with the loss of the vesicle tethering proteins GM130, p115, and Golgin-45 from the Golgi and compromises ribbon linking. Furthermore, GRASP55 and/or GRASP65 is not required for maintaining stacks or de novo assembly of stacked cisternae at the end of mitosis. These results demonstrate that both GRASPs are dispensable for Golgi stacking but are involved in maintaining the integrity of the Golgi ribbon together with GM130 and Golgin-45.  相似文献   

6.
The mammalian Golgi complex is comprised of a ribbon of stacked cisternal membranes often located in the pericentriolar region of the cell. Here, we report that during apoptosis the Golgi ribbon is fragmented into dispersed clusters of tubulo-vesicular membranes. We have found that fragmentation is caspase dependent and identified GRASP65 (Golgi reassembly and stacking protein of 65 kD) as a novel caspase substrate. GRASP65 is cleaved specifically by caspase-3 at conserved sites in its membrane distal COOH terminus at an early stage of the execution phase. Expression of a caspase-resistant form of GRASP65 partially preserved cisternal stacking and inhibited breakdown of the Golgi ribbon in apoptotic cells. Our results suggest that GRASP65 is an important structural component required for maintenance of Golgi apparatus integrity.  相似文献   

7.
Coat protein I (COPI) transport vesicles can be tethered to Golgi membranes by a complex of fibrous, coiled-coil proteins comprising p115, Giantin and GM130. p115 has been postulated to act as a bridge, linking Giantin on the vesicle to GM130 on the Golgi membrane. Here we show that the acidic COOH terminus of p115 mediates binding to both GM130 and Giantin as well as linking the two together. Phosphorylation of serine 941 within this acidic domain enhances the binding as well as the link between them. Phosphorylation is mediated by casein kinase II (CKII) or a CKII-like kinase. Surprisingly, the highly conserved NH(2)-terminal head domain of p115 is not required for the NSF (N-ethylmaleimide-sensitive fusion protein)-catalyzed reassembly of cisternae from mitotic Golgi fragments in a cell-free system. However, the ability of p115 to link GM130 to Giantin and the phosphorylation of p115 at serine 941 are required for NSF-catalyzed cisternal regrowth. p115 phosphorylation may be required for the transition from COPI vesicle tethering to COPI vesicle docking, an event that involves the formation of trans-SNARE [corrected] (trans-soluble NSF attachment protein [SNAP] receptor) complexes.  相似文献   

8.
Together with other Golgi matrix components, GRASP65 contributes to the stacking of Golgi cisternae in interphase cells. During mitosis, GRASP65 is heavily phosphorylated, and in turn, cisternal stacking is inhibited leading to the breakdown of the Golgi apparatus. Here we show that GRASP65 is phosphorylated on serine 277 in interphase cells, and this is strongly enhanced in response to the addition of serum or epidermal growth factor. This is directly mediated by ERK suggesting that GRASP65 has some role in growth factor signal transduction. Phosphorylation of Ser-277 is also dramatically increased during mitosis, however this is mediated by Cdk1 and not by ERK. The microinjection of recombinant GRASP65 without N-terminal myristoylation or a peptide fragment containing Ser-277 into the cytosol of normal rat kidney cells inhibits passage through mitosis. This effect is abolished when Ser-277 is replaced with alanine suggesting the phosphorylation of Ser-277 plays an important role in cell cycle regulation. The convergence of cell cycle regulation and growth factor signals on GRASP65 Ser-277 suggests that GRASP65 may function as a signal integrator controlling the cell growth.  相似文献   

9.
A direct role for GRASP65 as a mitotically regulated Golgi stacking factor   总被引:1,自引:0,他引:1  
Cell-free assays that mimic the disassembly and reassembly cycle of the Golgi apparatus during mitosis implicated GRASP65 as a mitotically regulated stacking factor. We now present evidence that GRASP65 is directly involved in stacking Golgi cisternae. GRASP65 is the major phosphorylation target in rat liver Golgi membranes of two mitotic kinases, cdc2-cyclin B and polo-like kinases, which alone will unstack Golgi membranes, generating single cisternae. Mitotic cells microinjected with antibodies to GRASP65 fail to form proper Golgi stacks after cell division. Beads coated with GRASP65 homodimers form extensive aggregates consistent with the formation of trans oligomers. These can be disaggregated using purified cdc2-cyclin B1 and polo-like kinases, and re-aggregated after dephosphorylation of GRASP65. Together, these data demonstrate that GRASP65 has the properties required to bind surfaces together in a mitotically regulated manner.  相似文献   

10.
At the onset of mitosis, the pericentriolar Golgi apparatus of mammalian cells is converted into small fragments, which are dispersed throughout the cytosol. The Golgi-associated protein GRASP65 is involved in this process. To address the role of GRASP65 in mitotic Golgi fragmentation, we depleted the protein from HeLa cells by RNAi. In the absence of GRASP65, the number of cisternae per Golgi stack is reduced without affecting the overall organization of Golgi membranes and protein transport. GRASP65-depleted cells entered mitosis, but accumulated in metaphase with condensed chromatin and multiple aberrant spindles and eventually died. Although Centrin2 and g-tubulin were detected in two of the spindle poles, the other spindle poles contained g-tubulin, but not Centrin2. Furthermore, we provide evidence that the expression of the C-terminus of GRASP65 interferes with entry of cells into mitosis. Our results suggest the requirement for GRASP65 in the regulation of spindle dynamics rather than a direct role in the stacking of Golgi cisternae. This novel function is in addition to the previously established negative role of GRASP65 at the G2/M transition, which is mediated by its C-terminus.  相似文献   

11.
Kuo A  Zhong C  Lane WS  Derynck R 《The EMBO journal》2000,19(23):6427-6439
Transforming growth factor-alpha (TGF-alpha) and related proteins represent a family of transmembrane growth factors with representatives in flies and worms. Little is known about the transport of TGF-alpha and other transmembrane growth factors to the cell surface and its regulation. p59 was purified as a cytoplasmic protein, which at endogenous levels associates with transmembrane TGF-alpha. cDNA cloning of p59 revealed a 452 amino acid sequence with two PDZ domains. p59 is myristoylated and palmitoylated, and associates with the Golgi system, where it co-localizes with TGF-alpha. Its first PDZ domain interacts with the C-terminus of transmembrane TGF-alpha and select transmembrane proteins. p59 is the human homolog of GRASP55, which is structurally related to GRASP65. GRASP55 and GRASP65 have been shown to play a role in stacking of the Golgi cisternae in vitro. C-terminal mutations of transmembrane TGF-alpha, which decrease or abolish the interaction with p59, also strongly impair cell surface expression of TGF-alpha. Our observations suggest a role for membrane tethering of p59/GRASP55 to select transmembrane proteins, including TGF-alpha, in maturation and transport to the cell surface.  相似文献   

12.
The Golgi apparatus in animal cells breaks down at the onset of mitosis and is later rebuilt in the two daughter cells. Two AAA ATPases, NSF and p97/VCP, have been implicated in regulating membrane fusion steps that lead to regrowth of Golgi cisternae from mitotic fragments. NSF dissociates complexes of SNARE proteins, thereby reactivating them to mediate membrane fusion. However, NSF has a second function in regulating SNARE pairing together with the ubiquitin-like protein GATE-16. p97/VCP, on the other hand, is involved in a cycle of ubiquitination and deubiquitination of an unknown target that governs Golgi membrane dynamics. Here, these findings are reviewed and discussed in the context of the increasingly evident role of ubiquitin in membrane traffic processes.  相似文献   

13.
The Golgi apparatus in animal cells breaks down at the onset of mitosis and is later rebuilt in the two daughter cells. Two AAA ATPases, NSF and p97/VCP, have been implicated in regulating membrane fusion steps that lead to regrowth of Golgi cisternae from mitotic fragments. NSF dissociates complexes of SNARE proteins, thereby reactivating them to mediate membrane fusion. However, NSF has a second function in regulating SNARE pairing together with the ubiquitin-like protein GATE-16. p97/VCP, on the other hand, is involved in a cycle of ubiquitination and deubiquitination of an unknown target that governs Golgi membrane dynamics. Here, these findings are reviewed and discussed in the context of the increasingly evident role of ubiquitin in membrane traffic processes.  相似文献   

14.
Cisternae of the Golgi apparatus adhere to each other to form stacks, which are aligned side by side to form the Golgi ribbon. Two proteins, GRASP65 and GRASP55, previously implicated in stacking of cisternae, are shown to be required for the formation of the Golgi ribbon.

IntroductionThe Golgi apparatus is an intermediate organelle along the secretory pathway that receives proteins and lipids (“cargo”) from the endoplasmic reticulum, covalently modifies them, and then exports them via transport vesicles for trafficking to the plasma membrane or other organelles. In most eukaryotic cells, disc-shaped membrane cisternae, each containing a distinct repertoire of cargo-processing enzymes, are stacked one on top of another to form the “Golgi stack,” a visual hallmark of the organelle (Fig. 1). The cisternae of the Golgi stack are polarized, with the compartment receiving endoplasmic reticulum–derived cargo termed the cis cisterna followed by the medial; trans; and finally, the trans-Golgi network. The physiological advantages conferred by stacking of Golgi cisternae are unclear, but it is thought to enhance the efficiencies of the sequential chemical modifications of glycoproteins and glycolipids during secretion. Cultured mammalian cells may possess more than 100 Golgi stacks, which are aligned side by side about the centrosome to form the “Golgi ribbon” (Fig. 1). Vesicles and tubules span the intervening, “noncompact” zones between stacks of cisternae, connecting analogous cisternae across the ribbon and thereby ensuring a homogeneous distribution of Golgi resident proteins among all cisternae. During mitosis, the Golgi ribbon is unlinked, the stacks are disassembled, and the cisternae are converted to vesicles and tubules; after cytokinesis, the process is reversed, and the Golgi is rebuilt. The dynamic nature of Golgi structure in interphase and mitotic cells implies the existence of a reversible mechanism that tethers Golgi cisternae to each other to form the stack and a mechanism that aligns and links the stacks into the ribbon.Open in a separate windowFigure 1.The organization of the Golgi apparatus in vertebrate cells. Individual stacks of Golgi cisternae are aligned side to side to form the Golgi ribbon. The GRASP65 and GRASP55 proteins are depicted to be enriched on the rims of the indicated cisternae within individual stacks of cisternae, where they are required to maintain the arrangement of stacks into the ribbon.GRASP proteins tether Golgi cisternae in vitroInvestigations into the molecular basis of Golgi cisterna stacking have ultimately focused attention on a handful of cytoplasmic proteins called “Golgins” and “GRASPs” that are associated with specific Golgi cisternae and interact with each other. Of particular interest are two related proteins GRASP65 and GRASP55 (respective systematic names GORASP1 and GORASP2), discovered by Warren and colleagues via in vitro reconstitution experiments, as capable of mediating stacking of Golgi cisternae (Barr et al., 1997; Shorter et al., 1999). Whereas GRASP65 localizes to the cis cisterna, GRASP55 localization favors medial/trans Golgi cisternae (Shorter et al., 1999); hence, these proteins could, in principle, tether cisternae to form a minimal Golgi stack. In these in vitro assays, perturbations (mutations, antibody interference) to either GRASP65 or GRASP55 inhibited stacking of reformed Golgi cisternae. Moreover, GRASP proteins are phosphorylated in mitosis just before vesiculation of Golgi cisternae, and preventing phosphorylation impairs the disassembly of the Golgi apparatus and mitotic progression (Wang et al., 2003). These findings underpin models of the Golgi stack where GRASP65 and GRASP55, along with Golgin proteins, constitute the core components of a cytoplasmic “matrix” of proteins that surround the cisternae, mediating their stacking as well as the tethering of transport vesicles to cisternae. Curiously, plant cells contain stacked Golgi cisternae, yet they do not express any GRASP or GRASP-related proteins. And some nonvertebrate organisms with stacked Golgi cisternae express just one GRASP-related protein, while the Golgi cisternae are not stacked in other nonvertebrate organisms (e.g., yeast) that express a single GRASP (Glick and Malhotra, 1998). Apparently, the presence or number of GRASP proteins expressed does not correlate with stacked cisternae.Whereas the results of in vitro biochemical assays underpin our conceptions of GRASP protein function, probing their roles in vivo has proven to be quite challenging. First, depletion/deletion of each individual GRASP protein is largely without effect on Golgi stack or ribbon formation, but a very complex phenotype results from depletion/deletion of both GRASP proteins. Thus, some reports conclude that the GRASP proteins function redundantly to stack cisternae (Bekier et al., 2017), while others conclude that the Golgi ribbon, not the stack per se, is perturbed upon loss of GRASP proteins (Puthenveedu et al., 2006; Feinstein and Linstedt, 2008; Xiang and Wang, 2010; Lee et al., 2014; Veenendaal et al., 2014). Recently, two papers published in the Journal of Cell Biology employed different methodologies to perturb GRASP protein functions in vivo (Grond et al., 2020; Zhang and Seemann, 2021), providing the most conclusive insight to date into the roles of GRASP proteins in Golgi structure.The Golgi ribbon is unlinked upon loss of GRASP proteinsRabouille and colleagues used traditional mouse gene knockout technology to delete GRASP65, finding that such mice are viable with no apparent physiological deficits or gross morphological perturbations of the Golgi (Veenendaal et al., 2014). In their recent study (Grond et al., 2020), GRASP55 was deleted in the GRASP65 null background, but double-knockout mice could not be obtained, consistent with GRASP proteins being at least partially physiologically redundant. Next, using a conditional knockout approach, double GRASP null cells were produced postnatally in the small intestine, and the Golgi of intestinal epithelial cells was examined. In these cells, stacked Golgi cisternae were observed, but their arrangement into a ribbon was compromised, a result corroborated by more detailed analysis of cells in organoid cultures. These findings are at odds with the conclusions of Wang and colleagues (Bekier et al., 2017), who used CRISPR-Cas9 gene editing technology to construct cultured mammalian cell lines that do not express GRASP65 and GRASP55. They found that the appearance of Golgi cisternae was grossly altered, resembling clusters of tubules and vesicles (“tubulovesicular clusters”) about swollen cisterna remnants that debatably appeared to be stacked. One possible reason for the disparities between these two studies is that Bekier et al. (2017) documented that loss of GRASP proteins in cultured mammalian cells also resulted in depletion of a subset of Golgin proteins (e.g., GM130, Golgin-45) from Golgi cisternae, so it was not possible to parse the specific contributions of GRASP proteins to Golgi structure.Analyses of siRNA-depleted and gene-edited cell lines and modified animals are often complicated by incomplete depletion of a query protein, unintended loss of other proteins, or compensatory processes that obscure loss-of-function effects. Notably, siRNA depletion of GM130, which is associated with GRASP65 on the cis cisterna, impairs secretory traffic from the endoplasmic reticulum to the Golgi apparatus, resulting in a reduction in the size of Golgi cisternae and diminished interstack connectivity possibly due to vesiculation of cisternae (Seemann et al., 2000; Puthenveedu et al., 2006). To minimize these drawbacks, Zhang and Seemann (2021) used gene editing to modify the GRASP65 and GRASP55 loci to append an inducible protein degradation domain to each protein in cultured mammalian cells, which was used to elicit degradation of the GRASP proteins within just 2 h. Hence, the acute effects of GRASP protein depletion could be determined before the onset of potentially confounding effects. Fluorescence recovery after photobleaching assays of a fluorescently tagged Golgi resident protein revealed that acute depletion of both GRASP65 and GRASP55 resulted in decreased mobility of the resident Golgi enzyme within the ribbon, indicating that connectivity of cisternae between stacks was compromised. Stacks of Golgi cisternae with proper cis–trans polarity were observed by electron and light microscopy, both shortly (∼2 h) after GRASP protein turnover was initiated, and after mitosis, indicating that GRASP proteins are not required to establish or to maintain the Golgi stacks. Importantly, the authors observed no changes in the levels of GRASP-associated proteins (e.g., GM130) when assayed shortly after initiating GRASP protein turnover, but the amounts of several GRASP-associated proteins were reduced after prolonged growth in the absence of GRASP proteins. The results are in general agreement with experiments by Jarvela and Linstedt (2014), who expressed GRASP65 and GRASP55 fusion proteins appended with “killer RFP” and used chomophore-assisted light inactivation to rapidly (1 min) ablate the proteins in cultured mammalian cells. Similar to Zhang and Seemann (2021), they observed that the Golgi ribbon was disassembled upon inactivation of GRASP proteins, but stacking of cisternae was unaffected. Taken together, these results conclusively show that acute depletion of GRASP65 and GRASP55 impairs lateral linking of stacked Golgi cisternae within the ribbon while not affecting stacking of cisternae.Conclusions and perspectivesA body of work now more than 20 years old has shown that GRASP65 and GRAPS55 are core structural components of a matrix of cytoplasmic proteins associated with Golgi cisternae; however, the Grond et al. (2020) and Zhang and Seemann (2021) reports now firmly establish that GRASP proteins are dispensable for stacking of Golgi cisterna and indicate that they are required for linking Golgi stacks within the ribbon. These new studies suggest that the integrity of the Golgi matrix critically depends on the presence of GRASP proteins, and their absence perturbs the balance of cargo flow through the Golgi, reducing the interstack exchange required to maintain connectivity of stacks within the ribbon. How might GRASP proteins facilitate linking of stacks within the Golgi ribbon? When the ribbon is disrupted (using the microtubule depolymerizing reagent nocodazole) and individual Golgi stacks are examined, GRASP65 and GRASP55 appear to be enriched at the rims of Golgi cisternae (Fig. 1; Tie et al., 2018). Hence, the GRASP proteins are positioned at the vesicle-rich interface between adjacent cisternal stacks. Grond et al. (2020) observed reductions in the size of Golgi cisternae in cells deleted of both GRASP proteins and speculated that this may be due to increased coatomer I vesicle formation at the rims of cisternae. In this view, GRASP proteins dampen vesicle flux at the rims of Golgi cisternae, a model supported by the observation that depletion of GRASP proteins leads to an increase in secretion rate (Wang et al., 2008). These new studies firmly shift our view of GRASP protein function away from the stacking of Golgi cisternae, and we look forward to new mechanistic insights into the roles of GRASP proteins in Golgi ribbon formation as well as in non–Golgi-dependent processes, such as unconventional protein secretion (Kinseth et al., 2007).  相似文献   

15.
The Golgi reassembly stacking protein (GRASP) family has been implicated in the stacking of Golgi cisternae and the regulation of Golgi disassembly/reassembly during mitosis in mammalian cells. GRASP65 is a dimer that can directly link adjacent surfaces through trans-oligomerization in a mitotically regulated manner. Here we show that the N-terminal GRASP domain (amino acids 1-201) is both necessary and sufficient for dimerization and trans-oligomerization but is not mitotically regulated. The C-terminal serine/proline-rich domain (amino acids 202-446) cannot dimerize nor can it link adjacent surfaces. It does, however, confer mitotic regulation on the GRASP domain through multiple sites phosphorylated by the mitotic kinases, cdc2/B1, and the polo-like kinase. Transient expression corroborated these results by showing that the GRASP domain alone inhibited mitotic fragmentation of the Golgi apparatus.  相似文献   

16.
GRASP65 (Golgi reassembly and stacking protein of 65 KDa) is a cis-Golgi protein with roles in Golgi structure, membrane trafficking and cell signalling. It is cleaved by caspase-3 early in apoptosis, promoting Golgi fragmentation. We now show that cleavage is needed for Fas-mediated apoptosis: expression of caspase-resistant GRASP65 protects cells, whereas expression of membrane proximal caspase-cleaved GRASP65 fragments dramatically sensitises cells. GRASP65 coordinates passage through the Golgi apparatus of proteins containing C-terminal hydrophobic motifs, via its tandem PDZ type ‘GRASP'' domains. Fas/CD95 contains a C-terminal leucine–valine pairing so its trafficking might be coordinated by GRASP65. Mutagenesis of the Fas/CD95 LV motif reduces the number of cells with Golgi-associated Fas/CD95, and generates a receptor that is more effective at inducing apoptosis; however, siRNA-mediated silencing or expression of mutant GRASP65 constructs do not alter the steady state distribution of Fas/CD95. We also find no evidence for a GRASP65–Fas/CD95 interaction at the molecular level. Instead, we find that the C-terminal fragments of GRASP65 produced following caspase cleavage are targeted to mitochondria, and ectopic expression of these sensitises HeLa cells to Fas ligand. Our data suggest that GRASP65 cleavage promotes Fas/CD95-mediated apoptosis via release of C-terminal fragments that act at the mitochondria, and we identify Bcl-XL as a candidate apoptotic binding partner for GRASP65.  相似文献   

17.
The Golgi apparatus is a highly complex organelle comprised of a stack of cisternal membranes on the secretory pathway from the ER to the cell surface. This structure is maintained by an exoskeleton or Golgi matrix constructed from a family of coiled-coil proteins, the golgins, and other peripheral membrane components such as GRASP55 and GRASP65. Here we find that TMP21, p24a, and gp25L, members of the p24 cargo receptor family, are present in complexes with GRASP55 and GRASP65 in vivo. GRASPs interact directly with the cytoplasmic domains of specific p24 cargo receptors depending on their oligomeric state, and mutation of the GRASP binding site in the cytoplasmic tail of one of these, p24a, results in it being transported to the cell surface. These results suggest that one function of the Golgi matrix is to aid efficient retention or sequestration of p24 cargo receptors and other membrane proteins in the Golgi apparatus.  相似文献   

18.
Membrane traffic between the endoplasmic reticulum (ER) and Golgi apparatus and through the Golgi apparatus is a highly regulated process controlled by members of the rab GTPase family. The GTP form of rab1 regulates ER to Golgi transport by interaction with the vesicle tethering factor p115 and the cis-Golgi matrix protein GM130, also part of a complex with GRASP65 important for the organization of cis-Golgi cisternae. Here, we find that a novel coiled-coil protein golgin-45 interacts with the medial-Golgi matrix protein GRASP55 and the GTP form of rab2 but not other Golgi rab proteins. Depletion of golgin-45 disrupts the Golgi apparatus and causes a block in secretory protein transport. These results demonstrate that GRASP55 and golgin-45 form a rab2 effector complex on medial-Golgi essential for normal protein transport and Golgi structure.  相似文献   

19.
The mammalian GRASPs (Golgi reassembly stacking proteins) GRASP65 and GRASP55 were first discovered more than a decade ago as factors involved in the stacking of Golgi cisternae. Since then, orthologues have been identified in many different organisms and GRASPs have been assigned new roles that may seem disconnected. In vitro, GRASPs have been shown to have the biochemical properties of Golgi stacking factors, but the jury is still out as to whether they act as such in vivo. In mammalian cells, GRASP65 and GRASP55 are required for formation of the Golgi ribbon, a structure which is fragmented in mitosis owing to the phosphorylation of a number of serine and threonine residues situated in its C-terminus. Golgi ribbon unlinking is in turn shown to be part of a mitotic checkpoint. GRASP65 also seems to be the key target of signalling events leading to re-orientation of the Golgi during cell migration and its breakdown during apoptosis. Interestingly, the Golgi ribbon is not a feature of lower eukaryotes, yet a GRASP homologue is present in the genome of Encephalitozoon cuniculi, suggesting they have other roles. GRASPs have no identified function in bulk anterograde protein transport along the secretory pathway, but some cargo-specific trafficking roles for GRASPs have been discovered. Furthermore, GRASP orthologues have recently been shown to mediate the unconventional secretion of the cytoplasmic proteins AcbA/Acb1, in both Dictyostelium discoideum and yeast, and the Golgi bypass of a number of transmembrane proteins during Drosophila development. In the present paper, we review the multiple roles of GRASPs.  相似文献   

20.
Directed cell migration requires the orientation of the Golgi and centrosome toward the leading edge. We show that stimulation of interphase cells with the mitogens epidermal growth factor or lysophosphatidic acid activates the extracellular signal–regulated kinase (ERK), which phosphorylates the Golgi structural protein GRASP65 at serine 277. Expression of a GRASP65 Ser277 to alanine mutant or a GRASP65 1–201 truncation mutant, neither of which can be phosphorylated by ERK, prevents Golgi orientation to the leading edge in a wound assay. We show that phosphorylation of GRASP65 with recombinant ERK leads to the loss of GRASP65 oligomerization and causes Golgi cisternal unstacking. Furthermore, preventing Golgi polarization by expressing mutated GRASP65 inhibits centrosome orientation, which is rescued upon disassembly of the Golgi structure by brefeldin A. We conclude that Golgi remodeling, mediated by phosphorylation of GRASP65 by ERK, is critical for the establishment of cell polarity in migrating cells.  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