SlideShare ist ein Scribd-Unternehmen logo
1 von 12
Downloaden Sie, um offline zu lesen
REVIEWS




                                   Bacterial cell division: assembly,
                                   maintenance and disassembly of
                                   the Z ring
                                   David W. Adams and Jeff Errington
                                   Abstract | Bacterial cell division is orchestrated by a tubulin homologue, FtsZ, which
                                   polymerizes to form a ring-like structure that is both a scaffold for the assembly of the
                                   bacterial cytokinetic machinery and, at least in part, a source of the energy for constriction.
                                   FtsZ assembly is tightly regulated, and a diverse repertoire of accessory proteins contributes
                                   to the formation of a functional division machine that is responsive to cell cycle status and
                                   environmental stress. In this Review, we describe the interaction of these proteins with FtsZ
                                   and discuss recent advances in our understanding of Z ring assembly.

                                  Cytokinesis in bacteria is achieved by a macromolecular             past decade (TABLE 1). Many of these proteins have roles
                                  machine called the divisome1–3. Divisome assembly is ini-           that are similar to those of the microtubule-associated
                                  tiated by polymerization of the tubulin homologue FtsZ              proteins (MAPs) of the Eukarya and affect the assem-
                                  into a ring-like structure that lies close to the cytoplasmic       bly, organization and stability of the Z ring, although
                                  membrane at the prospective division site4. In rod-shaped           none seems to be a true MAP homologue. Some of
                                  cells such as Escherichia coli and Bacillus subtilis, the divi-     these proteins are essential for survival but many are
                                  sion site usually lies at the midpoint of the rod, perpen-          not, and there is considerable functional overlap in the
                                  dicular to the long axis of the cell (FIG. 1a). Constriction        group. How then do these proteins ensure the correct
                                  of the ring gives rise to two equal daughter cells, each of         assembly of FtsZ into a Z ring that is both competent
                                  which is half the length of the parent cell. This ‘Z ring’          for cell division and responsive to cell cycle status and
                                  serves as a scaffold for the recruitment of the downstream          environmental stresses? This Review discusses the pro-
                                  components of the divisome and it persists throughout               teins that interact with FtsZ during the assembly of the
                                  division, guiding the synthesis, location and shape of              Z ring and highlights the recent progress in our under-
                                  the division septum5. FtsZ is almost universally conserved          standing of how these proteins intervene in the mecha-
                                  throughout the Bacteria (BOX 1); it is also present in the          nism of cell division and its regulation once the Z ring
                                  Euryarchaeota and has an active role in the division of             has been established.
                                  the plastids and mitochondria of several groups of the
                                  Eukarya6. FtsZ was recently shown to be an important new            FtsZ polymerization and the nature of the Z ring
                                  target for antibiotics, as specific inhibitors of FtsZ were         FtsZ is a cytoplasmic protein that is composed of two globu-
                                  found to protect against a lethal dose of Staphylococcus            lar domains separated by a central core helix (FIG. 2a). Despite
                                  aureus in a mouse model of infection7.                              the low level of sequence similarity, the tertiary structure
                                      Although the lifestyle and developmental behaviour              of FtsZ is remarkably similar to that of tubulin, supporting
                                  of some bacteria (for example, Caulobacter crescentus)              the theory that these proteins have common ances-
Centre for Bacterial Cell         dictates tight regulation of both FtsZ synthesis and                try 13–15. Furthermore, FtsZ polymerizes into tubulin-like
Biology, Institute for Cell and
                                  stability 8,9, in the two best studied bacteria, E. coli            protofilaments by the head-to-tail association of indi-
Molecular Biosciences,
Newcastle University,             and B. subtilis, no significant changes seem to occur in            vidual subunits16–19 (FIG. 2b). FtsZ, like tubulin, is a self-
Medical School, Framlington       the concentration of FtsZ, either throughout the cell               activating GTPase20–22: the insertion of the synergy loop
Place, Newcastle-upon-Tyne,       cycle or under different growth conditions10,11. In these           from the base of one subunit into the nucleotide-binding
NE2 4HH, UK.                      bacteria, cell division must therefore be regulated at              site of the subunit below places the catalytic residues close
Correspondence to J.E.
e-mail: jeff.errington@
                                  the level of FtsZ assembly 12 (BOX 2; FIG. 1b). Indeed, the         to the γ-phosphate, thereby allowing nucleotide hydro-
newcastle.ac.uk                   number and diversity of proteins that are known to                  lysis to proceed19,23. The GTPase activity of FtsZ therefore
doi:10.1038/nrmicro2198           interact directly with FtsZ has grown steadily over the             depends on FtsZ polymerization24.


642 | SEPTEMBER 2009 | VoluME 7                                                                                                    www.nature.com/reviews/micro

                                                        © 2009 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                    a                     Escherichia coli
                                                                                           Nucleoid occlusion
                                                                                                                                Bacillus subtilis

                                                                                                 Nucleoid
                                                                                              MinC gradient
                                                                                                   Z ring




                                    b
                                        Min   NO    Min          Min         NO      Min           Min      NO       NO   Min            Min        NO     NO     Min




                                                                                                                          Inhibitor-free zone
                                    Figure 1 | Cell division in rod-shaped bacteria. a | Two different modes of division. After chromosome replication and
                                    segregation into nucleoids the Z ring assembles at mid-cell. The ring then constricts to bring about division. Cell wall
                                                                                                                                           Nature Reviews | Microbiology
                                    synthesis follows the ring inwards. In Escherichia coli, synthesis of the division septum is accompanied by constriction of the
                                    outer membrane. In Bacillus subtilis, a cross wall of peptidoglycan initially divides the cell before it is degraded and
                                    remodelled to form the new, hemi-spherical cell poles. b | The spatial regulation of Z ring assembly. Nucleoid occlusion (NO),
                                    which is mediated by Noc (in B. subtilis) or SlmA (in E. coli), inhibits Z ring assembly close to the nucleoid. The Min system acts
                                    to prevent Z ring assembly at the cell poles (for simplicity, the Min dynamics in E. coli have been omitted). From left to right:
                                    in ‘newborn’ cells, both systems initially inhibit Z ring assembly throughout the cell; following cell elongation and chromosome
                                    replication, NO maintains inhibition in the cylindrical part of the cell; and finally, the progression of chromosome segregation
Cooperative assembly
                                    reveals an inhibitor-free region at mid-cell, allowing the Z ring to assemble.
Assembly that is characterized
by the increased affinity of
individual subunits for the
                                        In vitro, FtsZ displays cooperative assembly25–30 into a         remodelling throughout its existence, with rapid turnover
growing polymer rather than
for each other. Polymerization
                                    range of polymeric forms, from single-stranded protofila-            dynamics (in the order of 8 seconds) that depend on
displays a sigmoidal                ments to rings, bundles, tubules and sheets, depending               the intrinsic GTPase activity of the FtsZ polymer 10,40,41.
relationship with concentration     on the experimental conditions used16,17,31,32. However, the         Indeed, GTP hydrolysis promotes the disassembly of the
and has a defined critical          architecture of the Z ring that is formed in vivo is not well        protofilament and drives the highly dynamic nature of
concentration, below which no
significant assembly occurs.
                                    understood, and recent cryo-electron microscopy tomography           the FtsZ polymers that are observed both in the cell and
                                    of C. crescentus cells suggests that it might consist of a large     in vitro, as well as limiting the length of individual proto-
Cryo-electron microscopy            number of short, overlapping protofilaments, rather than             filaments30,34,40,41. Accordingly, FtsZ mutations that impair
tomography                          forming a continuous ring33. In vitro, FtsZ exhibits at least        GTP hydrolysis but not GTP binding stabilize polymers
A technique that allows the
                                    two important transitions during its assembly 34. The ini-           by reducing subunit turnover 41,42.
visualization of biological
molecules in a near-native
                                    tial assembly of single-stranded protofilaments occurs at                In addition to a regulatory function, nucleotide hydrol-
state. Unfixed samples are          a critical concentration of around 0.5–1 μM28,30,34,35, but          ysis has been proposed to have a force-generating role
flash frozen, held at cryogenic     there is a further transition at around 3 μM, which might            during septation, whereby a nucleotide-dependent tran-
temperature and visualized by       represent the assembly of higher-order structures with               sition from a straight, GTP-bound polymer to a curved,
transmission electron
microscopy. Tomography uses
                                    extensive lateral interactions between the protofilaments34.         GDP-bound polymer can transmit mechanical work to
a series of tilted images from      Puzzlingly, the FtsZ protofilament is only one subunit               the membrane33,43. However, the physiological role of this
which a three-dimensional           thick25,36, so the origin of this cooperativity remains              force remains controversial and is complicated by the
image of the sample can be          unclear. Recently, modelling has suggested that a mono-              fact that mutants that are severely impaired in nucleotide
deduced.
                                    mer isomerization step could be the source of this behav-            hydrolysis can still divide44,45. Moreover, recent work sug-
Fluorescence recovery after         iour 35–37. Importantly, the intracellular concentration of          gests that the relationship between the morphology of
photobleaching                      FtsZ greatly exceeds that of the critical concentration              polymers assembled in vitro and GTP hydrolysis is not as
A live-cell imaging technique       for assembly, at 3–10 μM10,38.                                       straightforward as was initially thought 46. Nevertheless,
used to monitor the                     FtsZ polymerization depends on GTP binding but                   the recent demonstration that Z rings can impart a force
intracellular dynamics of
fluorescently tagged molecules.
                                    not hydrolysis16. The combination of a partially solvent,            on a tubular membrane in vitro47, combined with the
A target region is irreversibly     accessible nucleotide-binding site and the abundance                 observation of both straight and curved filaments in vivo33,
photobleached by a laser and        of GTP in the cytoplasm means that GTP hydrolysis is                 reinforces the possibility of force generation by the Z ring.
then monitored for recovery of      probably a rate-limiting step in the polymerization cycle            In addition, mathematical modelling of Z ring dynam-
fluorescence. Dynamics are
                                    and, in contrast to tubulin, FtsZ polymers might contain             ics has shown that force generation could be achieved
expressed in terms of the
half-time for recovery (the time
                                    mainly GTP19,29,39. Fluorescence recovery after photobleaching       independently of FtsZ GTPase activity, with hydrolysis
taken to reach half of the          (FRAP), as well as other studies, has revealed that the              instead being required for subunit turnover and polymer
original fluorescence intensity).   Z ring is not a static structure: it undergoes constant              remodelling 35,48.


NATuRE REVIEwS | MiCrobiology                                                                                                    VoluME 7 | SEPTEMBER 2009 | 643

                                                          © 2009 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                                                                                      been found in the Bacteria. Nonetheless, the similarity
 Box 1 | FtsZ-independent division
                                                                                                      of the polymerization dynamics of FtsZ and tubulin has
                                                                                                      afforded valuable insights into how the unique regula-
                                                                                                      tory proteins that interact with FtsZ might modulate
                                                                                                      its polymerization. First, proteins can prevent the ini-
                                                                                                      tial assembly of FtsZ by preventing nucleotide binding,
                                                                                                      sequestering individual monomers or capping growing
                                                                                                      polymers. Second, proteins that promote polymeriza-
                                                                                                      tion or stabilize existing polymers could do so by con-
 The wide conservation of FtsZ throughout the Bacteria andNature Reviews | Microbiology
                                                               the euryarchaeotal branch              centrating FtsZ at the membrane, cross-linking separate
 of the Archaea, which is supported by the ever-increasing wealth of genome sequence                  protofilaments or facilitating the formation of stabilizing
 information, has cemented its position as an essential cell division protein. There are,             lateral interactions, as well as by preventing the disas-
 however, several intriguing examples of species in which FtsZ is absent, has been                    sembly of the polymer by inhibiting nucleotide hydrol-
 replaced or is simply no longer required for cell division.                                          ysis. Finally, proteins that disassemble or destabilize
 Missing FtsZ                                                                                         the polymer might do so by directly counteracting the
 Several distinct groups of bacteria, including Chlamydiae and Planctomycetes, as well                mechanisms used by positive-acting factors — that is, by
 as Ureaplasma urealyticum and Mycoplasma mobile, are unusual among bacteria in that                  stimulating GTPase activity, facilitating disassembly
 they lack a recognizable homologue of FtsZ52,124,125. Although it has been suggested                 by sequestering monomers or inhibiting the formation
 that Chlamydia spp., which are obligate intracellular parasites, might draw on a host                of lateral interactions.
 cell division machinery126, there is compelling evidence to suggest that they divide
 independently of their hosts and might instead use the subset of division genes that
 they contain to synthesize a peptidoglycan division septum127. However, it is not clear              Assembling the Z ring
 how this site is chosen or how any putative synthetic machinery is assembled in the                  A crucial element of Z ring assembly is the association
 absence of FtsZ. Planctomycetes and U. urealyticum are free-living organisms and so                  between FtsZ and the cell membrane. FtsZ does not
 must be capable of independent cell division, but to date little progress has been made              seem to have any direct affinity for the membrane, but
 in understanding how this might be accomplished.                                                     all models for Z ring formation require its attachment to
 replacing FtsZ                                                                                       the membrane, both to maintain its structural integrity
 Substantial progress has been made in understanding the division of the                              during septation and probably to transmit any constric-
 Crenarchaeota, which completely lack FtsZ, owing to the discovery of a new cell                      tive force to the membrane1,33,51,53. In E. coli there is good
 division machine128,129. The products of three cell cycle-regulated genes, cdvA, cdvB                evidence that two proteins, FtsA and ZipA, collaborate in
 and cdvC, constitute a protein complex that forms a contractile-ring structure                       anchoring FtsZ to the membrane54. Both of these proteins
 between segregated nucleoids in dividing cells that is essential for cell division.                  are essential for cell division54,55: depletion of either single
 Although CdvA is unique to the Crenarchaeota, CdvB and CdvC are homologous to                        protein results in the formation of elongated filaments
 proteins of the endosomal sorting complex required for transport (ESCRT) III complex
                                                                                                      with regularly assembled Z rings that are positioned
 of the Eukarya, which is involved in endosomal protein sorting, viral budding and
 membrane abscission during cytokinesis. These proteins are found in all but one order
                                                                                                      correctly between segregated nucleoids but are non-
 of the Crenarchaeota and are absent from the FtsZ-dependent Euryarchaeota.                           functional for division. when both proteins are removed,
                                                                                                      Z rings do not form54,56.
 life before FtsZ
 Recently, the L-form (wall-less derivatives of common bacteria) of Bacillus subtilis was
 found to propagate independently of FtsZ by a new mode of division that is defined by a              ZipA. ZipA is a bitopic protein that is composed of three
 remarkable extrusion–resolution process (see the figure, which shows phase-contrast                  domains: a transmembrane anchor, a long, basic arm
 images of division in L-form cells of B. subtilis) that discharges viable progeny130. This           that is rich in proline and glutamine residues and a
 might be related to the surprising finding that ftsZ can be deleted in Streptomyces                  large, globular C-terminal domain 55,57,58. ZipA has
 spp.131. It is possible that these sick mutants use a similar extrusion–resolution process to        a particularly rare membrane topology, as its sole trans-
 propagate. Although the mechanisms underlying extrusion–resolution remain unclear,                   membrane anchor is at the amino terminus and the
 the process could provide a glimpse into the distant past, to a time before the                      C-terminal domain is in the cytoplasm55,59. This unusual
 FtsZ-based division machinery had evolved.                                                           topology is shared by the division protein extra Z rings
 Scale bar represents 3mm. Images courtesy of P. Domínguez-Cuevas, Newcastle University, UK.          A (EzrA) in B. subtilis 1,60. However, these two proteins
                                                                                                      seem to have considerably different roles in cell division,
                                                                                                      so the potential importance of their shared membrane
                                       Although the carboxy-terminal domain of FtsZ is                topology is unclear.
                                   similar in its fold and acidic nature to that of tubulin, it           In the cell, ZipA is dispersed throughout the inner
                                   lacks the extended surface loops that are thought to aid           membrane until the initiation of cell division, when it is
                                   the assembly of the microtubule and its interaction with           recruited to the assembling Z ring by a direct interaction
                                   motor proteins49. However, FtsZ contains a unique, highly          with FtsZ55,56,61. The division block that occurs in cells
                                   conserved peptide (~9 amino acids) at its extreme C ter-           that are depleted of ZipA seems to be due to the failure
                                   minus; this site is thought to be unstructured and is dis-         of the Z ring to recruit FtsK, a protein that is essential for
                                   pensable for polymerization but essential for cell division,       the recruitment of downstream division proteins54,62. ZipA
                                   as it is the binding site for several division proteins50–52       interacts directly with the conserved C-terminal tail of
Bitopic protein                    (discussed below).                                                 FtsZ57–59,61,63. This interaction is mediated by the C-terminal
A protein that contains a single
transmembrane segment
                                       MAPs can alter the stability, bundling and disassembly         domain of ZipA, which is both necessary and sufficient
between the cytoplasmic and        of the tubulin polymer in response to cellular cues but,           for its recruitment to the Z ring59. Interestingly, the trans-
extracytoplasmic domains.          as mentioned above, to date no true homologues have                membrane domain does not seem to be simply an inert


644 | SEPTEMBER 2009 | VoluME 7                                                                                                    www.nature.com/reviews/micro

                                                        © 2009 Macmillan Publishers Limited. All rights reserved
REVIEWS

  Box 2 | Spatial regulation of cell division                                                            FtsA show that ZipA can function as a membrane tether
                                                                                                         for FtsZ51,54–56. The flexibility of the unstructured linker
  In both Bacillus subtilis and Escherichia coli, assembly of the Z ring — and thus the                  domain between the transmembrane anchor and the
  initiation of cell division — is regulated by two distinct but partially overlapping                   C-terminal domain is likely to be pivotal in this tethering
  systems (FIG. 1b). These systems have been reviewed in detail elsewhere but are briefly
                                                                                                         role, allowing the C-terminal domain to reach out from the
  described here132,133. First, nucleoid occlusion prevents the assembly of the Z ring over
                                                                                                         membrane and bind the C-terminus of FtsZ51,65.
  the nucleoid by the action of two unrelated DNA binding proteins, Noc in B. subtilis and
  SlmA in E. coli134,135. Although SlmA interacts directly with FtsZ in vitro, the exact                     ZipA is not widely conserved outside the Gamma-
  mechanism by which either protein inhibits the formation of the Z ring remains                         proteobacteria, which suggests that its role has either been
  unclear135. Second, the Min system contributes to spatial regulation by inhibiting Z ring              replaced or become redundant55. In support of the sec-
  assembly at the cell poles, preventing the generation of anucleate mini-cells. This is                 ond suggestion, a single amino acid substitution in FtsA
  achieved by creating a symmetrical concentration gradient of MinC and MinD,                            renders ZipA non-essential in E. coli, allowing efficient
  negative regulators of Z ring assembly, around the mid-cell position so that the                       growth and division in its absence66 (discussed below).
  time-averaged concentration is always highest at the cell poles. MinC interacts directly
  with FtsZ and is thought to prevent Z ring assembly at several levels by inhibiting                    FtsA. FtsA is widely conserved throughout the Bacteria
  lateral interactions and weakening longitudinal bonds, thereby compromising the
                                                                                                         and is often found upstream of ftsZ, in an operon within
  integrity of the assembled polymer92,93,110,136,137. In addition, in E. coli MinC has been
                                                                                                         the dcw cluster 67. The crystal structure of FtsA from
  shown to act early during the assembly of the Z ring, competing with FtsA for binding
  at the extreme carboxy-terminal tail of FtsZ138. The removal of FtsA further destabilizes              Thermotoga maritima showed that it is structurally related
  the assembling Z ring and concentrates the inhibitor in direct contact with its target138.             to actin and consists of two domains, each of which can be
    In E. coli topological specificity is imparted by the periodic oscillation of MinC and MinD          further divided into two subdomains68. In one structure,
  from pole to pole, a process that is driven by MinE139–141. In B. subtilis, two unrelated proteins,    the nucleotide-binding site, which is formed by an inter-
  DivIVA and MinJ, are thought to recruit MinC and MinD to division sites and retain it at the           domain cleft, was found to contain both ATP and a Mg 2+
  new cell poles142–147. Recruitment to the nascent division site allows MinC to prevent the             ion68, supporting reports that FtsA can bind ATP69–71.
  re-initiation of cell division and to ensure that only one division event occurs per cell cycle.       Interestingly, FtsA purified from Streptococcus pneumoniae
                                                                                                         can bind various nucleotide phosphates in a manner that
                                                                                                         is directly proportional to the length of the phosphate
                                    anchor, suggesting that it has a specific function in divi-          moiety; ATP and GTP are favoured over ADP and GDP,
                                    sion, perhaps by interacting with one or more of the many            which are favoured over AMP and GMP71. In B. subtilis,
                                    membrane-bound components of the divisome59 (BOX 3).                 FtsA has been shown to exist as a dimer in solution70,
                                        Structural studies have revealed that the C-terminal             and various two-hybrid assays have shown that FtsA can
                                    domain of ZipA consists of a six-stranded anti-parallel              interact with itself 72–76. Furthermore, FtsA from S. pneu-
                                    β-sheet laid against three α-helices, with a core domain             moniae has been reported to reversibly polymerize into
                                    containing a split β–α–β fold that is similar to the folds           corkscrew-like helices that are composed of pairs of paired
                                    found in many RNA-binding proteins57,58. The solvent-                protofilaments71. These polymers exhibited nucleotide-
                                    exposed face of this motif forms a shallow hydrophobic               dependent stability, forming extremely stable polymers
                                    cleft that extends across the domain57,58. The final 20 amino        in the presence of ATP and less stable polymers in the
                                    acids of the C-terminal domain of FtsZ are sufficient for the        presence of ADP71. This report suggests a role for nucle-
                                    interaction with ZipA, and a co-crystal structure of                 otide hydrolysis in the regulation of FtsA polymerization;
                                    the complex has been obtained57, which shows that the                however, ATPase activity was not found in S. pneumo-
                                    hydrophobic cleft of ZipA is occupied by the C-terminal              niae FtsA and has so far only been reported in FtsA from
                                    fragment of FtsZ57. This binding is driven by the burial of          B. subtilis70. Although the role of nucleotide binding is not
                                    exposed hydrophobic residues, and most of the binding                yet known, it is intriguing that mutation of residues that
                                    affinity is provided by only a few key residues, which are           have been shown to contact the nucleotide in the crystal
                                    highly conserved57,58.                                               structure abolishes the ability of FtsA to interact both with
                                        In vitro, the C terminus of ZipA promotes the assembly           itself and with FtsZ72. Structural modelling suggests that
                                    of FtsZ into thick bundles and sheets of protofilaments              FtsA dimerizes in a head-to-tail fashion, with the indi-
                                    that are aligned laterally 59,64. Consistent with this positive      vidual subunits rotated 180° to one another; however, the
                                    role in assembly and in the formation of stabilizing lateral         importance of this model remains unclear 77.
                                    interactions, twofold overproduction of ZipA suppresses                  In E. coli, FtsA interacts with the conserved extreme
                                    the division defect of cells bearing the thermosensitive             C terminus of FtsZ51,54,56. In both E. coli and B. subtilis the
                                    ftsZ84 mutation at the non-permissive temperature64.                 ratio of FtsZ to FtsA is approximately 5 to 1 (REFs 10,70),
                                        Although the mechanism by which ZipA organizes                   and maintenance of this balance is crucial for cell divi-
                                    FtsZ protofilaments is not known, it is clear that the indi-         sion78. This is exemplified by the fact that the toxic
                                    vidual domains of ZipA have separable functions in vivo,             overproduction of either protein can be counteracted
ftsZ84
                                    and several models describing the activity and physiologi-           by overproduction of the other 78. The ability of FtsA to sup-
A mutation that results in the
substitution of glycine to serine   cal role of ZipA have been proposed51,55,57,59,64. The globular      port the initial assembly of the Z ring without ZipA, as well
at position 105 in                  C-terminal domain might facilitate bundle formation either           as the ability of an ftsA gain-of-function mutant to bypass
Escherichia coli FtsZ. This         by a chaperone-like activity, through which it stabilizes the        the need for ZipA, suggested that FtsA can also tether the Z
substitution reduces both the       unstructured C-terminal tail, or by simply shielding                 ring to the inner membrane56,66. Indeed, the re-examination
GTP binding and the GTPase
activity of FtsZ in vitro and
                                    the solvent-exposed hydrophobic residues57. In addition, the         of a highly conserved motif at the extreme C terminus of
renders the protein                 essential nature of the transmembrane domain and                     FtsA revealed a membrane-targeting amphipathic helix
temperature sensitive in vivo.      the ability of the Z ring to assemble in the absence of              that is separated from the core protein by a flexible linker


NATuRE REVIEwS | MiCrobiology                                                                                                    VoluME 7 | SEPTEMBER 2009 | 645

                                                           © 2009 Macmillan Publishers Limited. All rights reserved
REVIEWS

 Table 1 | FtsZ and the proteins that modulate its polymerization
Protein role                                                 Size* Structural characterization                     In vitro data Essential? Conservation
                                                             (kDa)                                                 available?
Assembly of the Z ring
FtsZ    A structural subunit of the Z ring that is   40.2             A self-assembling GTPase that is             Yes              Yes‡            Widely conserved
        required for initiation of cell division and                  structurally related to tubulin
        serves as a scaffold for divisome assembly
FtsA    A principal membrane tether that             48               An ATP-binding protein that is               No§              Yes||           Widely conserved;
        is required for Z ring assembly,                              structurally related to actin but                                             often found in an
        organization of FtsZ polymers and                             that has the uniquely positioned                                              operon with ftsZ
        recruitment of downstream proteins                            subdomain 1C
ZipA    Promotes Z ring assembly and is a            36.3             A large globular domain that is              Yes              Yes             Gammaproteobacteria
        secondary membrane anchor                                     linked to a single amino-terminal TM
                                                                      domain by an extended linker
Regulators of Z ring dynamics
ZapA        A positive modulator of Z ring assembly          9.7      Forms a dimer through its extensive          Yes              No              Widely conserved
            and stability                                             coiled-coil region
ZapB        Has a redundant role in ensuring proper          9.5      A dimer consisting of a single               No§              No              A subset of
            Z ring assembly                                           coiled-coil region; self-assembles                                            Gammaproteobacteria
                                                                      into large polymer bundles
SepF      Has an overlapping role with FtsA in Z             17       NA                                           No               No¶             Gram-positive bacteria
          ring assembly and is required for proper                                                                                                  and Cyanobacteria
          septal morphology
EzrA      A negative regulator of Z ring assembly            64.8     Predicted to contain four coiled-coil        Yes              No              Gram-positive bacteria
          throughout the cell membrane that                           regions in its carboxyl terminus and                                          with a low GC content
          contributes to mid-cell Z ring dynamics                     a single N-terminal TM anchor
          and has a role in coordinating cell
          elongation with division
Cell cycle-responsive regulators
UgtP      A growth rate-dependent inhibitor of               43.4     A membrane-associated protein                Yes              No              A widely conserved
          cell division                                               that self-assembles into tightly                                              metabolic pathway
                                                                      wound spirals in vitro
SulA     A cell division inhibitor of the SOS                18.7     Forms a dimer, either face of which          Yes              No              Gammaproteobacteria
         response that prevents the assembly                          can interact with the T7 loop of FtsZ
         of new Z rings and facilitates the
         disassembly of existing Z rings
MciZ     Contributes to the inhibition of Z ring             4.8      A small peptide that is           Yes                         No              Bacillus spp.
         assembly following the initiation of                         predicted to bind close to the
         sporulation                                                  nucleotide-binding pocket of FtsZ
General regulators
ClpX     Helps to maintain the cytoplasmic pool              46.2     A hexameric substrate-recognition            Yes              No              Widely conserved
         of free FtsZ subunits                                        complex of ClpXP protease
 *The stated protein sizes are for Bacillus subtilis or, where appropriate, Escherichia coli and are calculated values, published at the SubtiList or Colibri World-Wide
 Web Servers. ‡This protein is not essential for viability in Streptomyces coelicolor. §In vitro data are only available on the proteins themselves, not on their potential
 to modulate FtsZ polymerization. ||This is essential in E. coli but not for vegetative cell division in B. subtilis. ¶This protein is essential in Synechococcus elongatus.
 EzrA, extra Z rings A; MciZ, mother cell inhibitor of FtsZ; NA, no data available; TM, transmembrane.


                                  region67. This amphipathic helix is essential for the function          characterized, little was known about the nature of the
                                  of FtsA under normal laboratory conditions: deletion of                 binding interface in FtsA until recently50. A screen for
                                  this region renders FtsA non-functional and causes                      ftsA mutations that cause the resultant protein to interact
                                  it to assemble into deleterious cytoplasmic rods67.                     with the membrane but not with FtsZ identified impor-
                                  Importantly, and in contrast to ZipA59, the membrane                    tant FtsA residues, including three charged residues that
                                  targeting region of FtsA can be replaced by various                     are strictly conserved and are clustered mainly on the sur-
                                  unrelated membrane-targeting determinants67,79.                         face of the 2B subdomain72. Substitutions of these resi-
                                      FtsA differs from actin in that it is missing the 1B                dues do not affect the ability of FtsA to self-interact, but
                                  domain and it has an unrelated subdomain called 1C68.                   they do abolish its ability to interact with FtsZ72. The 2B
                                  Deletion of 1C renders FtsA unable to interact with itself,             subdomain is likely to have additional functions in cell
                                  although it retains the ability to localize to the Z ring 73. The       division, as deletion of a small anti-parallel β-sheet at the
                                  inability of FtsA lacking the 1C domain to allow the pro-               apex of this domain seems to affect the correct placement
                                  gression of cell division has been explained by the finding             of the Z ring73. Furthermore, the ftsA* mutation is found
                                  that the 1C domain is involved in recruiting downstream                 in the same region66 (discussed below).
                                  components of the divisome to the Z ring 73,80.                             Aside from its role in Z ring assembly, FtsA is now
                                      Although the role of the extreme C terminus of                      known to affect the integrity of the Z ring at various stages
                                  FtsZ in the interaction with FtsA has been extensively                  of divisome assembly 67,72,74,81. The ftsA* gain-of-function


646 | SEPTEMBER 2009 | VoluME 7                                                                                                             www.nature.com/reviews/micro

                                                          © 2009 Macmillan Publishers Limited. All rights reserved
REVIEWS

a                                       Nucleotide-binding site          b                           Z ring assembly and polymer dynamics
                                                                                                     Many mainly non-essential proteins are thought to
         H7                                                                                          directly interact with FtsZ to regulate the dynamics of the
                                                                                                     Z ring that is assembled at mid-cell. Although individually
                                                                                                     these proteins are not required for Z ring assembly, their
                                                                                                     various roles in modulating the polymerization of FtsZ
                                                                                                     mean that the combined loss of certain regulatory proteins
                                                                                                     often produces a synthetic lethal division phenotype.

                                                                                                     ZapA. ZapA is a small protein of 85 amino acids that was
C-terminal                                                                                           identified in a genome-wide screen for proteins that,
domain                                                                                               when overexpressed, can overcome the lethality that is
                                                                                                     induced by MinD overexpression in B. subtilis86. In both
                                                                                                     B. subtilis and E. coli, ZapA is recruited to the divisome
                                                        N-terminal                                   early, probably by a direct interaction with FtsZ, where
                                                        domain
                                                                                                     it enhances both the assembly and the stability of the
                                                                                                     Z ring86. Cells lacking ZapA exhibit no phenotype in either
                                                                                                     organism86,87 but, similar to other non-essential regulatory
                                                                                                     proteins, this loss is conditionally lethal in various sensi-
                                   T7 loop
                                                                                                     tized backgrounds60,85,88, including in cells with artificially
Figure 2 | FtsZ polymerization. a | The crystal structure of a Bacillus subtilis FtsZ                lowered FtsZ levels86. Surprisingly, given its apparently
monomer bound to GTP-γS (Protein Data Bank entry 2RHO). FtsZ is composed of two                      positive role in Z ring assembly, null mutations in zapA
                                                                 Nature Reviews | Microbiology
independently folding domains and a central core helix, H7. The amino-terminal domain                are synthetically lethal when combined with null muta-
contains the tubulin signature motif, GGGTGTG, and forms the nucleotide-binding site.                tions in ezrA86. This synergism probably results from the
At the base of the carboxy-terminal domain, following H7, is the catalytic T7 or synergy             assembly of a less dynamic Z ring in the absence of EzrA
loop. The extreme C-terminal tail, which forms the binding site for several division                 (discussed below), which reduces the cytoplasmic pool of
proteins, is not visible in the crystal structure. b | FtsZ polymerizes by the head-to-tail          subunits that are available for exchange. This, combined
association of individual subunits, shown in blue and green, to form a single-stranded
                                                                                                     with the increased concentration of FtsZ that is required
protofilament with a longitudinal subunit repeat that is similar to that of tubulin.
                                                                                                     for assembly in the absence of ZapA, leads to the synthetic
Insertion of the T7 loop into the nucleotide-binding site (orange) of the subunit below
places two highly conserved aspartate residues in the vicinity of the FtsZ γ-phosphate;              lethal phenotype86.
these aspartate residues are thought to activate the GTPase activity of FtsZ by                          The crystal structure of ZapA revealed an anti-parallel
polarizing an attacking water molecule.                                                              tetramer that is composed of a pair of dimers which asso-
                                                                                                     ciate through extensive coiled-coil interactions along the
                                                                                                     C termini of the individual subunits89. Dimerization of
                                 mutation bypasses the need for ZipA and partially sup-              ZapA is mediated mainly by contacts between the small,
                                 presses the requirement for FtsK66. Cells bearing only              globular N-terminal domains89. In solution, oligomeri-
                                 ftsA* assemble Z rings earlier in the cell cycle and can            zation is constrained by concentration and the protein
                                 tolerate higher levels of MinC than wild-type cells, indi-          exists in a dimer–tetramer equilibrium. However, taking
                                 cating that both the initial assembly and the stability of          into account the predicted abundance of ZapA in vivo,
                                 the Z ring are enhanced by FtsA* (REF. 82). FtsA* seems             the dimer is more likely to be the physiologically relevant
                                 to confer enhanced stability on the Z ring at the level of          species86,89,90.
                                 initial assembly, as it affects neither the intrinsic dynamics          In vitro, ZapA specifically interacts with FtsZ and,
                                 of FtsZ subunits nor the proportion of FtsZ present in the          consistent with its positive role in polymerization in vivo,
                                 ring 82. In support of this model, the interaction of FtsZ          enhances both the initial polymerization of FtsZ and the
                                 with FtsA* is much stronger than the interaction with               stability of the resulting polymers86,89,90. ZapA does not
                                 wild-type FtsA82. In addition, a further gain-of-function           form higher-order structures under any conditions exam-
                                 mutation in FtsA that seems to act by stabilizing the Z ring        ined to date86,89,90. FtsZ polymers that have assembled in
                                 has recently been reported81.                                       the presence of equimolar amounts of ZapA form higher-
                                     The assertion that FtsA probably constitutes the princi-        order structures, such as branched networks that are
                                 pal membrane anchor for the Z ring is supported by several          composed of thick protofilament bundles86,89,90, which
                                 lines of evidence67,72 and most notably by the absence of rec-      are similar to those observed in the presence of artifi-
                                 ognizable homologues of ZipA in most bacterial lineages55.          cial bundling agents such as diethylaminoethyl dextran
                                 The situation in B. subtilis, however, is somewhat different.       and Ca2+ (REF. 91). The enhancement of polymerization and,
                                 FtsA is not essential for vegetative cell division, although        in particular, polymer stability is directly correlated with
                                 the cells are highly filamentous and the frequency of func-         the inhibition of FtsZ GTPase activity 86,90. Intriguingly,
                                 tional Z ring formation is severely reduced83–85. This is con-      one report identified a potential change in the confor-
                                 sistent with the proposed role of FtsA in promoting Z ring          mational state of the bound nucleotide, using the new
                                 integrity84. However, the ability of the Z ring to form, even       technique of linear dichroism to monitor changes in the
Synthetic lethal
                                 at low frequency, is puzzling and suggests that another             FtsZ polymer 90,91. The authors suggested that this con-
Lethality due to a combination   protein might also have a similar role to FtsA, albeit at a         formational change might be the result of a general activ-
of two non-lethal mutations.     reduced level of efficiency (discussed below).                      ity of bundling agents and proposed a model in which


NATuRE REVIEwS | MiCrobiology                                                                                                VoluME 7 | SEPTEMBER 2009 | 647

                                                       © 2009 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                                                                                 zapB are viable, although slightly elongated, and tend to
 Box 3 | Assembly of the divisome
                                                                                                 form filamentous cells that are blocked in cell division. In
a                                                b                                               the absence of ZapB, Z ring formation is both reduced in
                                                                   PBP1   PBP2B                  frequency and altered in morphology, and short, tight-




                                                                                             DivIC
                                                                                             FtsL
                                                                                   DivIB         pitched helices are frequently formed. In vitro, ZapB
            EzrA                                            EzrA                                 dimers can interact through their termini to form large
                                                                                                 bundles. As ZapB is abundant (approximately 13,000 mol-
                                                                                                 ecules per cell), ZapB polymers might bridge shorter FtsZ
                                                                   GpsB
         FtsA                                             FtsA           FtsW                    protofilaments in the assembling Z ring to promote their
                                                                                                 organization into a functional ring. However, the con-
   ZapA                                            ZapA
                                                                                                 tribution of ZapB to Z ring assembly seems to be subtle,
                         SepF
                                      FtsZ
                                                                        SepF
                                                                                     FtsZ        as mutation of zapB generates a striking division pheno-
                             SepF                                           SepF                 type only in bacteria carrying the ftsZ84 mutation, not
                                                                                                 in bacteria with a range of other cell division mutations,
Following the relief of spatial regulation as the cell grows and its chromosomes segregate,      including of zapA. The interaction of ZapB with FtsZ or
                                                                   Nature Reviews | Microbiology
the Z ring forms at mid-cell, tethered to the inner face of the cytoplasmic membrane. It is      its potential to modulate FtsZ polymerization in vitro has
not clear whether there are early intermediates in ring formation, but dynamic helical
                                                                                                 not yet been reported.
 polymers of FtsZ have been described, which suggests that the Z ring forms by the
 collapse of a spiral intermediate148–151. Once assembled, the Z ring begins the process of
 recruiting the downstream components of the divisome.                                                 EzrA. EzrA was identified in B. subtilis as a negative
   In Bacillus subtilis, assembly of the divisome occurs by a two-step mechanism, in which             regulator of Z ring assembly in a screen for extragenic
 the early-assembling proteins (see the figure, part a) are directly recruited to the Z ring           suppressors of a temperature-sensitive ftsZ mutant
 by FtsZ and, after a substantial delay, are followed by the concerted and interdependent              tagged with green fluorescent protein (ftsZ-GFP)60, but
 assembly of the later-assembling proteins (see the figure, part b)1,152. In Escherichia coli,         it probably has multiple functions in division. EzrA is
 classical localization studies had suggested an essentially linear pathway of divisome                an integral membrane protein and is predicted to have an
 assembly153. However, recent work has shown that this process probably occurs by the                  N-terminal transmembrane anchor and a cytoplas-
 sequential assembly of at least three subcomplexes2,154. Similarly to B. subtilis, a time             mic C-terminal domain that consists of four coiled-coil
 delay between the assembly of the Z ring and the recruitment of the later-assembling
                                                                                                       regions60,95. This domain structure is conserved through-
 proteins has been detected155.
   Following the completion of divisome assembly and, possibly, an as-yet-undiscovered                 out the low-GC-content Gram-positive bacteria in which
 signal, the Z ring constricts ahead of the leading edge of the newly synthesized division             it is present 95. EzrA is constitutively expressed and is
 septum. Finally, the daughter cells separate, and this involves the action of specialized             abundant, with an estimated 10,000–20,000 molecules per
 peptidoglycan hydrolases. The overlap between invagination and cell separation varies                 cell95,96. It is distributed throughout the plasma membrane
 between organisms: in B. subtilis a complete cross wall is usually formed before cell                 of elongating cells and then recruited to the Z ring by a
 separation begins, whereas in E. coli the two processes occur almost simultaneously,                  recently identified patch of seven amino acids, known as
 leading to a characteristic furrow at the active division site (FIG. 1a). These distinctions          the QNR patch, that follows the last coiled-coil region in the
 probably relate to the differing cell envelope structures of Gram-positive and                        C-terminal region60,97. ezrA mutants have an increased
 Gram-negative bacteria but might also reflect flexibility in the relative timing of septal            frequency of Z ring formation and additional Z rings are
 wall hydrolysis, which is tightly coupled to septum synthesis in E. coli but is much more
                                                                                                       often observed at one or both cell poles, which suggests
 separable in B. subtilis.
                                                                                                       a Min-like function in preventing Z ring assembly at cell
 EzrA, extra Z rings A; GpsB, guiding PBP1-shuttling protein B; PBP1, penicillin-binding protein 1;    poles42,60,95,97. In support of its role as a negative regulator,
 PBP2B, penicillin-binding protein 2B.
                                                                                                       a lack of EzrA lowers both the concentration of FtsZ that
                                                                                                       is required for Z ring assembly and the ability of MinC
                                 ZapA binding at a site proximal to the active site favours            and MinD overproduction to inhibit this process42,60,97. In
                                 an alternate conformation of GTP, rendering it less ame-              addition, twofold overproduction of EzrA in a sensitized
                                 nable to hydrolysis and thereby stabilizing the polymer 90.           genetic background is sufficient to efficiently block Z ring
                                 A second, not necessarily mutually exclusive model sug-               assembly 95. Taken together, these reports suggest that
                                 gests that the multimeric nature of ZapA might allow                  FtsZ assembly is stabilized in the absence of EzrA.
                                 it to cross-link individual protofilaments and, through                    Despite the apparent contradiction posed by the pres-
                                 this bridging activity, help to promote the assembly of               ence of a negative regulator of FtsZ at the divisome, it has
                                 FtsZ bundles by strengthening lateral interactions86,89.              been proposed that EzrA participates in maintaining the
                                 orthologues of ZapA are found throughout the Bacteria,                dynamic nature of the medial Z ring 40; indeed, several
                                 and its ability to promote the formation of stable bun-               studies point to an active role for EzrA at mid-cell60,97,98.
                                 dles of FtsZ has been shown in three different organ-                 Cells lacking EzrA are notably longer than wild-type cells,
                                 isms86,89,90. The possibility that ZapA acts positively in            which suggests that their cell division is delayed60,97,98. one
                                 Z ring formation is supported by its ability to antagonize            group has suggested that this delay results from the recruit-
                                 the inhibitory activity of MinC in vitro92,93.                        ment of other division proteins to additional polar Z rings,
                                                                                                       resulting in a dilution of the available pool of limiting divi-
                                 ZapB. ZapB is a small protein of 81 amino acids that was              sion proteins. In support of this model, they found that the
                                 first identified in E. coli and has an anti-parallel coiled-          overproduction of Ftsl could suppress the increased cell
                                 coil dimeric structure94. ZapB is recruited early to the              length phenotype98. However, another group found that
                                 assembling Z ring, and this process requires FtsZ but                 ezrA-null cells were longer than wild-type cells even under
                                 neither FtsA nor ZipA. Cells bearing a null mutation in               growth conditions that do not support the formation of


648 | SEPTEMBER 2009 | VoluME 7                                                                                                     www.nature.com/reviews/micro

                                                         © 2009 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                 polar Z rings, which led them to propose that the delay                  The defective septa pointed to a role for SepF in the
                                 in cell division results from the increased stability of the         period following Z ring completion, during the constric-
                                 medial Z ring 60,97. In support of this model, it was shown          tion period of cell division88. In support of this hypothesis,
                                 that the cell length phenotype, as well as the ability to over-      Z rings can assemble in the absence of SepF and EzrA and
                                 come the inhibition caused by MinC and MinD, correlated              can recruit the late-assembling division protein PBP2B
                                 with the extent to which EzrA could localize to the Z ring;          but cannot initiate septum synthesis88. This model also
                                 further support came from the finding that in the absence            explains the observed increase in cell length, as it predicts
                                 of EzrA, the turnover of FtsZ in the Z ring is reduced41,97.         a delay in cell division in cells that lack SepF, owing to
                                     Several groups have now established that there is a              the synthesis of thick septa88. However, sepF is also syn-
                                 direct interaction between EzrA and FtsZ in vitro using              thetically lethal with ftsA, which suggests that it might
                                 a wide range of techniques and, in support of bacte-                 act early during the assembly of the Z ring 85. Indeed,
                                 rial two-hybrid analyses, have found that EzrA can self-             Z ring assembly is abolished in cells lacking both proteins;
                                 interact 95,99,100. using both sedimentation analysis and            instead, FtsZ forms loose spiral structures in the inter-
                                 90°-angle light scattering, EzrA has been found to inhibit           nucleoid spaces of these cells85. In strong support for a
                                 FtsZ polymerization in a concentration-dependent man-                role in Z ring assembly, the overproduction of SepF in an
                                 ner, with a concurrent fourfold increase in the critical FtsZ        ftsA-null background can restore both normal Z ring for-
                                 concentration that is required for Z ring assembly 95,99,100.        mation and cell growth rate in a concentration-dependent
                                 The use of a fluorescently conjugated nucleotide analogue            manner 85.
                                 revealed that EzrA inhibits FtsZ polymerization at two                   SepF is conserved throughout the Gram-positive
                                 distinct levels99: first, at the monomeric level, by reducing        bacteria. In S. pneumoniae it forms part of the ftsAZ
                                 the affinity of FtsZ for GTP; and second, at the polymeric           operon85,88,103, and its absence causes gross morphological
                                 level, by increasing the rate of GTP hydrolysis, potentially         changes, including septal defects103. Perhaps more
                                 by stabilizing the transition state of the hydrolysis reaction,      importantly, SepF is essential in the cyanobacterium
                                 and thereby promoting the disassembly of FtsZ polymers.              Synechococcus elongatus, which does not have an identifi-
                                 Although only modest, the increased rate of hydrolysis               able homologue of FtsA104. The two disparate models dis-
                                 is supported by the kinetics of the reaction, which show             cussed above could be reconciled if we consider that the
                                 statistically significant increases in both Kcat and Km. In all      constriction of an incorrectly assembled Z ring leads to a
                                 cases, the in vitro data showed maximal activity of EzrA at          morphological abnormality of the septum (M. Gündoğdu,
                                 a 1/1 to 2/1 molar ratio of EzrA to FtsZ95,99,100. In addition, it   personal communication). SepF overproduction allows
                                 has recently been shown that EzrA modulates FtsZ assem-              essentially wild-type growth and division in the absence
                                 bly primarily by interacting with the conserved extreme              of FtsA, a fact that contrasts with the overlapping roles of
                                 C terminus of FtsZ100, and that the conserved patch respon-          ZipA and FtsA in E. coli (see previous section) and that
                                 sible for the mid-cell localization of EzrA is not necessary         has important implications for the role of FtsA in this
                                 for its inhibitory activity97. This implies that EzrA can            organism85. FtsA is not essential in B. subtilis 83,84, which
                                 either interact with FtsZ at more than one site or, more             suggests that it does not recruit any essential downstream
                                 probably, that the QNR patch mediates an interaction with            proteins and that a correctly formed Z ring is sufficient
                                 another core member of the divisome. one notable dis-                for their recruitment and for efficient division. Finally, as
                                 crepancy in the in vitro data is that only one study showed          FtsA probably has a structural role in tethering the Z ring
                                 that EzrA can disassemble preformed FtsZ polymers100.                to the inner membrane, these findings also predict that
                                     Recently, EzrA has been shown to have a third role: the          another division protein, such as EzrA or SepF itself, can
                                 coordination of the elongation–division cycle along with             efficiently support this function.
                                 a newly identified component of the divisome, guiding
                                 PBP1-shuttling protein B (GpsB)101,102. In this pathway,             ClpX. Recently, ClpX, the substrate recognition complex of
                                 EzrA was shown to be primarily involved in the recruit-              the ClpXP protease, was identified as a negative regulator
                                 ment of the bifunctional penicillin-binding protein 1                of Z ring assembly in B. subtilis, during a screen for sup-
                                 (PBP1) to the divisome and, in agreement with cell biol-             pressors of the temperature-sensitive ftsZ–GFP mutant105.
                                 ogy data, was found to interact with the transmembrane               Consistent with ClpX functioning as a negative regulator,
                                 domain of PBP1 (REF. 101).                                           a clpX-null mutation also suppresses the severe division
                                                                                                      defect that is associated with MinC and MinD overpro-
                                 SepF. Recently, two groups independently identified                  duction105,106. Moreover, despite producing only a modest
                                 SepF as a new component of the divisome in B. sub-                   increase in cell length in a wild-type strain, twofold over-
                                 tilis that is required for correct septal morphology 85,88.          production of ClpX is sufficient to completely block Z ring
                                 In vivo, SepF interacts both with itself and with FtsZ,              assembly in a temperature-sensitive ftsZ background, even
                                 and its recruitment to the Z ring is dependent on FtsZ               at the permissive temperature105. In vitro, ClpX specifically
90°-angle light scattering
A real-time assay that           but not on the later-assembling proteins85,88. In addi-              interacts with FtsZ and efficiently inhibits FtsZ polymeri-
measures the amount of light     tion, SepF can be isolated from cells in complex with                zation in a concentration-dependent manner, independ-
that is scattered by FtsZ        FtsZ, FtsA, EzrA and ZapA85. sepF-null mutants are                   ently of both its ATP-dependent chaperone activity and its
polymers. The increase in        viable and have only a mild defect in cell division85,88.            proteolytically active partner, ClpP105,106. This inhibition
signal is proportional to the
extent of Z ring assembly, and
                                 However, the septa of these cells are abnormally thick               is not accompanied by any change in the GTPase activ-
the method can also be used      and, in some cases, cell separation is initiated before              ity that is associated with FtsZ polymerization, which
to follow disassembly.           septal completion88.                                                 suggests that ClpX is active against FtsZ polymers105.


NATuRE REVIEwS | MiCrobiology                                                                                                VoluME 7 | SEPTEMBER 2009 | 649

                                                        © 2009 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                    Although the mechanism of inhibition remains unclear,                explains both the observed inhibition of FtsZ GTPase
                                    the wide conservation of ClpX throughout the Bacteria                activity 112,113,115 by SulA and the finding that FtsZ mutants
                                    supports the proposal that ClpX could be a general                   that are impaired in GTPase activity exhibit resistance
                                    regulator of Z ring assembly 105.                                    to SulA, with the degree of resistance correlating with
                                                                                                         the extent of their remaining GTPase activities45,115–117. The
                                    Responding to cell cycle status                                      key to understanding this finding came with the dis-
                                    UgtP. Consistent with previous data showing that cell                covery that the dynamic nature of FtsZ polymers that is
                                    division in B. subtilis is subject to growth rate-dependent          observed both in the cell and in vitro34,40,41 depends on
                                    regulation11, ugtP was recently identified by weart et al. as        GTP hydrolysis30. Therefore, when SulA binds to FtsZ
                                    a growth rate-dependent inhibitor of Z ring assembly 107.            monomers and removes them from the pool that is avail-
                                    ugtP is a terminal sugar transferase and uses uDP–glucose            able for exchange, ongoing nucleotide hydrolysis in the
                                    in the synthesis of the diglucosyl diacylglycerol anchor of          polymer promotes its rapid disassembly. This is consist-
                                    lipoteichoic acid. It acts as a growth rate-dependent inhibi-        ent with reports that SulA can disassemble FtsZ polymers
                                    tor of cell division during growth in nutrient-rich media,           both in the cell and in vitro. Similarly, mutants of FtsZ that
                                    but is downregulated under nutrient-limiting conditions.             are impaired in GTP hydrolysis but not GTP binding form
                                    ugtP associates reversibly with the divisome, and the                stable polymers, and so the rapid turnover of subunits
                                    transition between the divisome and sequestration into               does not occur, rendering them resistant to sequestration
                                    apparently random foci is controlled by the availabil-               by SulA42,45. Interestingly, SulA was found to exist as a
                                    ity of uDP–glucose, which is supplied by the upstream                dimer both in the crystal structure and in solution, which
                                    members of the pathway, uTP–glucose-1-phosphate                      suggests that it could cross-link FtsZ polymers and allow
                                    uridylyltransferase B (GtaB) and phosphoglucomutase A                substoichiometric levels of the protein to rapidly inhibit
                                    (PgcA). ugtP is present at approximately 2,400 molecules             Z ring formation113. However, a recent report found that in
                                    per cell and at a physiologically relevant molar ratio was           E. coli at least, and consistent with the proposed sequestra-
                                    shown to be a potent inhibitor of FtsZ assembly by dis-              tion model, SulA is effective only once it reaches a critical
                                    rupting the formation of the stabilizing lateral interactions        concentration35.
                                    between protofilaments107. The inhibitory activity of ugtP               SulA is not well conserved, but recently two proteins
                                    is partially dependent on the availability of uDP–glucose,           responsible for the SoS-induced division arrest in
                                    although the main role of substrate availability seems               B. subtilis and Corynebacterium glutamicum (YneA and
                                    likely to be in stimulating the re-localization of ugtP to           DivS, respectively) have been identified118,119. These pro-
                                    the divisome.                                                        teins are not homologous to SulA or to each other. The
                                        The authors proposed a model in which ugtP responds              assembly of the Z ring is inhibited on the production of
                                    to nutrient availability, as sensed by the supply of uDP–            DivS119, but YneA seems to act at a later stage in cell divi-
                                    glucose, to coordinate cell division with the growth rate.           sion, possibly by preventing the recruitment of Ftsl or
                                    They suggest that ugtP localizes to the Z ring to delay              DivIC98,118. The presence of a lysin motif (lysM) pepti-
                                    cell division until the cells reach a sufficient length to           doglycan-binding domain in YneA is intriguing, but its
                                    allow completion of the multi-fork DNA replication that              importance is not yet understood118.
                                    is typical of rapidly growing cells. A similar cell length
                                    phenotype in E. coli cells lacking the PgcA homologue,               MciZ. Recently, a 40-amino acid peptide, mother cell
                                    Pgm, suggests that this might be a conserved mechanism               inhibitor of FtsZ (MciZ), was identified as an inhibitor of
                                    for coordinating cell size and division throughout the               Z ring assembly that is produced during B. subtilis sporu-
                                    Bacteria107.                                                         lation120. Cells lacking MciZ exhibit a markedly increased
                                                                                                         frequency of Z ring formation during the later stages of
                                    SulA. SulA is a cell division inhibitor that is produced in          sporulation and, consistent with a role as a negative regu-
                                    response to DNA damage, as part of the SoS response,                 lator of FtsZ, artificial expression of mciZ during vegeta-
                                    in E. coli 108,109 and rapidly stalls cell division by both pre-     tive growth prevents Z ring assembly 120. In vitro, MciZ
                                    venting the assembly of nascent Z rings and facilitating             binds specifically to FtsZ and is a potent inhibitor of FtsZ
                                    the disassembly of existing Z rings, including those that            polymerization120. A large reduction in the GTPase activ-
                                    are actively constricting 110–112.                                   ity of FtsZ accompanies this inhibitory activity, suggest-
                                        SulA is a potent inhibitor of FtsZ polymerization                ing that MciZ acts at the level of the monomer to inhibit
                                    in vitro35,110,112–115, as it increases the critical concentration   the initial assembly of the FtsZ polymer. There is evidence
                                    that is necessary for assembly almost fivefold without               to support a model proposing that MciZ binds at a site
                                    affecting the cooperative nature of assembly 35. Recent              proximal to the nucleotide-binding pocket of FtsZ, so as
                                    structural studies revealed that SulA interacts with the             to occlude GTP binding and FtsZ polymerization. First,
                                    base of the C-terminal domain of FtsZ, completely mask-              an MciZ-resistant mutant of FtsZ that bears a substitu-
                                    ing the catalytic synergy loop113. SulA is therefore thought         tion close to the nucleotide-binding site has been isolated,
Lipoteichoic acid                   to act by binding to FtsZ monomers, sequestering them                identifying the probable peptide-binding site. Second, the
A class of teichoic acids that is   and thereby increasing the apparent critical concentra-              kinetics of GTPase activity in the presence of MciZ dis-
anchored in the cell                tion112,113. In support of this mode of action, when FtsZ            played a significant increase in Km, which is characteris-
membrane. Teichoic acids are
anionic polymers that are
                                    is in molar excess it efficiently assembles even in the              tic of competitive inhibition120. Interestingly, the mode
ubiquitous in the cell walls of     presence of SulA35, arguing against a filament-capping               of action of MciZ is similar to that recently proposed for
Gram-positive bacteria.             mechanism for SulA113,115. A sequestration model also                a chemical inhibitor of FtsZ assembly 121, reinforcing the


650 | SEPTEMBER 2009 | VoluME 7                                                                                                      www.nature.com/reviews/micro

                                                           © 2009 Macmillan Publishers Limited. All rights reserved
REVIEWS

                   Cytoplasmic species                                     Membrane-tethered species                         has been made in understanding how Z ring assembly
           SOS response         Sporulation                                             Rapid growth                         is both coordinated with the cell cycle and rendered
                                                                                                                             responsive to cellular and environmental stresses. As
                                                      FtsA, SepF             ZapA, ZapB
                      SulA     MciZ                    and ZipA               and SepF UgtP                                  we have seen, there is considerable overlap and redun-
                                                                                                                             dancy present in the regulation of the Z ring (FIG. 3) and,
                                                                                                                             as a result, it is likely that further, non-essential regulators
FtsZ                                                                                                                         remain to be discovered. Although, as we have described,
                                                                                                                             the regulation of the initial assembly and the stability
                    ClpX and EzrA                         EzrA                          EzrA                                 of the Z ring is now reasonably well understood, there is as
       Monomer                         Multimeric                      Growing                            Organized          yet no consensus on how the Z ring senses the completion
       pool                            species                         polymer                            polymer            of the divisome, initiates constriction and eventually dis-
Figure 3 | regulation of FtsZ assembly by accessory proteins. A large number of                                              assembles. Similarly, the emerging model of Z ring forma-
accessory proteins act in concert to regulate cell division at various levels of FtsZ assembly.                              tion following the collapse of a spiral intermediate might
Before division, FtsZ is thought to exist as a cytoplasmic population of Reviews | Microbiology
                                                                   Nature monomers and
                                                                                                                             also involve additional regulators or might simply be the
multimeric species of various lengths. Inhibitors of FtsZ assembly, such as SulA in Escherichia                              result of passive remodelling, driven by the relief of spatial
coli (Ec), which sequesters monomers, or mother cell inhibitor of FtsZ (MciZ) in Bacillus subtilis                           regulation. Although to date no true bacterial homologues
(Bs), which prevents GTP binding, act on the monomer pool to prevent Z ring assembly and                                     of MAPs have been found, as we advance our understand-
also facilitate Z ring disassembly owing to the rapid exchange of FtsZ subunits in the Z ring                                ing of division proteins, tentative functional parallels can
with those in the cytoplasm. The monomer pool that is available for exchange is maintained                                   be drawn and further characterization of these proteins
by both ClpX, which seems to prevent further assembly of FtsZ, and extra Z rings A (EzrABs),
                                                                                                                             might reveal additional relationships with the MAPs122.
which reduces the initial assembly of FtsZ and promotes its disassembly from the medial Z
ring. Following the relief of spatial regulation, the Z ring assembles at mid-cell and is tethered                               The recent in vitro reconstitution of contractile Z rings
to the inner face of the cytoplasmic membrane by FtsA or ZipAEc, which, along with ZapA, also                                in tubular liposomes as well as the assembly of a MinD–
contribute to the stability of the Z ring. Furthermore, early-assembling accessory proteins                                  MinE gradient on the surface of a membrane are prom-
such as ZapBEc and SepFBs also have potential roles in organizing the ultrastructure of the Z                                ising steps forward47,123 that offer the potential to study
ring; SepF seems to make a substantial contribution to this process in Bacillus subtilis and                                 the regulation of Z rings using in vitro systems, rather
might itself act as a membrane tether. During rapid growth, further ‘fine tuning’ of the Z ring is                           than simply studying the effects of individual proteins
carried out by UgtPBs, which delays Z ring completion until cells reach an appropriate size.                                 on the bulk polymer. Although this will undoubtedly be
                                                                                                                             technically challenging, we must remain cautious when
                                         notion that an understanding of the cellular mechanisms                             drawing conclusions on the basis of the effects of single
                                         that regulate FtsZ assembly is likely to inform future work                         accessory proteins on FtsZ polymers. In the cell, we also
                                         in developing clinically relevant inhibitors of bacterial                           need to take into account the effects of membrane bind-
                                         cytokinesis.                                                                        ing, the presence of numerous other regulatory proteins
                                                                                                                             at the Z ring and the crowded nature of the cytoplasmic
                                         Future perspectives                                                                 environment. The advances that we anticipate making in
                                         The past few years have seen the discovery of many addi-                            understanding Z ring assembly and its regulation prom-
                                         tional factors that are involved in Z ring formation and                            ise to make this an enlightening and exciting field in the
                                         modulating polymer dynamics. Moreover, much progress                                coming years.

1.     Errington, J., Daniel, R. A. & Scheffers, D. J. Cytokinesis          and ZipA during the Escherichia coli cell cycle.               20. de Boer, P., Crossley, R. & Rothfield, L. The essential
       in bacteria. Microbiol. Mol. Biol. Rev. 67, 52–65 (2003).            J. Bacteriol. 185, 3344–3351 (2003).                               bacterial cell-division protein FtsZ is a GTPase. Nature
2.     Goehring, N. W. & Beckwith, J. Diverse paths to midcell:      11.    Weart, R. B. & Levin, P. A. Growth rate-dependent                  359, 254–256 (1992).
       assembly of the bacterial cell division machinery. Curr.             regulation of medial FtsZ ring formation. J. Bacteriol.        21. RayChaudhuri, D. & Park, J. T. Escherichia coli cell-
       Biol. 15, R514–R526 (2005).                                          185, 2826–2834 (2003).                                             division gene ftsZ encodes a novel GTP-binding protein.
3.     Harry, E., Monahan, L. & Thompson, L. Bacterial cell          12.    Romberg, L. & Levin, P. A. Assembly dynamics of the                Nature 359, 251–254 (1992).
       division: the mechanism and its precison. Int. Rev. Cytol.           bacterial cell division protein FtsZ: poised at the edge of    22. Mukherjee, A., Dai, K. & Lutkenhaus, J. Escherichia coli
       253, 27–94 (2006).                                                   stability. Annu. Rev. Microbiol. 57, 125–154 (2003).               cell division protein FtsZ is a guanine nucleotide binding
4.     Bi, E. F. & Lutkenhaus, J. FtsZ ring structure associated     13.    Löwe, J. & Amos, L. A. Crystal structure of the bacterial          protein. Proc. Natl Acad. Sci. USA 90, 1053–1057
       with division in Escherichia coli. Nature 354, 161–164               cell-division protein FtsZ. Nature 391, 203–206                    (1993).
       (1991).                                                              (1998).                                                        23. Scheffers, D. J., de Wit, J. G., den Blaauwen, T. &
5.     Addinall, S. G. & Lutkenhaus, J. FtsZ-spirals and -arcs       14.    Nogales, E., Wolf, S. G. & Downing, K. H. Structure of the         Driessen, A. J. Substitution of a conserved aspartate
       determine the shape of the invaginating septa in some                αβ tubulin dimer by electron crystallography. Nature               allows cation-induced polymerization of FtsZ. FEBS Lett.
       mutants of Escherichia coli. Mol. Microbiol. 22,                     391, 199–203 (1998).                                               494, 34–37 (2001).
       231–237 (1996).                                               15.    Erickson, H. P. FtsZ, a prokaryotic homolog of tubulin?        24. Scheffers, D. J., de Wit, J. G., den Blaauwen, T. &
6.     Margolin, W. FtsZ and the division of prokaryotic cells              Cell 80, 367–370 (1995).                                           Driessen, A. J. GTP hydrolysis of cell division protein FtsZ:
       and organelles. Nature Rev. Mol. Cell Biol. 6,                16.    Mukherjee, A. & Lutkenhaus, J. Guanine nucleotide-                 evidence that the active site is formed by the association
       862–871 (2005).                                                      dependent assembly of FtsZ into filaments. J. Bacteriol.           of monomers. Biochemistry 41, 521–529 (2002).
7.     Haydon, D. J. et al. An inhibitor of FtsZ with potent and            176, 2754–2758 (1994).                                         25. Romberg, L., Simon, M. & Erickson, H. P. Polymerization
       selective anti-staphylococcal activity. Science 321,          17.    Erickson, H. P., Taylor, D. W., Taylor, K. A. & Bramhill, D.       of Ftsz, a bacterial homolog of tubulin: is assembly
       1673–1675 (2008).                                                    Bacterial cell division protein FtsZ assembles into                cooperative? J. Biol. Chem. 276, 11743–11753 (2001).
       This paper describes the validation of FtsZ as a                     protofilament sheets and minirings, structural homologs        26. Caplan, M. R. & Erickson, H. P. Apparent cooperative
       clinically relevant target for new antibiotics.                      of tubulin polymers. Proc. Natl Acad. Sci. USA 93,                 assembly of the bacterial cell division protein FtsZ
8.     Kelly, A. J., Sackett, M. J., Din, N., Quardokus, E. &               519–523 (1996).                                                    demonstrated by isothermal titration calorimetry. J. Biol.
       Brun, Y. V. Cell cycle-dependent transcriptional and          18.    Löwe, J. & Amos, L. A. Tubulin-like protofilaments in              Chem. 278, 13784–13788 (2003).
       proteolytic regulation of FtsZ in Caulobacter. Genes Dev.            Ca2+-induced FtsZ sheets. EMBO J. 18, 2364–2371                27. Chen, Y., Bjornson, K., Redick, S. D. & Erickson, H. P. A
       12, 880–893 (1998).                                                  (1999).                                                            rapid fluorescence assay for FtsZ assembly indicates
9.     Quardokus, E., Din, N. & Brun, Y. V. Cell cycle regulation    19.    Oliva, M. A., Cordell, S. C. & Löwe, J. Structural insights        cooperative assembly with a dimer nucleus. Biophys. J.
       and cell type-specific localization of the FtsZ division             into FtsZ protofilament formation. Nature Struct. Mol.             88, 505–514 (2005).
       initiation protein in Caulobacter. Proc. Natl Acad. Sci.             Biol. 11, 1243–1250 (2004).                                    28. Wang, X. & Lutkenhaus, J. The FtsZ protein of Bacillus
       USA 93, 6314–6319 (1996).                                            The work described in references 18 and 19 provides                subtilis is localized at the division site and has GTPase
10.    Rueda, S., Vicente, M. & Mingorance, J. Concentration                evidence that the protofilaments of FtsZ and tubulin               activity that is dependent upon FtsZ concentration. Mol.
       and assembly of the division ring proteins FtsZ, FtsA,               make similar longitudinal contacts.                                Microbiol. 9, 435–442 (1993).




NATuRE REVIEwS | MiCrobiology                                                                                                                             VoluME 7 | SEPTEMBER 2009 | 651

                                                                     © 2009 Macmillan Publishers Limited. All rights reserved
Bacterial cell division-_assembly_maintenance_and_disassembly_of_the_z_ring
Bacterial cell division-_assembly_maintenance_and_disassembly_of_the_z_ring

Weitere ähnliche Inhalte

Was ist angesagt?

Cells, prokaryotes and eukaryotes
Cells, prokaryotes and eukaryotesCells, prokaryotes and eukaryotes
Cells, prokaryotes and eukaryoteslhedward
 
DNA repair and recombination
DNA repair and recombinationDNA repair and recombination
DNA repair and recombinationKAUSHAL SAHU
 
Bronchopulmonary segments and cell cycle
Bronchopulmonary segments and cell cycleBronchopulmonary segments and cell cycle
Bronchopulmonary segments and cell cyclechdx
 
Replicacion y reparacion del dna modifi
Replicacion y reparacion del dna modifiReplicacion y reparacion del dna modifi
Replicacion y reparacion del dna modifiSayra Medellín
 
Introtocells 111109074946-phpapp01
Introtocells 111109074946-phpapp01Introtocells 111109074946-phpapp01
Introtocells 111109074946-phpapp01joy000 renojo
 
Natural transformation
Natural transformation Natural transformation
Natural transformation Lauren Welker
 
Types of dna recombination
Types of dna recombinationTypes of dna recombination
Types of dna recombinationAimanNisar4
 
Cell structures and functions
Cell structures and functionsCell structures and functions
Cell structures and functionsJohnJoseph Gabata
 
BIO - Chapter 2-5 - Cells, Movement of Substances, Nutrients and Enzymes
BIO - Chapter 2-5 - Cells, Movement of Substances, Nutrients and EnzymesBIO - Chapter 2-5 - Cells, Movement of Substances, Nutrients and Enzymes
BIO - Chapter 2-5 - Cells, Movement of Substances, Nutrients and EnzymesSofian Muhd
 
Unit 3 recombination
Unit 3 recombinationUnit 3 recombination
Unit 3 recombinationDhanya G
 
12 cellcycle text
12  cellcycle text12  cellcycle text
12 cellcycle text1slid
 
Chapter 12: Cell Cycle
Chapter 12: Cell CycleChapter 12: Cell Cycle
Chapter 12: Cell CycleAngel Vega
 
Apoptosis in Bacteria - Programmed cell death in prokaryotic cells
Apoptosis in Bacteria - Programmed cell death in prokaryotic cellsApoptosis in Bacteria - Programmed cell death in prokaryotic cells
Apoptosis in Bacteria - Programmed cell death in prokaryotic cellsRivkah Epstein
 
Celldiscoveryianjones
CelldiscoveryianjonesCelldiscoveryianjones
Celldiscoveryianjonesijones78
 

Was ist angesagt? (19)

Cells, prokaryotes and eukaryotes
Cells, prokaryotes and eukaryotesCells, prokaryotes and eukaryotes
Cells, prokaryotes and eukaryotes
 
Nucleus report
Nucleus reportNucleus report
Nucleus report
 
DNA repair and recombination
DNA repair and recombinationDNA repair and recombination
DNA repair and recombination
 
Bronchopulmonary segments and cell cycle
Bronchopulmonary segments and cell cycleBronchopulmonary segments and cell cycle
Bronchopulmonary segments and cell cycle
 
Replicacion y reparacion del dna modifi
Replicacion y reparacion del dna modifiReplicacion y reparacion del dna modifi
Replicacion y reparacion del dna modifi
 
Introtocells 111109074946-phpapp01
Introtocells 111109074946-phpapp01Introtocells 111109074946-phpapp01
Introtocells 111109074946-phpapp01
 
Natural transformation
Natural transformation Natural transformation
Natural transformation
 
Types of dna recombination
Types of dna recombinationTypes of dna recombination
Types of dna recombination
 
Organelles
OrganellesOrganelles
Organelles
 
Organelles
OrganellesOrganelles
Organelles
 
Cell structures and functions
Cell structures and functionsCell structures and functions
Cell structures and functions
 
BIO - Chapter 2-5 - Cells, Movement of Substances, Nutrients and Enzymes
BIO - Chapter 2-5 - Cells, Movement of Substances, Nutrients and EnzymesBIO - Chapter 2-5 - Cells, Movement of Substances, Nutrients and Enzymes
BIO - Chapter 2-5 - Cells, Movement of Substances, Nutrients and Enzymes
 
Types of Recombinase
Types of Recombinase Types of Recombinase
Types of Recombinase
 
Unit 3 recombination
Unit 3 recombinationUnit 3 recombination
Unit 3 recombination
 
12 cellcycle text
12  cellcycle text12  cellcycle text
12 cellcycle text
 
Chapter 12: Cell Cycle
Chapter 12: Cell CycleChapter 12: Cell Cycle
Chapter 12: Cell Cycle
 
Apoptosis in Bacteria - Programmed cell death in prokaryotic cells
Apoptosis in Bacteria - Programmed cell death in prokaryotic cellsApoptosis in Bacteria - Programmed cell death in prokaryotic cells
Apoptosis in Bacteria - Programmed cell death in prokaryotic cells
 
Ocular Biology
Ocular BiologyOcular Biology
Ocular Biology
 
Celldiscoveryianjones
CelldiscoveryianjonesCelldiscoveryianjones
Celldiscoveryianjones
 

Ähnlich wie Bacterial cell division-_assembly_maintenance_and_disassembly_of_the_z_ring

bacterial cell division
bacterial cell divisionbacterial cell division
bacterial cell divisionSHUBHANGI2509
 
Eukaryotic Cell Functions
Eukaryotic Cell FunctionsEukaryotic Cell Functions
Eukaryotic Cell FunctionsKimberly Thomas
 
Describe The Relationship Between Organelles And Eukaryotes
Describe The Relationship Between Organelles And EukaryotesDescribe The Relationship Between Organelles And Eukaryotes
Describe The Relationship Between Organelles And EukaryotesPaula Smith
 
Presentation3 - Microbio
Presentation3 - MicrobioPresentation3 - Microbio
Presentation3 - Microbiohiimtia
 
Microbial Growth.pptx...............................
Microbial Growth.pptx...............................Microbial Growth.pptx...............................
Microbial Growth.pptx...............................Shraddha Jaiswal
 
Cell cycle and mitosis
Cell cycle and mitosisCell cycle and mitosis
Cell cycle and mitosisJosh Lamberte
 
Dinamica membrana 3decadas_cellmbr-vereb2003
Dinamica membrana 3decadas_cellmbr-vereb2003Dinamica membrana 3decadas_cellmbr-vereb2003
Dinamica membrana 3decadas_cellmbr-vereb2003Tamara Jorquiera
 
Plant cell expansion: scaling the wall
Plant cell expansion: scaling the wall Plant cell expansion: scaling the wall
Plant cell expansion: scaling the wall Frédéric NICOL, phD
 
Bacterial Cell Differentiation
Bacterial Cell DifferentiationBacterial Cell Differentiation
Bacterial Cell DifferentiationN Poorin
 

Ähnlich wie Bacterial cell division-_assembly_maintenance_and_disassembly_of_the_z_ring (20)

bacterial cell division
bacterial cell divisionbacterial cell division
bacterial cell division
 
Bacterial cell division
Bacterial cell divisionBacterial cell division
Bacterial cell division
 
Sprint Final 09 B
Sprint Final 09 BSprint Final 09 B
Sprint Final 09 B
 
Chapt 04
Chapt 04Chapt 04
Chapt 04
 
Eukaryotic Cell Functions
Eukaryotic Cell FunctionsEukaryotic Cell Functions
Eukaryotic Cell Functions
 
Describe The Relationship Between Organelles And Eukaryotes
Describe The Relationship Between Organelles And EukaryotesDescribe The Relationship Between Organelles And Eukaryotes
Describe The Relationship Between Organelles And Eukaryotes
 
Presentation3 - Microbio
Presentation3 - MicrobioPresentation3 - Microbio
Presentation3 - Microbio
 
Chapt 04
Chapt 04Chapt 04
Chapt 04
 
Microbial Growth.pptx...............................
Microbial Growth.pptx...............................Microbial Growth.pptx...............................
Microbial Growth.pptx...............................
 
Cell cycle and mitosis
Cell cycle and mitosisCell cycle and mitosis
Cell cycle and mitosis
 
Lindo mitosis 140309070845-phpapp01
Lindo mitosis 140309070845-phpapp01Lindo mitosis 140309070845-phpapp01
Lindo mitosis 140309070845-phpapp01
 
Centrioles
CentriolesCentrioles
Centrioles
 
Centrioles[1]
Centrioles[1]Centrioles[1]
Centrioles[1]
 
Abt seminor
Abt seminorAbt seminor
Abt seminor
 
Cytoskeleton
CytoskeletonCytoskeleton
Cytoskeleton
 
Dinamica membrana 3decadas_cellmbr-vereb2003
Dinamica membrana 3decadas_cellmbr-vereb2003Dinamica membrana 3decadas_cellmbr-vereb2003
Dinamica membrana 3decadas_cellmbr-vereb2003
 
Plant cell expansion: scaling the wall
Plant cell expansion: scaling the wall Plant cell expansion: scaling the wall
Plant cell expansion: scaling the wall
 
Bacterial Cell Differentiation
Bacterial Cell DifferentiationBacterial Cell Differentiation
Bacterial Cell Differentiation
 
Cell cycle
Cell cycleCell cycle
Cell cycle
 
Cell biology.
Cell biology.Cell biology.
Cell biology.
 

Kürzlich hochgeladen

WordPress Websites for Engineers: Elevate Your Brand
WordPress Websites for Engineers: Elevate Your BrandWordPress Websites for Engineers: Elevate Your Brand
WordPress Websites for Engineers: Elevate Your Brandgvaughan
 
Scanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL CertsScanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL CertsRizwan Syed
 
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024BookNet Canada
 
Nell’iperspazio con Rocket: il Framework Web di Rust!
Nell’iperspazio con Rocket: il Framework Web di Rust!Nell’iperspazio con Rocket: il Framework Web di Rust!
Nell’iperspazio con Rocket: il Framework Web di Rust!Commit University
 
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)Mark Simos
 
Search Engine Optimization SEO PDF for 2024.pdf
Search Engine Optimization SEO PDF for 2024.pdfSearch Engine Optimization SEO PDF for 2024.pdf
Search Engine Optimization SEO PDF for 2024.pdfRankYa
 
TeamStation AI System Report LATAM IT Salaries 2024
TeamStation AI System Report LATAM IT Salaries 2024TeamStation AI System Report LATAM IT Salaries 2024
TeamStation AI System Report LATAM IT Salaries 2024Lonnie McRorey
 
TrustArc Webinar - How to Build Consumer Trust Through Data Privacy
TrustArc Webinar - How to Build Consumer Trust Through Data PrivacyTrustArc Webinar - How to Build Consumer Trust Through Data Privacy
TrustArc Webinar - How to Build Consumer Trust Through Data PrivacyTrustArc
 
CloudStudio User manual (basic edition):
CloudStudio User manual (basic edition):CloudStudio User manual (basic edition):
CloudStudio User manual (basic edition):comworks
 
Merck Moving Beyond Passwords: FIDO Paris Seminar.pptx
Merck Moving Beyond Passwords: FIDO Paris Seminar.pptxMerck Moving Beyond Passwords: FIDO Paris Seminar.pptx
Merck Moving Beyond Passwords: FIDO Paris Seminar.pptxLoriGlavin3
 
"Subclassing and Composition – A Pythonic Tour of Trade-Offs", Hynek Schlawack
"Subclassing and Composition – A Pythonic Tour of Trade-Offs", Hynek Schlawack"Subclassing and Composition – A Pythonic Tour of Trade-Offs", Hynek Schlawack
"Subclassing and Composition – A Pythonic Tour of Trade-Offs", Hynek SchlawackFwdays
 
Leverage Zilliz Serverless - Up to 50X Saving for Your Vector Storage Cost
Leverage Zilliz Serverless - Up to 50X Saving for Your Vector Storage CostLeverage Zilliz Serverless - Up to 50X Saving for Your Vector Storage Cost
Leverage Zilliz Serverless - Up to 50X Saving for Your Vector Storage CostZilliz
 
Anypoint Exchange: It’s Not Just a Repo!
Anypoint Exchange: It’s Not Just a Repo!Anypoint Exchange: It’s Not Just a Repo!
Anypoint Exchange: It’s Not Just a Repo!Manik S Magar
 
What's New in Teams Calling, Meetings and Devices March 2024
What's New in Teams Calling, Meetings and Devices March 2024What's New in Teams Calling, Meetings and Devices March 2024
What's New in Teams Calling, Meetings and Devices March 2024Stephanie Beckett
 
"ML in Production",Oleksandr Bagan
"ML in Production",Oleksandr Bagan"ML in Production",Oleksandr Bagan
"ML in Production",Oleksandr BaganFwdays
 
DevoxxFR 2024 Reproducible Builds with Apache Maven
DevoxxFR 2024 Reproducible Builds with Apache MavenDevoxxFR 2024 Reproducible Builds with Apache Maven
DevoxxFR 2024 Reproducible Builds with Apache MavenHervé Boutemy
 
Commit 2024 - Secret Management made easy
Commit 2024 - Secret Management made easyCommit 2024 - Secret Management made easy
Commit 2024 - Secret Management made easyAlfredo García Lavilla
 
Streamlining Python Development: A Guide to a Modern Project Setup
Streamlining Python Development: A Guide to a Modern Project SetupStreamlining Python Development: A Guide to a Modern Project Setup
Streamlining Python Development: A Guide to a Modern Project SetupFlorian Wilhelm
 
"Debugging python applications inside k8s environment", Andrii Soldatenko
"Debugging python applications inside k8s environment", Andrii Soldatenko"Debugging python applications inside k8s environment", Andrii Soldatenko
"Debugging python applications inside k8s environment", Andrii SoldatenkoFwdays
 

Kürzlich hochgeladen (20)

WordPress Websites for Engineers: Elevate Your Brand
WordPress Websites for Engineers: Elevate Your BrandWordPress Websites for Engineers: Elevate Your Brand
WordPress Websites for Engineers: Elevate Your Brand
 
Scanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL CertsScanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL Certs
 
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
 
E-Vehicle_Hacking_by_Parul Sharma_null_owasp.pptx
E-Vehicle_Hacking_by_Parul Sharma_null_owasp.pptxE-Vehicle_Hacking_by_Parul Sharma_null_owasp.pptx
E-Vehicle_Hacking_by_Parul Sharma_null_owasp.pptx
 
Nell’iperspazio con Rocket: il Framework Web di Rust!
Nell’iperspazio con Rocket: il Framework Web di Rust!Nell’iperspazio con Rocket: il Framework Web di Rust!
Nell’iperspazio con Rocket: il Framework Web di Rust!
 
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
 
Search Engine Optimization SEO PDF for 2024.pdf
Search Engine Optimization SEO PDF for 2024.pdfSearch Engine Optimization SEO PDF for 2024.pdf
Search Engine Optimization SEO PDF for 2024.pdf
 
TeamStation AI System Report LATAM IT Salaries 2024
TeamStation AI System Report LATAM IT Salaries 2024TeamStation AI System Report LATAM IT Salaries 2024
TeamStation AI System Report LATAM IT Salaries 2024
 
TrustArc Webinar - How to Build Consumer Trust Through Data Privacy
TrustArc Webinar - How to Build Consumer Trust Through Data PrivacyTrustArc Webinar - How to Build Consumer Trust Through Data Privacy
TrustArc Webinar - How to Build Consumer Trust Through Data Privacy
 
CloudStudio User manual (basic edition):
CloudStudio User manual (basic edition):CloudStudio User manual (basic edition):
CloudStudio User manual (basic edition):
 
Merck Moving Beyond Passwords: FIDO Paris Seminar.pptx
Merck Moving Beyond Passwords: FIDO Paris Seminar.pptxMerck Moving Beyond Passwords: FIDO Paris Seminar.pptx
Merck Moving Beyond Passwords: FIDO Paris Seminar.pptx
 
"Subclassing and Composition – A Pythonic Tour of Trade-Offs", Hynek Schlawack
"Subclassing and Composition – A Pythonic Tour of Trade-Offs", Hynek Schlawack"Subclassing and Composition – A Pythonic Tour of Trade-Offs", Hynek Schlawack
"Subclassing and Composition – A Pythonic Tour of Trade-Offs", Hynek Schlawack
 
Leverage Zilliz Serverless - Up to 50X Saving for Your Vector Storage Cost
Leverage Zilliz Serverless - Up to 50X Saving for Your Vector Storage CostLeverage Zilliz Serverless - Up to 50X Saving for Your Vector Storage Cost
Leverage Zilliz Serverless - Up to 50X Saving for Your Vector Storage Cost
 
Anypoint Exchange: It’s Not Just a Repo!
Anypoint Exchange: It’s Not Just a Repo!Anypoint Exchange: It’s Not Just a Repo!
Anypoint Exchange: It’s Not Just a Repo!
 
What's New in Teams Calling, Meetings and Devices March 2024
What's New in Teams Calling, Meetings and Devices March 2024What's New in Teams Calling, Meetings and Devices March 2024
What's New in Teams Calling, Meetings and Devices March 2024
 
"ML in Production",Oleksandr Bagan
"ML in Production",Oleksandr Bagan"ML in Production",Oleksandr Bagan
"ML in Production",Oleksandr Bagan
 
DevoxxFR 2024 Reproducible Builds with Apache Maven
DevoxxFR 2024 Reproducible Builds with Apache MavenDevoxxFR 2024 Reproducible Builds with Apache Maven
DevoxxFR 2024 Reproducible Builds with Apache Maven
 
Commit 2024 - Secret Management made easy
Commit 2024 - Secret Management made easyCommit 2024 - Secret Management made easy
Commit 2024 - Secret Management made easy
 
Streamlining Python Development: A Guide to a Modern Project Setup
Streamlining Python Development: A Guide to a Modern Project SetupStreamlining Python Development: A Guide to a Modern Project Setup
Streamlining Python Development: A Guide to a Modern Project Setup
 
"Debugging python applications inside k8s environment", Andrii Soldatenko
"Debugging python applications inside k8s environment", Andrii Soldatenko"Debugging python applications inside k8s environment", Andrii Soldatenko
"Debugging python applications inside k8s environment", Andrii Soldatenko
 

Bacterial cell division-_assembly_maintenance_and_disassembly_of_the_z_ring

  • 1. REVIEWS Bacterial cell division: assembly, maintenance and disassembly of the Z ring David W. Adams and Jeff Errington Abstract | Bacterial cell division is orchestrated by a tubulin homologue, FtsZ, which polymerizes to form a ring-like structure that is both a scaffold for the assembly of the bacterial cytokinetic machinery and, at least in part, a source of the energy for constriction. FtsZ assembly is tightly regulated, and a diverse repertoire of accessory proteins contributes to the formation of a functional division machine that is responsive to cell cycle status and environmental stress. In this Review, we describe the interaction of these proteins with FtsZ and discuss recent advances in our understanding of Z ring assembly. Cytokinesis in bacteria is achieved by a macromolecular past decade (TABLE 1). Many of these proteins have roles machine called the divisome1–3. Divisome assembly is ini- that are similar to those of the microtubule-associated tiated by polymerization of the tubulin homologue FtsZ proteins (MAPs) of the Eukarya and affect the assem- into a ring-like structure that lies close to the cytoplasmic bly, organization and stability of the Z ring, although membrane at the prospective division site4. In rod-shaped none seems to be a true MAP homologue. Some of cells such as Escherichia coli and Bacillus subtilis, the divi- these proteins are essential for survival but many are sion site usually lies at the midpoint of the rod, perpen- not, and there is considerable functional overlap in the dicular to the long axis of the cell (FIG. 1a). Constriction group. How then do these proteins ensure the correct of the ring gives rise to two equal daughter cells, each of assembly of FtsZ into a Z ring that is both competent which is half the length of the parent cell. This ‘Z ring’ for cell division and responsive to cell cycle status and serves as a scaffold for the recruitment of the downstream environmental stresses? This Review discusses the pro- components of the divisome and it persists throughout teins that interact with FtsZ during the assembly of the division, guiding the synthesis, location and shape of Z ring and highlights the recent progress in our under- the division septum5. FtsZ is almost universally conserved standing of how these proteins intervene in the mecha- throughout the Bacteria (BOX 1); it is also present in the nism of cell division and its regulation once the Z ring Euryarchaeota and has an active role in the division of has been established. the plastids and mitochondria of several groups of the Eukarya6. FtsZ was recently shown to be an important new FtsZ polymerization and the nature of the Z ring target for antibiotics, as specific inhibitors of FtsZ were FtsZ is a cytoplasmic protein that is composed of two globu- found to protect against a lethal dose of Staphylococcus lar domains separated by a central core helix (FIG. 2a). Despite aureus in a mouse model of infection7. the low level of sequence similarity, the tertiary structure Although the lifestyle and developmental behaviour of FtsZ is remarkably similar to that of tubulin, supporting of some bacteria (for example, Caulobacter crescentus) the theory that these proteins have common ances- Centre for Bacterial Cell dictates tight regulation of both FtsZ synthesis and try 13–15. Furthermore, FtsZ polymerizes into tubulin-like Biology, Institute for Cell and stability 8,9, in the two best studied bacteria, E. coli protofilaments by the head-to-tail association of indi- Molecular Biosciences, Newcastle University, and B. subtilis, no significant changes seem to occur in vidual subunits16–19 (FIG. 2b). FtsZ, like tubulin, is a self- Medical School, Framlington the concentration of FtsZ, either throughout the cell activating GTPase20–22: the insertion of the synergy loop Place, Newcastle-upon-Tyne, cycle or under different growth conditions10,11. In these from the base of one subunit into the nucleotide-binding NE2 4HH, UK. bacteria, cell division must therefore be regulated at site of the subunit below places the catalytic residues close Correspondence to J.E. e-mail: jeff.errington@ the level of FtsZ assembly 12 (BOX 2; FIG. 1b). Indeed, the to the γ-phosphate, thereby allowing nucleotide hydro- newcastle.ac.uk number and diversity of proteins that are known to lysis to proceed19,23. The GTPase activity of FtsZ therefore doi:10.1038/nrmicro2198 interact directly with FtsZ has grown steadily over the depends on FtsZ polymerization24. 642 | SEPTEMBER 2009 | VoluME 7 www.nature.com/reviews/micro © 2009 Macmillan Publishers Limited. All rights reserved
  • 2. REVIEWS a Escherichia coli Nucleoid occlusion Bacillus subtilis Nucleoid MinC gradient Z ring b Min NO Min Min NO Min Min NO NO Min Min NO NO Min Inhibitor-free zone Figure 1 | Cell division in rod-shaped bacteria. a | Two different modes of division. After chromosome replication and segregation into nucleoids the Z ring assembles at mid-cell. The ring then constricts to bring about division. Cell wall Nature Reviews | Microbiology synthesis follows the ring inwards. In Escherichia coli, synthesis of the division septum is accompanied by constriction of the outer membrane. In Bacillus subtilis, a cross wall of peptidoglycan initially divides the cell before it is degraded and remodelled to form the new, hemi-spherical cell poles. b | The spatial regulation of Z ring assembly. Nucleoid occlusion (NO), which is mediated by Noc (in B. subtilis) or SlmA (in E. coli), inhibits Z ring assembly close to the nucleoid. The Min system acts to prevent Z ring assembly at the cell poles (for simplicity, the Min dynamics in E. coli have been omitted). From left to right: in ‘newborn’ cells, both systems initially inhibit Z ring assembly throughout the cell; following cell elongation and chromosome replication, NO maintains inhibition in the cylindrical part of the cell; and finally, the progression of chromosome segregation Cooperative assembly reveals an inhibitor-free region at mid-cell, allowing the Z ring to assemble. Assembly that is characterized by the increased affinity of individual subunits for the In vitro, FtsZ displays cooperative assembly25–30 into a remodelling throughout its existence, with rapid turnover growing polymer rather than for each other. Polymerization range of polymeric forms, from single-stranded protofila- dynamics (in the order of 8 seconds) that depend on displays a sigmoidal ments to rings, bundles, tubules and sheets, depending the intrinsic GTPase activity of the FtsZ polymer 10,40,41. relationship with concentration on the experimental conditions used16,17,31,32. However, the Indeed, GTP hydrolysis promotes the disassembly of the and has a defined critical architecture of the Z ring that is formed in vivo is not well protofilament and drives the highly dynamic nature of concentration, below which no significant assembly occurs. understood, and recent cryo-electron microscopy tomography the FtsZ polymers that are observed both in the cell and of C. crescentus cells suggests that it might consist of a large in vitro, as well as limiting the length of individual proto- Cryo-electron microscopy number of short, overlapping protofilaments, rather than filaments30,34,40,41. Accordingly, FtsZ mutations that impair tomography forming a continuous ring33. In vitro, FtsZ exhibits at least GTP hydrolysis but not GTP binding stabilize polymers A technique that allows the two important transitions during its assembly 34. The ini- by reducing subunit turnover 41,42. visualization of biological molecules in a near-native tial assembly of single-stranded protofilaments occurs at In addition to a regulatory function, nucleotide hydrol- state. Unfixed samples are a critical concentration of around 0.5–1 μM28,30,34,35, but ysis has been proposed to have a force-generating role flash frozen, held at cryogenic there is a further transition at around 3 μM, which might during septation, whereby a nucleotide-dependent tran- temperature and visualized by represent the assembly of higher-order structures with sition from a straight, GTP-bound polymer to a curved, transmission electron microscopy. Tomography uses extensive lateral interactions between the protofilaments34. GDP-bound polymer can transmit mechanical work to a series of tilted images from Puzzlingly, the FtsZ protofilament is only one subunit the membrane33,43. However, the physiological role of this which a three-dimensional thick25,36, so the origin of this cooperativity remains force remains controversial and is complicated by the image of the sample can be unclear. Recently, modelling has suggested that a mono- fact that mutants that are severely impaired in nucleotide deduced. mer isomerization step could be the source of this behav- hydrolysis can still divide44,45. Moreover, recent work sug- Fluorescence recovery after iour 35–37. Importantly, the intracellular concentration of gests that the relationship between the morphology of photobleaching FtsZ greatly exceeds that of the critical concentration polymers assembled in vitro and GTP hydrolysis is not as A live-cell imaging technique for assembly, at 3–10 μM10,38. straightforward as was initially thought 46. Nevertheless, used to monitor the FtsZ polymerization depends on GTP binding but the recent demonstration that Z rings can impart a force intracellular dynamics of fluorescently tagged molecules. not hydrolysis16. The combination of a partially solvent, on a tubular membrane in vitro47, combined with the A target region is irreversibly accessible nucleotide-binding site and the abundance observation of both straight and curved filaments in vivo33, photobleached by a laser and of GTP in the cytoplasm means that GTP hydrolysis is reinforces the possibility of force generation by the Z ring. then monitored for recovery of probably a rate-limiting step in the polymerization cycle In addition, mathematical modelling of Z ring dynam- fluorescence. Dynamics are and, in contrast to tubulin, FtsZ polymers might contain ics has shown that force generation could be achieved expressed in terms of the half-time for recovery (the time mainly GTP19,29,39. Fluorescence recovery after photobleaching independently of FtsZ GTPase activity, with hydrolysis taken to reach half of the (FRAP), as well as other studies, has revealed that the instead being required for subunit turnover and polymer original fluorescence intensity). Z ring is not a static structure: it undergoes constant remodelling 35,48. NATuRE REVIEwS | MiCrobiology VoluME 7 | SEPTEMBER 2009 | 643 © 2009 Macmillan Publishers Limited. All rights reserved
  • 3. REVIEWS been found in the Bacteria. Nonetheless, the similarity Box 1 | FtsZ-independent division of the polymerization dynamics of FtsZ and tubulin has afforded valuable insights into how the unique regula- tory proteins that interact with FtsZ might modulate its polymerization. First, proteins can prevent the ini- tial assembly of FtsZ by preventing nucleotide binding, sequestering individual monomers or capping growing polymers. Second, proteins that promote polymeriza- tion or stabilize existing polymers could do so by con- The wide conservation of FtsZ throughout the Bacteria andNature Reviews | Microbiology the euryarchaeotal branch centrating FtsZ at the membrane, cross-linking separate of the Archaea, which is supported by the ever-increasing wealth of genome sequence protofilaments or facilitating the formation of stabilizing information, has cemented its position as an essential cell division protein. There are, lateral interactions, as well as by preventing the disas- however, several intriguing examples of species in which FtsZ is absent, has been sembly of the polymer by inhibiting nucleotide hydrol- replaced or is simply no longer required for cell division. ysis. Finally, proteins that disassemble or destabilize Missing FtsZ the polymer might do so by directly counteracting the Several distinct groups of bacteria, including Chlamydiae and Planctomycetes, as well mechanisms used by positive-acting factors — that is, by as Ureaplasma urealyticum and Mycoplasma mobile, are unusual among bacteria in that stimulating GTPase activity, facilitating disassembly they lack a recognizable homologue of FtsZ52,124,125. Although it has been suggested by sequestering monomers or inhibiting the formation that Chlamydia spp., which are obligate intracellular parasites, might draw on a host of lateral interactions. cell division machinery126, there is compelling evidence to suggest that they divide independently of their hosts and might instead use the subset of division genes that they contain to synthesize a peptidoglycan division septum127. However, it is not clear Assembling the Z ring how this site is chosen or how any putative synthetic machinery is assembled in the A crucial element of Z ring assembly is the association absence of FtsZ. Planctomycetes and U. urealyticum are free-living organisms and so between FtsZ and the cell membrane. FtsZ does not must be capable of independent cell division, but to date little progress has been made seem to have any direct affinity for the membrane, but in understanding how this might be accomplished. all models for Z ring formation require its attachment to replacing FtsZ the membrane, both to maintain its structural integrity Substantial progress has been made in understanding the division of the during septation and probably to transmit any constric- Crenarchaeota, which completely lack FtsZ, owing to the discovery of a new cell tive force to the membrane1,33,51,53. In E. coli there is good division machine128,129. The products of three cell cycle-regulated genes, cdvA, cdvB evidence that two proteins, FtsA and ZipA, collaborate in and cdvC, constitute a protein complex that forms a contractile-ring structure anchoring FtsZ to the membrane54. Both of these proteins between segregated nucleoids in dividing cells that is essential for cell division. are essential for cell division54,55: depletion of either single Although CdvA is unique to the Crenarchaeota, CdvB and CdvC are homologous to protein results in the formation of elongated filaments proteins of the endosomal sorting complex required for transport (ESCRT) III complex with regularly assembled Z rings that are positioned of the Eukarya, which is involved in endosomal protein sorting, viral budding and membrane abscission during cytokinesis. These proteins are found in all but one order correctly between segregated nucleoids but are non- of the Crenarchaeota and are absent from the FtsZ-dependent Euryarchaeota. functional for division. when both proteins are removed, Z rings do not form54,56. life before FtsZ Recently, the L-form (wall-less derivatives of common bacteria) of Bacillus subtilis was found to propagate independently of FtsZ by a new mode of division that is defined by a ZipA. ZipA is a bitopic protein that is composed of three remarkable extrusion–resolution process (see the figure, which shows phase-contrast domains: a transmembrane anchor, a long, basic arm images of division in L-form cells of B. subtilis) that discharges viable progeny130. This that is rich in proline and glutamine residues and a might be related to the surprising finding that ftsZ can be deleted in Streptomyces large, globular C-terminal domain 55,57,58. ZipA has spp.131. It is possible that these sick mutants use a similar extrusion–resolution process to a particularly rare membrane topology, as its sole trans- propagate. Although the mechanisms underlying extrusion–resolution remain unclear, membrane anchor is at the amino terminus and the the process could provide a glimpse into the distant past, to a time before the C-terminal domain is in the cytoplasm55,59. This unusual FtsZ-based division machinery had evolved. topology is shared by the division protein extra Z rings Scale bar represents 3mm. Images courtesy of P. Domínguez-Cuevas, Newcastle University, UK. A (EzrA) in B. subtilis 1,60. However, these two proteins seem to have considerably different roles in cell division, so the potential importance of their shared membrane Although the carboxy-terminal domain of FtsZ is topology is unclear. similar in its fold and acidic nature to that of tubulin, it In the cell, ZipA is dispersed throughout the inner lacks the extended surface loops that are thought to aid membrane until the initiation of cell division, when it is the assembly of the microtubule and its interaction with recruited to the assembling Z ring by a direct interaction motor proteins49. However, FtsZ contains a unique, highly with FtsZ55,56,61. The division block that occurs in cells conserved peptide (~9 amino acids) at its extreme C ter- that are depleted of ZipA seems to be due to the failure minus; this site is thought to be unstructured and is dis- of the Z ring to recruit FtsK, a protein that is essential for pensable for polymerization but essential for cell division, the recruitment of downstream division proteins54,62. ZipA as it is the binding site for several division proteins50–52 interacts directly with the conserved C-terminal tail of Bitopic protein (discussed below). FtsZ57–59,61,63. This interaction is mediated by the C-terminal A protein that contains a single transmembrane segment MAPs can alter the stability, bundling and disassembly domain of ZipA, which is both necessary and sufficient between the cytoplasmic and of the tubulin polymer in response to cellular cues but, for its recruitment to the Z ring59. Interestingly, the trans- extracytoplasmic domains. as mentioned above, to date no true homologues have membrane domain does not seem to be simply an inert 644 | SEPTEMBER 2009 | VoluME 7 www.nature.com/reviews/micro © 2009 Macmillan Publishers Limited. All rights reserved
  • 4. REVIEWS Box 2 | Spatial regulation of cell division FtsA show that ZipA can function as a membrane tether for FtsZ51,54–56. The flexibility of the unstructured linker In both Bacillus subtilis and Escherichia coli, assembly of the Z ring — and thus the domain between the transmembrane anchor and the initiation of cell division — is regulated by two distinct but partially overlapping C-terminal domain is likely to be pivotal in this tethering systems (FIG. 1b). These systems have been reviewed in detail elsewhere but are briefly role, allowing the C-terminal domain to reach out from the described here132,133. First, nucleoid occlusion prevents the assembly of the Z ring over membrane and bind the C-terminus of FtsZ51,65. the nucleoid by the action of two unrelated DNA binding proteins, Noc in B. subtilis and SlmA in E. coli134,135. Although SlmA interacts directly with FtsZ in vitro, the exact ZipA is not widely conserved outside the Gamma- mechanism by which either protein inhibits the formation of the Z ring remains proteobacteria, which suggests that its role has either been unclear135. Second, the Min system contributes to spatial regulation by inhibiting Z ring replaced or become redundant55. In support of the sec- assembly at the cell poles, preventing the generation of anucleate mini-cells. This is ond suggestion, a single amino acid substitution in FtsA achieved by creating a symmetrical concentration gradient of MinC and MinD, renders ZipA non-essential in E. coli, allowing efficient negative regulators of Z ring assembly, around the mid-cell position so that the growth and division in its absence66 (discussed below). time-averaged concentration is always highest at the cell poles. MinC interacts directly with FtsZ and is thought to prevent Z ring assembly at several levels by inhibiting FtsA. FtsA is widely conserved throughout the Bacteria lateral interactions and weakening longitudinal bonds, thereby compromising the and is often found upstream of ftsZ, in an operon within integrity of the assembled polymer92,93,110,136,137. In addition, in E. coli MinC has been the dcw cluster 67. The crystal structure of FtsA from shown to act early during the assembly of the Z ring, competing with FtsA for binding at the extreme carboxy-terminal tail of FtsZ138. The removal of FtsA further destabilizes Thermotoga maritima showed that it is structurally related the assembling Z ring and concentrates the inhibitor in direct contact with its target138. to actin and consists of two domains, each of which can be In E. coli topological specificity is imparted by the periodic oscillation of MinC and MinD further divided into two subdomains68. In one structure, from pole to pole, a process that is driven by MinE139–141. In B. subtilis, two unrelated proteins, the nucleotide-binding site, which is formed by an inter- DivIVA and MinJ, are thought to recruit MinC and MinD to division sites and retain it at the domain cleft, was found to contain both ATP and a Mg 2+ new cell poles142–147. Recruitment to the nascent division site allows MinC to prevent the ion68, supporting reports that FtsA can bind ATP69–71. re-initiation of cell division and to ensure that only one division event occurs per cell cycle. Interestingly, FtsA purified from Streptococcus pneumoniae can bind various nucleotide phosphates in a manner that is directly proportional to the length of the phosphate anchor, suggesting that it has a specific function in divi- moiety; ATP and GTP are favoured over ADP and GDP, sion, perhaps by interacting with one or more of the many which are favoured over AMP and GMP71. In B. subtilis, membrane-bound components of the divisome59 (BOX 3). FtsA has been shown to exist as a dimer in solution70, Structural studies have revealed that the C-terminal and various two-hybrid assays have shown that FtsA can domain of ZipA consists of a six-stranded anti-parallel interact with itself 72–76. Furthermore, FtsA from S. pneu- β-sheet laid against three α-helices, with a core domain moniae has been reported to reversibly polymerize into containing a split β–α–β fold that is similar to the folds corkscrew-like helices that are composed of pairs of paired found in many RNA-binding proteins57,58. The solvent- protofilaments71. These polymers exhibited nucleotide- exposed face of this motif forms a shallow hydrophobic dependent stability, forming extremely stable polymers cleft that extends across the domain57,58. The final 20 amino in the presence of ATP and less stable polymers in the acids of the C-terminal domain of FtsZ are sufficient for the presence of ADP71. This report suggests a role for nucle- interaction with ZipA, and a co-crystal structure of otide hydrolysis in the regulation of FtsA polymerization; the complex has been obtained57, which shows that the however, ATPase activity was not found in S. pneumo- hydrophobic cleft of ZipA is occupied by the C-terminal niae FtsA and has so far only been reported in FtsA from fragment of FtsZ57. This binding is driven by the burial of B. subtilis70. Although the role of nucleotide binding is not exposed hydrophobic residues, and most of the binding yet known, it is intriguing that mutation of residues that affinity is provided by only a few key residues, which are have been shown to contact the nucleotide in the crystal highly conserved57,58. structure abolishes the ability of FtsA to interact both with In vitro, the C terminus of ZipA promotes the assembly itself and with FtsZ72. Structural modelling suggests that of FtsZ into thick bundles and sheets of protofilaments FtsA dimerizes in a head-to-tail fashion, with the indi- that are aligned laterally 59,64. Consistent with this positive vidual subunits rotated 180° to one another; however, the role in assembly and in the formation of stabilizing lateral importance of this model remains unclear 77. interactions, twofold overproduction of ZipA suppresses In E. coli, FtsA interacts with the conserved extreme the division defect of cells bearing the thermosensitive C terminus of FtsZ51,54,56. In both E. coli and B. subtilis the ftsZ84 mutation at the non-permissive temperature64. ratio of FtsZ to FtsA is approximately 5 to 1 (REFs 10,70), Although the mechanism by which ZipA organizes and maintenance of this balance is crucial for cell divi- FtsZ protofilaments is not known, it is clear that the indi- sion78. This is exemplified by the fact that the toxic vidual domains of ZipA have separable functions in vivo, overproduction of either protein can be counteracted ftsZ84 and several models describing the activity and physiologi- by overproduction of the other 78. The ability of FtsA to sup- A mutation that results in the substitution of glycine to serine cal role of ZipA have been proposed51,55,57,59,64. The globular port the initial assembly of the Z ring without ZipA, as well at position 105 in C-terminal domain might facilitate bundle formation either as the ability of an ftsA gain-of-function mutant to bypass Escherichia coli FtsZ. This by a chaperone-like activity, through which it stabilizes the the need for ZipA, suggested that FtsA can also tether the Z substitution reduces both the unstructured C-terminal tail, or by simply shielding ring to the inner membrane56,66. Indeed, the re-examination GTP binding and the GTPase activity of FtsZ in vitro and the solvent-exposed hydrophobic residues57. In addition, the of a highly conserved motif at the extreme C terminus of renders the protein essential nature of the transmembrane domain and FtsA revealed a membrane-targeting amphipathic helix temperature sensitive in vivo. the ability of the Z ring to assemble in the absence of that is separated from the core protein by a flexible linker NATuRE REVIEwS | MiCrobiology VoluME 7 | SEPTEMBER 2009 | 645 © 2009 Macmillan Publishers Limited. All rights reserved
  • 5. REVIEWS Table 1 | FtsZ and the proteins that modulate its polymerization Protein role Size* Structural characterization In vitro data Essential? Conservation (kDa) available? Assembly of the Z ring FtsZ A structural subunit of the Z ring that is 40.2 A self-assembling GTPase that is Yes Yes‡ Widely conserved required for initiation of cell division and structurally related to tubulin serves as a scaffold for divisome assembly FtsA A principal membrane tether that 48 An ATP-binding protein that is No§ Yes|| Widely conserved; is required for Z ring assembly, structurally related to actin but often found in an organization of FtsZ polymers and that has the uniquely positioned operon with ftsZ recruitment of downstream proteins subdomain 1C ZipA Promotes Z ring assembly and is a 36.3 A large globular domain that is Yes Yes Gammaproteobacteria secondary membrane anchor linked to a single amino-terminal TM domain by an extended linker Regulators of Z ring dynamics ZapA A positive modulator of Z ring assembly 9.7 Forms a dimer through its extensive Yes No Widely conserved and stability coiled-coil region ZapB Has a redundant role in ensuring proper 9.5 A dimer consisting of a single No§ No A subset of Z ring assembly coiled-coil region; self-assembles Gammaproteobacteria into large polymer bundles SepF Has an overlapping role with FtsA in Z 17 NA No No¶ Gram-positive bacteria ring assembly and is required for proper and Cyanobacteria septal morphology EzrA A negative regulator of Z ring assembly 64.8 Predicted to contain four coiled-coil Yes No Gram-positive bacteria throughout the cell membrane that regions in its carboxyl terminus and with a low GC content contributes to mid-cell Z ring dynamics a single N-terminal TM anchor and has a role in coordinating cell elongation with division Cell cycle-responsive regulators UgtP A growth rate-dependent inhibitor of 43.4 A membrane-associated protein Yes No A widely conserved cell division that self-assembles into tightly metabolic pathway wound spirals in vitro SulA A cell division inhibitor of the SOS 18.7 Forms a dimer, either face of which Yes No Gammaproteobacteria response that prevents the assembly can interact with the T7 loop of FtsZ of new Z rings and facilitates the disassembly of existing Z rings MciZ Contributes to the inhibition of Z ring 4.8 A small peptide that is Yes No Bacillus spp. assembly following the initiation of predicted to bind close to the sporulation nucleotide-binding pocket of FtsZ General regulators ClpX Helps to maintain the cytoplasmic pool 46.2 A hexameric substrate-recognition Yes No Widely conserved of free FtsZ subunits complex of ClpXP protease *The stated protein sizes are for Bacillus subtilis or, where appropriate, Escherichia coli and are calculated values, published at the SubtiList or Colibri World-Wide Web Servers. ‡This protein is not essential for viability in Streptomyces coelicolor. §In vitro data are only available on the proteins themselves, not on their potential to modulate FtsZ polymerization. ||This is essential in E. coli but not for vegetative cell division in B. subtilis. ¶This protein is essential in Synechococcus elongatus. EzrA, extra Z rings A; MciZ, mother cell inhibitor of FtsZ; NA, no data available; TM, transmembrane. region67. This amphipathic helix is essential for the function characterized, little was known about the nature of the of FtsA under normal laboratory conditions: deletion of binding interface in FtsA until recently50. A screen for this region renders FtsA non-functional and causes ftsA mutations that cause the resultant protein to interact it to assemble into deleterious cytoplasmic rods67. with the membrane but not with FtsZ identified impor- Importantly, and in contrast to ZipA59, the membrane tant FtsA residues, including three charged residues that targeting region of FtsA can be replaced by various are strictly conserved and are clustered mainly on the sur- unrelated membrane-targeting determinants67,79. face of the 2B subdomain72. Substitutions of these resi- FtsA differs from actin in that it is missing the 1B dues do not affect the ability of FtsA to self-interact, but domain and it has an unrelated subdomain called 1C68. they do abolish its ability to interact with FtsZ72. The 2B Deletion of 1C renders FtsA unable to interact with itself, subdomain is likely to have additional functions in cell although it retains the ability to localize to the Z ring 73. The division, as deletion of a small anti-parallel β-sheet at the inability of FtsA lacking the 1C domain to allow the pro- apex of this domain seems to affect the correct placement gression of cell division has been explained by the finding of the Z ring73. Furthermore, the ftsA* mutation is found that the 1C domain is involved in recruiting downstream in the same region66 (discussed below). components of the divisome to the Z ring 73,80. Aside from its role in Z ring assembly, FtsA is now Although the role of the extreme C terminus of known to affect the integrity of the Z ring at various stages FtsZ in the interaction with FtsA has been extensively of divisome assembly 67,72,74,81. The ftsA* gain-of-function 646 | SEPTEMBER 2009 | VoluME 7 www.nature.com/reviews/micro © 2009 Macmillan Publishers Limited. All rights reserved
  • 6. REVIEWS a Nucleotide-binding site b Z ring assembly and polymer dynamics Many mainly non-essential proteins are thought to H7 directly interact with FtsZ to regulate the dynamics of the Z ring that is assembled at mid-cell. Although individually these proteins are not required for Z ring assembly, their various roles in modulating the polymerization of FtsZ mean that the combined loss of certain regulatory proteins often produces a synthetic lethal division phenotype. ZapA. ZapA is a small protein of 85 amino acids that was C-terminal identified in a genome-wide screen for proteins that, domain when overexpressed, can overcome the lethality that is induced by MinD overexpression in B. subtilis86. In both B. subtilis and E. coli, ZapA is recruited to the divisome N-terminal early, probably by a direct interaction with FtsZ, where domain it enhances both the assembly and the stability of the Z ring86. Cells lacking ZapA exhibit no phenotype in either organism86,87 but, similar to other non-essential regulatory proteins, this loss is conditionally lethal in various sensi- T7 loop tized backgrounds60,85,88, including in cells with artificially Figure 2 | FtsZ polymerization. a | The crystal structure of a Bacillus subtilis FtsZ lowered FtsZ levels86. Surprisingly, given its apparently monomer bound to GTP-γS (Protein Data Bank entry 2RHO). FtsZ is composed of two positive role in Z ring assembly, null mutations in zapA Nature Reviews | Microbiology independently folding domains and a central core helix, H7. The amino-terminal domain are synthetically lethal when combined with null muta- contains the tubulin signature motif, GGGTGTG, and forms the nucleotide-binding site. tions in ezrA86. This synergism probably results from the At the base of the carboxy-terminal domain, following H7, is the catalytic T7 or synergy assembly of a less dynamic Z ring in the absence of EzrA loop. The extreme C-terminal tail, which forms the binding site for several division (discussed below), which reduces the cytoplasmic pool of proteins, is not visible in the crystal structure. b | FtsZ polymerizes by the head-to-tail subunits that are available for exchange. This, combined association of individual subunits, shown in blue and green, to form a single-stranded with the increased concentration of FtsZ that is required protofilament with a longitudinal subunit repeat that is similar to that of tubulin. for assembly in the absence of ZapA, leads to the synthetic Insertion of the T7 loop into the nucleotide-binding site (orange) of the subunit below places two highly conserved aspartate residues in the vicinity of the FtsZ γ-phosphate; lethal phenotype86. these aspartate residues are thought to activate the GTPase activity of FtsZ by The crystal structure of ZapA revealed an anti-parallel polarizing an attacking water molecule. tetramer that is composed of a pair of dimers which asso- ciate through extensive coiled-coil interactions along the C termini of the individual subunits89. Dimerization of mutation bypasses the need for ZipA and partially sup- ZapA is mediated mainly by contacts between the small, presses the requirement for FtsK66. Cells bearing only globular N-terminal domains89. In solution, oligomeri- ftsA* assemble Z rings earlier in the cell cycle and can zation is constrained by concentration and the protein tolerate higher levels of MinC than wild-type cells, indi- exists in a dimer–tetramer equilibrium. However, taking cating that both the initial assembly and the stability of into account the predicted abundance of ZapA in vivo, the Z ring are enhanced by FtsA* (REF. 82). FtsA* seems the dimer is more likely to be the physiologically relevant to confer enhanced stability on the Z ring at the level of species86,89,90. initial assembly, as it affects neither the intrinsic dynamics In vitro, ZapA specifically interacts with FtsZ and, of FtsZ subunits nor the proportion of FtsZ present in the consistent with its positive role in polymerization in vivo, ring 82. In support of this model, the interaction of FtsZ enhances both the initial polymerization of FtsZ and the with FtsA* is much stronger than the interaction with stability of the resulting polymers86,89,90. ZapA does not wild-type FtsA82. In addition, a further gain-of-function form higher-order structures under any conditions exam- mutation in FtsA that seems to act by stabilizing the Z ring ined to date86,89,90. FtsZ polymers that have assembled in has recently been reported81. the presence of equimolar amounts of ZapA form higher- The assertion that FtsA probably constitutes the princi- order structures, such as branched networks that are pal membrane anchor for the Z ring is supported by several composed of thick protofilament bundles86,89,90, which lines of evidence67,72 and most notably by the absence of rec- are similar to those observed in the presence of artifi- ognizable homologues of ZipA in most bacterial lineages55. cial bundling agents such as diethylaminoethyl dextran The situation in B. subtilis, however, is somewhat different. and Ca2+ (REF. 91). The enhancement of polymerization and, FtsA is not essential for vegetative cell division, although in particular, polymer stability is directly correlated with the cells are highly filamentous and the frequency of func- the inhibition of FtsZ GTPase activity 86,90. Intriguingly, tional Z ring formation is severely reduced83–85. This is con- one report identified a potential change in the confor- sistent with the proposed role of FtsA in promoting Z ring mational state of the bound nucleotide, using the new integrity84. However, the ability of the Z ring to form, even technique of linear dichroism to monitor changes in the Synthetic lethal at low frequency, is puzzling and suggests that another FtsZ polymer 90,91. The authors suggested that this con- Lethality due to a combination protein might also have a similar role to FtsA, albeit at a formational change might be the result of a general activ- of two non-lethal mutations. reduced level of efficiency (discussed below). ity of bundling agents and proposed a model in which NATuRE REVIEwS | MiCrobiology VoluME 7 | SEPTEMBER 2009 | 647 © 2009 Macmillan Publishers Limited. All rights reserved
  • 7. REVIEWS zapB are viable, although slightly elongated, and tend to Box 3 | Assembly of the divisome form filamentous cells that are blocked in cell division. In a b the absence of ZapB, Z ring formation is both reduced in PBP1 PBP2B frequency and altered in morphology, and short, tight- DivIC FtsL DivIB pitched helices are frequently formed. In vitro, ZapB EzrA EzrA dimers can interact through their termini to form large bundles. As ZapB is abundant (approximately 13,000 mol- ecules per cell), ZapB polymers might bridge shorter FtsZ GpsB FtsA FtsA FtsW protofilaments in the assembling Z ring to promote their organization into a functional ring. However, the con- ZapA ZapA tribution of ZapB to Z ring assembly seems to be subtle, SepF FtsZ SepF FtsZ as mutation of zapB generates a striking division pheno- SepF SepF type only in bacteria carrying the ftsZ84 mutation, not in bacteria with a range of other cell division mutations, Following the relief of spatial regulation as the cell grows and its chromosomes segregate, including of zapA. The interaction of ZapB with FtsZ or Nature Reviews | Microbiology the Z ring forms at mid-cell, tethered to the inner face of the cytoplasmic membrane. It is its potential to modulate FtsZ polymerization in vitro has not clear whether there are early intermediates in ring formation, but dynamic helical not yet been reported. polymers of FtsZ have been described, which suggests that the Z ring forms by the collapse of a spiral intermediate148–151. Once assembled, the Z ring begins the process of recruiting the downstream components of the divisome. EzrA. EzrA was identified in B. subtilis as a negative In Bacillus subtilis, assembly of the divisome occurs by a two-step mechanism, in which regulator of Z ring assembly in a screen for extragenic the early-assembling proteins (see the figure, part a) are directly recruited to the Z ring suppressors of a temperature-sensitive ftsZ mutant by FtsZ and, after a substantial delay, are followed by the concerted and interdependent tagged with green fluorescent protein (ftsZ-GFP)60, but assembly of the later-assembling proteins (see the figure, part b)1,152. In Escherichia coli, it probably has multiple functions in division. EzrA is classical localization studies had suggested an essentially linear pathway of divisome an integral membrane protein and is predicted to have an assembly153. However, recent work has shown that this process probably occurs by the N-terminal transmembrane anchor and a cytoplas- sequential assembly of at least three subcomplexes2,154. Similarly to B. subtilis, a time mic C-terminal domain that consists of four coiled-coil delay between the assembly of the Z ring and the recruitment of the later-assembling regions60,95. This domain structure is conserved through- proteins has been detected155. Following the completion of divisome assembly and, possibly, an as-yet-undiscovered out the low-GC-content Gram-positive bacteria in which signal, the Z ring constricts ahead of the leading edge of the newly synthesized division it is present 95. EzrA is constitutively expressed and is septum. Finally, the daughter cells separate, and this involves the action of specialized abundant, with an estimated 10,000–20,000 molecules per peptidoglycan hydrolases. The overlap between invagination and cell separation varies cell95,96. It is distributed throughout the plasma membrane between organisms: in B. subtilis a complete cross wall is usually formed before cell of elongating cells and then recruited to the Z ring by a separation begins, whereas in E. coli the two processes occur almost simultaneously, recently identified patch of seven amino acids, known as leading to a characteristic furrow at the active division site (FIG. 1a). These distinctions the QNR patch, that follows the last coiled-coil region in the probably relate to the differing cell envelope structures of Gram-positive and C-terminal region60,97. ezrA mutants have an increased Gram-negative bacteria but might also reflect flexibility in the relative timing of septal frequency of Z ring formation and additional Z rings are wall hydrolysis, which is tightly coupled to septum synthesis in E. coli but is much more often observed at one or both cell poles, which suggests separable in B. subtilis. a Min-like function in preventing Z ring assembly at cell EzrA, extra Z rings A; GpsB, guiding PBP1-shuttling protein B; PBP1, penicillin-binding protein 1; poles42,60,95,97. In support of its role as a negative regulator, PBP2B, penicillin-binding protein 2B. a lack of EzrA lowers both the concentration of FtsZ that is required for Z ring assembly and the ability of MinC ZapA binding at a site proximal to the active site favours and MinD overproduction to inhibit this process42,60,97. In an alternate conformation of GTP, rendering it less ame- addition, twofold overproduction of EzrA in a sensitized nable to hydrolysis and thereby stabilizing the polymer 90. genetic background is sufficient to efficiently block Z ring A second, not necessarily mutually exclusive model sug- assembly 95. Taken together, these reports suggest that gests that the multimeric nature of ZapA might allow FtsZ assembly is stabilized in the absence of EzrA. it to cross-link individual protofilaments and, through Despite the apparent contradiction posed by the pres- this bridging activity, help to promote the assembly of ence of a negative regulator of FtsZ at the divisome, it has FtsZ bundles by strengthening lateral interactions86,89. been proposed that EzrA participates in maintaining the orthologues of ZapA are found throughout the Bacteria, dynamic nature of the medial Z ring 40; indeed, several and its ability to promote the formation of stable bun- studies point to an active role for EzrA at mid-cell60,97,98. dles of FtsZ has been shown in three different organ- Cells lacking EzrA are notably longer than wild-type cells, isms86,89,90. The possibility that ZapA acts positively in which suggests that their cell division is delayed60,97,98. one Z ring formation is supported by its ability to antagonize group has suggested that this delay results from the recruit- the inhibitory activity of MinC in vitro92,93. ment of other division proteins to additional polar Z rings, resulting in a dilution of the available pool of limiting divi- ZapB. ZapB is a small protein of 81 amino acids that was sion proteins. In support of this model, they found that the first identified in E. coli and has an anti-parallel coiled- overproduction of Ftsl could suppress the increased cell coil dimeric structure94. ZapB is recruited early to the length phenotype98. However, another group found that assembling Z ring, and this process requires FtsZ but ezrA-null cells were longer than wild-type cells even under neither FtsA nor ZipA. Cells bearing a null mutation in growth conditions that do not support the formation of 648 | SEPTEMBER 2009 | VoluME 7 www.nature.com/reviews/micro © 2009 Macmillan Publishers Limited. All rights reserved
  • 8. REVIEWS polar Z rings, which led them to propose that the delay The defective septa pointed to a role for SepF in the in cell division results from the increased stability of the period following Z ring completion, during the constric- medial Z ring 60,97. In support of this model, it was shown tion period of cell division88. In support of this hypothesis, that the cell length phenotype, as well as the ability to over- Z rings can assemble in the absence of SepF and EzrA and come the inhibition caused by MinC and MinD, correlated can recruit the late-assembling division protein PBP2B with the extent to which EzrA could localize to the Z ring; but cannot initiate septum synthesis88. This model also further support came from the finding that in the absence explains the observed increase in cell length, as it predicts of EzrA, the turnover of FtsZ in the Z ring is reduced41,97. a delay in cell division in cells that lack SepF, owing to Several groups have now established that there is a the synthesis of thick septa88. However, sepF is also syn- direct interaction between EzrA and FtsZ in vitro using thetically lethal with ftsA, which suggests that it might a wide range of techniques and, in support of bacte- act early during the assembly of the Z ring 85. Indeed, rial two-hybrid analyses, have found that EzrA can self- Z ring assembly is abolished in cells lacking both proteins; interact 95,99,100. using both sedimentation analysis and instead, FtsZ forms loose spiral structures in the inter- 90°-angle light scattering, EzrA has been found to inhibit nucleoid spaces of these cells85. In strong support for a FtsZ polymerization in a concentration-dependent man- role in Z ring assembly, the overproduction of SepF in an ner, with a concurrent fourfold increase in the critical FtsZ ftsA-null background can restore both normal Z ring for- concentration that is required for Z ring assembly 95,99,100. mation and cell growth rate in a concentration-dependent The use of a fluorescently conjugated nucleotide analogue manner 85. revealed that EzrA inhibits FtsZ polymerization at two SepF is conserved throughout the Gram-positive distinct levels99: first, at the monomeric level, by reducing bacteria. In S. pneumoniae it forms part of the ftsAZ the affinity of FtsZ for GTP; and second, at the polymeric operon85,88,103, and its absence causes gross morphological level, by increasing the rate of GTP hydrolysis, potentially changes, including septal defects103. Perhaps more by stabilizing the transition state of the hydrolysis reaction, importantly, SepF is essential in the cyanobacterium and thereby promoting the disassembly of FtsZ polymers. Synechococcus elongatus, which does not have an identifi- Although only modest, the increased rate of hydrolysis able homologue of FtsA104. The two disparate models dis- is supported by the kinetics of the reaction, which show cussed above could be reconciled if we consider that the statistically significant increases in both Kcat and Km. In all constriction of an incorrectly assembled Z ring leads to a cases, the in vitro data showed maximal activity of EzrA at morphological abnormality of the septum (M. Gündoğdu, a 1/1 to 2/1 molar ratio of EzrA to FtsZ95,99,100. In addition, it personal communication). SepF overproduction allows has recently been shown that EzrA modulates FtsZ assem- essentially wild-type growth and division in the absence bly primarily by interacting with the conserved extreme of FtsA, a fact that contrasts with the overlapping roles of C terminus of FtsZ100, and that the conserved patch respon- ZipA and FtsA in E. coli (see previous section) and that sible for the mid-cell localization of EzrA is not necessary has important implications for the role of FtsA in this for its inhibitory activity97. This implies that EzrA can organism85. FtsA is not essential in B. subtilis 83,84, which either interact with FtsZ at more than one site or, more suggests that it does not recruit any essential downstream probably, that the QNR patch mediates an interaction with proteins and that a correctly formed Z ring is sufficient another core member of the divisome. one notable dis- for their recruitment and for efficient division. Finally, as crepancy in the in vitro data is that only one study showed FtsA probably has a structural role in tethering the Z ring that EzrA can disassemble preformed FtsZ polymers100. to the inner membrane, these findings also predict that Recently, EzrA has been shown to have a third role: the another division protein, such as EzrA or SepF itself, can coordination of the elongation–division cycle along with efficiently support this function. a newly identified component of the divisome, guiding PBP1-shuttling protein B (GpsB)101,102. In this pathway, ClpX. Recently, ClpX, the substrate recognition complex of EzrA was shown to be primarily involved in the recruit- the ClpXP protease, was identified as a negative regulator ment of the bifunctional penicillin-binding protein 1 of Z ring assembly in B. subtilis, during a screen for sup- (PBP1) to the divisome and, in agreement with cell biol- pressors of the temperature-sensitive ftsZ–GFP mutant105. ogy data, was found to interact with the transmembrane Consistent with ClpX functioning as a negative regulator, domain of PBP1 (REF. 101). a clpX-null mutation also suppresses the severe division defect that is associated with MinC and MinD overpro- SepF. Recently, two groups independently identified duction105,106. Moreover, despite producing only a modest SepF as a new component of the divisome in B. sub- increase in cell length in a wild-type strain, twofold over- tilis that is required for correct septal morphology 85,88. production of ClpX is sufficient to completely block Z ring In vivo, SepF interacts both with itself and with FtsZ, assembly in a temperature-sensitive ftsZ background, even and its recruitment to the Z ring is dependent on FtsZ at the permissive temperature105. In vitro, ClpX specifically 90°-angle light scattering A real-time assay that but not on the later-assembling proteins85,88. In addi- interacts with FtsZ and efficiently inhibits FtsZ polymeri- measures the amount of light tion, SepF can be isolated from cells in complex with zation in a concentration-dependent manner, independ- that is scattered by FtsZ FtsZ, FtsA, EzrA and ZapA85. sepF-null mutants are ently of both its ATP-dependent chaperone activity and its polymers. The increase in viable and have only a mild defect in cell division85,88. proteolytically active partner, ClpP105,106. This inhibition signal is proportional to the extent of Z ring assembly, and However, the septa of these cells are abnormally thick is not accompanied by any change in the GTPase activ- the method can also be used and, in some cases, cell separation is initiated before ity that is associated with FtsZ polymerization, which to follow disassembly. septal completion88. suggests that ClpX is active against FtsZ polymers105. NATuRE REVIEwS | MiCrobiology VoluME 7 | SEPTEMBER 2009 | 649 © 2009 Macmillan Publishers Limited. All rights reserved
  • 9. REVIEWS Although the mechanism of inhibition remains unclear, explains both the observed inhibition of FtsZ GTPase the wide conservation of ClpX throughout the Bacteria activity 112,113,115 by SulA and the finding that FtsZ mutants supports the proposal that ClpX could be a general that are impaired in GTPase activity exhibit resistance regulator of Z ring assembly 105. to SulA, with the degree of resistance correlating with the extent of their remaining GTPase activities45,115–117. The Responding to cell cycle status key to understanding this finding came with the dis- UgtP. Consistent with previous data showing that cell covery that the dynamic nature of FtsZ polymers that is division in B. subtilis is subject to growth rate-dependent observed both in the cell and in vitro34,40,41 depends on regulation11, ugtP was recently identified by weart et al. as GTP hydrolysis30. Therefore, when SulA binds to FtsZ a growth rate-dependent inhibitor of Z ring assembly 107. monomers and removes them from the pool that is avail- ugtP is a terminal sugar transferase and uses uDP–glucose able for exchange, ongoing nucleotide hydrolysis in the in the synthesis of the diglucosyl diacylglycerol anchor of polymer promotes its rapid disassembly. This is consist- lipoteichoic acid. It acts as a growth rate-dependent inhibi- ent with reports that SulA can disassemble FtsZ polymers tor of cell division during growth in nutrient-rich media, both in the cell and in vitro. Similarly, mutants of FtsZ that but is downregulated under nutrient-limiting conditions. are impaired in GTP hydrolysis but not GTP binding form ugtP associates reversibly with the divisome, and the stable polymers, and so the rapid turnover of subunits transition between the divisome and sequestration into does not occur, rendering them resistant to sequestration apparently random foci is controlled by the availabil- by SulA42,45. Interestingly, SulA was found to exist as a ity of uDP–glucose, which is supplied by the upstream dimer both in the crystal structure and in solution, which members of the pathway, uTP–glucose-1-phosphate suggests that it could cross-link FtsZ polymers and allow uridylyltransferase B (GtaB) and phosphoglucomutase A substoichiometric levels of the protein to rapidly inhibit (PgcA). ugtP is present at approximately 2,400 molecules Z ring formation113. However, a recent report found that in per cell and at a physiologically relevant molar ratio was E. coli at least, and consistent with the proposed sequestra- shown to be a potent inhibitor of FtsZ assembly by dis- tion model, SulA is effective only once it reaches a critical rupting the formation of the stabilizing lateral interactions concentration35. between protofilaments107. The inhibitory activity of ugtP SulA is not well conserved, but recently two proteins is partially dependent on the availability of uDP–glucose, responsible for the SoS-induced division arrest in although the main role of substrate availability seems B. subtilis and Corynebacterium glutamicum (YneA and likely to be in stimulating the re-localization of ugtP to DivS, respectively) have been identified118,119. These pro- the divisome. teins are not homologous to SulA or to each other. The The authors proposed a model in which ugtP responds assembly of the Z ring is inhibited on the production of to nutrient availability, as sensed by the supply of uDP– DivS119, but YneA seems to act at a later stage in cell divi- glucose, to coordinate cell division with the growth rate. sion, possibly by preventing the recruitment of Ftsl or They suggest that ugtP localizes to the Z ring to delay DivIC98,118. The presence of a lysin motif (lysM) pepti- cell division until the cells reach a sufficient length to doglycan-binding domain in YneA is intriguing, but its allow completion of the multi-fork DNA replication that importance is not yet understood118. is typical of rapidly growing cells. A similar cell length phenotype in E. coli cells lacking the PgcA homologue, MciZ. Recently, a 40-amino acid peptide, mother cell Pgm, suggests that this might be a conserved mechanism inhibitor of FtsZ (MciZ), was identified as an inhibitor of for coordinating cell size and division throughout the Z ring assembly that is produced during B. subtilis sporu- Bacteria107. lation120. Cells lacking MciZ exhibit a markedly increased frequency of Z ring formation during the later stages of SulA. SulA is a cell division inhibitor that is produced in sporulation and, consistent with a role as a negative regu- response to DNA damage, as part of the SoS response, lator of FtsZ, artificial expression of mciZ during vegeta- in E. coli 108,109 and rapidly stalls cell division by both pre- tive growth prevents Z ring assembly 120. In vitro, MciZ venting the assembly of nascent Z rings and facilitating binds specifically to FtsZ and is a potent inhibitor of FtsZ the disassembly of existing Z rings, including those that polymerization120. A large reduction in the GTPase activ- are actively constricting 110–112. ity of FtsZ accompanies this inhibitory activity, suggest- SulA is a potent inhibitor of FtsZ polymerization ing that MciZ acts at the level of the monomer to inhibit in vitro35,110,112–115, as it increases the critical concentration the initial assembly of the FtsZ polymer. There is evidence that is necessary for assembly almost fivefold without to support a model proposing that MciZ binds at a site affecting the cooperative nature of assembly 35. Recent proximal to the nucleotide-binding pocket of FtsZ, so as structural studies revealed that SulA interacts with the to occlude GTP binding and FtsZ polymerization. First, base of the C-terminal domain of FtsZ, completely mask- an MciZ-resistant mutant of FtsZ that bears a substitu- ing the catalytic synergy loop113. SulA is therefore thought tion close to the nucleotide-binding site has been isolated, Lipoteichoic acid to act by binding to FtsZ monomers, sequestering them identifying the probable peptide-binding site. Second, the A class of teichoic acids that is and thereby increasing the apparent critical concentra- kinetics of GTPase activity in the presence of MciZ dis- anchored in the cell tion112,113. In support of this mode of action, when FtsZ played a significant increase in Km, which is characteris- membrane. Teichoic acids are anionic polymers that are is in molar excess it efficiently assembles even in the tic of competitive inhibition120. Interestingly, the mode ubiquitous in the cell walls of presence of SulA35, arguing against a filament-capping of action of MciZ is similar to that recently proposed for Gram-positive bacteria. mechanism for SulA113,115. A sequestration model also a chemical inhibitor of FtsZ assembly 121, reinforcing the 650 | SEPTEMBER 2009 | VoluME 7 www.nature.com/reviews/micro © 2009 Macmillan Publishers Limited. All rights reserved
  • 10. REVIEWS Cytoplasmic species Membrane-tethered species has been made in understanding how Z ring assembly SOS response Sporulation Rapid growth is both coordinated with the cell cycle and rendered responsive to cellular and environmental stresses. As FtsA, SepF ZapA, ZapB SulA MciZ and ZipA and SepF UgtP we have seen, there is considerable overlap and redun- dancy present in the regulation of the Z ring (FIG. 3) and, as a result, it is likely that further, non-essential regulators FtsZ remain to be discovered. Although, as we have described, the regulation of the initial assembly and the stability ClpX and EzrA EzrA EzrA of the Z ring is now reasonably well understood, there is as Monomer Multimeric Growing Organized yet no consensus on how the Z ring senses the completion pool species polymer polymer of the divisome, initiates constriction and eventually dis- Figure 3 | regulation of FtsZ assembly by accessory proteins. A large number of assembles. Similarly, the emerging model of Z ring forma- accessory proteins act in concert to regulate cell division at various levels of FtsZ assembly. tion following the collapse of a spiral intermediate might Before division, FtsZ is thought to exist as a cytoplasmic population of Reviews | Microbiology Nature monomers and also involve additional regulators or might simply be the multimeric species of various lengths. Inhibitors of FtsZ assembly, such as SulA in Escherichia result of passive remodelling, driven by the relief of spatial coli (Ec), which sequesters monomers, or mother cell inhibitor of FtsZ (MciZ) in Bacillus subtilis regulation. Although to date no true bacterial homologues (Bs), which prevents GTP binding, act on the monomer pool to prevent Z ring assembly and of MAPs have been found, as we advance our understand- also facilitate Z ring disassembly owing to the rapid exchange of FtsZ subunits in the Z ring ing of division proteins, tentative functional parallels can with those in the cytoplasm. The monomer pool that is available for exchange is maintained be drawn and further characterization of these proteins by both ClpX, which seems to prevent further assembly of FtsZ, and extra Z rings A (EzrABs), might reveal additional relationships with the MAPs122. which reduces the initial assembly of FtsZ and promotes its disassembly from the medial Z ring. Following the relief of spatial regulation, the Z ring assembles at mid-cell and is tethered The recent in vitro reconstitution of contractile Z rings to the inner face of the cytoplasmic membrane by FtsA or ZipAEc, which, along with ZapA, also in tubular liposomes as well as the assembly of a MinD– contribute to the stability of the Z ring. Furthermore, early-assembling accessory proteins MinE gradient on the surface of a membrane are prom- such as ZapBEc and SepFBs also have potential roles in organizing the ultrastructure of the Z ising steps forward47,123 that offer the potential to study ring; SepF seems to make a substantial contribution to this process in Bacillus subtilis and the regulation of Z rings using in vitro systems, rather might itself act as a membrane tether. During rapid growth, further ‘fine tuning’ of the Z ring is than simply studying the effects of individual proteins carried out by UgtPBs, which delays Z ring completion until cells reach an appropriate size. on the bulk polymer. Although this will undoubtedly be technically challenging, we must remain cautious when notion that an understanding of the cellular mechanisms drawing conclusions on the basis of the effects of single that regulate FtsZ assembly is likely to inform future work accessory proteins on FtsZ polymers. In the cell, we also in developing clinically relevant inhibitors of bacterial need to take into account the effects of membrane bind- cytokinesis. ing, the presence of numerous other regulatory proteins at the Z ring and the crowded nature of the cytoplasmic Future perspectives environment. The advances that we anticipate making in The past few years have seen the discovery of many addi- understanding Z ring assembly and its regulation prom- tional factors that are involved in Z ring formation and ise to make this an enlightening and exciting field in the modulating polymer dynamics. Moreover, much progress coming years. 1. Errington, J., Daniel, R. A. & Scheffers, D. J. Cytokinesis and ZipA during the Escherichia coli cell cycle. 20. de Boer, P., Crossley, R. & Rothfield, L. The essential in bacteria. Microbiol. Mol. Biol. Rev. 67, 52–65 (2003). J. Bacteriol. 185, 3344–3351 (2003). bacterial cell-division protein FtsZ is a GTPase. Nature 2. Goehring, N. W. & Beckwith, J. Diverse paths to midcell: 11. Weart, R. B. & Levin, P. A. Growth rate-dependent 359, 254–256 (1992). assembly of the bacterial cell division machinery. Curr. regulation of medial FtsZ ring formation. J. Bacteriol. 21. RayChaudhuri, D. & Park, J. T. Escherichia coli cell- Biol. 15, R514–R526 (2005). 185, 2826–2834 (2003). division gene ftsZ encodes a novel GTP-binding protein. 3. Harry, E., Monahan, L. & Thompson, L. Bacterial cell 12. Romberg, L. & Levin, P. A. Assembly dynamics of the Nature 359, 251–254 (1992). division: the mechanism and its precison. Int. Rev. Cytol. bacterial cell division protein FtsZ: poised at the edge of 22. Mukherjee, A., Dai, K. & Lutkenhaus, J. Escherichia coli 253, 27–94 (2006). stability. Annu. Rev. Microbiol. 57, 125–154 (2003). cell division protein FtsZ is a guanine nucleotide binding 4. Bi, E. F. & Lutkenhaus, J. FtsZ ring structure associated 13. Löwe, J. & Amos, L. A. Crystal structure of the bacterial protein. Proc. Natl Acad. Sci. USA 90, 1053–1057 with division in Escherichia coli. Nature 354, 161–164 cell-division protein FtsZ. Nature 391, 203–206 (1993). (1991). (1998). 23. Scheffers, D. J., de Wit, J. G., den Blaauwen, T. & 5. Addinall, S. G. & Lutkenhaus, J. FtsZ-spirals and -arcs 14. Nogales, E., Wolf, S. G. & Downing, K. H. Structure of the Driessen, A. J. Substitution of a conserved aspartate determine the shape of the invaginating septa in some αβ tubulin dimer by electron crystallography. Nature allows cation-induced polymerization of FtsZ. FEBS Lett. mutants of Escherichia coli. Mol. Microbiol. 22, 391, 199–203 (1998). 494, 34–37 (2001). 231–237 (1996). 15. Erickson, H. P. FtsZ, a prokaryotic homolog of tubulin? 24. Scheffers, D. J., de Wit, J. G., den Blaauwen, T. & 6. Margolin, W. FtsZ and the division of prokaryotic cells Cell 80, 367–370 (1995). Driessen, A. J. GTP hydrolysis of cell division protein FtsZ: and organelles. Nature Rev. Mol. Cell Biol. 6, 16. Mukherjee, A. & Lutkenhaus, J. Guanine nucleotide- evidence that the active site is formed by the association 862–871 (2005). dependent assembly of FtsZ into filaments. J. Bacteriol. of monomers. Biochemistry 41, 521–529 (2002). 7. Haydon, D. J. et al. An inhibitor of FtsZ with potent and 176, 2754–2758 (1994). 25. Romberg, L., Simon, M. & Erickson, H. P. Polymerization selective anti-staphylococcal activity. Science 321, 17. Erickson, H. P., Taylor, D. W., Taylor, K. A. & Bramhill, D. of Ftsz, a bacterial homolog of tubulin: is assembly 1673–1675 (2008). Bacterial cell division protein FtsZ assembles into cooperative? J. Biol. Chem. 276, 11743–11753 (2001). This paper describes the validation of FtsZ as a protofilament sheets and minirings, structural homologs 26. Caplan, M. R. & Erickson, H. P. Apparent cooperative clinically relevant target for new antibiotics. of tubulin polymers. Proc. Natl Acad. Sci. USA 93, assembly of the bacterial cell division protein FtsZ 8. Kelly, A. J., Sackett, M. J., Din, N., Quardokus, E. & 519–523 (1996). demonstrated by isothermal titration calorimetry. J. Biol. Brun, Y. V. Cell cycle-dependent transcriptional and 18. Löwe, J. & Amos, L. A. Tubulin-like protofilaments in Chem. 278, 13784–13788 (2003). proteolytic regulation of FtsZ in Caulobacter. Genes Dev. Ca2+-induced FtsZ sheets. EMBO J. 18, 2364–2371 27. Chen, Y., Bjornson, K., Redick, S. D. & Erickson, H. P. A 12, 880–893 (1998). (1999). rapid fluorescence assay for FtsZ assembly indicates 9. Quardokus, E., Din, N. & Brun, Y. V. Cell cycle regulation 19. Oliva, M. A., Cordell, S. C. & Löwe, J. Structural insights cooperative assembly with a dimer nucleus. Biophys. J. and cell type-specific localization of the FtsZ division into FtsZ protofilament formation. Nature Struct. Mol. 88, 505–514 (2005). initiation protein in Caulobacter. Proc. Natl Acad. Sci. Biol. 11, 1243–1250 (2004). 28. Wang, X. & Lutkenhaus, J. The FtsZ protein of Bacillus USA 93, 6314–6319 (1996). The work described in references 18 and 19 provides subtilis is localized at the division site and has GTPase 10. Rueda, S., Vicente, M. & Mingorance, J. Concentration evidence that the protofilaments of FtsZ and tubulin activity that is dependent upon FtsZ concentration. Mol. and assembly of the division ring proteins FtsZ, FtsA, make similar longitudinal contacts. Microbiol. 9, 435–442 (1993). NATuRE REVIEwS | MiCrobiology VoluME 7 | SEPTEMBER 2009 | 651 © 2009 Macmillan Publishers Limited. All rights reserved