首页 | 官方网站   微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Lipase maturation factor 1 (LMF1) is predicted to be a polytopic protein localized to the endoplasmic reticulum (ER) membrane. It functions in the post-translational attainment of enzyme activity for both lipoprotein lipase and hepatic lipase. By using transmembrane prediction methods in mouse and human orthologs, models of LMF1 topology were constructed and tested experimentally. Employing a tagging strategy that used insertion of ectopic glycan attachment sites and terminal fusions of green fluorescent protein, we established a five-transmembrane model, thus dividing LMF1 into six domains. Three domains were found to face the cytoplasm (the amino-terminal domain and loops B and D), and the other half was oriented to the ER lumen (loops A and C and the carboxyl-terminal domain). This representative model shows the arrangement of an evolutionarily conserved domain within LMF1 (DUF1222) that is essential to lipase maturation. DUF1222 comprises four of the six domains, with the two largest ones facing the ER lumen. We showed for the first time, using several naturally occurring variants featuring DUF1222 truncations, that Lmf1 interacts physically with lipoprotein lipase and hepatic lipase and localizes the lipase interaction site to loop C within DUF1222. We discuss the implication of our results with regard to lipase maturation and DUF1222 domain structure.  相似文献   

2.
Transmembrane proteins are synthesized and folded in the endoplasmic reticulum (ER), an interconnected network of flattened sacs or tubes. Up to now, this organelle has eluded a detailed analysis of the dynamics of its constituents, mainly due to the complex three-dimensional morphology within the cellular cytosol, which precluded high-resolution, single-molecule microscopy approaches. Recent evidences, however, pointed out that there are multiple interaction sites between ER and the plasma membrane, rendering total internal reflection microscopy of plasma membrane proximal ER regions feasible. Here we used single-molecule fluorescence microscopy to study the diffusion of the human serotonin transporter at the ER and the plasma membrane. We exploited the single-molecule trajectories to map out the structure of the ER close to the plasma membrane at subdiffractive resolution. Furthermore, our study provides a comparative picture of the diffusional behavior in both environments. Under unperturbed conditions, the majority of proteins showed similar mobility in the two compartments; at the ER, however, we found an additional 15% fraction of molecules moving with 25-fold faster mobility. Upon degradation of the actin skeleton, the diffusional behavior in the plasma membrane was strongly influenced, whereas it remained unchanged in the ER.Live-cell microscopy and three-dimensional electron tomography has boosted our understanding of endoplasmic reticulum (ER) dynamics and morphology. Proteins have been identified which regulate the formation of cisternae versus tubelike membranes, and the contacts between ER and the various cellular organelles have been studied in detail (1). Little information, however, is available when it comes to protein dynamics and organization within the ER membrane. Its complex three-dimensional topology hampers standard diffraction-limited fluorescence microscopy approaches: in fluorescence recovery after photobleaching, for example, the obtained diffusion coefficients can be several-folds off, if the ER morphology is not correctly taken into account (2). A method is therefore needed which allows for resolving molecular movements on length scales below the typical dimensions of the ER structures.In principle, single-molecule tracking would provide the required spatial resolution due to the high precision in localizing the moving point emitters: localization errors of <40 nm can be easily achieved (3). This technique has given rise to multiple studies, in which the paths of the diffusing objects were used to make conclusions on the properties of the environment; particularly, the plasma membrane has become a favorite target for such investigations, yielding precise determinations of the diffusion coefficients of a variety of membrane proteins or lipids (4).Here, we report what is, to our knowledge, the first application of single-molecule tracking for a comparative study of the diffusion dynamics of a membrane protein at the ER versus the plasma membrane. As the protein of interest, we chose the human serotonin transporter (SERT): it is a polytopic membrane protein containing 12 transmembrane domains, with both C- and N-termini residing in the cytoplasm. Stable SERT oligomers of various degrees were observed to coexist in the plasma membrane (5). Functionally, SERT (6) is a pivotal element in shaping serotonergic neurotransmission: SERT-mediated high-affinity uptake of released serotonin clears the synaptic cleft and supports refilling of vesicular stores (7). Wild-type SERT (SERT-wt) is efficiently targeted to the presynaptic plasma membrane, whereas the truncation of its C-terminus (SERT-ΔC30) retains the mutant protein in the ER (8). The N-terminal mGFP- and eYFP-fusion constructs of the two versions of SERT thus allowed us to specifically address SERT located at the ER (eYFP-SERT-ΔC30) or at the plasma membrane (mGFP-SERT-wt (7)).Our experiments were performed at 37°C on proteins heterologously expressed in CHO cells. Total internal reflection (TIR) illumination afforded a reduction in background fluorescence and allowed for selective imaging of single mGFP-SERT-wt molecules at the cells’ plasma membrane or single eYFP-SERT-ΔC30 molecules at plasma membrane-proximal ER (Fig. 1 and see the Supporting Material). TIR was particularly crucial for single-molecule imaging of the ER-retained mutant, where out-of-focus background would surpass the weak single-molecule signals in epi-illumination.Open in a separate windowFigure 1Schematics of the plasma membrane (PM) and a part of the ER containing mGFP-SERT-wt or the ER-retained eYFP-SERT-ΔC30 mutant, respectively. Both can be excited by total internal reflection fluorescence (TIRF) excitation. Experiments were carried out either on cells expressing mGFP-SERT-wt or eYFP-SERT-ΔC30.For both mutants, the majority of molecules were mobile: in fluorescence-recovery-after-photobleaching experiments we observed a mobile fraction of 82 ± 8% for mGFP-SERT-wt and 91 ± 4% for eYFP-SERT-ΔC30. For single-molecule tracking, the high surface density of signals was reduced by completely photobleaching a rectangular part of the cell in epi-illumination; after a brief recovery period, a few single-molecule signals had entered the bleached area and could be monitored and tracked at high signal/noise using TIR excitation. Samples were illuminated stroboscopically for till = 2 ms, and movies of 500 frames were recorded with a delay of tdel = 6 ms; the short delay times ensured that even rapidly diffusing molecules hardly reached the borders of the ER tubes between two consecutive frames. This illumination protocol was run for 20 times per cell, yielding ∼2500 trajectories per cell.The single-molecule localizations were first used to map those areas that are accessible to the diffusing proteins. eYFP-SERT-ΔC30 showed distinct hotspots, representing plasma membrane-proximal ER, excitable by the evanescent field (Fig. 2 A). These hotspots hardly moved within the timescale of an experiment (tens of minutes, see Fig. S1 in the Supporting Material); indeed, remarkable ER stability was previously observed using superresolution microscopy (9). In contrast, a rather homogeneous distribution was observed for mGFP-SERT-wt in the plasma membrane (Fig. 2 B).Open in a separate windowFigure 2Superresolution and tracking data at the ER and the plasma membrane. Superresolution images are shown for the ER-retained SERT mutant eYFP-SERT-ΔC30 (A) and for mGFP-SERT-wt in the plasma membrane (B). (C and D) Diffusion coefficients of eYFP-SERT-ΔC30 (C) and mGFP-SERT-wt (D) are shown as normalized histograms before (blue) and after (red) Cytochalasin D treatment. Data were fitted by Gaussian mobility distributions (see Table S1 in the Supporting Material for the fit results).Next, we compared the mobility of the observed proteins. Single-molecule localizations were linked to trajectories as described in Gao and Kilfoil (10), and the apparent diffusion coefficient, D, of each molecule was estimated from the first two points of the mean-square displacement membrane. The distribution of log10 D showed a pronounced single peak (Fig. 2 D). It could be well fitted by a linear combination of two Gaussian functions, with the major fraction (85%) characterized by Dwt = 0.30 μm2/s; a broad shoulder to the left indicates the presence of proteins that are immobilized during the observation period. In contrast, the mobility of the ER-retained mutant showed a substantially different distribution, containing two clearly visible peaks (Fig. 2 C). We fitted the data with a three-component Gaussian model: the main fraction (82%) behaved similar to SERT at the plasma membrane, with DΔC30 = 0.32 μm2/s. In addition, a large fraction (15%) with high mobility of DΔC30 = 7.8 μm2/s and a minor fraction (3%) with low mobility was observed. The proteins responded as expected to degradation of the actin membrane skeleton (red bars in Fig. 2, C and D): at the plasma membrane, the mobility of mGFP-SERT-wt increased 4.6-fold (mean values), whereas at the ER membrane there was only a minor change for eYFP-SERT-ΔC30 mobility (1.06-fold increase; note that the ER is not connected to actin filaments (11)).The observation of a high mobility subfraction at the ER membrane is surprising. In general, the presence of obstacles—irrespective of whether randomly distributed or clustered, mobile or immobile—reduces the diffusivity of mobile tracers in a membrane (12). It is generally assumed that the high protein density in cell membranes is responsible for the rather low fluidity when compared to synthetic membranes (compare, e.g., Saxton and Jacobson (13) with Weiss et al. (14)). Interestingly, the observed diffusion constant of 7.8 μm2/s is of similar order as the mobility determined for various proteins in synthetic lipid membranes (14). It is thus tempting to hypothesize the presence of extended protein-depleted regions of higher fluidity within the ER membrane; such membrane domains were indeed observed already at the plasma membrane (15). We were also concerned, however, that protein degradation fragments could have contributed to our data: the three-dimensional mobility of an 85-kDa protein is ∼10 μm2/s (16), similar to the high mobility diffusion constant of eYFP-SERT-ΔC30.We tested the two explanations by analyzing the spatial distribution of fast (DΔC30 > 1 μm2/s) versus slow trajectories (DΔC30 < 1 μm2/s) of eYFP-SERT-ΔC30 (Fig. 3). Both types of trajectories clustered in the same regions, and no segregation into ER subdomains was observable at the resolved length scales. This finding—on the one hand—disfavors freely diffusing protein fragments as the origin of the high mobility fraction. On the other hand, it calls for further experiments to identify the origin of the fast and the slow mobility subfraction. Interestingly, when analyzing all eYFP-SERT-ΔC30 trajectories we found that 80% of the molecules showed diffusion confined to domains of 230-nm radius (see Fig. S2). This size is clearly smaller than the lateral extensions of the visible ER regions observed in Fig. 3. The finding indicates domain formation at the ER membrane; domains are averaged out in Fig. 3 due to the long recording times. Note that free diffusion was observed for mGFP-SERT-wt at the plasma membrane (5).Open in a separate windowFigure 3Ripley’s K function analysis of the different mobility fractions in the ER. For the cell presented in Fig. 2, the first position of every slow (D < 1 μm2/s; red) and fast (D > 1 μm2/s; blue) trajectory was plotted in panel A. Contour lines indicate regions of ER attachment to the plasma membrane. In panel B, the point-correlation function L(r)−r is plotted for the slow (red) and fast (blue) fraction. Furthermore, the correlation between fast versus slow is plotted (green). All three curves show a peak at ∼450 nm, which agrees with the extensions of the ER attachment zones.In conclusion, we have shown that single-molecule tracking is feasible for constituents of the ER membrane. We found a surprising diffusion behavior of SERT resulting in the following:
  • 1.A slow fraction showing mobility reminiscent of protein diffusion in the plasma membrane, likely reflecting SERT diffusing in protein-crowded regions of the ER membrane; and
  • 2.A fast fraction showing 25-fold faster diffusion kinetics.
This likely represents diffusion in altered ER membrane environments, possibly of different lipid or protein composition. Given the fact that synthesis of virtually all membrane proteins and most lipids proceeds at the ER membrane, ER heterogeneity at the nanoscale due to the continuous synthesis activity and selection for correct folding appears highly plausible.  相似文献   

3.
Transmembrane proteins are synthesized and folded in the endoplasmic reticulum (ER), an interconnected network of flattened sacs or tubes. Up to now, this organelle has eluded a detailed analysis of the dynamics of its constituents, mainly due to the complex three-dimensional morphology within the cellular cytosol, which precluded high-resolution, single-molecule microscopy approaches. Recent evidences, however, pointed out that there are multiple interaction sites between ER and the plasma membrane, rendering total internal reflection microscopy of plasma membrane proximal ER regions feasible. Here we used single-molecule fluorescence microscopy to study the diffusion of the human serotonin transporter at the ER and the plasma membrane. We exploited the single-molecule trajectories to map out the structure of the ER close to the plasma membrane at subdiffractive resolution. Furthermore, our study provides a comparative picture of the diffusional behavior in both environments. Under unperturbed conditions, the majority of proteins showed similar mobility in the two compartments; at the ER, however, we found an additional 15% fraction of molecules moving with 25-fold faster mobility. Upon degradation of the actin skeleton, the diffusional behavior in the plasma membrane was strongly influenced, whereas it remained unchanged in the ER.  相似文献   

4.
Hundreds of eukaryotic membrane proteins are anchored to membranes by a single transmembrane domain at their carboxyl terminus. Many of these tail-anchored (TA) proteins are posttranslationally targeted to the endoplasmic reticulum (ER) membrane for insertion by the guided-entry of TA protein insertion (GET) pathway. In recent years, most of the components of this conserved pathway have been biochemically and structurally characterized. Get3 is the pathway-targeting factor that uses nucleotide-linked conformational changes to mediate the delivery of TA proteins between the GET pretargeting machinery in the cytosol and the transmembrane pathway components in the ER. Here we focus on the mechanism of the yeast GET pathway and make a speculative analogy between its membrane insertion step and the ATPase-driven cycle of ABC transporters.The mechanism of membrane protein insertion into the endoplasmic reticulum (ER) has been extensively studied for many years (Shao and Hegde 2011). From this work, the signal recognition particle (SRP)/Sec61 pathway has emerged as a textbook example of a cotranslational membrane insertion mechanism (Grudnik et al. 2009). The SRP binds a hydrophobic segment (either a cleavable amino-terminal signal sequence or a transmembrane domain) immediately after it emerges from the ribosomal exit tunnel. This results in a translational pause that persists until SRP engages its receptor in the ER and delivers the ribosome-nascent chain complex to the Sec61 channel. Last, the Sec61 channel enables protein translocation into the ER lumen along with partitioning of hydrophobic transmembrane domains into the lipid bilayer through the Sec61 lateral gate (Rapoport 2007).Approximately 5% of all eukaryotic membrane proteins have an ER targeting signal in a single carboxy-terminal transmembrane domain that emerges from the ribosome exit tunnel following completion of protein synthesis and is not recognized by the SRP (Stefanovic and Hegde 2007). Nonetheless, because hydrophobic peptides in the cytoplasm are prone to aggregation and subject to degradation by quality control systems (Hessa et al. 2011), these tail-anchored (TA) proteins still have to be specifically recognized, shielded from the aqueous environment, and guided to the ER membrane for insertion. In the past five years, the guided-entry of TA proteins (GET) pathway has come to prominence as the major machinery for performing these tasks and the enabler of many key cellular processes mediated by TA proteins including vesicle fusion, membrane protein insertion, and apoptosis. This research has rapidly yielded biochemical and structural insights (and2)2) into many of the GET pathway components (Hegde and Keenan 2011; Chartron et al. 2012a; Denic 2012). In particular, Get3 is an ATPase that uses metabolic energy to bridge recognition of TA proteins by upstream pathway components with TA protein recruitment to the ER for membrane insertion. However, the precise mechanisms of nucleotide-dependent TA protein binding to Get3 and how the GET pathway inserts tail anchors into the membrane are still poorly understood. Here, we provide an overview of the budding yeast GET pathway with emphasis on mechanistic insights that have come from structural studies of its membrane-associated steps and make a speculative juxtaposition with the ABC transporter mechanism.

Table 1.

A catalog of GET pathway component structures
ComponentRole in the pathwayPDB ID
Sgt2Component of the pretargeting complex that delivers TA proteins to Get3; dimer interacts with Get4/Get5, contains TPR repeats that interact with Hsps3SZ7
Get5Component of the pretargeting complex that delivers TA proteins to Get3; dimer interacts with Get4 via amino-terminal domain and with Sgt2 via its ubiquitin-like domain2LNZ
3VEJ
2LO0
Get4Component of the pretargeting complex that delivers TA proteins to Get3; interacts with Get3 via amino-terminal domain and with Get4 via carboxy-terminal domain3LPZ
3LKU
3WPV
Get3ATPase that binds the TA protein; dimer interacts with the pretargeting complex in the cytosol, and with Get1/2 at the ER membraneTable 2
Get1ER receptor for Get3; integral ER membrane
protein, three TMDs; forms a complex with Get2
3SJA, 3SJB
3SJC, 3ZS8
3VLC, 3B2E
Get2ER receptor for Get3; integral ER membrane
protein, three TMDs; forms a complex with Get1
3SJD
3ZS9
Open in a separate windowTA, tail anchored; TPR, tetratricopeptide repeat; TMDs, transmembrane domains.

Table 2.

An itemized list of published Get3 structures with associated nucleotides and conformation nomenclature
OrganismNucleotideConformationPDB IDReferences
Get3
Schizosaccharomyces pombeNoneOpen2WOOMateja et al. 2009
Saccharomyces cerevisiaeNoneOpen3H84Hu et al. 2009
3A36Yamagata et al. 2010
Aspergillus fumigatusADPOpen3IBGSuloway et al. 2009
S. cerevisiaeADPOpen3A37Yamagata et al. 2010
Debaryomyces hanseniiADPClosed3IO3Hu et al. 2009
Chaetomium thermophilumAMPPNP-Mg2+Closed3IQWBozkurt et al. 2009
C. thermophilumADP-Mg2+Closed3IQXBozkurt et al. 2009
S. cerevisiaeADP•AlF4-Mg2+Fully closed2WOJMateja et al. 2009
Methanothermobacter thermautotrophicusADP•AlF4-Mg2+Fully closed3ZQ6Sherill et al. 2011
Methanococcus jannaschiiADP•AlF4-Mg2+Tetrameric3UG6Suloway et al. 2012
3UG7
Get3/Get2cyto
S. cerevisiaeADP-Mg2+Closed3SJDStefer et al. 2011
S. cerevisiaeADP•AlF4-Mg2+Closed3ZS9Mariappan et al. 2011
Get3/Get1cyto
S. cerevisiaeNoneSemiopen3SJCStefer et al. 2011
S. cerevisiaeADPSemiopen3VLCKubota et al. 2012
S. cerevisiaeNoneOpen3SJAStefer et al. 2011
3SJBStefer et al. 2011
3ZS8Mariappan et al. 2011
ADPOpen3B2EKubota et al. 2012
Open in a separate windowADP, adenosine diphosphate.  相似文献   

5.
Peroxisome biogenesis initiates at the endoplasmic reticulum (ER) and maturation allows for the formation of metabolically active organelles. Yet, peroxisomes can also multiply by growth and division. Several proteins, called peroxins, are known to participate in these processes but little is known about their organization to orchestrate peroxisome proliferation. Here, we demonstrate that regulation of peroxisome proliferation relies on the integrity of the tubular ER network. Using a dual track SILAC-based quantitative interaction proteomics approach, we established a comprehensive network of stable as well as transient interactions of the peroxin Pex30p, an integral membrane protein. Through association with merely ER resident proteins, in particular with proteins containing a reticulon homology domain, and with other peroxins, Pex30p designates peroxisome contact sites at ER subdomains. We show that Pex30p traffics through the ER and segregates in punctae to which peroxisomes specifically append, and we ascertain its transient interaction with all subunits of the COPI coatomer complex suggesting the involvement of a vesicle-mediated transport. We establish that the membrane protein Pex30p facilitates the connection of peroxisomes to the ER. Taken together, our data indicate that Pex30p-containing protein complexes act as focal points from which peroxisomes can form and that the tubular ER architecture organized by the reticulon homology proteins Rtn1p, Rtn2p and Yop1p controls this process.All nucleated cells contain essential round-shaped organelles called peroxisomes, whose function is mainly associated with lipid metabolism (1). Depending on the cellular requirements, the size, number, and protein content of these single membrane-bound organelles can vary widely. Although peroxisomes are dispensable for unicellular species such as yeasts, they are essential for the development of multicellular organisms (2, 3). In human, mutations in PEX genes lead to defects in peroxisome function or formation and are associated with the development of lethal pathologies (4). These PEX genes code for proteins, called peroxins, which are involved in peroxisome assembly and maintenance (5).Two major routes seem to lead to peroxisome formation, namely, de novo biogenesis and growth/division of pre-existing peroxisomes. The division pathway operates with proteins of the Pex11 family and requires fission factors shared with mitochondria (6). Studies in yeast and mammalian cells revealed that through the action of the protein Pex3p peroxisome precursors can also originate from the endoplasmic reticulum (ER)1 and, via import of membrane and matrix proteins, mature into fully functional organelles (7, 8). Furthermore, several peroxisomal membrane proteins were shown to migrate to peroxisomes via the ER (7, 9, 10). The molecular mechanism underlying the biogenic pathway of peroxisome formation has not been clarified so far. Recent data based on cell-free vesicle-budding reactions, however, demonstrated that several peroxisomal proteins traffic from the ER to peroxisomes in a COPII vesicle-independent manner (11). These observations point to the existence of vesicular events to mediate the transport of peroxisomal membrane proteins from the ER. In fact, analysis of secretory mutant yeast cells already suggest that part of the ER-associated secretory machinery is involved in peroxisome biogenesis (12).The de novo biogenesis of peroxisomes and the growth/division pathways are usually seen as independent routes; however, these events may be coordinated and, thus, intimately linked. Indeed, peroxisomes need to acquire membrane components to proliferate and it has been proposed that their binding to the cell cortex or to the cytoskeleton allows their partitioning and segregation during cell division (1315).Among the proteins required for assembly of peroxisomes, the membrane proteins Pex23p and Pex24p play essential roles in the yeast Yarrowia lipolytica (16, 17). Homologs of these two proteins in Saccharomyces cerevisiae are Pex30p, Pex31p, and Pex32p, all containing at least one transmembrane domain and a dysferlin domain as common structural motifs, as well as Pex28p and Pex29p. In S. cerevisiae, these proteins seem to negatively control peroxisomal size and number (18, 19). Interestingly, Pex30p seems to exhibit species-specific differences in the regulation of peroxisome proliferation. While the lack of Pex30p in S. cerevisiae leads to an increase in the number of normal-sized peroxisomes (18), in Pichia pastoris its absence correlates with the appearance of fewer and clustered peroxisomes (20). Although peroxisomes are highly versatile organelles, under given conditions their total number per cell remains fairly constant owing to the delicate balance of proliferation, inheritance and degradation (21, 22). The question is: what are the molecular mechanisms responsible for the spatiotemporal organization of these events?Here, we present data obtained from a dual approach based on quantitative interaction proteomics using stable isotope labeling with amino acids in cell culture (SILAC) (23, 24) and live-cell imaging, revealing for the first time the dynamic interaction network around Pex30p and its function in the organization of ER-to-peroxisome membrane associations. We report the existence of a macromolecular membrane protein complex that acts as a hub for the regulation of peroxisome proliferation and movement. Our data suggest a direct role for the tubular cortical ER and the reticulon homology proteins Rtn1p, Rtn2p, and Yop1p in the regulation of peroxisome biogenesis. Furthermore, as an initially cortical-ER localized protein that interacts with reticulon homology proteins, Pex30p is shown in this work to establish contacts between ER tubules and peroxisomes and to specifically traffic through the ER. In summary, our data reveal a central role for Pex30p in the formation of ER-to-peroxisomes associations that appear to be involved in the coordination of peroxisome biogenesis and maintenance.  相似文献   

6.
7.
The endoplasmic reticulum (ER) glycoprotein HMG-CoA reductase (HMGR) catalyzes the rate-limiting step in sterols biosynthesis. Mammalian HMGR is ubiquitinated and degraded by the proteasome when sterols accumulate in cells, representing the best example for metabolically controlled ER-associated degradation (ERAD). This regulated degradation involves the short-lived ER protein Insig-1. Here, we investigated the dislocation of these ERAD substrates to the cytosol en route to proteasomal degradation. We show that the tagged HMGR membrane region, HMG350-HA, the endogenous HMGR, and Insig-1-Myc, all polytopic membrane proteins, dislocate to the cytosol as intact full-length polypeptides. Dislocation of HMG350-HA and Insig-1-Myc requires metabolic energy and involves the AAA-ATPase p97/VCP. Sterols stimulate HMG350-HA and HMGR release to the cytosol concurrent with removal of their N-glycan by cytosolic peptide:N-glycanase. Sterols neither accelerate dislocation nor stimulate deglycosylation of ubiquitination-defective HMG350-HA(K89 + 248R) mutant. Dislocation of HMG350-HA depends on Insig-1-Myc, whose dislocation and degradation are sterol independent. Coimmunoprecipitation experiments demonstrate sterol-stimulated association between HMG350-HA and Insig-1-Myc. Sterols do not enhance binding to Insig-1-Myc of HMG350-HA mutated in its sterol-sensing domain or of HMG350-HA(K89 + 248R). Wild-type HMG350-HA and Insig-1-Myc coimmunoprecipitate from the soluble fraction only when both proteins were coexpressed in the same cell, indicating their encounter before or during dislocation, raising the possibility that they are dislocated as a tightly bound complex.  相似文献   

8.
The activation of a high affinity Ca2+ influx system (HACS) in the plasma membrane is required for survival of yeast cells exposed to natural or synthetic inhibitors of essential processes (secretory protein folding or sterol biosynthesis) in the endoplasmic reticulum (ER). The mechanisms linking ER stress to HACS activation are not known. Here we show that Kch1, a recently identified low affinity K+ transporter in the plasma membrane of Saccharomyces cerevisiae, is up-regulated in response to several ER stressors and necessary for HACS activation. The activation of HACS required extracellular K+ and was also dependent on the high affinity K+ transporters Trk1 and Trk2. However, a paralog of Kch1 termed Kch2 was not expressed and not necessary for HACS activation in these conditions. The pathogenic yeast Candida albicans carries only one homolog of Kch1/Kch2, and homozygous knock-out mutants were similarly deficient in the activation of HACS during the responses to tunicamycin. However, the Kch1 homolog was not necessary for HACS activation or cell survival in response to several clinical antifungals (azoles, allylamines, echinocandins) that target the ER or cell wall. Thus, Kch1 family proteins represent a conserved linkage between HACS and only certain classes of ER stress in these yeasts.  相似文献   

9.
There is mounting evidence for the organization and compartmentation of signaling molecules at the plasma membrane. We find that hormone-sensitive adenylyl cyclase activity is enriched in a subset of regulatory G protein-containing fractions of the plasma membrane. These subfractions resemble, in low buoyant density, structures of the plasma membrane termed caveolae. Immunofluorescence experiments revealed a punctate pattern of G protein α and β subunits, consistent with concentration of these proteins at distinct sites on the plasma membrane. Partial coincidence of localization of G protein α subunits with caveolin (a marker for caveolae) was observed by double immunofluorescence. Results of immunogold electron microscopy suggest that some G protein is associated with invaginated caveolae, but most of the protein resides in irregular structures of the plasma membrane that could not be identified morphologically. Because regulated adenylyl cyclase activity is present in low-density subfractions of plasma membrane from a cell type (S49 lymphoma) that does not express caveolin, this protein is not required for organization of the adenylyl cyclase system. The data suggest that hormone-sensitive adenylyl cyclase systems are localized in a specialized subdomain of the plasma membrane that may optimize the efficiency and fidelity of signal transduction.  相似文献   

10.
Nascent chains are known to be targeted to the endoplasmic reticulum membrane either by a signal recognition particle (SRP)-dependent co-translational or by an SRP-independent post-translational translocation route depending on signal sequences. Using a set of model and cellular proteins carrying an N-terminal signal anchor sequence of controlled hydrophobicity and yeast mutant strains defective in SRP or Sec62 function, the hydrophobicity-dependent targeting efficiency and targeting pathway preference were systematically evaluated. Our results suggest that an SRP-dependent co-translational and an SRP-independent post-translational translocation are not mutually exclusive for signal anchor proteins and that moderately hydrophobic ones require both SRP and Sec62 for proper targeting and translocation to the endoplasmic reticulum. Further, defect in Sec62 selectively reduced signal sequences inserted in an Nin-Cout (type II) membrane topology, implying an undiscovered role of Sec62 in regulating the orientation of the signal sequence in an early stage of translocation.  相似文献   

11.
The ankyrin and SOCS (suppressor of cytokine signaling) box (ASB) family of proteins function as the substrate recognition subunit in a subset of Elongin-Cullin-SOCS (ECS) E3 ubiquitin ligases. Despite counting 18 members in humans, the identity of the physiological targets of the Asb proteins remains largely unexplored. To increase our understanding of the function of ASB proteins, we conducted a family-wide SILAC (stable isotope labeling by amino acids in cell culture)-based protein/protein interaction analysis. This investigation led to the identification of novel as well as known ASB-associated proteins like Cullin 5 and Elongins B/C. We observed that several proteins can be bound by more than one Asb protein. The additional exploration of this phenomenon demonstrated that ASB-Cullin 5 complexes can oligomerize and provides evidence that Cullin 5 forms heterodimeric complexes with the Cullin 4a-DDB1 complex. We also demonstrated that ASB11 is a novel endoplasmic reticulum-associated ubiquitin ligase with the ability to interact and promote the ubiquitination of Ribophorin 1, an integral protein of the oligosaccharyltransferase (OST) glycosylation complex. Moreover, expression of ASB11 can increase Ribophorin 1 protein turnover in vivo. In summary, we provide a comprehensive protein/protein interaction data resource that can aid the biological and functional characterization of ASB ubiquitin ligases.  相似文献   

12.
Abstract: Various glycolipid-binding toxins are internalized from the cell surface to the Golgi apparatus. Prominent among these is cholera toxin (CT), which consists of a pentameric B subunit that binds to ganglioside GM1 and an A subunit that mediates toxicity. We now demonstrate that rhodamine (Rh)-CT can be further internalized from the Golgi apparatus to the endoplasmic reticulum (ER) in cultured hippocampal neurons and in neuroblastoma N18TG-2 cells and that the A subunit is essential for retrograde transport to the ER. In addition, the rate of internalization of Rh-CT to the Golgi apparatus and ER decreases dramatically as hippocampal neurons mature. The Golgi apparatus was labeled in almost all 1-day-old neurons after <1 h of incubation with Rh-CT but was labeled in <10% of 14-day-old neurons after 1 h. During the first 14 days in culture, there was a 15-fold increase in the number of 125I-CT-binding sites per cell, indicating that the decrease in the rate of internalization of Rh-CT is not due to reduced levels of cell surface GM1 in older neurons. These results imply that the rate of retrograde transport of CT from the plasma membrane to the Golgi apparatus and ER is regulated during neuronal development and differentiation.  相似文献   

13.
Apolipoprotein B (ApoB) is the only protein component of the low density lipoproteins (LDL) in plasma. It is a glycoprotein with a molecular mass of about 550 kDa (4536 amino acids) containing 16 N-glycans. We have studied the interaction of ApoB with two lectin-like chaperones of the Endoplasmic Reticulum (ER)—Calnexin (CN) and Calreticulin (CR). Using a co-immunoprecipitation approach we observed that newly synthesized ApoB associates with CN and CR. The interaction was transient; within 30–60 min after synthesis bulk of newly formed ApoB dissociated. Using McA Rh7777 cells expressing an N-terminal fragment of ApoB we found that inhibition of glucosidases in the ER prevented the association of CN and CR to newly synthesized ApoB. The results showed that like for association with other glycoprotein substrates, trimming of glucose residues was essential for ApoB binding to CN and CR.  相似文献   

14.
Nonenveloped viruses undergo conformational changes that enable them to bind to, disrupt, and penetrate a biological membrane leading to successful infection. We assessed whether cytosolic factors play any role in the endoplasmic reticulum (ER) membrane penetration of the nonenveloped SV40. We find the cytosolic SGTA-Hsc70 complex interacts with the ER transmembrane J-proteins DnaJB14 (B14) and DnaJB12 (B12), two cellular factors previously implicated in SV40 infection. SGTA binds directly to SV40 and completes ER membrane penetration. During ER-to-cytosol transport of SV40, SGTA disengages from B14 and B12. Concomitant with this, SV40 triggers B14 and B12 to reorganize into discrete foci within the ER membrane. B14 must retain its ability to form foci and interact with SGTA-Hsc70 to promote SV40 infection. Our results identify a novel role for a cytosolic chaperone in the membrane penetration of a nonenveloped virus and raise the possibility that the SV40-induced foci represent cytosol entry sites.  相似文献   

15.
Copines are highly conserved proteins with lipid-binding activities found in animals, plants, and protists. They contain two calcium-dependent phospholipid binding C2 domains at the amino terminus and a VWA domain at the carboxyl terminus. The biological roles of most copines are not understood and the biochemical properties required for their functions are largely unknown. The Arabidopsis copine gene BON1/CPN1 is a negative regulator of cell death and defense responses. Here we probed the potential biochemical activities of BON1 through mutagenic studies. We found that mutations of aspartates in the C2 domains did not alter plasma membrane localization but compromised BON1 activity. Mutation at putative myristoylation residue glycine 2 altered plasma membrane localization of BON1 and rendered BON1 inactive. Mass spectrometry analysis of BON1 further suggests that the N-peptide of BON1 is modified. Furthermore, mutations that affect the interaction between BON1 and its functional partner BAP1 abolished BON1 function. This analysis reveals an unanticipated regulation of copine protein localization and function by calcium and lipid modification and suggests an important role in protein-protein interaction for the VWA domain of copines.  相似文献   

16.
17.
Ras GTPases signal by orchestrating a balance among several effector pathways, of which those driven by the GTPases RalA and RalB are essential to Ras oncogenic functions. RalA and RalB share the same effectors but support different aspects of oncogenesis. One example is the importance of active RalA in anchorage-independent growth and membrane raft trafficking. This study has shown a new post-translational modification of Ral GTPases: nondegradative ubiquitination. RalA (but not RalB) ubiquitination increases in anchorage-independent conditions in a caveolin-dependent manner and when lipid rafts are endocytosed. Forcing RalA mono-ubiquitination (by expressing a protein fusion consisting of ubiquitin fused N-terminally to RalA) leads to RalA enrichment at the plasma membrane and increases raft exposure. This study suggests the existence of an ubiquitination/de-ubiquitination cycle superimposed on the GDP/GTP cycle of RalA, involved in the regulation of RalA activity as well as in membrane raft trafficking.  相似文献   

18.
In addition to its well-known role in recognition by the proteasome, ubiquitin-conjugation is also involved in downregulation of membrane receptors, transporters and channels. In most cases, ubiquitination of these plasma membrane proteins leads to their internalization followed by targeting to the lysosome/vacuole for degradation. A crucial role in ubiquitination of many plasma membrane proteins appears to be played by ubiquitin-protein ligases of the Nedd4/Rsp5p family. All family members carry an N-terminal Ca2+-dependent lipid/protein binding (C2) domain, two to four WW domains and a C-terminal catalytic Hect-domain. Nedd4 is involved in downregulation of the epithelial Na+ channel, by binding of its WW domains to specific PY motifs of the channel. Rsp5p, the unique family member in S. cerevisiae, is involved in ubiquitin-dependent endocytosis of a great number of yeast plasma membrane proteins. These proteins lack apparent PY motifs, but carry acidic sequences, and/or phosphorylated-based sequences that might be important, directly or indirectly, for their recognition by Rsp5p. In contrast to polyubiquitination leading to proteasomal recognition, a number of Rsp5p targets carry few ubiquitins per protein, and moreover with a different ubiquitin linkage. Accumulating evidence suggests that, at least in yeast, ubiquitin itself may constitute an internalization signal, recognized by a hypothetical receptor. Recent data also suggest that Nedd4/Rsp5p might play a role in the endocytic process possibly involving its C2 domain, in addition to its role in ubiquitinating endocytosed proteins. Recieved: 19 January 2000/Revised: 6 April 2000  相似文献   

19.
Coatomer is the soluble precursor of the COPI coat (coat protein I) involved in traffic among membranes of the endoplasmic reticulum and the Golgi apparatus. We report herein that neomycin precipitates coatomer from cell extracts and from purified coatomer preparations. Precipitation first increased and then decreased as the neomycin concentration increased, analogous to the precipitation of a polyvalent antigen by divalent antibodies. This suggested that neomycin cross-linked coatomer into large aggregates and implies that coatomer has two or more binding sites for neomycin. A variety of other aminoglycoside antibiotics precipitated coatomer, or if they did not precipitate, they interfered with the ability of neomycin to precipitate. Coatomer is known to interact with a motif (KKXX) containing adjacent lysine residues at the carboxyl terminus of the cytoplasmic domains of some membrane proteins resident in the endoplasmic reticulum. All of the antibiotics that interacted with coatomer contain at least two close amino groups, suggesting that the antibiotics might be interacting with the di-lysine binding site of coatomer. Consistent with this idea, di-lysine itself blocked the interaction of antibiotics with coatomer. Moreover, di-lysine and antibiotics each blocked the coating of Golgi membranes by coatomer. These data suggest that certain aminoglycoside antibiotics interact with di-lysine binding sites on coatomer and that coatomer contains at least two of these di-lysine binding sites.  相似文献   

20.
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-23

京公网安备 11010802026262号