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.

Background

Bacterial cell division is an essential process driven by the formation of a Z-ring structure, as a cytoskeletal scaffold at the mid-cell, followed by the recruitment of various proteins which form the divisome. The cell division interactome reflects the complement of different interactions between all divisome proteins. To date, only two cell division interactomes have been characterized, in Escherichia coli and in Streptococcus pneumoniae. The cell divison proteins encoded by Neisseria gonorrhoeae include FtsZ, FtsA, ZipA, FtsK, FtsQ, FtsI, FtsW, and FtsN. The purpose of the present study was to characterize the cell division interactome of N. gonorrhoeae using several different methods to identify protein-protein interactions. We also characterized the specific subdomains of FtsA implicated in interactions with FtsZ, FtsQ, FtsN and FtsW.

Results

Using a combination of bacterial two-hybrid (B2H), glutathione S-transferase (GST) pull-down assays, and surface plasmon resonance (SPR), nine interactions were observed among the eight gonococcal cell division proteins tested. ZipA did not interact with any other cell division proteins. Comparisons of the N. gonorrhoeae cell division interactome with the published interactomes from E. coli and S. pneumoniae indicated that FtsA-FtsZ and FtsZ-FtsK interactions were common to all three species. FtsA-FtsW and FtsK-FtsN interactions were only present in N. gonorrhoeae. The 2A and 2B subdomains of FtsANg were involved in interactions with FtsQ, FtsZ, and FtsN, and the 2A subdomain was involved in interaction with FtsW.

Conclusions

Results from this research indicate that N. gonorrhoeae has a distinctive cell division interactome as compared with other microorganisms.
  相似文献   

2.
FtsZ is a highly conserved protein that performs one of the key roles in cell division of most prokaryotes. At the present time it is believed that the main role of FtsZ in the cells of the microorganisms studied (Escherichia coli, Bacillus subtilis, etc.) is to control the process of cell wall synthesis at the site of future septum. In this regard, the presence of FtsZ in the cells of mycoplasmas – bacteria that lack the cell wall, looks intriguing. In the present work we investigated the influence of FtsZ proteins of three mycoplasmas – Mycoplasma hominis, Mycoplasma gallisepticum and Acholeplasma laidlawii – on the cytokinesis of E. coli. FtsZ proteins were able to interact with the E. coli division machinery, including inhibiting the cytokinesis process, while demonstrating various patterns of distribution in the cell. The data obtained support the hypothesis that FtsZ plays a significant role in the division of mycoplasma cells.  相似文献   

3.
The FtsZ protein, a bacterial tubulin homolog, is one of the key proteins in bacterial cell division, forming a contractile Z-ring in the middle of the dividing cell. In the present study, immunofluorescence staining, in combination with the localization microscopy method, was used for visualization of the structures formed by unlabelled FtsZ in Escherrichia coli cells. The techniques employed allowed reconstruction of the multistep mechanism of formation of FtsZ structures during the cytokinesis process. New data were obtained confirming the hypothesis that FtsZ is a helixlike structure that constricts during the division, producing constriction between the daughter cells.  相似文献   

4.
In this review we have tried to describe proteins and supermolecular structures which take part in the division of bacterial cell. The principal cell division protein of the most of prokaryotes is FtsZ, a homologue of eukaryotic tubulin. FtsZ just as tubulin is capable to bind and hydrolyze GTP. The division of bacterial cell begins with forming of so called divisome. The basis of such divisome is a contractile ring (Z ring); the ring encircles the cell about midcell. Z ring consists of a bundle of laterally bound protofilaments, which have been formed as a result of FtsZ polymerization. Z ring is rigidly bounded to cytozolic side of inner membrane with participation of FtsA, ZipA, FtsW and many other cell division proteins of divisome. The ring directs the process of cytokinesis transmitting power of constriction to membrane. Primary structures of members of the family of prokaryotic FtsZs differ from eukaryotic tubulines significantly except the region, where the site of GTP binding is placed. There is high degree of homology between structures of these proteins in the region. FtsZ is one of the most conservative proteins in prokaryotes, but ftsZ genes have not been found in completely sequenced genomes of several species of microorganisms. There are 2 species of mycoplasmas (Ureaplasma parvum and Mycoplasma mobile), Prostecobacter dejongeii, 10 species of chlamydia and 5 species of archaea among them. So these organisms divide without FtsZ. There are many open reading frames which encode proteins with unknown functions in genomes of U. parvum and M. mobile. The comparison of primary structures of these hypothetical proteins with structures of cell division proteins did not allow researchers to find similar proteins among them. We suppose that the process of cell division of these organisms should recruit proteins with function similar to FtsZ and having homologous with FtsZ or other cell division proteins spatial structures.  相似文献   

5.
Bacterial cell division is orchestrated by the divisome, a protein complex centered on the tubulin homolog FtsZ. FtsZ polymerizes into a dynamic ring that defines the division site, recruits downstream proteins, and directs peptidoglycan synthesis to drive constriction. Recent studies have documented treadmilling of FtsZ polymer clusters both in cells and in vitro. Emerging evidence suggests that FtsZ dynamics are regulated largely by intrinsic properties of FtsZ itself and by the membrane anchoring protein FtsA. Although FtsZ dynamics are broadly required for Z-ring assembly, their role(s) during constriction may vary among bacterial species. These recent advances set the stage for future studies to investigate how FtsZ dynamics are physically and/or functionally coupled to peptidoglycan metabolic enzymes to direct efficient division.  相似文献   

6.
FtsZ is an essential cell division protein in Escherichia coli, and its localization, filamentation, and bundling at the mid-cell are required for Z-ring stability. Once assembled, the Z-ring recruits a series of proteins that comprise the bacterial divisome. Zaps (FtsZ-associated proteins) stabilize the Z-ring by increasing lateral interactions between individual filaments, bundling FtsZ to provide a scaffold for divisome assembly. The x-ray crystallographic structure of E. coli ZapA was determined, identifying key structural differences from the existing ZapA structure from Pseudomonas aeruginosa, including a charged α-helix on the globular domains of the ZapA tetramer. Key helix residues in E. coli ZapA were modified using site-directed mutagenesis. These ZapA variants significantly decreased FtsZ bundling in protein sedimentation assays when compared with WT ZapA proteins. Electron micrographs of ZapA-bundled FtsZ filaments showed the modified ZapA variants altered the number of FtsZ filaments per bundle. These in vitro results were corroborated in vivo by expressing the ZapA variants in an E. coli ΔzapA strain. In vivo, ZapA variants that altered FtsZ bundling showed an elongated phenotype, indicating improper cell division. Our findings highlight the importance of key ZapA residues that influence the extent of FtsZ bundling and that ultimately affect Z-ring formation in dividing cells.  相似文献   

7.
8.
Cytokinesis in bacteria is accomplished by a ring-shaped cell-division complex (the Z-ring). The primary component of the Z-ring is FtsZ, a filamentous tubulin homolog that serves as a scaffold for the recruitment of other cell-division-related proteins. FtsZ forms filaments and bundles. In the cell, it has been suggested that FtsZ filaments form the arcs of the ring and are aligned in the cell-circumferential direction. Using polarized fluorescence microscopy in live Escherichia coli cells, we measure the structural organization of FtsZ filaments in the Z-ring. The data suggest a disordered organization: a substantial portion of FtsZ filaments are aligned in the cell-axis direction. FtsZ organization in the Z-ring also appears to depend on the bacterial species. Taken together, the unique arrangement of FtsZ suggests novel unexplored mechanisms in bacterial cell division.  相似文献   

9.
Cytokinesis in bacteria is accomplished by a ring-shaped cell-division complex (the Z-ring). The primary component of the Z-ring is FtsZ, a filamentous tubulin homolog that serves as a scaffold for the recruitment of other cell-division-related proteins. FtsZ forms filaments and bundles. In the cell, it has been suggested that FtsZ filaments form the arcs of the ring and are aligned in the cell-circumferential direction. Using polarized fluorescence microscopy in live Escherichia coli cells, we measure the structural organization of FtsZ filaments in the Z-ring. The data suggest a disordered organization: a substantial portion of FtsZ filaments are aligned in the cell-axis direction. FtsZ organization in the Z-ring also appears to depend on the bacterial species. Taken together, the unique arrangement of FtsZ suggests novel unexplored mechanisms in bacterial cell division.  相似文献   

10.

Background

Cell division in Bacillus subtilis takes place precisely at midcell, through the action of Noc, which prevents division from occurring over the nucleoids, and the Min system, which prevents cell division from taking place at the poles. Originally it was thought that the Min system acts directly on FtsZ, preventing the formation of a Z-ring and, therefore, the formation of a complete cytokinetic ring at the poles. Recently, a new component of the B. subtilis Min system was identified, MinJ, which acts as a bridge between DivIVA and MinCD.

Methodology/Principal Findings

We used fluorescence microscopy and molecular genetics to examine the molecular role of MinJ. We found that in the absence of a functional Min system, FtsA, FtsL and PBP-2B remain associated with completed division sites. Evidence is provided that MinCDJ are responsible for the failure of these proteins to localize properly, indicating that MinCDJ can act on membrane integral components of the divisome.

Conclusions/Significance

Taken together, we postulate that the main function of the Min system is to prevent minicell formation adjacent to recently completed division sites by promoting the disassembly of the cytokinetic ring, thereby ensuring that cell division occurs only once per cell cycle. Thus, the role of the Min system in rod-shaped bacteria seems not to be restricted to an inhibitory function on FtsZ polymerization, but can act on different levels of the divisome.  相似文献   

11.
Bacterial cell division is mediated by a multi-protein machine known as the "divisome", which assembles at the site of cell division. Formation of the divisome starts with the polymerization of the tubulin-like protein FtsZ into a ring, the Z-ring. Z-ring formation is under tight control to ensure bacteria divide at the right time and place. Several proteins bind to the Z-ring to mediate its membrane association and persistence throughout the division process. A conserved stretch of amino acids at the C-terminus of FtsZ appears to be involved in many interactions with other proteins. Here, we describe a novel pull-down assay to look for binding partners of the FtsZ C-terminus, using a HaloTag affinity tag fused to the C-terminal 69 amino acids of B. subtilis FtsZ. Using lysates of Escherichia coli overexpressing several B. subtilis cell division proteins as prey we show that the FtsZ C-terminus specifically pulls down SepF, but not EzrA or MinC, and that the interaction depends on a conserved 16 amino acid stretch at the extreme C-terminus. In a reverse pull-down SepF binds to full-length FtsZ but not to a FtsZΔC16 truncate or FtsZ with a mutation of a conserved proline in the C-terminus. We show that the FtsZ C-terminus is required for the formation of tubules from FtsZ polymers by SepF rings. An alanine-scan of the conserved 16 amino acid stretch shows that many mutations affect SepF binding. Combined with the observation that SepF also interacts with the C-terminus of E. coli FtsZ, which is not an in vivo binding partner, we propose that the secondary and tertiary structure of the FtsZ C-terminus, rather than specific amino acids, are recognized by SepF.  相似文献   

12.
Cytokinesis in bacteria depends upon the contractile Z ring, which is composed of dynamic polymers of the tubulin homolog FtsZ as well as other membrane-associated proteins such as FtsA, a homolog of actin that is required for membrane attachment of the Z ring and its subsequent constriction. Here we show that a previously characterized hypermorphic mutant FtsA (FtsA*) partially disassembled FtsZ polymers in vitro. This effect was strictly dependent on ATP or ADP binding to FtsA* and occurred at substoichiometric levels relative to FtsZ, similar to cellular levels. Nucleotide-bound FtsA* did not affect FtsZ GTPase activity or the critical concentration for FtsZ assembly but was able to disassemble preformed FtsZ polymers, suggesting that FtsA* acts on FtsZ polymers. Microscopic examination of the inhibited FtsZ polymers revealed a transition from long, straight polymers and polymer bundles to mainly short, curved protofilaments. These results indicate that a bacterial actin, when activated by adenine nucleotides, can modify the length distribution of bacterial tubulin polymers, analogous to the effects of actin-depolymerizing factor/cofilin on F-actin.Bacterial cell division requires a large number of proteins that colocalize to form a putative protein machine at the cell membrane (1). This machine, sometimes called the divisome, recruits enzymes to synthesize the septum cell wall and to initiate and coordinate the invagination of the cytoplasmic membrane (and in Gram-negative bacteria, the outer membrane). The most widely conserved and key protein for this process is FtsZ, a homolog of tubulin that forms a ring structure called the Z ring, which marks the site of septum formation (2, 3). Like tubulin, FtsZ assembles into filaments with GTP but does not form microtubules (4). The precise assembly state and conformation of these FtsZ filaments at the division ring is not clear, although recent electron tomography work suggests that the FtsZ ring consists of multiple short filaments tethered to the membrane at discrete junctures (5), which may represent points along the filaments bridged by membrane anchor proteins.In Escherichia coli, two of these anchor proteins are known. One of these, ZipA, is not well conserved but is an essential protein in E. coli. ZipA binds to the C-terminal tail of FtsZ (68), and purified ZipA promotes bundling of FtsZ filaments in vitro (9, 10). The other, FtsA, is also essential in E. coli and is more widely conserved among bacterial species. FtsA is a member of the HSP70/actin superfamily (11, 12), and like ZipA, it interacts with the C-terminal tail of FtsZ (7, 1315). FtsA can self-associate (16, 17) and bind ATP (12, 18), but reports of ATPase activity vary, with Bacillus subtilis FtsA having high activity (19) and Streptococcus pneumoniae FtsA exhibiting no detectable activity (20). There are no reports of any other in vitro activities of FtsA, including effects on FtsZ assembly.Understanding how FtsA affects FtsZ assembly is important because FtsA has a number of key activities in the cell. It is required for recruitment of a number of divisome proteins (21, 22) and helps to tether the Z ring to the membrane via a C-terminal membrane-targeting sequence (23). FtsA, like ZipA and other divisome proteins, is necessary to activate the contraction of the Z ring (24, 25). In E. coli, the FtsA:FtsZ ratio is crucial for proper cell division, with either too high or too low a ratio inhibiting septum formation (26, 27). This ratio is roughly 1:5, with ∼700 molecules of FtsA and 3200 molecules of FtsZ per cell (28), which works out to concentrations of 1–2 and 5–10 μm, respectively.Another interesting property of FtsA is that single residue alterations in the protein can result in significant enhancement of divisome activity. For example, the R286W mutation of FtsA, also called FtsA*, can substitute for the native FtsA and divide the cell. However, this mutant FtsA causes E. coli cells to divide at less than 80% of their normal length (29) and allows efficient division of E. coli cells in the absence of ZipA (30), indicating that it has gain-of-function activity. FtsA* and other hypermorphic mutations such as E124A and I143L can also increase division activity in cells lacking other essential divisome components (3133). The R286W and E124A mutants of FtsA also bypass the FtsA:FtsZ ratio rule, allowing cell division to occur at higher ratios than with WT2 FtsA. This may be because the altered FtsA proteins self-associate more readily than WT FtsA, which may cause different changes in FtsZ assembly state as compared with WT FtsA (17, 34).In this study, we use an in vitro system with purified FtsZ and a purified tagged version of FtsA* to elucidate the role of FtsA in activating constriction of the Z ring in vivo. We show that FtsA*, at physiological concentrations in the presence of ATP or ADP, has significant effects on the assembly of FtsZ filaments.  相似文献   

13.
Grapevine trunk diseases (Eutypa dieback, esca and Botryosphaeria dieback) are caused by a complex of xylem-inhabiting fungi, which severely reduce yields in vineyards. Botryosphaeria dieback is associated with Botryosphaeriaceae. In order to develop effective strategies against Botryosphaeria dieback, we investigated the molecular basis of grapevine interactions with a virulent species, Neofusicoccum parvum, and a weak pathogen, Diplodia seriata. We investigated defenses induced by purified secreted fungal proteins within suspension cells of Vitis (Vitis rupestris and Vitis vinifera cv. Gewurztraminer) with putative different susceptibility to Botryosphaeria dieback. Our results show that Vitis cells are able to detect secreted proteins produced by Botryosphaeriaceae, resulting in a rapid alkalinization of the extracellular medium and the production of reactive oxygen species. Concerning early defense responses, N. parvum proteins induced a more intense response compared to D. seriata. Early and late defense responses, i.e., extracellular medium alkalinization, cell death, and expression of PR defense genes were stronger in V. rupestris compared to V. vinifera, except for stilbene production. Secreted Botryosphaeriaceae proteins triggered a high accumulation of δ-viniferin in V. vinifera suspension cells. Artificial inoculation assays on detached canes with N. parvum and D. seriata showed that the development of necrosis is reduced in V. rupestris compared to V. vinifera cv. Gewurztraminer. This may be related to a more efficient induction of defense responses in V. rupestris, although not sufficient to completely inhibit fungal colonization. Overall, our work shows a specific signature of defense responses depending on the grapevine genotype and the fungal species.  相似文献   

14.
Assembly of the tubulin-like cytoskeletal protein FtsZ into a ring structure at midcell establishes the location of the nascent division sites in prokaryotes. However, it is not yet known how the assembly and contraction of the Z ring are regulated, especially in cyanobacteria, the environmentally crucial organisms for which only one FtsZ partner protein, ZipN, has been described so far. Here, we characterized SepF and Ftn6, two novel septal proteins, in the spherical-celled strain Synechocystis PCC 6803. Both proteins were found to be indispensable to Synechocystis sp. strain PCC 6803. The depletion of both SepF and Ftn6 resulted in delayed cytokinesis and the generation of giant cells but did not prevent FtsZ polymerization, as shown by the visualization of green fluorescent protein (GFP)-tagged FtsZ polymers. These GFP-tagged Z-ring-like structures often appeared to be abnormal, because these reporter cells respond to the depletion of either SepF or Ftn6 with an increased abundance of total, natural, and GFP-tagged FtsZ proteins. In agreement with their septal localization, we found that both SepF and Ftn6 interact physically with FtsZ. Finally, we showed that SepF, but not Ftn6, stimulates the formation and/or stability of FtsZ polymers in vitro.Binary fission of a mother cell to form two daughter cells is a widely conserved cell proliferation mechanism. In nearly all bacteria, cell division is initiated by the polymerization into a ring-like structure at midcell of the tubulin homolog GTPase protein FtsZ, which is also found in some archae, as well as in plastids and some mitochondria (for reviews, see references 7, 21, and 33). The Z-ring is subsequently used as a scaffold for recruitment of downstream factors that execute the synthesis of the division septum. The assembly of this complex, also referred to as the divisome, has been thoroughly investigated in studies of the rod-shaped model organisms Escherichia coli and Bacillus subtilis) (for reviews, see references 3, 4, 7, 9, 11, 19, and 21). In E. coli, more than 10 different proteins are required for the progression and completion of cell division. They are designated Fts proteins because their depletion leads to filamentation of the bacteria, and they are recruited to the division site in the following sequential order: FtsZ→FtsA/ZipA/ZapB→FtsK→FtsQ and FtsL/FtsB→FtsW→FtsI and FtsN.The stability of the FtsZ protofilaments is thought to be important for assembly of the septal Z ring. Four FtsZ-interacting proteins have been shown to promote FtsZ polymerization and/or Z-ring stabilization, namely, ZapA and ZipA (found only in gammaproteobacteria), FtsA (an actin-like protein), and SepF (not found in gammaproteobacteria) (10, 31). Both FtsA and ZipA assemble at the Z-ring early and participate in its anchorage to the inner face of the cytoplasmic membrane of the cell. They also participate in the recruitment of the downstream cytokinetic factor FtsK. Subsequently, the recruitment of FtsQ and the FtsB/FtsL complex allow the progressive assembly of downstream factors (FtsW, FtsI, and FtsN) involved in synthesis of the septal cell wall (7).By contrast, the negative regulatory proteins MinCDE, DivIVA, EzrA, SulA, and Noc operate in the destabilization and positioning of the Z-ring at midcell (7, 21, 30), sometimes through a direct interaction with FtsZ (SulA, MinC, and ErzA).Little is known concerning cell division in cyanobacteria, in spite of their crucial importance to the biosphere (5, 27, 34) and their interest for biotechnologists (1, 6, 32). Cyanobacteria are also attractive because many species (such as E. coli and B. subtilis) exhibit a cylindrical morphology with a well-defined middle, whereas many others have a spherical shape (29) and thus possess an infinite number of potential division planes at the point of greatest cell diameter. Furthermore, as the progenitor of the chloroplasts (8), cyanobacteria can be of help for deciphering the stromal chloroplastic division machinery (33). Interestingly, several cell division factors occurring in E. coli and B. subtilis have been shown (FtsZ, MinCDE, and SulA) or proposed (FtsE, FtsI, FtsQ, and FtsW) to be conserved in cyanobacteria (23, 26) and chloroplasts (which lack MinC) (33). In contrast, ftsA, ftsB, zipA, ftsK, ftsL, ftsN, and zapA have not been detected in cyanobacteria.So far, cyanobacterial cytokinesis has mainly been investigated using the two unicellular species Synechococcus sp. strain PCC 7942 (rod shaped; hereafter S. elongatus) and Synechocystis sp. strain PCC 6803 (spherical-celled; hereafter Synechocystis sp.) and the filamentous strain Anabaena PCC 7120, all of which possess a fully sequenced genome (http://genome.kazusa.or.jp/cyanobase/) that is easily manipulated (16). Both FtsZ and ZipN/Ftn2/Arc6, a protein occurring only in cyanobacteria (ZipN [alternative name, Ftn2]) and plant chloroplasts (Arc6), were found to be crucial for cytokinesis (17, 23, 26) and to physically interact with each other (20, 23). We also reported that the MinCDE system participates in determining the correct positioning of the septal Z ring at midcell (23). In addition, it has recently been shown in studies of Synechococcus sp. that inactivation of both the cdv2 gene (an orthologue of the gene encoding B.subtilis sepF) and the ftn6 gene (present in only some cyanobacteria) promotes filamentation, though their role in cell division has yet to be characterized (16, 26).In a continuous effort to characterize the divisome machine of Synechocystis sp., we have used a combination of in vivo and in vitro techniques for thorough analysis of the SepF and Ftn6 proteins. We report here that both SepF and Ftn6 are crucial cytokinetic proteins that localize at the division site at midcell and whose depletion leads to the formation of giant cells that remain spherical. In agreement with their septal localization, both SepF and Ftn6 were found to interact physically with FtsZ; also, SepF, but not Ftn6, was found to stimulate the formation and/or stability of FtsZ polymers.  相似文献   

15.
How bacteria coordinate cell growth with division is not well understood. Bacterial cell elongation is controlled by actin–MreB while cell division is governed by tubulin–FtsZ. A ring‐like structure containing FtsZ (the Z ring) at mid‐cell attracts other cell division proteins to form the divisome, an essential protein assembly required for septum synthesis and cell separation. The Z ring exists at mid‐cell during a major part of the cell cycle without contracting. Here, we show that MreB and FtsZ of Escherichia coli interact directly and that this interaction is required for Z ring contraction. We further show that the MreB–FtsZ interaction is required for transfer of cell‐wall biosynthetic enzymes from the lateral to the mature divisome, allowing cells to synthesise the septum. Our observations show that bacterial cell division is coupled to cell elongation via a direct and essential interaction between FtsZ and MreB.  相似文献   

16.
In Escherichia coli cell division is driven by the tubulin-like GTPase, FtsZ, which forms the cytokinetic Z-ring. The Z-ring serves as a dynamic platform for the assembly of the multiprotein divisome, which catalyzes membrane cleavage to create equal daughter cells. Several proteins effect FtsZ assembly, thereby providing spatiotemporal control over cell division. One important class of FtsZ interacting/regulatory proteins is the Z-ring-associated proteins, Zaps, which typically modulate Z-ring formation by increasing lateral interactions between FtsZ protofilaments. Strikingly, these Zap proteins show no discernable sequence similarity, suggesting that they likely harbor distinct structures and mechanisms. The 19.8-kDa ZapC in particular shows no homology to any known protein. To gain insight into ZapC function, we determined its structure to 2.15 Å and performed genetic and biochemical studies. ZapC is a monomer composed of two domains, an N-terminal α/β region and a C-terminal twisted β barrel-like domain. The structure contains two pockets, one on each domain. The N-domain pocket is lined with residues previously implicated to be important for ZapC function as an FtsZ bundler. The adjacent C-domain pocket contains a hydrophobic center surrounded by conserved basic residues. Mutagenesis analyses indicate that this pocket is critical for FtsZ binding. An extensive FtsZ binding surface is consistent with the fact that, unlike many FtsZ regulators, ZapC binds the large FtsZ globular core rather than C-terminal tail, and the presence of two adjacent pockets suggests possible mechanisms for ZapC-mediated FtsZ bundling.  相似文献   

17.
Juarez JR  Margolin W 《The EMBO journal》2012,31(10):2235-2236
EMBO J 31 10, 2249–2260 (2012); published online March302012Once thought to exist only in eukaryotic cells, the highly conserved bacterial cytoskeleton is now known to function analogously to its eukaryotic counterparts, particularly in cell shape and division. For instance, the actin-like MreB protein and its homologs are important to maintain cell shape in many rod-shaped bacteria, probably by organizing how peptidoglycan is synthesized. FtsZ, a tubulin homolog, forms a scaffold for the cytokinetic ring, or divisome, by GTP-dependent polymerization into protofilaments. In this issue of The EMBO Journal, Szwedziak et al (2012) reveal the first crystal structures of cell division protein FtsA polymerizing into actin-like filaments, along with in vivo evidence that this self-interaction is crucial for proper cell division.FtsA is an actin homolog required for cytokinesis in many bacterial species and has several key roles in cell division, including helping to tether FtsZ to the cytoplasmic membrane via a membrane-targeting sequence (MTS), recruiting other essential proteins to the divisome, and perhaps promoting divisome constriction (de Boer, 2010). Szwedziak et al (2012) recapitulate the FtsZ-FtsA-membrane association in vitro using liposomes with FtsZ and FtsA proteins from Thermotoga maritima. To get a closer look at the FtsA-FtsZ interface, the authors co-crystallize FtsA with the carboxy-terminal tail of FtsZ, which is known to interact with FtsA. Intriguingly, the crystal reveals an FtsA homodimer. Contrary to the previous bioinformatics model of FtsA self-interaction that proposed a 180° rotation between the two subunits (Carettoni et al, 2003), the FtsA-FtsA interface in the crystal structure shows no rotation, similar to F-actin. Szwedziak et al (2012) also show that FtsA can form longer, actin-like polymers in the presence of non-hydrolysable ATP or on lipid monolayers. These results are surprising because FtsA has a divergent subdomain architecture compared to other actin-family proteins (van den Ent and Löwe).A critical question now is whether FtsA needs to form polymers in vivo to function properly. Purified Streptococcus pneumoniae FtsA assembles into large polymers that are not like F-actin, and it remains unclear if these structures are relevant in vivo (Krupka et al, 2012). Wild-type FtsA proteins do not form detectable filaments in cells, but C-terminal truncations of FtsA that remove the MTS form polymers quite readily in cells when overproduced, although they are not functional (Pichoff and Lutkenhaus, 2007). Even so, starting with an MTS truncation derivative of FtsA to visualize in vivo polymers, Szwedziak et al (2012) design site-directed mutants of Bacillus subtilis FtsA based on the FtsA-FtsA interface of their crystals; these fail to assemble into polymers in vivo. Using a similar MTS truncation derivative, Pichoff et al (2012) created random mutations in Escherichia coli FtsA, and found that those mapping to the same interface found by Szwedziak et al (2012) also disrupted polymer formation. Together, these data suggest that these residues are needed for FtsA self-interaction. Perplexingly, when these mutants were subsequently tested for functionality in the context of full-length FtsA, the results were mixed. Pichoff et al (2012) showed that FtsA mutants deficient for self-interaction in E. coli have a gain-of-function phenotype, whereas Szwedziak et al (2012) report that analogous mutants in B. subtilis FtsA suffer a loss of function. These results support the idea that FtsA self-association is related to its activity (Shiomi and Margolin, 2007), yet understanding how self-interaction regulates FtsA function clearly requires further study.The ability of eukaryotic cytoskeletal proteins to form long polymers is essential to their function, but the physiological relevance of long polymer formation by bacterial cytoskeletal proteins is now a topic of debate (Figure 1). For example, it has been hypothesized that FtsZ protofilaments wrap around the entire circumference of the cell to form the cytokinetic ring. However, recent studies using photoactivated localization microscopy (PALM) and electron cryotomography reveal a different model in which FtsZ forms a series of very short polymers that overlap to encompass the diameter of the cell (Li et al, 2007; Fu et al, 2010). MreB was also originally thought to form long-range helical polymers extending the length of the cell, but recent data obtained with more sophisticated microscopic techniques suggest that MreB is distributed in patches that move circumferentially and independently (White and Gober, 2012). It is not yet clear which of these models represents the true cellular architecture of MreB, although it is likely that some degree of MreB polymerization is still needed for function. It is notable that other bacterial homologs of actin and tubulin used for generating scaffolds or partitioning plasmid DNA, but not for essential cellular processes such as cell division and growth, tend to form long polymers that extend throughout the cell (Pogliano, 2008). The continued combined use of microscopic, biochemical, and genetic methods, as demonstrated by Szwedziak et al (2012) will enhance future understanding of ancestral tubulin and actin proteins in prokaryotes.Open in a separate windowFigure 1Bacterial actin and tubulin filaments involved in cell growth and division. (A) MreB (purple) has long been thought of as a spiral filament twisting along the cell length to control cell shape. Likewise, FtsZ protofilaments (blue) were once thought to wrap around the cell midpoint to organize the divisome. (B) Recent work using high-resolution microscopy has revealed that long cytoskeletal filaments are more likely to be short patches of polymers. The present work by Szwedziak et al (2012) has added FtsA actin-like filaments (green) to the model of possible divisome architecture.  相似文献   

18.
E. coli JM109?envC?nlpD deleted with genes envC and nlpD responsible for degrading peptidoglycan (PG) led to long filamentous cell shapes. When cell fission ring location genes minC and minD of Escherichia coli were deleted, E. coli JM109?minCD changed the cell growth pattern from binary division to multiple fissions. Bacterial morphology can be further engineered by overexpressing sulA gene resulting in inhibition on FtsZ, thus generating very long cellular filaments. By overexpressing sulA in E. coli JM109?envC?nlpD and E. coli JM109?minCD harboring poly(3-hydroxybutyrate) (PHB) synthesis operon phbCAB encoded in plasmid pBHR68, respectively, both engineered cells became long filaments and accumulated more PHB compared with the wild-type. Under same shake flask growth conditions, E. coli JM109?minCD (pBHR68) overexpressing sulA grown in multiple fission pattern accumulated approximately 70 % PHB in 9 g/L cell dry mass (CDM), which was significantly higher than E. coli JM109?envC?nlpD and the wild type, that produced 7.6 g/L and 8 g/L CDM containing 64 % and 51 % PHB, respectively. Results demonstrated that a combination of the new division pattern with elongated shape of E. coli improved PHB production. This provided a new vision on the enhanced production of inclusion bodies.  相似文献   

19.

Background

Cell division in Bacillus subtilis occurs precisely at midcell. Positional control of cell division is exerted by two mechanisms: nucleoid occlusion, through Noc, which prevents division through nucleoids, and the Min system, where the combined action of the MinC, D and J proteins prevents formation of the FtsZ ring at cell poles or recently completed division sites.

Methodology/Principal Findings

We used a genetic screen to identify mutations in ftsZ that confer resistance to the lethal overexpression of the MinC/MinD division inhibitor. The FtsZ mutants were purified and found to polymerize to a similar or lesser extent as wild type FtsZ, and all mutants displayed reduced GTP hydrolysis activity indicative of a reduced polymerization turnover. We found that even though the mutations conferred in vivo resistance to MinC/D, the purified FtsZ mutants did not display strong resistance to MinC in vitro.

Conclusions/Significance

Our results show that in B. subtilis, overproduction of MinC can be countered by mutations that alter FtsZ polymerization dynamics. Even though it would be very likely that the FtsZ mutants found depend on other Z-ring stabilizing proteins such as ZapA, FtsA or SepF, we found this not to be the case. This indicates that the cell division process in B. subtilis is extremely robust.  相似文献   

20.
The ribosome-binding GTPase HflX is required for manganese homeostasis in E. coli. While under normal conditions ?hflX cells behave like wild type E. coli with respect to growth pattern and morphology, deletion of hflX makes E. coli cells extremely sensitive to manganese, characterized by arrested cell growth and filamentation. Here we demonstrate that upon complementation by hflX, manganese stress is relieved. In phenotypic studies done in a manganese-rich environment, ?hflX cells were highly sensitive to antibiotics that bind the penicillin binding protein 3 (PBP3), suggesting that the manganese stress led to impaired peptidoglycan biosynthesis. An irregular distribution of dark bands of constriction along filaments, delocalization of the dark bands from midcell towards poles and subpoles, lack of septum formation and arrested cell division were observed in ?hflX cells under manganese stress. However, chromosome replication and segregation of nucleoids were unaffected under these conditions, as observed from confocal microscopy imaging and FACS studies. We conclude that absence of HflX leads to manganese accumulation in E. coli cells, affecting cell septum formation, probably by modulating the activity of the cell division protein PBP3 (FtsI), a major component of the divisome apparatus. We propose that HflX acts as a gatekeeper, regulating the influx of manganese into the cell.  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