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 62 毫秒
1.
During clathrin‐mediated endocytosis (CME), actin assembly provides force to drive vesicle internalization. Members of the Wiskott–Aldrich syndrome protein (WASP) family play a fundamental role stimulating actin assembly. WASP family proteins contain a WH2 motif that binds globular actin (G‐actin) and a central‐acidic motif that binds the Arp2/3 complex, thus promoting the formation of branched actin filaments. Yeast WASP (Las17) is the strongest of five factors promoting Arp2/3‐dependent actin polymerization during CME. It was suggested that this strong activity may be caused by a putative second G‐actin‐binding motif in Las17. Here, we describe the in vitro and in vivo characterization of such Las17 G‐actin‐binding motif (LGM) and its dependence on a group of conserved arginine residues. Using the yeast two‐hybrid system, GST‐pulldown, fluorescence polarization and pyrene‐actin polymerization assays, we show that LGM binds G‐actin and is necessary for normal Arp2/3‐mediated actin polymerization in vitro. Live‐cell fluorescence microscopy experiments demonstrate that LGM is required for normal dynamics of actin polymerization during CME. Further, LGM is necessary for normal dynamics of endocytic machinery components that are recruited at early, intermediate and late stages of endocytosis, as well as for optimal endocytosis of native CME cargo. Both in vitro and in vivo experiments show that LGM has relatively lower potency compared to the previously known Las17 G‐actin‐binding motif, WH2. These results establish a second G‐actin‐binding motif in Las17 and advance our knowledge on the mechanism of actin assembly during CME.   相似文献   

2.
Shigella flexneri is an intracellular pathogen that disseminates in colonic epithelial cells through actin‐based motility and formation of membrane protrusions at cell–cell contacts, that project into adjacent cells and resolve into vacuoles, from which the pathogen escapes, thereby achieving cell‐to‐cell spread. Actin nucleation at the bacterial pole relies on the recruitment of the nucleation‐promoting factor N‐WASP, which activates the actin nucleator ARP2/3. In cells, the vast majority of N‐WASP exists as a complex with WIP. The involvement of WIP in N‐WASP‐dependent actin‐based motility of various pathogens, including vaccinia virus and S. flexneri, has been highly controversial. Here, we show that WIPF2 was the only WIP family member expressed in the human colonic epithelial cell line HT‐29, and its depletion impaired S. flexneri dissemination. WIPF2 depletion increased the number of cytosolic bacteria lacking actin tails (non‐motile) and decreased the velocity of motile bacteria. This correlated with a decrease in the recruitment of N‐WASP to the bacterial pole, and among N‐WASP‐positive bacteria, a decrease in actin tail‐positive bacteria, suggesting that WIPF2 is required for N‐WASP recruitment and activation at the bacterial pole. In addition, when motile bacteria formed protrusions, WIPF2 depletion decreased the number of membrane protrusions that successfully resolved into vacuoles.  相似文献   

3.
Cargo sorting and membrane carrier initiation in recycling endosomes require appropriately coordinated actin dynamics. However, the mechanism underlying the regulation of actin organization during recycling transport remains elusive. Here we report that the loss of PTRN‐1/CAMSAP stalled actin exchange and diminished the cytosolic actin structures. Furthermore, we found that PTRN‐1 is required for the recycling of clathrin‐independent cargo hTAC‐GFP. The N‐terminal calponin homology (CH) domain and central coiled‐coils (CC) region of PTRN‐1 can synergistically sustain the flow of hTAC‐GFP. We identified CYK‐1/formin as a binding partner of PTRN‐1. The N‐terminal GTPase‐binding domain (GBD) of CYK‐1 serves as the binding interface for the PTRN‐1 CH domain. The presence of the PTRN‐1 CH domain promoted CYK‐1‐mediated actin polymerization, which suggests that the PTRN‐1‐CH:CYK‐1‐GBD interaction efficiently relieves autoinhibitory interactions within CYK‐1. As expected, the overexpression of the CYK‐1 formin homology domain 2 (FH2) substantially restored actin structures and partially suppressed the hTAC‐GFP overaccumulation phenotype in ptrn‐1 mutants. We conclude that the PTRN‐1 CH domain is required to stimulate CYK‐1 to facilitate actin dynamics during endocytic recycling.  相似文献   

4.
Clathrin‐mediated endocytosis is a fundamental transport pathway that depends on numerous protein‐protein interactions. Testing the importance of the adaptor protein‐clathrin interaction for coat formation and progression of endocytosis in vivo has been difficult due to experimental constrains. Here, we addressed this question using the yeast clathrin adaptor Sla1, which is unique in showing a cargo endocytosis defect upon substitution of 3 amino acids in its clathrin‐binding motif (sla1AAA) that disrupt clathrin binding. Live‐cell imaging showed an impaired Sla1‐clathrin interaction causes reduced clathrin levels but increased Sla1 levels at endocytic sites. Moreover, the rate of Sla1 recruitment was reduced indicating proper dynamics of both clathrin and Sla1 depend on their interaction. sla1AAA cells showed a delay in progression through the various stages of endocytosis. The Arp2/3‐dependent actin polymerization machinery was present for significantly longer time before actin polymerization ensued, revealing a link between coat formation and activation of actin polymerization. Ultimately, in sla1AAA cells a larger than normal actin network was formed, dramatically higher levels of various machinery proteins other than clathrin were recruited, and the membrane profile of endocytic invaginations was longer. Thus, the Sla1‐clathrin interaction is important for coat formation, regulation of endocytic progression and membrane bending.   相似文献   

5.
Background: The actin-related proteins Arp2 and Arp3 are part of a seven-protein complex which is localized in the lamellipodia of a variety of cell types, and in actin-rich spots of unknown function. The Arp2/3 complex enhances actin nucleation and causes branching and crosslinking of actin filaments in vitro; in vivo it is thought to drive the formation of lamellipodia and to be a control center for actin-based motility. The Wiskott–Aldrich syndrome protein, WASP, is an adaptor protein implicated in the transmission of signals from tyrosine kinase receptors and small GTPases to the actin cytoskeleton. Scar1 is a member of a new family of proteins related to WASP, and it may also have a role in regulating the actin cytoskeleton. Scar1 is the human homologue of Dictyostelium Scar1, which is thought to connect G-protein-coupled receptors to the actin cytoskeleton. The mammalian Scar family contains at least four members. We have examined the relationships between WASP, Scar1, and the Arp2/3 complex.Results: We have identified WASP and its relative Scar1 as proteins that interact with the Arp2/3 complex. We have used deletion analysis to show that both WASP and Scar1 interact with the p21 subunit of the Arp2/3 complex through their carboxyl termini. Overexpression of carboxy-terminal fragments of Scar1 or WASP in cells caused a disruption in the localization of the Arp2/3 complex and, concomitantly, induced a complete loss of lamellipodia and actin spots. The induction of lamellipodia by platelet-derived growth factor was also suppressed by overexpression of the fragment of Scar1 that binds to the Arp2/3 complex.Conclusions: We have identified a conserved sequence domain in proteins of the WASP family that binds to the Arp2/3 complex. Overexpression of this domain in cells disrupts the localization of the Arp2/3 complex and inhibits lamellipodia formation. Our data suggest that WASP-related proteins may regulate the actin cytoskeleton through the Arp2/3 complex.  相似文献   

6.
Actin polymerization essential for endocytic internalization in budding yeast is controlled by four nucleation promoting factors (NPFs) that each exhibits a unique dynamic behavior at endocytic sites. How each NPF functions and is regulated to restrict actin assembly to late stages of endocytic internalization is not known. Quantitative analysis of NPF biochemical activities, and genetic analysis of recruitment and regulatory mechanisms, defined a linear pathway in which protein composition changes at endocytic sites control actin assembly and function. We show that yeast WASP initiates actin assembly at endocytic sites and that this assembly and the recruitment of a yeast WIP-like protein by WASP recruit a type I myosin with both NPF and motor activities. Importantly, type I myosin motor and NPF activities are separable, and both contribute to endocytic coat inward movement, which likely represents membrane invagination. These results reveal a mechanism in which actin nucleation and myosin motor activity cooperate to promote endocytic internalization.  相似文献   

7.
The major virulence determinant of Legionella pneumophila is the type IVB secretion system (T4BSS), which delivers approximately 330 effector proteins into the host cell to modulate various cellular processes. However, the functions of most effector proteins remain unclear. WipA, an effector, was the first phosphotyrosine phosphatase of Legionella with unknown function. In this study, we found that WipA induced relatively strong growth defects in yeast in a phosphatase activity‐dependent manner. Phosphoproteomics data showed that WipA was likely involved into endocytosis, FcγR‐mediated phagocytosis, tight junction, and regulation of actin cytoskeleton pathways. Western blotting further confirmed WipA dephosphorylates several proteins associated with actin polymerisation, such as p‐N‐WASP, p‐ARP3, p‐ACK1, and p‐NCK1. Thus, we hypothesised that WipA targets N‐WASP/ARP2/3 complex signalling pathway, leading to disturbance of actin polymerisation. Indeed, we demonstrated that WipA inhibits host F‐actin polymerisation by reducing the G‐actin to F‐actin transition during L. penumophila infection. Furthermore, the intracellular proliferation of wipA/legK2 double mutant was significantly impaired at the late stage of infection, although the absence of WipA does not confer any further effect on actin polymerisation to the legK2 mutant. Collectively, this study provides unique insights into the WipA‐mediated regulation of host actin polymerisation and assists us to elucidate the pathogenic mechanisms of L. pnuemophila infection.  相似文献   

8.
Bartonella henselae enters human endothelial cells (ECs) by two alternative routes: either by endocytosis, giving rise to Bartonella‐containing vacuoles or by invasome‐mediated internalization. Only the latter process depends on the type IV secretion system VirB/VirD4 and involves the formation of cell surface‐associated bacterial aggregates, which get engulfed by EC membranes in an F‐actin‐dependent manner, eventually resulting in their complete internalization. Here, we report that among the VirB/VirD4‐translocated effector proteins BepA‐BepG only BepG is required for triggering invasome‐mediated internalization. Expression of BepG in the Bep‐deficient ΔbepA–G mutant restored invasome‐mediated internalization. Likewise, ectopic expression of BepG in ECs also restored invasome‐mediated internalization of the ΔbepA–G mutant, while no discernable cytoskeletal rearrangements were triggered in uninfected cells. Rather, BepG inhibited endocytic uptake of B. henselae into Bartonella‐containing vacuoles and other endocytic processes, that is, invasin‐mediated uptake of Yersinia enterocolitica and uptake of inert microspheres. BepG thus triggers invasome‐mediated internalization primarily by inhibiting bacterial endocytosis. Bacteria accumulating on the cell surface then induce locally the F‐actin rearrangements characteristic for the invasome. These cytoskeletal changes encompass both the rearrangement of pre‐existing F‐actin fibres and the de novo polymerization of cortical F‐actin in the periphery of the invasome by Rac1/Scar1/WAVE‐ and Cdc42/WASP‐dependent pathways that involve the recruitment of the Arp2/3 complex.  相似文献   

9.
The bacterial pathogen Listeria monocytogenes induces internalization into mammalian cells and uses actin‐based motility to spread within tissues. Listeria accomplishes this intracellular life cycle by exploiting or antagonizing several host GTPases. Internalization into human cells is mediated by the bacterial surface proteins InlA or InlB. These two modes of uptake each require a host actin polymerization pathway comprised of the GTPase Rac1, nucleation promotion factors, and the Arp2/3 complex. In addition to Rac1, InlB‐mediated internalization involves inhibition of the GTPase Arf6 and participation of Dynamin and septin family GTPases. After uptake, Listeria is encased in host phagosomes. The bacterial protein GAPDH inactivates the human GTPase Rab5, thereby delaying phagosomal acquisition of antimicrobial properties. After bacterial‐induced destruction of the phagosome, cytosolic Listeria uses the surface protein ActA to stimulate actin‐based motility. The GTPase Dynamin 2 reduces the density of microtubules that would otherwise limit bacterial movement. Cell‐to‐cell spread results when motile Listeria remodel the host plasma membrane into protrusions that are engulfed by neighbouring cells. The human GTPase Cdc42, its activator Tuba, and its effector N‐WASP form a complex with the potential to restrict Listeria protrusions. Bacteria overcome this restriction through two microbial factors that inhibit Cdc42‐GTP or Tuba/N‐WASP interaction.  相似文献   

10.
Arp2/3 complex is an important actin filament nucleator that creates branched actin filament networks required for formation of lamellipodia and endocytic actin structures. Cellular assembly of branched actin networks frequently requires multiple Arp2/3 complex activators, called nucleation promoting factors (NPFs). We recently presented a mechanism by which cortactin, a weak NPF, can displace a more potent NPF, N-WASP, from nascent branch junctions to synergistically accelerate nucleation. The distinct roles of these NPFs in branching nucleation are surprising given their similarities. We biochemically dissected these two classes of NPFs to determine how their Arp2/3 complex and actin interacting segments modulate their influences on branched actin networks. We find that the Arp2/3 complex-interacting N-terminal acidic sequence (NtA) of cortactin has structural features distinct from WASP acidic regions (A) that are required for synergy between the two NPFs. Our mutational analysis shows that differences between NtA and A do not explain the weak intrinsic NPF activity of cortactin, but instead that cortactin is a weak NPF because it cannot recruit actin monomers to Arp2/3 complex. We use TIRF microscopy to show that cortactin bundles branched actin filaments using actin filament binding repeats within a single cortactin molecule, but that N-WASP antagonizes cortactin-mediated bundling. Finally, we demonstrate that multiple WASP family proteins synergistically activate Arp2/3 complex and determine the biochemical requirements in WASP proteins for synergy. Our data indicate that synergy between WASP proteins and cortactin may play a general role in assembling diverse actin-based structures, including lamellipodia, podosomes, and endocytic actin networks.  相似文献   

11.
WASP‐family proteins are known to promote assembly of branched actin networks by stimulating the filament‐nucleating activity of the Arp2/3 complex. Here, we show that WASP‐family proteins also function as polymerases that accelerate elongation of uncapped actin filaments. When clustered on a surface, WASP‐family proteins can drive branched actin networks to grow much faster than they could by direct incorporation of soluble monomers. This polymerase activity arises from the coordinated action of two regulatory sequences: (i) a WASP homology 2 (WH2) domain that binds actin, and (ii) a proline‐rich sequence that binds profilin–actin complexes. In the absence of profilin, WH2 domains are sufficient to accelerate filament elongation, but in the presence of profilin, proline‐rich sequences are required to support polymerase activity by (i) bringing polymerization‐competent actin monomers in proximity to growing filament ends, and (ii) promoting shuttling of actin monomers from profilin–actin complexes onto nearby WH2 domains. Unoccupied WH2 domains transiently associate with free filament ends, preventing their growth and dynamically tethering the branched actin network to the WASP‐family proteins that create it. Collaboration between WH2 and proline‐rich sequences thus strikes a balance between filament growth and tethering. Our work expands the number of critical roles that WASP‐family proteins play in the assembly of branched actin networks to at least three: (i) promoting dendritic nucleation; (ii) linking actin networks to membranes; and (iii) accelerating filament elongation.  相似文献   

12.
Cryptococcus neoformans is rich in polysaccharides of the cell wall and capsule. Dectin‐2 recognizes high‐mannose polysaccharides and plays a central role in the immune response to fungal pathogens. Previously, we demonstrated Dectin‐2 was involved in the activation of dendritic cells upon stimulation with C. neoformans, suggesting the existence of a ligand recognized by Dectin‐2. In the present study, we examined the cell wall structures of C. neoformans contributing to the Dectin‐2‐mediated activation of immune cells. In a NFAT‐GFP reporter assay of the reported cells expressing Dectin‐2, the lysates, but not the whole yeast cells, of an acapsular strain of C. neoformans (Cap67) delivered Dectin‐2‐mediated signaling. This activity was detected in the supernatant of β‐glucanase‐treated Cap67 and more strongly in the semi‐purified polysaccharides of this supernatant using ConA‐affinity chromatography (ConA‐bound fraction), in which a large amount of saccharides, but not protein, were detected. Treatment of this supernatant with periodic acid and the addition of excessive mannose, but not glucose or galactose, strongly inhibited this activity. The ConA‐bound fraction of the β‐glucanase‐treated Cap67 supernatant was bound to Dectin‐2‐Fc fusion protein in a dose‐dependent manner and strongly induced the production of interleukin‐12p40 and tumour necrosis factor‐α by dendritic cells; this was abrogated under the Dectin‐2‐deficient condition. Finally, 98 kDa mannoprotein (MP98) derived from C. neoformans showed activation of the reporter cells expressing Dectin‐2. These results suggested that a ligand with mannose moieties may exist in the cell walls and play a critical role in the activation of dendritic cells during infection with C. neoformans.  相似文献   

13.
We analyzed plant‐derived α1,4‐fucosyltransferase (FucTc) homologs by reporter fusions and focused on representatives of the Brassicaceae and Solanaceae. Arabidopsis thaliana AtFucTc‐green fluorescent protein (GFP) or tomato LeFucTc‐GFP restored Lewis‐a formation in a fuctc mutant, confirming functionality in the trans‐Golgi. AtFucTc‐GFP partly accumulated at the nuclear envelope (NE) not observed for other homologs or truncated AtFucTc lacking the N‐terminus or catalytic domain. Analysis of At/LeFucTc‐GFP swap constructs with exchanged cytosolic, transmembrane and stalk (CTS), or only the CT regions, revealed that sorting information resides in the membrane anchor. Other domains of AtFuctc also contribute, since amino‐acid changes in the CT region strongly reduced but did not abolish NE localization. By contrast, two N‐terminal GFP copies did, indicating localization at the inner nuclear membrane (INM). Tunicamycin treatment of AtFucTc‐GFP abolished NE localization and enhanced overlap with an endosomal marker, suggesting involvement of N‐glycosylation. Yet neither expression in protoplasts of Arabidopsis N‐glycosylation mutants nor elimination of the N‐glycosylation site in AtFucTc prevented perinuclear accumulation. Disruption of endoplasmic reticulum (ER)‐to‐Golgi transport by co‐expression of Sar1(H74L) trapped tunicamycin‐released AtFucTc‐GFP in the ER, however, without NE localization. Since recovery after tunicamycin‐washout required de novo‐protein synthesis, our analyses suggest that AtFucTc localizes to the NE/INM due to interaction with an unknown (glyco)protein.   相似文献   

14.
WASP is a remodeler of the actin cytoskeleton, but its mechanistic contribution to neutrophil migration is unclear. In this issue, Brunetti et al. (2021. J. Cell Biol. https://doi.org/10.1083/jcb.202104046) show that WASP is recruited to substrate-induced membrane deformations near the cell front, where it induces Arp2/3 complex–mediated local actin assembly to direct migration.

Neutrophils are the most abundant type of white blood cells in humans and constitute an important first line of defense of the innate immune system. Neutrophils exit the blood stream in response to chemoattractants that signal danger in the form of damage, infection, or inflammation, ultimately removing dangerous particles by phagocytosis. Efficient migration is crucial to properly execute such functions, but how precisely the coordination of polarity establishment and complex shape changes needed for migratory processes occurs in these cells is just beginning to be elucidated. The dynamic remodeling of actin filaments, which can differentially assemble and disassemble at both ends, is key to the development of pulling and pushing forces below the plasma membrane of most eukaryotic cells or on the surfaces of their intracellular organelles (1).Actin filaments are organized into either bundles or networks, with networks most commonly generated and maintained by the continuous filament branching activity of the actin-related protein 2/3 (Arp2/3) complex (2). Arp2/3 complex–driven branching in actin networks has often been associated with the development of pushing forces, even in a mechano-responsive fashion (3, 4), and less so linked with pulling or traction forces as developed, for instance, by adhesions. The heteropentameric Arp2/3 complex, which catalyzes the formation of daughter filaments off the sides of mother filaments, is intrinsically inactive, but can be activated by nucleation promoting factors (NPFs). The canonical NPFs include the Wiskott-Aldrich syndrome protein (WASP), its ubiquitous orthologue neural WASP (N-WASP), three WASP family verprolin-homologous protein (WAVE) isoforms as well as WASH, WHAMM, and JMY (5). The C-termini of all these factors physically interact with the Arp2/3 complex, driving it into an active conformation, and aid daughter filament polymerization by adding actin monomers onto Arp-2 and -3, forming the base of the branch. Coordinating the branching of daughter filaments by Arp2/3 complex and their elongation appears to be a major function of NPFs such as WASP. The role of WASP in stimulating actin assembly and the observation that patients with Wiskott-Aldrich syndrome harboring mutations in this gene have white blood cells that are unable to reach the sites of infections (6) points to WASP as a regulator of guided migration of immune cells. In this issue, Brunetti et al. establish the precise cellular function by which WASP controls neutrophil migration: WASP is recruited to sites of inward membrane deformations to stimulate the formation of spot-like actin structures that aid in the adhesion to or in the grabbing onto extracellular structures of high diversity and flexibility such as extracellular matrix fibrils, thus linking substrate topology, cell polarity, and migration (7).The authors first determined the spatiotemporal dynamics of EGFP-tagged, endogenous WASP in the human neutrophil–like cell line HL-60 using total internal reflection fluorescence microscopy. WASP formed puncta that were largely devoid of clathrin accumulation, thus likely not embodying endocytic pits, and exogenously triggered by submicron-sized beads, microfabricated ridges, or collagen fibers. Substratum-associated WASP puncta exhibited adhesive functions, as integrin inactivation by ion chelation largely dissociated them, stimulating their sliding and centripetal displacement. Surprisingly, the prominent WASP accumulation to beads below the plasma membrane was biased to the front half of the cell, and coincided with a gradient of activity of its major activator, the Rho GTPase Cdc42 from front to rear. This observation led Brunetti et al. to postulate WASP as a factor linking substrate topology to cell polarization and effective migration.A closer look at the recruitment of WASP to inward plasma membrane invaginations (positive membrane curvature) stimulated by submicron-sized beads revealed two patterns of association: either following the bead bodies as U-shaped accumulations or to the necks of invaginations reminiscent of endocytic pits, which could reflect later stages of bead–plasma membrane interactions. Although the mechanistic significance of these distinct association patterns remains elusive, it was clear that WASP favored associations at the plasma membrane with sites of comparably high, positive curvature (small, 100-nm-diameter beads more attractive than larger beads, for instance) and in a punctate fashion (even along ridges). In our view, this much diverges from WAVE isoforms exhibiting a prominent, more linear association with the rapidly protruding membranes of negative curvature in lamellipodia (8). Finally, the authors showed that Cdc42 loss-of-function diminished WASP puncta at the cell front, and that lack of WASP reduced actin puncta formation and the frequency of Arp2/3 complex accumulation at beads. WASP-null cells migrated less in the direction of ridges, although their perpendicular movement across the nanopatterns was not affected—indeed, it was increased. These observations connect the role of WASP in reading substrate topology to Arp2/3-dependent actin filament branching and network formation. The findings of Brunetti et al. appear to be of broader impact and relevance than just being a human neutrophil-specific phenomenon—a separate study has recently shown that sliding, WASP-dependent, punctate adhesion sites in murine dendritic cells and T cells, which are triggered in response to mechanical load, polymerize actin networks orthogonally to the plasma membrane, and aid in squeezing and dragging these cells through dense tissues (9).Taking all this together, the study by Brunetti et al. highlights that at least immune cells primarily respond to mechanical impact and indentations of their plasma membranes with rapid accumulation of WASP and Arp2/3 complex–dependent actin assembly, thereby pushing back or grabbing extracellular material to push and pull themselves forward during migration through the complex, dense, and three-dimensional environments on their way through tissues (Fig. 1).Open in a separate windowFigure 1.Neutrophils use WASP puncta to crawl like mountain climbers. Punctate accumulation of WASP and consequently F-actin brings about friction points that support pushing and pulling during cell translocation. In the front half of the cell, formation of these points strictly depends on Cdc42 mediating WASP activation and focal actin assembly. Even on substrates that would allow continuous adhesion (ridges or collagen fibers), WASP is recruited in a punctate fashion and preferentially to the front half of the cell that is dominated by Cdc42 activity signaling to cell polarity. Upon contact, movable objects like beads lead to a strong WASP/actin response potentially culminating in object envelopment.Interestingly, according to Brunetti et al., mechanisms of WASP/N-WASP recruitment to inward membrane deformations might even be conserved during early stages of formation of podosomes and invadopodia, which are WASP- and N-WASP–dependent, both protrusive and adhesive structures operating in matrix degradation of hematopoietic and cancer cells, respectively (10). This hypothesis, however, remains to be experimentally validated. Furthermore, additional questions remain unsolved, including the precise signals, aside from Cdc42, contributing to WASP accumulation and turnover at the plasma membrane. Where does the bias for focal, punctate WASP accumulation come from? WASP family NPFs are unlikely to themselves harbor curvature-sensing activities, so which are the decisive factors for interaction with positively curved membranes? How is specificity brought about? The F-BAR domain-containing TOCA family proteins would have been potential candidates, but surprisingly, disruption of two of their most prominent members, FBP17 and CIP4, did not cause severe defects, as Brunetti et al. showed in this study. Future work will surely improve our insights into the differential sorting and subcellular functions of distinct NPFs in both immune and other cells.  相似文献   

15.
Arp2/3 complex nucleates branched actin filaments that drive processes like endocytosis and lamellipodial protrusion. WISH/DIP/SPIN90 (WDS) proteins form a class of Arp2/3 complex activators or nucleation promoting factors (NPFs) that, unlike WASP family NPFs, activate Arp2/3 complex without requiring preformed actin filaments. Therefore, activation of Arp2/3 complex by WDS proteins is thought to produce the initial actin filaments that seed branching nucleation by WASP-bound Arp2/3 complexes. However, whether activation of Arp2/3 complex by WDS proteins is important for the initiation of branched actin assembly in cells has not been directly tested. Here, we used structure-based point mutations of the Schizosaccharomyces pombe WDS protein Dip1 to test the importance of its Arp2/3-activating activity in cells. Six of thirteen Dip1 mutants caused severe defects in Arp2/3 complex activation in vitro, and we found a strong correlation between the ability of mutants to activate Arp2/3 complex and to rescue endocytic actin assembly defects caused by deleting Dip1. These data support a model in which Dip1 activates Arp2/3 complex to produce actin filaments that initiate branched actin assembly at endocytic sites. Dip1 mutants that synergized with WASP in activating Arp2/3 complex in vitro showed milder defects in cells compared to those that did not, suggesting that in cells the two NPFs may coactivate Arp2/3 complex to initiate actin assembly. Finally, the mutational data reveal important complementary electrostatic contacts at the Dip1–Arp2/3 complex interface and corroborate the previously proposed wedge model, which describes how Dip1 binding triggers structural changes that activate Arp2/3 complex.  相似文献   

16.
17.
At the leading edge of migrating cells, protrusive forces are developed by the assembly of actin filaments organised in a lamellipodial dendritic array at the front and a more distal lamellar linear array. Whether these two arrays are distinct or functionally linked and how they contribute to cell migration is an open issue. Tropomyosin severely inhibits lamellipodium formation and facilitates the lamellar array while enhancing migration, by a mechanism that is not understood. Here we show that the complex in vivo effects of tropomyosin are recapitulated in the reconstituted propulsion of neural Wiskott–Aldrich syndrome protein (N‐WASP)‐functionalised beads, which is based on the sole formation of a dendritic array of actin‐related protein (Arp)2/3‐branched filaments. Actin‐depolymerising factor (ADF) and tropomyosin control the length of the actin tail. By competing with Arp2/3 during filament branching, tropomyosin displays opposite effects on propulsion depending on the surface density of N‐WASP. Tropomyosin binding to the dendritic array is facilitated following filament debranching, causing its enrichment at the rear of the actin tail, like in vivo. These results unveil the mechanism by which tropomyosin generates two morphologically and dynamically segregated actin networks from a single one.  相似文献   

18.
Cell death induced by over‐activation of glutamate receptors occurs in different neuropathologies. Cholesterol depletors protect from neurotoxic over‐activation of glutamate receptors, and we have recently reported that this neuroprotection is associated with a reduction of the N‐methyl‐D ‐aspartate subtype of glutamate receptors in detergent‐resistant membrane domains (DRM). In the present study we used comparative proteomics to further identify which proteins, besides the N‐methyl‐D ‐aspartate receptor, change its percentage of association to DRM after treatment of neurons with simvastatin. We detected 338 spots in neuronal DRM subjected to 2‐DE; eleven of these spots changed its intensity after treatment with simvastatin. All 11 differential spots showed reduced intensity in simvastatin‐treated samples and were identified as adipocyte plasma membrane associated protein, enolase, calretinin, coronin 1a, f‐actin capping protein α1, f‐actin capping protein α2, heat shock cognate protein 71, malate dehydrogenase, n‐myc downregulated gene 1, prohibitin 2, Rab GDP dissociation inhibitor, translationally controlled tumor protein and voltage dependent anion selective channel protein 1. The proteins tested colocalized with the lipid raft marker caveolin‐1. Interestingly, the proteins we have identified in the present study had been previously reported to play a role in cell fate and, thus, they might represent novel targets for neuroprotection.  相似文献   

19.
Myosins‐I are conserved proteins that bear an N‐terminal motor head followed by a Tail Homology 1 (TH1) lipid‐binding domain. Some myosins‐I have an additional C‐terminal extension (Cext) that promotes Arp2/3 complex‐dependent actin polymerization. The head and the tail are separated by a neck that binds calmodulin or calmodulin‐related light chains. Myosins‐I are known to participate in actin‐dependent membrane remodelling. However, the molecular mechanisms controlling their recruitment and their biochemical activities in vivo are far from being understood. In this study, we provided evidence suggesting the existence of an inhibitory interaction between the TH1 domain of the yeast myosin‐I Myo5 and its Cext. The TH1 domain prevented binding of the Myo5 Cext to the yeast WIP homologue Vrp1, Myo5 Cext‐induced actin polymerization and recruitment of the Myo5 Cext to endocytic sites. Our data also indicated that calmodulin dissociation from Myo5 weakened the interaction between the neck and TH1 domains and the Cext. Concomitantly, calmodulin dissociation triggered Myo5 binding to Vrp1, extended the myosin‐I lifespan at endocytic sites and activated Myo5‐induced actin polymerization.  相似文献   

20.
Phytosterols are classified into C24‐ethylsterols and C24‐methylsterols according to the different C24‐alkylation levels conferred by two types of sterol methyltransferases (SMTs). The first type of SMT (SMT1) is widely conserved, whereas the second type (SMT2) has diverged in charophytes and land plants. The Arabidopsis smt2 smt3 mutant is defective in the SMT2 step, leading to deficiency in C24‐ethylsterols while the C24‐methylsterol pathway is unchanged. smt2 smt3 plants exhibit severe dwarfism and abnormal development throughout their life cycle, with irregular cell division followed by collapsed cell files. Preprophase bands are occasionally formed in perpendicular directions in adjacent cells, and abnormal phragmoplasts with mislocalized KNOLLE syntaxin and tubulin are observed. Defects in auxin‐dependent processes are exemplified by mislocalizations of the PIN2 auxin efflux carrier due to disrupted cell division and failure to distribute PIN2 asymmetrically after cytokinesis. Although endocytosis of PIN2–GFP from the plasma membrane (PM) is apparently unaffected in smt2 smt3, strong inhibition of the endocytic recycling is associated with a remarkable reduction in the level of PIN2–GFP on the PM. Aberrant localization of the cytoplasmic linker associated protein (CLASP) and microtubules is implicated in the disrupted endocytic recycling in smt2 smt3. Exogenous C24‐ethylsterols partially recover lateral root development and auxin distribution in smt2 smt3 roots. These results indicate that C24‐ethylsterols play a crucial role in division plane determination, directional auxin transport, and polar growth. It is proposed that the divergence of SMT2 genes together with the ability to produce C24‐ethylsterols were critical events to achieve polarized growth in the plant lineage.  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