Combating anti-microbial resistance by developing alternative strategies is the need of the hour. Cell division, particularly FtsZ, is being extensively studied for its potential as an alternative target for anti-bacterial therapy. Bacillus subtilis and Escherichia coli are the two well-studied models for research on FtsZ, the leader protein of the cell division machinery. As representatives of gram-positive and gram-negative bacteria, respectively, these organisms have provided an extensive outlook into the process of cell division in rod-shaped bacteria. However, research on other shapes of bacteria, like cocci and ovococci, lags behind that of model rods. Even though most regions of FtsZ show sequence and structural conservation throughout bacteria, the differences in FtsZ functioning and interacting partners establish several different modes of division in different bacteria. In this review, we compare the features of FtsZ and cell division in the model rods B. subtilis and E. coli and the four pathogens: Staphylococcus aureus, Streptococcus pneumoniae, Mycobacterium tuberculosis, and Pseudomonas aeruginosa. Reviewing several recent articles on these pathogenic bacteria, we have highlighted the functioning of FtsZ, the unique roles of FtsZ-associated proteins, and the cell division processes in them. Further, we provide a detailed look at the anti-FtsZ compounds discovered and their target bacteria, emphasizing the need for elucidation of the anti-FtsZ mechanism of action in different bacteria. Current challenges and opportunities in the ongoing journey of identifying potent anti-FtsZ drugs have also been described.

Most bacteria divide by binary fission. One bacterial cell splitting into two daughter cells seems to be a simple process, but the research in this field spanning decades demonstrates the complexity of the process. Bacterial cell division involves a myriad of protein complexes that are spatiotemporally regulated in accordance with environmental, chemical, and intrinsic factors, all taking place in about 30 min!

A central protein that is responsible for orchestrating the cell division process is FtsZ, filamentous-temperature sensitive mutant Z. It is the bacterial homolog of the tubulin, the eukaryotic cytoskeletal protein [1]. When the bacterial cell is primed for division, FtsZ self-assembles into a dynamic structure at the mid-cell region called the Z-ring. Formed of several FtsZ polymers discontinuously bundled into treadmilling filaments, the Z-ring is responsible for the assembly of the divisome and the peptidoglycan (PG) synthesis complex at the site of cell division (Figure 1) [2–5].

FtsZ as the central player in different modes of cell division

Figure 1
FtsZ as the central player in different modes of cell division

(A) Division in model rods like E. coli and B. subtilis is carried out by the coordination of the elongasome machinery at different sites in the cell and by the divisome complex in the mid-cell region. On the direction of several mid-cell site markers, some associated with the nucleoid separation, FtsZ assembles into a central Z-ring in coordination with early divisome proteins to establish the divisome complex. The Z-ring coordinates septum formation and interacts with late divisome proteins to begin the septum peptidoglycan (PG) synthesis. Peripheral PG synthesis is carried out by the elongasome. Finally, membrane constriction leads to cytokinesis resulting in the formation of two daughter cells. (B) In the absence of elongasome machinery, cell division in cocci like S. aureus occurs through FtsZ-led division machinery only. Z-ring formation at the mid-cell site follows the septum formation in a centripetal pattern towards the centre of the cell. PG synthesis takes place in the septal region alone and final constriction leads to pinching off the two hemispherical lobes to give two daughter cells. The daughter cells immediately bulge out to form cocci each with one-half of the parent cell wall. (C) In ovococci like S. pneumoniae, the division is initiated by the assembly of initiation complex - FtsZ in a tight complex with early divisome proteins at the septal region. Cells have both elongasome machinery and divisome machinery present at the septal region, the coordination between the two leads to the formation of the septum and PG synthesis in the vicinity of the septum. The initiation complex assembles at the division site of the future daughter cells over the undivided nucleoid, thus giving rise to overlapping division cycles and cells mostly observed in diplococci state. (D) Asymmetrical cell division observed in M. tuberculosis is established by polar growth at one of the poles of the cell occurring at a faster rate than other. More Wag31, a protein that is associated with promoting polar growth, assembles at the old pole (established at previous division cycle) and coordinates accelerated growth (depicted in cyan). While growth at the new pole lags behind, the cell division process begins. Wag31 is also found to interact with FtsZ at the septum. The septum formation and membrane constriction proceed, and final ‘V-snapping’ results in the formation of two unequal daughter cells. The new poles are established after separation, while the other poles are now designated ‘old’ poles. (Images were created using BioRender.com)

Figure 1
FtsZ as the central player in different modes of cell division

(A) Division in model rods like E. coli and B. subtilis is carried out by the coordination of the elongasome machinery at different sites in the cell and by the divisome complex in the mid-cell region. On the direction of several mid-cell site markers, some associated with the nucleoid separation, FtsZ assembles into a central Z-ring in coordination with early divisome proteins to establish the divisome complex. The Z-ring coordinates septum formation and interacts with late divisome proteins to begin the septum peptidoglycan (PG) synthesis. Peripheral PG synthesis is carried out by the elongasome. Finally, membrane constriction leads to cytokinesis resulting in the formation of two daughter cells. (B) In the absence of elongasome machinery, cell division in cocci like S. aureus occurs through FtsZ-led division machinery only. Z-ring formation at the mid-cell site follows the septum formation in a centripetal pattern towards the centre of the cell. PG synthesis takes place in the septal region alone and final constriction leads to pinching off the two hemispherical lobes to give two daughter cells. The daughter cells immediately bulge out to form cocci each with one-half of the parent cell wall. (C) In ovococci like S. pneumoniae, the division is initiated by the assembly of initiation complex - FtsZ in a tight complex with early divisome proteins at the septal region. Cells have both elongasome machinery and divisome machinery present at the septal region, the coordination between the two leads to the formation of the septum and PG synthesis in the vicinity of the septum. The initiation complex assembles at the division site of the future daughter cells over the undivided nucleoid, thus giving rise to overlapping division cycles and cells mostly observed in diplococci state. (D) Asymmetrical cell division observed in M. tuberculosis is established by polar growth at one of the poles of the cell occurring at a faster rate than other. More Wag31, a protein that is associated with promoting polar growth, assembles at the old pole (established at previous division cycle) and coordinates accelerated growth (depicted in cyan). While growth at the new pole lags behind, the cell division process begins. Wag31 is also found to interact with FtsZ at the septum. The septum formation and membrane constriction proceed, and final ‘V-snapping’ results in the formation of two unequal daughter cells. The new poles are established after separation, while the other poles are now designated ‘old’ poles. (Images were created using BioRender.com)

Close modal

The growing problem of anti-microbial resistance (AMR) has triggered the extensive search for novel antibiotics and alternative strategies for antibacterial therapy. Cell division in bacteria is an essential process presenting the central player- FtsZ, as an attractive alternative target for the design of antibiotics. Several anti-FtsZ compounds have been discovered and their mechanisms have been explored [1,6,7]. However, no FtsZ-targeting agent has been translated to the clinical stage. What are the scientists missing?

The rod-shaped bacteria, Bacillus subtilis and Escherichia coli, have been the model organisms for extensive studies on their cell shape and cell division processes. Most of the information on FtsZ’s role in cell division comes from these rods with widespread in vivo and in vitro studies [8–10]. Several recent reviews have covered the vast research in the field of FtsZ with extensive descriptions of FtsZ structure and function, and its potential as an antibiotic target [8–22]. The dynamics of FtsZ and its regulation inside the cells is an ever-evolving field of research with new information added every day. But only a few of these are arising from FtsZ from other bacteria that include pathogenic species.

Even though FtsZ is a conserved protein throughout the bacterial species, there are several important differences even between B. subtilis and E. coli with respect to its regulation, interaction with other proteins, and FtsZ dynamics. These two organisms, although both rod-shaped, show dissimilarities within their FtsZ-mediated cell division process, demonstrating the need for understanding the cell division process at the species level. Considering the literature on bacterial pathogens and their cell shapes ranging from rods, and cocci, to ovoid, there is a significant gap in the knowledge of FtsZ and cell division in these organisms.

In this review, we highlight the differences in FtsZ characteristics between model bacteria, E. coli and B. subtilis, and some of the pathogenic bacteria, Staphylococcus aureus, Streptococcus pneumoniae, Mycobacterium tuberculosis, and Pseudomonas aeruginosa. We detail the known cell division and cell shape determinants of the bacteria in this review, focusing on FtsZ and its partners. We emphasize that a conserved protein like FtsZ, shows differential behavior and interactions in different bacteria leading us to ask the question—How ‘conserved’ is FtsZ? Our goal is to use the pathogenic bacteria as representatives to enumerate unique and dissimilar aspects of cell division to encourage the study of anti-FtsZ agents in the pathogenic bacteria. This would further lead to fast-tracking the development of alternative targets for anti-bacterial therapy and in time, bridge the gap between drug discovery and in vivo efficacy for clinical translation.

Cell division in rod-shaped bacteria is coordinated by Elongasome-Divisome interplay

The model organisms, B. subtilis and E. coli are gram-positive and gram-negative rods, respectively, with an average division time of 20 min. A nosocomial pathogen Pseudomonas aeruginosa is an encapsulated gram-negative rod. It is a wound-related infectious agent also causing bacteremia, septic shock, and urinary tract infections. Recently, carbapenem-resistant P. aeruginosa has been listed for critical need for development of alternative strategies for treatment against it [23]. The presence of the lipopolysaccharide outer membrane in the bacteria allows it to restrict the entry of antibiotics, contributing to AMR development. Efflux pumps and antibiotic-inactivating enzymes are other factors [23]. P. aeruginosa being a rod exhibits a similar cell division behavior as E. coli and B. subtilis. Even though homologs of several proteins in E. coli are found in P. aeruginosa, their functional characterization lags far behind that of the model rods.

A typical rod, like B. subtilis, has two machineries for cell division and cell elongation (Figure 1A). The protein complex responsible for septation and division called the divisome, and the protein complex responsible for peripheral elongation that determines the shape of the bacteria, called the elongasome, coordinate with each other during the cell division cycle. Cell elongation complex comprises MreB, the primary protein responsible for recruiting proteins such as MreC, MreD, and penicillin-binding proteins (PBP) to various foci along the cell. MreB forms a patchy network of filaments that undergo circumferential motion and coordinates with the peptidoglycan synthases [24–26]. In E. coli, MreB and FtsZ have been reported to be involved in murein biosynthesis through independent pathways [27]. Cell division is coordinated with chromosome separation with the help of the FtsK/SpoIIE family of DNA translocases [28]. The division site selection is greatly influenced by Nucleoid Occlusion (NO) and the Min proteins. The NO protein factors noc in B. subtilis and slmA in E. coli prevent the assembly of the divisome near the nucleoid [29,30]. An FtsZ-inhibitor protein, MinC, and its activator MinD are recruited to the mid-cell and also to the cell poles via interaction with MinJ and DivIVA in B. subtilis [31]. In contrast, the position of Z-ring assembly in E. coli is determined by an ‘oscillatory’ motion of the MinCDE complex [32]. FtsZ then assembles onto the mid-cell site with help of membrane-associated proteins. In B. subtilis it is FtsA, while E. coli requires dual membrane anchors, ZipA and FtsA [16,33,34]. EzrA and SepF, only identified in gram-positive bacteria, also play a role in the membrane association of FtsZ [35,36]. Throughout the division process, FtsZ interacts with several proteins classified as early and late divisome proteins. Early divisome proteins in B. subtilis that interact with FtsZ include FtsA, ZapA, SepF, and EzrA [37]. EzrA, SepF, and ZapA have been recently shown to be essential for the condensation of FtsZ filaments into the Z-ring [38]. Late divisome proteins include GpsB, FtsL, DivIB, FtsW, Pbp2B, and DivIVA [37]. In E. coli, FtsA, FtsE, and Zap proteins form the early divisome proteins and during later stages of cell division, FtsZ interacts with FtsK, FtsQ, FtsL, and FtsB, followed by FtsW with FtsI [39–41]. Polymerization of FtsZ and formation of the Z-ring are controlled by the tight regulation and coordination between early and late divisome proteins. The FtsZ polymers undergo treadmilling around the Z-ring to distribute the PG synthases forming the septum [4,5,42]. The rate of FtsZ treadmilling directly influences the septum PG distribution in both E. coli and B. subtilis and septum synthesis only in B. subtilis [4,5]. The difference is attributed to the latter’s requirement for a thicker cell wall [43]. The activation of cell wall synthesis drives septum formation and membrane invagination. It was recently shown that early divisome protein, FtsA interacts with late divisome proteins such as FtsW to activate septal PG synthesis [44]. Final constriction at the septum results in cytokinesis leading to the separation of the daughter cells.

Cocci have a single coordinator—the divisome complex

A gram-positive coccus, S. aureus, is an opportunistic pathogen which is a leading cause of bacteremia and infective endocarditis. The treatment of these infections, along with serious skin and soft tissue infections it causes, is threatened by a rising number of multi-drug resistant strains of the bacterium [45]. Multi-drug resistant S. aureus (MRSA) evades antibiotic treatment by synthesizing β-lactamase and PBP2a, which confer the resistance to β-lactam antibiotics [46].

In the cell division of cocci like S. aureus, the striking difference with model rods is the absence of an elongation machinery or MreB homologs (Figure 1B). The PG synthesis takes place mostly at the site of the division, i.e., the septum coordinated by FtsZ, leading to orthogonal division over several cell division cycles [47–49]. FtsW-PBP1 and RodA-PBP3 pairs are reported to regulate the septal and lateral PG synthesis, respectively [50]. Another recently discovered protein conserved in the family is SmdA, bearing roles in cell division and cell morphology [51]. In the absence of the Min system, the Noc system plays the role in the division site selection [52]. An early divisome protein GpsB from S. aureus was reported to have a direct cell division-modulating activity, unlike the homolog in B. subtilis [53]. It was recently shown to also regulate the wall teichoic acid biosynthesis [54]. CozEa and CozEb proteins, which have a homolog in S. pneumoniae, are found to interact with EzrA and regulate cell division [55]. Cytokinesis was proposed to be biphasic with a second, faster constriction process at the septum being independent of FtsZ function and likely dependent on PG deposition [56]. The septum formation takes place in a centripetal manner without elongation of the bacteria till the walls are pinched off after constriction. Immediately, the septum is reshaped into a curved hemisphere to give two daughter cocci [57,58]. The switch from PG synthesis at the cell periphery to the septum for a new cycle of cell division is directed by the lipid transporter protein MurJ [56]. This transition would take place after the Z-ring assembly and initiation of cell division, when MurJ is recruited by the late divisome complex [56].

The in-between bacterial morphology: ovococci

Another opportunistic pathogen S. pneumoniae, a gram-positive ovococcoid bacterium, resides in the human respiratory tract. It causes potentially life-threatening infections like pneumonia and meningitis. More than a hundred serotypes of the bacterium are discovered so far, contributing immensely to the growing anti-microbial resistance problem. Like S. aureus, horizontal gene transfer in S. pneumoniae has allowed the bacteria to develop mutations in PBP’s thereby evading antibiotics [59]. Several recent reports on S. pneumoniae cell division have contributed to the understanding of ovococci morphology [60–62].

In S. pneumoniae, the cell division time takes longer than the rods, approximately 30 min and the dividing cells mostly exist in pairs (also called diplococci) [63]. Min and Noc systems for division site selection and MreB homolog for directing the elongation are all absent in S. pneumoniae [60]. The striking feature of this ovococcoid bacterium is that the cell division cycle begins even before the completion of the previous cell division cycle [62,64]. The nascent equatorial rings are formed at the mid-cell region of the daughter cells even before they are separated from each other, over the undivided nucleoid (Figure 1C). This seemingly overlapping process of cell division cycles may lead to chains of dividing bacteria in a ‘streptococci’.

A mid-cell anchored protein, MapZ is positioned at the mid-cell by the new peptidoglycan synthesis acting as a guide for the movement of FtsZ and its associating proteins, from the septal ring to the equatorial ring to initiate a new cycle of cell division [65–67]. It was recently discovered that CcrZ is responsible for the regulation of DNA replication and the regulation of FtsZ and the Z-ring during cell division initiation [68]. FtsZ associating proteins FtsA and EzrA are essential for S. pneumoniae, being part of the initiation complex localizing with FtsZ throughout the cell division process [67,69]. It has been suggested that the role of EzrA in Z-ring assembly and PG synthesis might be central to overall cell division in S. pneumoniae [69]. ZapA and its interacting partner ZapJ are also shown to be FtsZ-regulators [69]. FtsZ treadmilling drives septal PG synthesis in concert with the FtsW-PBP2x complex [67]. The rate of FtsZ treadmilling and septal PG synthesis rate are independent, unlike the case with B. subtilis [67]. The position of the peripheral PG synthesis in Streptococci is different to that of these rods—it is positioned at the septal region and not at the side wall [70]. The two complexes for septal and peripheral PG synthesis assemble at the mid-cell region, and during division form concentric circles, with the septal PG synthesis taking place at the inner closer ring while peripheral PG synthesis takes place on the outer ring [71]. The peripheral elongation of S. pneumoniae is carried out by the elongasome complex. This complex, driven by the RodA-PBP2b pair of proteins, consists of CozE, MreC, MreD, and RodZ [60,61,72]. Out of the many models that describe the ovoid pneumococcal division, evidence supports the model where the two complexes for peripheral and septal peptidoglycan synthesis are assembled at the mid-cell forming one large protein complex driving both growth and division of S. pneumoniae.

An atypical form of bacterial cell division: asymmetric cell division

One of the most notorious pathogens is M. tuberculosis, an intracellular acid-fast rod. M. tuberculosis causes tuberculosis which is the leading cause of infection-related deaths with a death toll of 1.5 million each year [73]. M. tuberculosis is also associated with several pulmonary diseases and auto-immune disorders like sarcoidosis and lung cancer [74]. Apart from the absence of the common virulence factors and the latency period inside the host cells, the treatment of this highly adaptable pathogen is challenged by growing antimicrobial resistance. A possible reason for the requirement of longer periods of antibiotic treatment is the asymmetric cell division exhibited by the bacterium making the population heterogeneous [75].

Even though M. tuberculosis is a rod-shaped bacterium, it has some distinct features which have resulted in the knowledge gap in understanding its cell division. A key characteristic feature of Mycobacterial cells is the cell wall composed of mycolic acid, arabinogalactan, and the PG, which makes it a very thick layer [76]. Typically requiring approximately 18–24 h to divide, M. tuberculosis requires a special cohort of proteins that enable its division which is very different than typical rods B. subtilis and E. coli [77,78]. The geometry of cell division is asymmetrical (Figure 1D) exhibited by a polar growth, i.e., it takes place mostly at the poles of the bacterium rather than the side walls [75–78]. Unlike in B. subtilis where DivIVA regulates septum formation, its homolog in M. tuberculosis, Wag31 directs and regulates the polar growth [79]. Recent discovery revealed that a protein LamA maintains the asymmetry in mycobacteria and interacts with Wag31 [80]. In the absence of the MreB homolog, several pieces of evidence suggest that the timing of elongation and division machinery may be independent of each other [77,81]. The division site for Z-ring assembly is not regulated by the Noc or the Min system, as they are both absent in Mycobacteria. The division site is suggested to be present as troughs within the cell surface that have been inherited from one or two cell division cycles prior [82]. SepF is the only early divisome protein known to interact with FtsZ in M. tuberculosis, even though several late divisome proteins such as FtsW, showing a significant role in mycobacterial cell division, are present similar to B. subtilis [77,83,84]. FtsA and ZipA are both absent in M. tuberculosis, with studies proposing that FtsW might substitute their roles within this bacterium [84,85]. Recent studies have shown that a transmembrane protein unique to the family, CrgA, interacts with FtsZ and plays an important role in several steps of the cell division process [86]. CrgA also shows interactions with CwsA, a protein specific to mycobacteria that regulates Wag31 functioning [87]. WhmD and Rv0954 are other proteins that interact with the Z-ring and/or regulate the cell cycle in the mycobacterium species [88–90]. Cell wall deposition by the PBPs and their partners happens at a faster rate at the one pole, indicated as the old pole, inherited from the mother during the previous cell cycle [76]. Finally ‘V-snapping’ of the septum divides the cell into two, with the daughter cells of uneven sizes-the old-poled daughter bigger than the new-poled daughter cell [76–78]. The new cell cycle begins with the newly separated cell wall becoming the new poles of the daughter cells. Asymmetrical cell division gives the bacteria a survival advantage due to heterogeneity in the population, in the absence of which the bacteria is more susceptible to antibiotics [80].

Asymmetrical cell division is observed in other bacteria as well. It was recently reported that in gram-negative bacteria that exhibit asymmetrical division, short filaments of FtsZ were enough to begin the constriction process rather than the assembly of the entire Z-ring, unlike observed in other asymmetrically dividing bacteria [91].

Structure of FtsZ

As seen in the previous section, FtsZ is the central player in cell division that warrants deeper investigation at the protein level. Amino acid sequence alignment of all the different FtsZs in this review as well as the eukaryotic homolog of FtsZ, tubulin, is shown in Figure 2 [92–94]. FtsZ bears little sequence similarity with tubulin, barring the GTP binding region called the G-box motif [1]. However, the homology between FtsZ and tubulin is established because of the highly similar secondary structures of the two proteins [1]. FtsZ from Methanocaldococcus jannaschii was the first FtsZ to be crystallized in 1998 [95]. Very similar to tubulin, the crystal structure of FtsZ shows an N-terminal core domain bearing the GTP-binding site and a C-terminal core domain bearing the T7 loop (Figure 3). Rossmann folding establishes an arrangement of a core of β-sheets alternating with α-helices surrounding it as seen in Figure 3A [96,97]. The H7 helix separates the N- and C-termini cores. The active site is formed when the T7 loop of one FtsZ monomer interacts with the GTP binding domain of the adjacent monomer, forming an end-to-end connection. Single-stranded filament assembly is promoted by the conformational switch from low affinity to higher affinity for polymer formation, which allows the establishment of cooperativity [98–102]. The conformational switch is characterized by the relative rotation of the C-terminal T7 loop by ∼27o with respect to the N-terminal domain, pulling the H7 helix downwards [98,101]. By using tryptophan fluorescence as a detector of the change in the environment of T7 loop region, this conformational switch was recently observed in FtsZ from S. pneumoniae [102].

Sequence alignment of FtsZ and eukaryotic tubulin

Figure 2
Sequence alignment of FtsZ and eukaryotic tubulin

Clustal Omega tool was used to align the amino acid sequences of human α-tubulin isoform 1A (α-tubulin, 451 residues), M. jannaschii FtsZ (MjFtsZ, 364 residues), E. coli FtsZ (EcFtsZ, 383 residues), P. aeruginosa FtsZ (PaFtsZ, 394 residues), M. tuberculosis FtsZ (MtbFtsZ, 379 residues), S. pneumoniae FtsZ (SpnFtsZ, 419 residues), B. subtilis FtsZ (BsFtsZ, 382 residues), and S. aureus FtsZ (SaFtsZ, 390 residues). The residues that show identity are marked in blue using Jalview. N-terminal and C-terminal domains separated by the T7 loop region is indicated in blue, red, and yellow lines, respectively, above the sequence alignment. The G-box GTP binding motif and the C-terminal tails are highlighted.

Figure 2
Sequence alignment of FtsZ and eukaryotic tubulin

Clustal Omega tool was used to align the amino acid sequences of human α-tubulin isoform 1A (α-tubulin, 451 residues), M. jannaschii FtsZ (MjFtsZ, 364 residues), E. coli FtsZ (EcFtsZ, 383 residues), P. aeruginosa FtsZ (PaFtsZ, 394 residues), M. tuberculosis FtsZ (MtbFtsZ, 379 residues), S. pneumoniae FtsZ (SpnFtsZ, 419 residues), B. subtilis FtsZ (BsFtsZ, 382 residues), and S. aureus FtsZ (SaFtsZ, 390 residues). The residues that show identity are marked in blue using Jalview. N-terminal and C-terminal domains separated by the T7 loop region is indicated in blue, red, and yellow lines, respectively, above the sequence alignment. The G-box GTP binding motif and the C-terminal tails are highlighted.

Close modal

Structure of FtsZ protein

Figure 3
Structure of FtsZ protein

(A) The crystal structure of M. jannaschii FtsZ showing the N-terminal domain in blue and the C-terminal domain in red separated by the H7 helix and T7 loop (yellow). N-terminal domain shows GTP bound to the structure. PDB: 2VAP (B) Superimposition of the X-ray crystal structure of M. jannaschii 2VAP with AlphaFold prediction for the same shown in purple. (C) Superimposition of AlphaFold predictions of B. subtilis FtsZ (BsFtsZ) in green, E. coli FtsZ (EcFtsZ) in blue, S. aureus FtsZ (SaFtsZ) in magenta, S. pneumoniae FtsZ (SpnFtsZ) in orange, M. tuberculosis FtsZ (MtbFtsZ) in grey and P. aeruginosa FtsZ (PaFtsZ) in pink. (D) Intrinsically disordered C-terminal shown to be highly flexible compared to the compact, highly conserved core region.

Figure 3
Structure of FtsZ protein

(A) The crystal structure of M. jannaschii FtsZ showing the N-terminal domain in blue and the C-terminal domain in red separated by the H7 helix and T7 loop (yellow). N-terminal domain shows GTP bound to the structure. PDB: 2VAP (B) Superimposition of the X-ray crystal structure of M. jannaschii 2VAP with AlphaFold prediction for the same shown in purple. (C) Superimposition of AlphaFold predictions of B. subtilis FtsZ (BsFtsZ) in green, E. coli FtsZ (EcFtsZ) in blue, S. aureus FtsZ (SaFtsZ) in magenta, S. pneumoniae FtsZ (SpnFtsZ) in orange, M. tuberculosis FtsZ (MtbFtsZ) in grey and P. aeruginosa FtsZ (PaFtsZ) in pink. (D) Intrinsically disordered C-terminal shown to be highly flexible compared to the compact, highly conserved core region.

Close modal

As important domains forming the active site, the N-terminal regions and the C-terminal core domains are highly conserved in FtsZ (Figure 2). This indicates that the FtsZ-FtsZ interaction to form a polymer is conserved for all FtsZ. This makes up for approximately 80% of the residues in the amino acid sequence of FtsZ. However, several assembly-related characteristics arise for FtsZ from different bacteria. These are usually observed in spectroscopy and electron microscopy studies of protofilaments formed in vitro [103–106]. Further, functional differences like treadmilling properties and interactions with associated proteins arise, as seen in the previous section. Apart from the cellular factors contributing to these differences, the differences in the FtsZ behavior at the structural level arise from the C-terminal end domain, specifically the much less conserved C-terminal tail region.

C-terminal tail of FtsZ

With the recent advent of the AI-predicted modeling of protein structures through AlphaFold, it has become easier to visualize these disordered C-terminal tails relative to the defined structure of the core domains [107,108]. To demonstrate the accuracy of the AlphaFold prediction, the crystal structure of FtsZ in Figure 3A is superimposed with the AlphaFold prediction (Figure 3B). For B. subtilis FtsZ (BsFtsZ), E. coli FtsZ (EcFtsZ), S. aureus FtsZ (SaFtsZ), S. pneumoniae FtsZ (SpnFtsZ), M. tuberculosis FtsZ (MtbFtsZ), and P. aeruginosa FtsZ (PaFtsZ), AlphaFold predictions are used (Figure 3C). Superimposition of the structures shows that the core domains of N- and C-termini show highly conserved structural similarity. The differences arise in the disordered C-terminal tail (CTT) region (Figure 3D).

A comparison of the FtsZ sequence and structure between the model rods and the four pathogenic organisms is shown in Figure 4. The disordered highly variable region starts after the ‘ATG’ peptide which occurs at the 308-312 residues number in the amino acid sequence for the FtsZs considered. Sequence alignment shows that the sequences up to ‘ATG’ show more than 60% similarity. The C-terminal tail region is variable in length ranging from 68 to 104 residues (Figure 4). This CTT region has less than 20% similarity between the sequences. The CTT region is made up of a long C-terminal linker (CTL) region, a conserved C-terminal conserved peptide (CTC) and a C-terminal variable (CTV) region. The CTC region is an 8-residue stretch with fully conserved aspartic acid, proline, and phenylalanine residues. The rest of the 5 residues show more than 50% similarity between the organisms. The CTV region has 1-10 residues, with a net positive charge in gram-positive organisms and a net neutral charge in gram-negative organisms.

Schematic of conservation of FtsZ sequence and structure in model organisms and pathogens

Figure 4
Schematic of conservation of FtsZ sequence and structure in model organisms and pathogens

The sequence and structural similarity between B. subtilis FtsZ (BsFtsZ) in green, E. coli FtsZ (EcFtsZ), S. aureus FtsZ (SaFtsZ), S. pneumoniae FtsZ (SpnFtsZ), M. tuberculosis FtsZ (MtbFtsZ), and P. aeruginosa FtsZ (PaFtsZ) are compared. The core region of FtsZ is composed of a conserved N-terminal domain with a GTP (Shown as a blue circle) binding region and the C-terminal core region separated by the H7 helix. The intrinsically disordered tail begins after ‘ATG’ stretch of amino acids around 308-312 in the sequence. This intrinsically disordered region is composed of the C-terminal linker (CTL) followed by the C-terminal conserved (CTC) and C-terminal variable (CTV) regions. The CTL and CTV of different FtsZ are of variable length and show very less sequence similarity with each other.

Figure 4
Schematic of conservation of FtsZ sequence and structure in model organisms and pathogens

The sequence and structural similarity between B. subtilis FtsZ (BsFtsZ) in green, E. coli FtsZ (EcFtsZ), S. aureus FtsZ (SaFtsZ), S. pneumoniae FtsZ (SpnFtsZ), M. tuberculosis FtsZ (MtbFtsZ), and P. aeruginosa FtsZ (PaFtsZ) are compared. The core region of FtsZ is composed of a conserved N-terminal domain with a GTP (Shown as a blue circle) binding region and the C-terminal core region separated by the H7 helix. The intrinsically disordered tail begins after ‘ATG’ stretch of amino acids around 308-312 in the sequence. This intrinsically disordered region is composed of the C-terminal linker (CTL) followed by the C-terminal conserved (CTC) and C-terminal variable (CTV) regions. The CTL and CTV of different FtsZ are of variable length and show very less sequence similarity with each other.

Close modal

Structure of the C-terminal tail of FtsZ

Even though several crystal structures of FtsZ have been so far elucidated, the C-terminal end region has not been modeled through crystallography, indicating its disordered nature. However, several groups have shown that binding of the CTC with several FtsZ-associated proteins stabilizes the region to different conformations. From adopting a β-strand and helical conformation upon binding to ZipA [109], an extended conformation when bound to SlmA [100] and SepF [110], to interacting with FtsA [111] and ZapD [112] in a helical conformation, X-ray crystal structures of FtsZ bound to several accessory proteins shows different conformations of the CTC region. However, without any protein binding to this linker region, CTT remains an intrinsically disordered region, as recently shown by NMR spectroscopy of E. coli FtsZ [113]. Replacing the disordered region with rigid conformations results in slower assembly of FtsZ and aberrant GTPase activity emphasizing that the unstructured nature of the CTT is critical for its function [114].

The function of the C-terminal tail of FtsZ

The flexible CTL is shown to be important for the protofilament formation and lateral interactions of FtsZ in vitro, and for FtsZ’s attachment to the membrane and proper cell division in vivo [114–116]. While the sequence of this disordered region did not affect the functioning of FtsZ, the flexibility, net charge, and length were important factors for CTL function [114]. As seen for EcFtsZ, any disordered peptide of the length between 43 and 95 amino acids forms a viable linker, while longer or shorter peptides do not allow cell division to occur [115]. Increasing the linker peptide length to more than 100 residues in BsFtsZ also leads to perturbation of position of Z-ring and frequency of cell division [114]. Studies indicate that CTL might be linked to the generation of force during constriction and membrane invagination [116]. However, the exact role of the CTL in FtsZ function remains elusive.

While the CTL acts as a ‘spacer’ region for higher-order assemblies of FtsZ, the CTC and CTV stretch acts as a ‘sticker’ for interactions of the C-terminal tail with the core [117,118]. CTL shows auto-inhibitory functions preventing higher-order assemblies of FtsZ and the CTC and CTV stretch of BsFtsZ were shown to be important for the autoregulation of GTP hydrolysis and polymerization [117]. Specifically, the CTV was indicated for an important role in lateral interactions and bundling of B. subtilis FtsZ protofilaments in vitro, the deletion of which results in disrupted cell division [119]. The net charge of CTV also seemed to affect the bundling propensity of the protein [119]. However, the same effect was not observed for E. coli FtsZ, which bears a neutrally charged CTV [119]. The significance of this difference in the net charge of CTV of FtsZ in gram-positive and gram-negative organisms remains to be explored.

The CTC and CTV regions are together called the ‘grappling hook peptide’ due to their association with other proteins of the divisome in the cytoplasm and the plasma membrane [18,114]. Proteins that are associated with the positioning of the Z-ring, such as MinC [120–122], SlmA [29,123,124], and MapZ [125] are shown to bind to this extreme C-terminal region of FtsZ. This region also enables the binding of early divisome proteins, FtsA [126–128], EzrA [129,130], SepF [131], ZipA [109,132–134], and the Zap proteins [135,136] to FtsZ.

SpnFtsZ has an unusually long C-terminal tail with 104 residues (Figure 4). Considering that FtsZ binds to EzrA and FtsA throughout the cell cycle [69] and also binds to MapZ during the initiation of cell division [65], one can speculate that the longer length may be required to accommodate all these proteins bound to FtsZ C-terminal peptide at the same time. As shown in a visual review of S. pneumoniae [61], visualization of the arrangement of the proteins within the Z-ring and their interactions may help to understand the significance of the C-terminal linker lengths and flexibility. It would be interesting to determine whether this C-terminal region and its binding to FtsZ-interacting partners have any influence on the timing of cell division and establishment of pathogenicity/virulence of the bacteria.

Insights from C-terminal tails of eukaryotic tubulin

Little is known about the C-terminal tail of FtsZ, compared with the C-terminal tail of tubulin, a region that is also disordered and highly variable within various tubulin isoforms. Unlike FtsZ, CTT of tubulin is usually shorter in length with 10–20 residues across isoforms and is highly negatively charged (Figure 2) [137]. The variations within the CTT of tubulin are established for fine-tuning of different biochemical functions of the protein without disturbing the core that is essential for polymerization [137]. CTT is a region important for post-translational modifications of the protein that establishes the regulatory mechanism for microtubule binding motors [137–139]. Whether FtsZ’s C-terminal tail plays any such role in the regulation of the bacterial cell cycle remains to be explored. However, like the CTT of FtsZ that binds to FtsZ-associated proteins, the C-terminal tails of tubulin are also important for the binding of several microtubule-associated proteins. In the case of tubulin, the conformations of the CTT region bound to different proteins remain largely unexplored [140]. Cues from the behavior of CTT from tubulin and FtsZ would aid in understanding the biological functions of this versatile and significant region of polymer proteins.

Inhibition of cell division and FtsZ as an alternative strategy for anti-bacterial therapy

The disruption of the tight regulation of cell division in bacteria has been identified as an attractive strategy in the discovery of novel antibiotics. One of the highly sought-after targets is FtsZ, the inhibition of which leads to the cell division block. As described above, FtsZ is at the forefront of a plethora of interactions throughout the cell division process. Anti-FtsZ agents would disrupt FtsZ assembly leading to the collapse of the Z-ring, associated protein interactions, and peptidoglycan assembly. In the absence of a proper cell division machinery, the cells become defective after several rounds of elongation without division, a phenotype known as filamentous cells (Figure 5), and finally lyse.

Targeting cell division and FtsZ for anti-bacterial therapy

Figure 5
Targeting cell division and FtsZ for anti-bacterial therapy

Z-ring dynamics during normal cell division drive the successful separation of daughter cells after cytokinesis. The inhibition of FtsZ by anti-bacterial agents leads to the disruption of the Z-ring dynamics, finally leading to cell division block usually visualized as a filamentous phenotype of the elongated but undivided cells. DIC images of Bacillus subtilis 168 cells bear scale bar of 5 μm (unpublished data) (Cartoon images were created using BioRender.com)

Figure 5
Targeting cell division and FtsZ for anti-bacterial therapy

Z-ring dynamics during normal cell division drive the successful separation of daughter cells after cytokinesis. The inhibition of FtsZ by anti-bacterial agents leads to the disruption of the Z-ring dynamics, finally leading to cell division block usually visualized as a filamentous phenotype of the elongated but undivided cells. DIC images of Bacillus subtilis 168 cells bear scale bar of 5 μm (unpublished data) (Cartoon images were created using BioRender.com)

Close modal

Recent research on the small molecules that bind to FtsZ and inhibit its functioning through various mechanisms, has shown that targeting FtsZ is a highly useful strategy for anti-bacterial therapy. Broad-spectrum antibiotics can be developed against FtsZ, one of the most conserved proteins in bacteria. Several excellent reviews have enumerated the vast number of anti-FtsZ agents discovered and their mechanism of action [6,7,11,15,18,141–145]. A schematic of the different effects of anti-FtsZ agents on FtsZ assembly is depicted in Figure 6.

Mechanism of action of anti-FtsZ drugs

Figure 6
Mechanism of action of anti-FtsZ drugs

Anti-FtsZ agents can affect FtsZ in one or more of the following ways: (i) disrupting the monomer-monomer interaction at the active site and inhibiting polymerization, (ii) increasing or decreasing GTPase activity leading to a reduced net addition of monomers on to the polymer, (iii) inhibiting lateral interactions preventing bundling of protofilaments or promoting the lateral interactions and stabilize the polymers, and (iv) inducing aggregation of filaments or of monomers.

Figure 6
Mechanism of action of anti-FtsZ drugs

Anti-FtsZ agents can affect FtsZ in one or more of the following ways: (i) disrupting the monomer-monomer interaction at the active site and inhibiting polymerization, (ii) increasing or decreasing GTPase activity leading to a reduced net addition of monomers on to the polymer, (iii) inhibiting lateral interactions preventing bundling of protofilaments or promoting the lateral interactions and stabilize the polymers, and (iv) inducing aggregation of filaments or of monomers.

Close modal

Binding pockets of FtsZ

FtsZ has two important binding pockets that have been identified [1,21]. The nucleotide-binding pocket and the inter-domain cleft are shown in Figure 7. Due to the similarity of the nucleotide-binding pocket of FtsZ to that of the eukaryotic tubulin, it is more advantageous to design inhibitors to FtsZ that bind to the inter-domain cleft [1,21]. It is to be noted that during polymerization these two regions come in close contact with each other forming the active site. This indicates that the surrounding areas of the protein are yet to be explored and targeted by small molecules.

Chemical structures of anti-FtsZ agents

Figure 7
Chemical structures of anti-FtsZ agents

Chemical structures of natural and synthetic anti-bacterial agents that target FtsZ have been labeled in blue for those that bind near the nucleotide-binding pocket and red for those that bind near the inter-domain cleft and the T7 loop. The structures labeled in black are given for those compounds whose binding site on FtsZ is not determined yet. Some labels are shortened denotations from Table 1. For further descriptions of the compounds please see Table 1.

Figure 7
Chemical structures of anti-FtsZ agents

Chemical structures of natural and synthetic anti-bacterial agents that target FtsZ have been labeled in blue for those that bind near the nucleotide-binding pocket and red for those that bind near the inter-domain cleft and the T7 loop. The structures labeled in black are given for those compounds whose binding site on FtsZ is not determined yet. Some labels are shortened denotations from Table 1. For further descriptions of the compounds please see Table 1.

Close modal

Along with their structures provided in Figure 7, Table 1 provides a list of anti-FtsZ agents discovered, particularly with activity against pathogens S. aureus, S. pneumoniae, M. tuberculosis, P. aeruginosa, and gram-negative bacteria. The anti-FtsZ agents have been divided according to their binding sites as nucleotide-binding domain binders, inter-domain cleft binders, and unknown (for those drugs that have no information regarding their binding site available).

Table 1
List of anti-FtsZ agents with their source, binding region, mode of action, and activity on bacteria
SourceAnti-FtsZ agentMode of action on FtsZAnti-bacterial activityReferences
Nucleotide-binding domain binders 
Plant-derived Sanguinarine Inhibits bundling and assembly of EcFtsZ protofilament B. subtilis
IC50: 3 ± 1 µM
E. coli
IC50: 14 ± 2.3 µM 
[175
 Berberine Inhibits assembly and GTPase activity of EcFtsZ and SaFtsZ S. aureus
MIC: 128 µg/ml
B. subtilis
MIC: 128 µg/ml
E. coli
MIC: N/A 
[176,177
 Curcumin Increases GTPase activity of EcFtsZ E. coli
MIC80: 100 µM
B. subtilis
MIC:100 µM 
[178
Algae-derived Chrysophaentin A Inhibits polymerization and GTPase activity of EcFtsZ, BsFtsZ, and SaFtsZ S. aureus
MIC: 2 µg/ml
B. subtilis, E. coli
MIC: N/A 
[179–181
Gammaproteo-bacteria Aminodeoxyguanosine triphosphate (ADG-TP) Inhibits EcFtsZ polymerization and its localization during cell division E. coli
MIC: 4–16 µg/ml
Klebsiella pneumonia
MIC: 16 µg/ml 
[182
Synthetic compounds GAL core compound 14 Inhibits GTPase activity of PaFtsZ P. aeruginosa
IC50: 0.6 mM
S. aureus
IC50: 0.6 mM 
[183
 4-Aminofurazan derivative A189 Inhibits assembly of EcFtsZ and cell division E. coli
MIC: 64 µg/ml
S. aureus
MIC: 16 µg/ml 
[184
 Zantrin (Z1) Perturbs EcFtsZ assembly E. coli
MIC: 20 µM
B. subtilis
MIC: 1.25 µM
P. aeruginosa
MIC; 40 µM
S. pneumoniae
MIC: 0.312 µM
S. aureus
MIC: 2.5 µM 
[185
 UCM44 Enhances BsFtsZ assembly B. subtilis
MIC: 16 mg/L
S. aureus
MIC: 16 mg/L
S. pneumoniae
MIC: 32 mg/L
P. aeruginosa
MIC: 64 mg/L
E. coli
MIC: 128 mg/L 
[153
 Benzofuroquinolinium derivative 5 Inhibition of GTPase activity and polymerization of EcFtsZ B. subtilis
MIC: 0.25 µg/ml
S. aureus*
MIC: 0.25–1 µg/ml
E. coli
MIC: 1 µg/ml
P. aeruginosa
MIC: 4 µg/ml 
[186
 Pyrimidine-quinuclidine derivative 1 Inhibits GTPase activity of SaFtsZ S. aureus
MIC: 897.1 µM
E. coli
MIC: 449 µM 
[187
 2,4-Disubstituted-6-thiophenyl- pyrimidine Bb2 Inhibits polymerization and GTPase activity of SaFtsZ B. subtilis
MIC: 2 µg/ml
S. aureus
MIC: 2 µg/ml
E. coli
MIC: 128 µg/ml 
[188
 Biphenyl derivative 28 Inhibits filament dynamics of BsFtsZ B. subtilis
MIC: 5 µM
S. aureus
MIC: 7 µM 
[189
Inter-domain cleft binders 
Plant-derived Cinnamaldehyde Inhibits polymerization and GTPase activity of EcFtsZ E. coli
MIC: 1000 mg/L
B. subtilis
MIC: 500 mg/L
S. aureus
MIC: 250 mg/L 
[190
 Plumbagin Inhibits assembly and GTPase activity of BsFtsZ B. subtilis
MIC: 29 µM
M. smegmatis
MIC: 31 µM 
[147
 Scopoletin (coumarin) Inhibits polymerization and GTPase activity of EcFtsZ M. tuberculosis
MIC: 42 µg/ml
E. coli
MIC: N/A 
[191,192
 Gancaonin I (polyketide) Inhibits GTPase activity of BsFtsZ B. subtilis
MIC: 5 µM 
[193
 p-Coumaric acid Inhibits GTPase activity of EcFtsZ S. pneumoniae
MIC: 20 µg/ml
S. aureus
MIC: 20 µg/ml
B. subtilis
MIC: 20 µg/ml
E. coli
MIC: 80 µg/ml 
[194,195
Actinobacterium Doxorubicin Inhibits assembly and GTPase activity of EcFtsZ E. coli
MIC: 20 µM
B. subtilis
MIC: 10 µM
S. aureus
MIC: 5 µM 
[196
Synthetic compounds BT-benzo-29 Inhibits GTPase activity and assembly of BsFtsZ B. subtilis
IC50: 1 ± 0.2 μM
M. smegmatis
IC50: 1.6 ± 0.4 μM 
[197
 PC190723 Induces bundling of BsFtsZ and SaFtsZ and reduces GTPase activity. S. aureus*
MIC: 0.5–1 µg/ml
B. subtilis
MIC: 1 µg/ml 
[148,149,156
 TXA707 Enhances SaFtsZ assembly S. aureus*
MIC: 2–4 µg/ml 
[158
 CCR-11 Inhibits assembly and GTPase activity of BsFtsZ B. subtilis
MIC: 3 µM 
[143
 SB-RA-2001 Promote assembly and bundling of BsFtsZ and inhibits its GTPase activity M. tuberculosis
MIC: 60 μM
B. subtilis
MIC: 38 µM 
[198
 9-Phenoxy Berberine derivative 2 Inhibits GTPase activity and polymerization of SaFtsZ B. subtilis
MIC: 4 µg/ml
S. aureus
MIC: 2 µg/ml
E. coli
MIC: 32 µg/ml 
[177
 Thiazole Orange Derivative 1 Inhibits GTPase activity and increases the bundling of SaFtsZ and EcFtsZ protofilament B. subtilis
MIC: 1.5 µg/ml
S. aureus*
MIC: 1.5–3 µg/ml
E. coli
MIC: 3 µg/ml
P. aeruginosa
MIC: 6 µg/ml 
[199
 1,3 diketone 3 Inhibits SpnFtsZ polymerization and induces its aggregation B. subtilis
MIC: 11 µM
S. pneumoniae
MIC: 2 µM 
[200
 Thiazole-quinolinium derivative 4a4 Enhances SaFtsZ assembly S. aureus*
MIC: 1–2 µg/ml
B. subtilis
MIC: 1 µg/ml
E. coli*
MIC: 0.5–1 µg/ml
P. aeruginosa
MIC: 64 µg/ml 
[201
 Benzo[d]imidazole-2- carboxamide derivative 1o Inhibits the polymerization of BsFtsZ B. subtilis
MIC: 1.56 µg/ml
M. tuberculosis H37
MIC: 0.78 µg/ml 
[202
 Cinnamaldehyde derivative LXC 36 Inhibits GTPase activity and polymerization of AbFtsZ A. baumannii
MIC: 32 µg/ml 
[203
Unknown binding sites 
Plant Totarol Inhibits assembly and GTPase activity of MtbFtsZ B. subtilis
MIC: 2 µM
M. tuberculosis
MIC: 16 µg/ml 
[204,205
Fungal Viriditoxin Inhibits polymerization and GTPase activity of EcFtsZ S. aureus*
MIC: 4–8 µg/ml
S. pneumoniae*
MIC: 2–32 µg/ml
E. coli
MIC: >64 µg/ml 
[206
Synthesized (but naturally occurring) Vitamin K3 Increases GTPase activity and inhibits the polymerization of SpnFtsZ B. subtilis
MIC: 35 µM
S. pneumoniae
MIC: 35 µM 
[146
 Dichamenetin Inhibits GTPase activity of EcFtsZ S. aureus
MIC: 1.7 µM
B. subtilis
MIC: 1.7 µM
M. smegmatis
MIC: 3.4 µM
E. coli
MIC: N/A 
[207
Synthetic compounds OTBA Promotes assembly and bundling of EcFtsZ and BsFtsZ, decreases the GTPase activity B. subtilis
MIC: 2 µM
E. coli
MIC: N/A 
[208
 2d carbocyclic curcumin Enhances GTPase activity of BsFtsZ and inhibits the assembly B. subtilis
MIC: 0.7 mg/L
S. aureus
MIC: 2 mg/L
Streptococcus pyogenes
MIC: 2 mg/L 
[209
 SRI-3072 (2-alkoxycarbonylamin-o- pyridine) Inhibits polymerization and GTPase activity of MtbFtsZ M. tuberculosis
MIC: 0.25 mg/L 
[210
 TXA6101 Inhibits EcFtsZ polymerization S. aureus (MRSA Clinical Isolate MPW020)
MIC: 0.125 µg/ml
E. coli
MIC: 0.25 µg/mL
K. pneumoniae
MIC: 4 µg/ml
S. sonnei
MIC: 0.25 µg/ml 
[162,163
 TXA6129 Inhibits EcFtsZ polymerization E. coli
MIC - 0.5 µg/mL
K. pneumoniae
MIC: 8 µg/ml
Shigella sonnei
MIC: 0.25 µg/ml 
[163
SourceAnti-FtsZ agentMode of action on FtsZAnti-bacterial activityReferences
Nucleotide-binding domain binders 
Plant-derived Sanguinarine Inhibits bundling and assembly of EcFtsZ protofilament B. subtilis
IC50: 3 ± 1 µM
E. coli
IC50: 14 ± 2.3 µM 
[175
 Berberine Inhibits assembly and GTPase activity of EcFtsZ and SaFtsZ S. aureus
MIC: 128 µg/ml
B. subtilis
MIC: 128 µg/ml
E. coli
MIC: N/A 
[176,177
 Curcumin Increases GTPase activity of EcFtsZ E. coli
MIC80: 100 µM
B. subtilis
MIC:100 µM 
[178
Algae-derived Chrysophaentin A Inhibits polymerization and GTPase activity of EcFtsZ, BsFtsZ, and SaFtsZ S. aureus
MIC: 2 µg/ml
B. subtilis, E. coli
MIC: N/A 
[179–181
Gammaproteo-bacteria Aminodeoxyguanosine triphosphate (ADG-TP) Inhibits EcFtsZ polymerization and its localization during cell division E. coli
MIC: 4–16 µg/ml
Klebsiella pneumonia
MIC: 16 µg/ml 
[182
Synthetic compounds GAL core compound 14 Inhibits GTPase activity of PaFtsZ P. aeruginosa
IC50: 0.6 mM
S. aureus
IC50: 0.6 mM 
[183
 4-Aminofurazan derivative A189 Inhibits assembly of EcFtsZ and cell division E. coli
MIC: 64 µg/ml
S. aureus
MIC: 16 µg/ml 
[184
 Zantrin (Z1) Perturbs EcFtsZ assembly E. coli
MIC: 20 µM
B. subtilis
MIC: 1.25 µM
P. aeruginosa
MIC; 40 µM
S. pneumoniae
MIC: 0.312 µM
S. aureus
MIC: 2.5 µM 
[185
 UCM44 Enhances BsFtsZ assembly B. subtilis
MIC: 16 mg/L
S. aureus
MIC: 16 mg/L
S. pneumoniae
MIC: 32 mg/L
P. aeruginosa
MIC: 64 mg/L
E. coli
MIC: 128 mg/L 
[153
 Benzofuroquinolinium derivative 5 Inhibition of GTPase activity and polymerization of EcFtsZ B. subtilis
MIC: 0.25 µg/ml
S. aureus*
MIC: 0.25–1 µg/ml
E. coli
MIC: 1 µg/ml
P. aeruginosa
MIC: 4 µg/ml 
[186
 Pyrimidine-quinuclidine derivative 1 Inhibits GTPase activity of SaFtsZ S. aureus
MIC: 897.1 µM
E. coli
MIC: 449 µM 
[187
 2,4-Disubstituted-6-thiophenyl- pyrimidine Bb2 Inhibits polymerization and GTPase activity of SaFtsZ B. subtilis
MIC: 2 µg/ml
S. aureus
MIC: 2 µg/ml
E. coli
MIC: 128 µg/ml 
[188
 Biphenyl derivative 28 Inhibits filament dynamics of BsFtsZ B. subtilis
MIC: 5 µM
S. aureus
MIC: 7 µM 
[189
Inter-domain cleft binders 
Plant-derived Cinnamaldehyde Inhibits polymerization and GTPase activity of EcFtsZ E. coli
MIC: 1000 mg/L
B. subtilis
MIC: 500 mg/L
S. aureus
MIC: 250 mg/L 
[190
 Plumbagin Inhibits assembly and GTPase activity of BsFtsZ B. subtilis
MIC: 29 µM
M. smegmatis
MIC: 31 µM 
[147
 Scopoletin (coumarin) Inhibits polymerization and GTPase activity of EcFtsZ M. tuberculosis
MIC: 42 µg/ml
E. coli
MIC: N/A 
[191,192
 Gancaonin I (polyketide) Inhibits GTPase activity of BsFtsZ B. subtilis
MIC: 5 µM 
[193
 p-Coumaric acid Inhibits GTPase activity of EcFtsZ S. pneumoniae
MIC: 20 µg/ml
S. aureus
MIC: 20 µg/ml
B. subtilis
MIC: 20 µg/ml
E. coli
MIC: 80 µg/ml 
[194,195
Actinobacterium Doxorubicin Inhibits assembly and GTPase activity of EcFtsZ E. coli
MIC: 20 µM
B. subtilis
MIC: 10 µM
S. aureus
MIC: 5 µM 
[196
Synthetic compounds BT-benzo-29 Inhibits GTPase activity and assembly of BsFtsZ B. subtilis
IC50: 1 ± 0.2 μM
M. smegmatis
IC50: 1.6 ± 0.4 μM 
[197
 PC190723 Induces bundling of BsFtsZ and SaFtsZ and reduces GTPase activity. S. aureus*
MIC: 0.5–1 µg/ml
B. subtilis
MIC: 1 µg/ml 
[148,149,156
 TXA707 Enhances SaFtsZ assembly S. aureus*
MIC: 2–4 µg/ml 
[158
 CCR-11 Inhibits assembly and GTPase activity of BsFtsZ B. subtilis
MIC: 3 µM 
[143
 SB-RA-2001 Promote assembly and bundling of BsFtsZ and inhibits its GTPase activity M. tuberculosis
MIC: 60 μM
B. subtilis
MIC: 38 µM 
[198
 9-Phenoxy Berberine derivative 2 Inhibits GTPase activity and polymerization of SaFtsZ B. subtilis
MIC: 4 µg/ml
S. aureus
MIC: 2 µg/ml
E. coli
MIC: 32 µg/ml 
[177
 Thiazole Orange Derivative 1 Inhibits GTPase activity and increases the bundling of SaFtsZ and EcFtsZ protofilament B. subtilis
MIC: 1.5 µg/ml
S. aureus*
MIC: 1.5–3 µg/ml
E. coli
MIC: 3 µg/ml
P. aeruginosa
MIC: 6 µg/ml 
[199
 1,3 diketone 3 Inhibits SpnFtsZ polymerization and induces its aggregation B. subtilis
MIC: 11 µM
S. pneumoniae
MIC: 2 µM 
[200
 Thiazole-quinolinium derivative 4a4 Enhances SaFtsZ assembly S. aureus*
MIC: 1–2 µg/ml
B. subtilis
MIC: 1 µg/ml
E. coli*
MIC: 0.5–1 µg/ml
P. aeruginosa
MIC: 64 µg/ml 
[201
 Benzo[d]imidazole-2- carboxamide derivative 1o Inhibits the polymerization of BsFtsZ B. subtilis
MIC: 1.56 µg/ml
M. tuberculosis H37
MIC: 0.78 µg/ml 
[202
 Cinnamaldehyde derivative LXC 36 Inhibits GTPase activity and polymerization of AbFtsZ A. baumannii
MIC: 32 µg/ml 
[203
Unknown binding sites 
Plant Totarol Inhibits assembly and GTPase activity of MtbFtsZ B. subtilis
MIC: 2 µM
M. tuberculosis
MIC: 16 µg/ml 
[204,205
Fungal Viriditoxin Inhibits polymerization and GTPase activity of EcFtsZ S. aureus*
MIC: 4–8 µg/ml
S. pneumoniae*
MIC: 2–32 µg/ml
E. coli
MIC: >64 µg/ml 
[206
Synthesized (but naturally occurring) Vitamin K3 Increases GTPase activity and inhibits the polymerization of SpnFtsZ B. subtilis
MIC: 35 µM
S. pneumoniae
MIC: 35 µM 
[146
 Dichamenetin Inhibits GTPase activity of EcFtsZ S. aureus
MIC: 1.7 µM
B. subtilis
MIC: 1.7 µM
M. smegmatis
MIC: 3.4 µM
E. coli
MIC: N/A 
[207
Synthetic compounds OTBA Promotes assembly and bundling of EcFtsZ and BsFtsZ, decreases the GTPase activity B. subtilis
MIC: 2 µM
E. coli
MIC: N/A 
[208
 2d carbocyclic curcumin Enhances GTPase activity of BsFtsZ and inhibits the assembly B. subtilis
MIC: 0.7 mg/L
S. aureus
MIC: 2 mg/L
Streptococcus pyogenes
MIC: 2 mg/L 
[209
 SRI-3072 (2-alkoxycarbonylamin-o- pyridine) Inhibits polymerization and GTPase activity of MtbFtsZ M. tuberculosis
MIC: 0.25 mg/L 
[210
 TXA6101 Inhibits EcFtsZ polymerization S. aureus (MRSA Clinical Isolate MPW020)
MIC: 0.125 µg/ml
E. coli
MIC: 0.25 µg/mL
K. pneumoniae
MIC: 4 µg/ml
S. sonnei
MIC: 0.25 µg/ml 
[162,163
 TXA6129 Inhibits EcFtsZ polymerization E. coli
MIC - 0.5 µg/mL
K. pneumoniae
MIC: 8 µg/ml
Shigella sonnei
MIC: 0.25 µg/ml 
[163

N/A, Not Available.

Ab FtsZ, Acinetobacter baumannii FtsZ; BsFtsZ, B. subtilis FtsZ; EcFtsZ, E. coli FtsZ; MtbFtsZ, M. tuberculosis FtsZ; PaFtsZ, P. aeruginosa FtsZ; SaFtsZ, S. aureus FtsZ; SpnFtsZ, S. pneumoniae FtsZ.

*

MIC ranges are mentioned where MIC values for more than one strain of the organism is determined.

MIC of the compound was determined when used in combination with efflux pump inhibitor PAβN (100 μg mL−1).

Differential effects of anti-FtsZ agents on FtsZ from different bacteria

As seen from Table 1, most of the studies on anti-FtsZ compounds show the mechanism of action demonstrated in the model rods—B. subtilis and E. coli and their FtsZs. The model organisms have provided an easy and reliable platform for the study and visualization of the activity of anti-FtsZ inside the cells. Several studies have demonstrated the effect of the drugs on SaFtsZ, but only a few of the studies have explored the effect of the compounds on MtbFtsZ, SpnFtsZ, and PaFtsZ (Table 1). While inhibition of GTPase activity and polymerization of EcFtsZ or BsFtsZ provide a reliable indication of the FtsZ-targeting action of the compounds, does the drug show the same effect on FtsZ from other bacteria? Several examples from these studies given below demonstrate that it is not always the case.

We have recently shown that anti-FtsZ agents might show different effects in different bacteria with different phenotypes of FtsZ inhibition. Vitamin K3, a biomolecule of the vitamin K family, was shown to display differential effects on the FtsZ inhibition in B. subtilis, S. pneumoniae, and E. coli [146]. By immunostaining against FtsZ, it was observed that Z-ring disruption was observed in both the bacteria upon treatment with Vitamin K3; however, in B. subtilis there were punctate spots of FtsZ observed in the DNA-free regions of the cells while in S. pneumoniae no such spots were seen. Further, Vitamin K3 did not show inhibition of E. coli or its FtsZ [146]. This goes to show that Vitamin K3 might have different binding pockets within different FtsZ, thereby showing different effects on them or none at all (as in the case of E. coli FtsZ). Another similar compound, plumbagin also showed inhibitory activity against only BsFtsZ while it did not affect EcFtsZ [147]. Several other drugs mentioned in Table 1, also do not affect EcFtsZ, while they show good activity against BsFtsZ.

Evidence of anti-FtsZ targeting agents having differential binding in different FtsZ also comes from the studies on PC190723 [148–151]. PC190723 was shown to be a potent inhibitor of S. aureus with an anti-FtsZ mode of action [148]. PC190723 stabilized the polymerization and reduced the GTPase activity of SaFtsZ and BsFtsZ, and did not show any effect on EcFtsZ, thereby showing PC190723 could be a gram-positive bacterial inhibitor [149]. PC190723 was tested against gram-positive Enterococcus faecalis and S. pneumoniae and their FtsZ [150]. It was observed that these gram-positive bacteria were resistant to PC190723 and their FtsZs were not affected by the drug [150]. Two amino acids are different in the binding pocket of FtsZ from E. faecalis and S. pneumoniae compared with FtsZ of B. subtilis and S. aureus which was attributed to the resistance of these bacteria [150]. Additionally, computational studies established that PC190723 had different binding pockets for different FtsZ from bacteria [151].

The differences in binding affinities of a drug towards different FtsZ also sheds a light on the differences in FtsZ binding pockets. An example would be of a guanidinomethyl biaryl compound 13, which shows similar binding affinities between SaFtsZ and EcFtsZ, but shows lower affinity toward FtsZ from E. faecalis [152]. Synthetic inhibitors UCM05 and UCM44 inhibited the polymerization of BsFtsZ, but, stabilized and induced the polymerization of FtsZ from M. jannaschii, even though the binding pockets were similar [153]. This indicates that the differential activity arose not at the binding stage, but during filament formation. Further, UCM05 and UCM44 did not show a significant effect on EcFtsZ [153].

Even though several anti-FtsZ agents have been identified, no drug has been translated to the clinics. Only one FtsZ-targeting agent TXA709, a bioavailable prodrug of TXA707, is currently undergoing clinical trials [154]. The roadblocks to transitioning of an anti-FtsZ agent from pre-clinical studies to the clinical stage include the lack of broad specificity and low in vivo efficacy. Only a few FtsZ inhibitors have been identified that target both gram-positive and gram-negative organisms. To address these challenges, there needs to be a closer inspection of the characteristics of FtsZ that can render it ‘druggable’.

Lessons from the pre-clinical development of an FtsZ-targeting anti-Staphylococcal drug

An FtsZ-targeting compound, PC190723 (3-[(6-chloro[1,3]thiazolo[5,4-b]pyridin-2-yl)methoxy]-2,6-difluorobenzamide) was discovered to be highly potent against methicillin-resistant Staphylococcus aureus (MRSA) [148,149]. Even though PC190723 and its derivates with increased bioavailability showed promising in vivo efficacy in mouse model, due to poor solubility issues the clinical development of this compound as an anti-staphylococcal drug was hindered [148,155]. A group with TAXIS Pharmaceuticals Ltd. designed the prodrug TXY436 with improved solubility [156] and TXY541 with improved stability, showed potent in vivo efficacy [157]. Further derivation resulted in the design of the prodrug TXA709 that was resistant to metabolic attack [158]. Its active drug TXA707 (Figure 7 and Table 1) is a potent bactericidal agent against multidrug-resistant S. aureus showing low toxicity to mammalian cells [158]. Efficient in vivo translation of the anti-staphylococcal activity demonstrated in murine infection models established TXA709 as a promising candidate for clinical development [159]. TXA709 recently underwent in-human Phase 1 clinical trials and was shown to have no serious adverse events [160]. It is currently being developed as a combinatorial drug with conventional antibiotics for the oral treatment of MRSA [161]. TXA6101 is another derivative that is effective against mutated FtsZ from MRSA that has developed resistance to TXA707 [162]. From the crystal structures of TXA707 and TXA6101 bound to FtsZ [162], it is interesting to note that TXA6101 shows structural flexibility compared to TXA707. This enables it to target the FtsZ from gram-negative bacteria as well, broadening the specificity of these compounds to previously less targeted class of bacteria with respect to anti-FtsZ therapy [163]. TXA6101 and TXA6129 (Figure 7 and Table 1) show activity against Enterobacteriaceae family of pathogens when used in combination with efflux pump inhibitors [163]. As seen with several anti-FtsZ agents in section 4.3, these compounds inhibit E. coli FtsZ polymerization and bundling, in contrast to PC190723 which stabilizes S. aureus FtsZ polymers [149,163].

Exploring FtsZ from pathogenic organisms for anti-FtsZ studies

The examples in the previous section, highlight the need to further understand the ‘conserved’ protein FtsZ, which seems to have a species-specific behavior. By considering only model rod-shaped organisms for elucidating the mechanism of action, there is a significant limitation to the type of effects of the drugs that are studied. The pathogens against whom these drugs need to be identified have several unique and dissimilar characteristics compared to model rods. Screening libraries of potential FtsZ inhibitors in pathogenic organisms will aid in the process of identifying successful candidates for pre-clinical studies. Even if the identified candidates do not show broad-spectrum activity, the established activity of the drug against the pathogen will serve as a platform for the direct development of narrow-spectrum antibiotics. With the alarming increase in antibiotic resistance, narrow-spectrum antibiotics will also become essential for urgent treatment [164].

Alternative binding pockets in FtsZ

Considering the limited binding pockets of FtsZ that have been explored, an interesting binding region would be the C-terminal tail region. Even though several regions of the CTT, like CTL and CTV, are not conserved, the intrinsically disordered region of CTT seems to be important for the functioning of the protein, and for its interactions with other proteins of the divisome. Inhibiting the flexibility of the CTT can be a potential strategy for inhibiting the binding of several accessory proteins to FtsZ. Given that targeting intrinsically disordered regions is challenging, several computational approaches that have yielded successful drugs for other intrinsically disordered proteins can help [165]. One such example is the targeted covalent inhibitors that have shown promising activity against the intrinsically disordered protein, tau [166]. A very recent study involved the design of several peptides with multiple arginine residues that bind to negatively charged residues of the tubulin C-terminal tail [167]. The binding of these macrocyclic peptides to CTT caused the promotion of tubulin polymerization and the shortening of the nucleation phase [167]. This study introduces a viable strategy for targeting the intrinsically disordered CTT of tubulin, that can inspire the development of new strategies for targeting the CTT of FtsZ. Several proteins are known to bind the CTT region of FtsZ, many of which could share the same binding regions. An important finding in this regard is the same binding pocket on Corynebacterium glutamicum FtsZ for two structurally and functionally different proteins SepF and SlmA [110]. This encourages the challenging endeavor of identifying anti-FtsZ agents that target the CTT to impair several cell division-related interactions in the cell.

New strategies

With the discovery of the conformational switch of FtsZ during polymerization, arresting the protein in one of the conformational states is an attractive strategy to target FtsZ [99,102,168,169]. With the inhibition of the conformational change, assembly of FtsZ is prevented, rendering the Z-ring non-dynamic. Inhibitors targeting the inter-domain cleft may serve as potential leads for the design of such inhibitors, that need to bind to the C-terminal domain of FtsZ involved in the conformational change.

Whatever may be the inherent differences between the assembly characteristics of FtsZ from different bacteria, the fact remains that FtsZ is a central player in the cell division mechanism that interacts with several different proteins that are regulated spatiotemporally based on the shape and type of bacteria. This expands the target of anti-bacterial action from just FtsZ to those essential FtsZ-associated proteins and their interacting regions. Also, designing inhibitors based on the interactions between FtsZ and its associated proteins is an emerging field of study [170–172]. However, more research must be done for a clearer understanding of the interaction of FtsZ and its associated proteins, structurally and functionally, before inhibitors can be designed based on it.

FtsZ from different bacteria show different characteristics in vitro, bundling type, GTPase activity, and binding to cations [103,173]. Differences in assembly behavior may also arise due to the C-terminal tail region [119]. The differences in structure and functionality of FtsZ extend to the myriad of functions that the cytoskeletal protein performs within the cells. The differences observed in gram-positive and gram-negative bacteria may arise due to the thicker cell wall of the former, requiring different rates for treadmilling of FtsZ and subsequent assembly of peptidoglycan in the septum. FtsZ treadmilling may also be highly influenced by the accessory proteins which have several unique bacteria-specific roles. It is noted that these differences arise even at the species level. For example, key differences between elongation and PG incorporation between M. tuberculosis and Mycobacterium smegmatis have been recently reported [174]. This further emphasizes that the cell division machinery operates in a species-specific manner. Through this review, we have given a glimpse into the important differences in the cell division machinery and FtsZ in some of the pathogenic species, to reiterate the urgent need for their study. Even though the rods B. subtilis and E. coli have been the backbone research models for cell division studies, the scientific community is now expanding the horizons to pathogenic organisms. More and more evidence of FtsZ being the central protein that has several previously unidentified functions throughout the cell growth is established every day, emphasizing that FtsZ is an excellent target for anti-bacterial therapy. Several anti-bacterial agents that target FtsZ would be tested and studied in pathogenic organisms. With the era of antimicrobial resistance looming over the world, anti-bacterial therapeutic research has much to gain from these studies.

The authors declare that there are no competing interests associated with the manuscript.

This work is supported by a [grant number 37(1727)/19/EMR-II] from Council of Scientific and Industrial Research, Government of India (to D.P.).

Rachana Rao Battaje: Conceptualization, Writing—original draft. Ravikant Piyush: Writing—original draft. Vidyadhar Pratap: Writing—original draft. Dulal Panda: Resources, Supervision, Writing—review & editing.

AMR

anti-microbial resistance

CTC

C-terminal conserved

CTL

C-terminal linker

CTT

C-terminal tail

CTV

C-terminal variable

MRSA

methicillin-resistant Staphylococcus aureus

PBP

penicillin-binding proteins

PG

peptidoglycan

1.
Battaje
R.R.
and
Panda
D.
(
2017
)
Lessons from bacterial homolog of tubulin, FtsZ for microtubule dynamics
.
Endocr. Relat. Cancer
24
,
T1
T21
[PubMed]
2.
Li
Z.
,
Trimble
M.J.
,
Brun
Y.V.
and
Jensen
G.J.
(
2007
)
The structure of FtsZ filaments in vivo suggests a force-generating role in cell division
.
EMBO J.
26
,
4694
4708
[PubMed]
3.
Holden
S.J.
,
Pengo
T.
,
Meibom
K.L.
,
Fernandez Fernandez
C.
,
Collier
J.
and
Manley
S.
(
2014
)
High throughput 3D super-resolution microscopy reveals caulobacter crescentus in vivo Z-ring organization
.
Proc. Natl. Acad. Sci.
111
,
4566
4571
4.
Bisson-Filho
A.W.
,
Hsu
Y.-P.
,
Squyres
G.R.
,
Kuru
E.
,
Wu
F.
,
Jukes
C.
et al.
(
2017
)
Treadmilling by FtsZ filaments drives peptidoglycan synthesis and bacterial cell division
.
Science
355
,
739
743
[PubMed]
5.
Yang
X.
,
Lyu
Z.
,
Miguel
A.
,
McQuillen
R.
,
Huang
K.C.
and
Xiao
J.
(
2017
)
GTPase activity-coupled treadmilling of the bacterial tubulin ftsz organizes septal cell wall synthesis
.
Science
355
,
744
747
[PubMed]
6.
Kapoor
S.
and
Panda
D.
(
2009
)
Targeting FtsZ for antibacterial therapy: a promising avenue
.
Expert Opin. Ther. Targets
13
,
1037
1051
[PubMed]
7.
Panda
D.
,
Bhattacharya
D.
,
Gao
Q.H.
,
Oza
P.M.
,
Lin
H.-Y.J.
,
Hawkins
B.
et al.
(
2016
)
Identification of agents targeting FtsZ assembly
.
Future Med. Chem.
8
,
1111
1132
[PubMed]
8.
Dewachter
L.
,
Verstraeten
N.
,
Fauvart
M.
and
Michiels
J.
(
2018
)
An Integrative view of cell cycle control in Escherichia coli
.
FEMS Microbiol. Rev.
42
,
116
136
[PubMed]
9.
Errington
J.
and
Wu
L.J.
(
2017
)
Cell cycle machinery in Bacillus subtilis
.
Subcell. Biochem.
67
101
[PubMed]
10.
Levin
P.A.
and
Janakiraman
A.
(
2021
)
Localization, assembly, and activation of the Escherichia coli cell division machinery
.
EcoSal Plus
9
,
eESP00222021
[PubMed]
11.
Hurley
K.A.
,
Santos
T.M.A.
,
Nepomuceno
G.M.
,
Huynh
V.
,
Shaw
J.T.
and
Weibel
D.B.
(
2016
)
Targeting the bacterial division protein FtsZ
.
J. Med. Chem.
6975
6998
[PubMed]
12.
Haeusser
D.P.
and
Margolin
W.
(
2016
)
Splitsville: structural and functional insights into the dynamic bacterial Z ring
.
Nat. Rev. Microbiol.
14
,
305
319
[PubMed]
13.
Ortiz
C.
,
Natale
P.
,
Cueto
L.
and
Vicente
M.
(
2016
)
The keepers of the ring: regulators of FtsZ assembly
.
FEMS Microbiol. Rev.
40
,
57
67
[PubMed]
14.
Wagstaff
J.
and
Löwe
J.
(
2018
)
Prokaryotic cytoskeletons: protein filaments organizing small cells
.
Nat. Rev. Microbiol.
16
,
187
201
[PubMed]
15.
Kusuma
K.D.
,
Payne
M.
,
Ung
A.T.
,
Bottomley
A.L.
and
Harry
E.J.
(
2019
)
FtsZ as an antibacterial target: status and guidelines for progressing this avenue
.
ACS Infect. Dis.
5
,
1279
1294
[PubMed]
16.
Du
S.
and
Lutkenhaus
J.
(
2019
)
At the heart of bacterial cytokinesis: the Z ring
.
Trends Microbiol.
27
,
781
791
[PubMed]
17.
Mateos-Gil
P.
,
Tarazona
P.
and
Vélez
M.
(
2019
)
Bacterial cell division: modeling FtsZ assembly and force generation from single filament experimental data
.
FEMS Microbiol. Rev.
43
,
73
87
[PubMed]
18.
Silber
N.
,
Matos De Opitz
C.L.
,
Mayer
C.
and
Sass
P.
(
2020
)
Cell division protein FtsZ: from structure and mechanism to antibiotic target
.
Fut. Microbiol.
801
831
[PubMed]
19.
McQuillen
R.
and
Xiao
J.
(
2020
)
Insights into the structure, function, and dynamics of the bacterial cytokinetic FtsZ-ring
.
Annu. Rev. Biophys.
309
341
[PubMed]
20.
Wang
M.
,
Fang
C.
,
Ma
B.
,
Luo
X.
and
Hou
Z.
(
2020
)
Regulation of cytokinesis: FtsZ and its accessory proteins
.
Curr. Genet.
43
49
[PubMed]
21.
Pradhan
P.
,
Margolin
W.
and
Beuria
T.K.
(
2021
)
Targeting the achilles heel of FtsZ: the interdomain cleft
.
Front. Microbiol.
732796
[PubMed]
22.
Barrows
J.M.
and
Goley
E.D.
(
2021
)
FtsZ dynamics in bacterial division: what, how, and why?
Curr. Opinion Cell Biol.
68
,
163
172
23.
Pang
Z.
,
Raudonis
R.
,
Glick
B.R.
,
Lin
T.J.
and
Cheng
Z.
(
2019
)
Antibiotic resistance in Pseudomonas aeruginosa: mechanisms and alternative therapeutic strategies
.
Biotechnol. Adv.
177
192
[PubMed]
24.
Salje
J.
,
van den Ent
F.
,
de Boer
P.
and
Löwe
J.
(
2011
)
Direct membrane binding by bacterial actin MreB
.
Mol. Cell
43
,
478
487
[PubMed]
25.
Reimold
C.
,
Soufo
H.J.D.
,
Dempwolff
F.
and
Graumann
P.L.
(
2013
)
Motion of variable-length MreB filaments at the bacterial cell membrane influences cell morphology
.
Mol. Biol. Cell
24
,
2340
2349
[PubMed]
26.
Olshausen
P.V.
,
Defeu Soufo
H.J.
,
Wicker
K.
,
Heintzmann
R.
,
Graumann
P.L.
and
Rohrbach
A.
(
2013
)
Superresolution imaging of dynamic MreB filaments in B. subtilis - a multiple-motor-driven transport?
Biophys. J.
105
,
1171
1181
[PubMed]
27.
Varma
A.
and
Young
K.D.
(
2009
)
In Escherichia coli, MreB and FtsZ direct the synthesis of lateral cell wall via independent pathways that require PBP 2
.
J. Bacteriol.
191
,
3526
3533
[PubMed]
28.
Kaimer
C.
and
Graumann
P.L.
(
2011
)
Players between the worlds: multifunctional DNA translocases
.
Curr. Opin. Microbiol.
14
,
719
725
[PubMed]
29.
Bernhardt
T.G.
and
De Boer
P.A.J.
(
2005
)
SlmA, a nucleoid-associated, FtsZ binding protein required for blocking septal ring assembly over chromosomes in E. coli
.
Mol. Cell
18
,
555
564
[PubMed]
30.
Wu
L.J.
and
Errington
J.
(
2004
)
Coordination of cell division and chromosome segregation by a nucleoid occlusion protein in Bacillus subtilis
.
Cell
117
,
915
925
[PubMed]
31.
Patrick
J.E.
and
Kearns
D.B.
(
2008
)
MinJ (YvjD) is a topological determinant of cell division in Bacillus subtilis
.
Mol. Microbiol.
70
,
1166
1179
[PubMed]
32.
Lutkenhaus
J.
(
2007
)
Assembly dynamics of the bacterial MinCDE system and spatial regulation of the z ring
.
Annu. Rev. Biochem.
539
562
[PubMed]
33.
Pichoff
S.
and
Lutkenhaus
J.
(
2002
)
Unique and overlapping roles for ZipA and FtsA in septal ring assembly in Escherichia coli
.
EMBO J.
21
,
685
693
[PubMed]
34.
Jensen
S.O.
,
Thompson
L.S.
and
Harry
E.J.
(
2005
)
Cell division in Bacillus subtilis: FtsZ and FtsA association is Z-ring independent, and FtsA is required for efficient midcell Z-ring assembly
.
J. Bacteriol.
187
,
6536
6544
[PubMed]
35.
Gündoǧdu
M.E.
,
Kawai
Y.
,
Pavlendova
N.
,
Ogasawara
N.
,
Errington
J.
,
Scheffers
D.J.
et al.
(
2011
)
Large ring polymers align FtsZ polymers for normal septum formation
.
EMBO J.
30
,
617
626
[PubMed]
36.
Haeusser
D.P.
,
Schwartz
R.L.
,
Smith
A.M.
,
Oates
M.E.
and
Levin
P.A.
(
2004
)
EzrA prevents aberrant cell division by modulating assembly of the cytoskeletal protein FtsZ
.
Mol. Microbiol.
52
,
801
814
[PubMed]
37.
Gamba
P.
,
Veening
J.W.
,
Saunders
N.J.
,
Hamoen
L.W.
and
Daniel
R.A.
(
2009
)
Two-step assembly dynamics of the Bacillus subtilis divisome
.
J. Bacteriol.
191
,
4186
4194
[PubMed]
38.
Squyres
G.R.
,
Holmes
M.J.
,
Barger
S.R.
,
Pennycook
B.R.
,
Ryan
J.
,
Yan
V.T.
et al.
(
2021
)
Single-molecule imaging reveals that Z-ring condensation is essential for cell division in Bacillus subtilis
.
Nat. Microbiol.
6
,
553
562
[PubMed]
39.
Goehring
N.W.
and
Beckwith
J.
(
2005
)
Diverse paths to midcell: assembly of the bacterial cell division machinery
.
Curr. Biol.
R514
R526
[PubMed]
40.
Aarsman
M.E.G.
,
Piette
A.
,
Fraipont
C.
,
Vinkenvleugel
T.M.F.
,
Nguyen-Distèche
M.
and
Den Blaauwen
T.
(
2005
)
Maturation of the Escherichia coli divisome occurs in two steps
.
Mol. Microbiol.
55
,
1631
1645
[PubMed]
41.
Du
S.
and
Lutkenhaus
J.
(
2017
)
Assembly and activation of the Escherichia coli divisome
.
Mol. Microbiol.
105
,
177
187
[PubMed]
42.
Loose
M.
and
Mitchison
T.J.
(
2014
)
The bacterial cell division proteins FtsA and FtsZ self-organize into dynamic cytoskeletal patterns
.
Nat. Cell Biol.
16
,
38
46
[PubMed]
43.
Schoenemann
K.M.
and
Margolin
W.
(
2017
)
Bacterial division: FtsZ treadmills to build a beautiful wall
.
Curr. Biol.
27
,
R301
R303
[PubMed]
44.
Park
K.T.
,
Pichoff
S.
,
Du
S.
and
Lutkenhaus
J.
(
2021
)
FtsA acts through FtsW to promote cell wall synthesis during cell division in Escherichia coli
.
Proc. Natl. Acad. Sci. U. S. A.
118
,
e2107210118
[PubMed]
45.
Tong
S.Y.C.
,
Davis
J.S.
,
Eichenberger
E.
,
Holland
T.L.
and
Fowler
V.G.J.
(
2015
)
Staphylococcus aureus infections: epidemiology, pathophysiology, clinical manifestations, and management
.
Clin. Microbiol. Rev.
28
,
603
661
[PubMed]
46.
Blázquez
B.
,
Llarrull
L.I.
,
Luque-Ortega
J.R.
,
Alfonso
C.
,
Boggess
B.
and
Mobashery
S.
(
2014
)
Regulation of the expression of the β-lactam antibiotic-resistance determinants in methicillin-resistant Staphylococcus aureus (MRSA)
.
Biochemistry
53
,
1548
1550
[PubMed]
47.
Pinho
M.G.
and
Errington
J.
(
2003
)
Dispersed mode of Staphylococcus Aureus cell wall synthesis in the absence of the division machinery
.
Mol. Microbiol.
50
,
871
881
[PubMed]
48.
Zapun
A.
,
Vernet
T.
and
Pinho
M.G.
(
2008
)
The different shapes of Cocci
.
FEMS Microbiol. Rev.
32
,
345
360
[PubMed]
49.
Pereira
A.R.
,
Hsin
J.
,
Król
E.
,
Tavares
A.C.
,
Flores
P.
,
Hoiczyk
E.
et al.
(
2016
)
FtsZ-dependent elongation of a coccoid bacterium
.
MBio
7
,
e00908
e00916
[PubMed]
50.
Reichmann
N.T.
,
Tavares
A.C.
,
Saraiva
B.M.
,
Jousselin
A.
,
Reed
P.
,
Pereira
A.R.
et al.
(
2019
)
SEDS-BPBP pairs direct lateral and septal peptidoglycan synthesis in Staphylococcus aureus
.
Nat. Microbiol.
4
,
1368
1377
[PubMed]
51.
Myrbråten
I.S.
,
Stamsås
G.A.
,
Chan
H.
,
Angeles
D.M.
,
Knutsen
T.M.
,
Salehian
Z.
et al.
(
2022
)
SmdA is a novel cell morphology determinant in Staphylococcus aureus
.
MBio
13
,
e0340421
[PubMed]
52.
Veiga
H.
,
Jorge
A.M.
and
Pinho
M.G.
(
2011
)
Absence of nucleoid occlusion effector noc impairs formation of orthogonal FtsZ rings during Staphylococcus aureus cell division
.
Mol. Microbiol.
80
,
1366
1380
[PubMed]
53.
Eswara
P.J.
,
Brzozowski
R.S.
,
Viola
M.G.
,
Graham
G.
,
Spanoudis
C.
,
Trebino
C.
et al.
(
2018
)
An essential Staphylococcus aureus cell division protein directly regulates Ftsz dynamics
.
Elife
7
,
e38856
[PubMed]
54.
Hammond
L.R.
,
Sacco
M.D.
,
Khan
S.J.
,
Spanoudis
C.
,
Hough-Neidig
A.
,
Chen
Y.
et al.
(
2022
)
GpsB coordinates cell division and cell surface decoration by wall teichoic acids in Staphylococcus aureus
.
Microbiol. Spectr.
10
,
e0141322
[PubMed]
55.
Stamsås
G.A.
,
Myrbråten
I.S.
,
Straume
D.
,
Salehian
Z.
,
Veening
J.W.
,
Håvarstein
L.S.
et al.
(
2018
)
CozEa and CozEb play overlapping and essential roles in controlling cell division in Staphylococcus aureus
.
Mol. Microbiol.
109
,
615
632
[PubMed]
56.
Monteiro
J.M.
,
Pereira
A.R.
,
Reichmann
N.T.
,
Saraiva
B.M.
,
Fernandes
P.B.
,
Veiga
H.
et al.
(
2018
)
Peptidoglycan synthesis drives an FtsZ-treadmilling-independent step of cytokinesis
.
Nature
554
,
528
532
[PubMed]
57.
Monteiro
J.M.
,
Fernandes
P.B.
,
Vaz
F.
,
Pereira
A.R.
,
Tavares
A.C.
,
Ferreira
M.T.
et al.
(
2015
)
Cell shape dynamics during the staphylococcal cell cycle
.
Nat. Commun.
6
,
8055
[PubMed]
58.
Matias
V.R.F.
and
Beveridge
T.J.
(
2007
)
Cryo-electron microscopy of cell division in Staphylococcus aureus reveals a mid-zone between nascent cross walls
.
Mol. Microbiol.
64
,
195
206
[PubMed]
59.
von Specht
M.
,
García Gabarrot
G.
,
Mollerach
M.
,
Bonofiglio
L.
,
Gagetti
P.
,
Kaufman
S.
et al.
(
2021
)
resistance to β-lactams in Streptococcus pneumoniae
.
Rev. Argent. Microbiol.
53
,
266
271
[PubMed]
60.
Briggs
N.S.
,
Bruce
K.E.
,
Naskar
S.
,
Winkler
M.E.
and
Roper
D.I.
(
2021
)
The pneumococcal divisome: dynamic control of Streptococcus pneumoniae cell division
.
Front. Microbiol.
737396
[PubMed]
61.
Engholm
D.H.
,
Kilian
M.
,
Goodsell
D.S.
,
Andersen
E.S.
and
Kjærgaard
R.S.
(
2017
)
A visual review of the human pathogen Streptococcus pneumoniae
.
FEMS Microbiol. Rev.
41
,
854
879
[PubMed]
62.
Massidda
O.
,
Nováková
L.
and
Vollmer
W.
(
2013
)
From models to pathogens: how much have we learned about Streptococcus pneumoniae cell division?
Environ. Microbiol.
15
,
3133
3157
63.
Todar
K.
(
2012
)
Todar's online textbook of bacteriology
.
Streptococcus Pneumoniae
64.
Garcia
P.S.
,
Simorre
J.-P.
,
Brochier-Armanet
C.
and
Grangeasse
C.
(
2016
)
Cell division of streptococcus pneumoniae: think positive!
.
Curr. Opin. Microbiol.
34
,
18
23
[PubMed]
65.
Fleurie
A.
,
Lesterlin
C.
,
Manuse
S.
,
Zhao
C.
,
Cluzel
C.
,
Lavergne
J.-P.
et al.
(
2014
)
MapZ marks the division sites and positions FtsZ rings in Streptococcus pneumoniae
.
Nature
516
,
259
262
[PubMed]
66.
Holečková
N.
,
Doubravová
L.
,
Massidda
O.
,
Molle
V.
,
Buriánková
K.
,
Benada
O.
et al.
(
2015
)
LocZ is a new cell division protein involved in proper septum placement in Streptococcus pneumoniae
.
Young Larry S., K.D.M., Ed., MBio
6
,
e01700
e01714
67.
Perez
A.J.
,
Cesbron
Y.
,
Shaw
S.L.
,
Bazan Villicana
J.
,
Tsui
H.-C.T.
,
Boersma
M.J.
et al.
(
2019
)
Movement dynamics of divisome proteins and PBP2x:FtsW in cells of Streptococcus pneumoniae
.
Proc. Natl. Acad. Sci.
116
,
3211
3220
68.
Gallay
C.
,
Sanselicio
S.
,
Anderson
M.E.
,
Soh
Y.M.
,
Liu
X.
,
Stamsås
G.A.
et al.
(
2021
)
CcrZ is a pneumococcal spatiotemporal cell cycle regulator that interacts with FtsZ and controls DNA replication by modulating the activity of DnaA
.
Nat. Microbiol.
6
,
1175
1187
[PubMed]
69.
Perez
A.J.
,
Villicana
J.B.
,
Tsui
H.C.T.
,
Danforth
M.L.
,
Benedet
M.
,
Massidda
O.
et al.
(
2021
)
FtsZ-ring regulation and cell division are mediated by essential EzrA and accessory proteins ZapA and ZapJ in Streptococcus pneumoniae
.
Front. Microbiol.
12
,
780864
[PubMed]
70.
Cadby
I.T.
and
Lovering
A.L.
(
2014
)
Molecular surveillance of the subtle septum: discovering a new mode of peptidoglycan synthesis in Streptococci
.
Mol. Microbiol.
94
,
1
4
[PubMed]
71.
Perez
A.J.
,
Boersma
M.J.
,
Bruce
K.E.
,
Lamanna
M.M.
,
Shaw
S.L.
,
Tsui
H.C.T.
et al.
(
2021
)
Organization of peptidoglycan synthesis in nodes and separate rings at different stages of cell division of Streptococcus pneumoniae
.
Mol. Microbiol.
115
,
1152
1169
[PubMed]
72.
Fenton
A.K.
,
El Mortaji
L.
,
Lau
D.T.C.
,
Rudner
D.Z.
and
Bernhardt
T.G.
(
2016
)
CozE is a member of the MreCD complex that directs cell elongation in Streptococcus pneumoniae
.
Nat. Microbiol.
2
,
16237
[PubMed]
73.
WHO
(
2022
)
Tuberculosis
.
World Heal. Organ.
74.
Chai
Q.
,
Zhang
Y.
and
Liu
C.H.
(
2018
)
Mycobacterium tuberculosis: an adaptable pathogen associated with multiple human diseases
.
Front. Cell. Infection Microbiol.
158
75.
Aldridge
B.B.
,
Fernandez-Suarez
M.
,
Heller
D.
,
Ambravaneswaran
V.
,
Irimia
D.
,
Toner
M.
et al.
(
2012
)
Asymmetry and aging of mycobacterial cells lead to variable growth and antibiotic susceptibility
.
Science
335
,
100
104
[PubMed]
76.
Kieser
K.J.
and
Rubin
E.J.
(
2014
)
How sisters grow apart: mycobacterial growth and division
.
Nat. Rev. Microbiol.
550
562
[PubMed]
77.
Baranowski
C.
,
Rego
E.H.
and
Rubin
E.J.
(
2019
)
The dream of a mycobacterium
.
Microbiol. Spectr.
7
,
7.2.38
78.
Logsdon
M.M.
and
Aldridge
B.B.
(
2018
)
Stable regulation of cell cycle events in mycobacteria: insights from inherently heterogeneous bacterial populations
.
Front. Microbiol.
9
,
514
[PubMed]
79.
Kang
C.M.
,
Nyayapathy
S.
,
Lee
J.Y.
,
Suh
J.W.
and
Husson
R.N.
(
2008
)
Wag31, a homologue of the cell division protein DivIVA, regulates growth, morphology and polar cell wall synthesis in mycobacteria
.
Microbiology
154
,
725
735
[PubMed]
80.
Hesper Rego
E.
,
Audette
R.E.
and
Rubin
E.J.
(
2017
)
Deletion of a mycobacterial divisome factor collapses single-cell phenotypic heterogeneity
.
Nature
546
,
153
157
[PubMed]
81.
Daniel
R.A.
and
Errington
J.
(
2003
)
Control of cell morphogenesis in bacteria: two distinct ways to make a rod-shaped cell
.
Cell
113
,
767
776
[PubMed]
82.
Eskandarian
H.A.
,
Odermatt
P.D.
,
Ven
J.X.Y.
,
Hannebelle
M.T.M.
,
Nievergelt
A.P.
,
Dhar
N.
et al.
(
2017
)
Division site selection linked to inherited cell surface wave troughs in mycobacteria
.
Nat. Microbiol.
2
,
17094
[PubMed]
83.
Gola
S.
,
Munder
T.
,
Casonato
S.
,
Manganelli
R.
and
Vicente
M.
(
2015
)
The essential role of sepf in mycobacterial division
.
Mol. Microbiol.
97
,
560
576
[PubMed]
84.
Datta
P.
,
Dasgupta
A.
,
Singh
A.K.
,
Mukherjee
P.
,
Kundu
M.
and
Basu
J.
(
2006
)
Interaction between FtsW and penicillin-binding protein 3 (PBP3) directs PBP3 to mid-cell, controls cell septation and mediates the formation of a trimeric complex involving FtsZ, FtsW and PBP3 in mycobacteria
.
Mol. Microbiol.
62
,
1655
1673
[PubMed]
85.
Rajagopalan
M.
,
Maloney
E.
,
Dziadek
J.
,
Poplawska
M.
,
Lofton
H.
,
Chauhan
A.
et al.
(
2005
)
Genetic evidence that mycobacterial FtsZ and FtsW proteins interact, and colocalize to the division site in Mycobacterium smegmatis
.
FEMS Microbiol. Lett.
250
,
9
17
[PubMed]
86.
Plocinski
P.
,
Ziolkiewicz
M.
,
Kiran
M.
,
Vadrevu
S.I.
,
Nguyen
H.B.
,
Hugonnet
J.
et al.
(
2011
)
Characterization of CrgA, a new partner of the Mycobacterium tuberculosis peptidoglycan polymerization complexes
.
J. Bacteriol.
193
,
3246
3256
[PubMed]
87.
Plocinski
P.
,
Arora
N.
,
Sarva
K.
,
Blaszczyk
E.
,
Qin
H.
,
Das
N.
et al.
(
2012
)
Mycobacterium tuberculosis CwsA interacts with CrgA and Wag31, and the CrgA-CwsA complex is involved in peptidoglycan synthesis and cell shape determination
.
J. Bacteriol.
194
,
6398
6409
[PubMed]
88.
Dhaked
H.P.S.
,
Bhattacharya
A.
,
Yadav
S.
,
Dantu
S.C.
,
Kumar
A.
and
Panda
D.
(
2016
)
Mutation of Arg191 in FtsZ impairs cytokinetic abscission of Bacillus Subtilis cells
.
Biochemistry
55
,
5754
5763
[PubMed]
89.
Gomez
J.E.
and
Bishai
W.R.
(
2000
)
WhmD Is an essential mycobacterial gene required for proper septation and cell division
.
Proc. Natl. Acad. Sci. U. S. A.
97
,
8554
8559
[PubMed]
90.
Wang
R.
and
Ehrt
S.
(
2021
)
Rv0954 Is a member of the mycobacterial cell division complex
.
Front. Microbiol.
12
,
626461
[PubMed]
91.
Yao
Q.
,
Jewett
A.I.
,
Chang
Y.
,
Oikonomou
C.M.
,
Beeby
M.
,
Iancu
C.V.
et al.
(
2017
)
Short FtsZ filaments can drive asymmetric cell envelope constriction at the onset of bacterial cytokinesis
.
EMBO J.
36
,
1577
1589
[PubMed]
92.
Waterhouse
A.M.
,
Procter
J.B.
,
Martin
D.M.A.
,
Clamp
M.
and
Barton
G.J.
(
2009
)
Jalview version 2-A multiple sequence alignment editor and analysis workbench
.
Bioinformatics
25
,
1189
1191
[PubMed]
93.
Madeira
F.
,
Pearce
M.
,
Tivey
A.R.N.
,
Basutkar
P.
,
Lee
J.
,
Edbali
O.
et al.
(
2022
)
Search and sequence analysis tools services from EMBL-EBI in 2022
.
Nucleic Acids Res.
50
,
W276
W279
[PubMed]
94.
Sievers
F.
,
Wilm
A.
,
Dineen
D.
,
Gibson
T.J.
,
Karplus
K.
,
Li
W.
et al.
(
2011
)
Fast, scalable generation of high-quality protein multiple sequence alignments using clustal omega
.
Mol. Syst. Biol.
7
,
539
95.
Löwe
J.
and
Amos
L.A.
(
1998
)
Crystal structure of the bacterial cell-division protein FtsZ
.
Nature
391
,
203
206
[PubMed]
96.
Oliva
M.A.
,
Trambaiolo
D.
and
Löwe
J.
(
2007
)
Structural insights into the conformational variability of FtsZ
.
J. Mol. Biol.
373
,
1229
1242
[PubMed]
97.
Shin
W.H.
and
Kihara
D.
(
2019
)
55 years of the Rossmann fold
.
Methods Mol. Biol.
1958
,
1
13
[PubMed]
98.
Matsui
T.
,
Yamane
J.
,
Mogi
N.
,
Yamaguchi
H.
,
Takemoto
H.
,
Yao
M.
et al.
(
2012
)
Structural reorganization of the bacterial cell-division protein FtsZ from Staphylococcus aureus
.
Acta Crystallogr. Sect. D Biol. Crystallogr.
68
,
1175
1188
99.
Elsen
N.L.
,
Lu
J.
,
Parthasarathy
G.
,
Reid
J.C.
,
Sharma
S.
,
Soisson
S.M.
et al.
(
2012
)
Mechanism of action of the cell-division inhibitor PC190723: modulation of FtsZ assembly cooperativity
.
J. Am. Chem. Soc.
134
,
12342
12345
[PubMed]
100.
Schumacher
M.A.
,
Ohashi
T.
,
Corbin
L.
and
Erickson
H.P.
(
2020
)
High-resolution crystal structures of Escherichia coli FtsZ bound to GDP and GTP
.
Acta Crystallogr. Sect. F Struct. Biol. Commun.
76
,
94
102
101.
Wagstaff
J.M.
,
Tsim
M.
,
Oliva
M.A.
,
García-Sanchez
A.
,
Kureisaite-Ciziene
D.
,
Andreu
J.M.
et al.
(
2017
)
A polymerization-associated structural switch in Ftsz that enables treadmilling of model filaments
.
MBio
8
,
e00254
e00317
[PubMed]
102.
Battaje
R.R.
,
Bhondwe
P.
,
Dhaked
H.P.S.
and
Panda
D.
(
2021
)
Evidence of conformational switch in Streptococcus pneumoniae FtsZ during polymerization
.
Protein Sci.
30
,
523
530
[PubMed]
103.
Jaiswal
R.
and
Panda
D.
(
2009
)
Differential assembly properties of Escherichia coli FtsZ and Mycobacterium tuberculosis FtsZ: an analysis using divalent calcium
.
J. Biochem.
146
,
733
742
[PubMed]
104.
Dhaked
H.P.S.
,
Ray
S.
,
Battaje
R.R.
,
Banerjee
A.
and
Panda
D.
(
2019
)
Regulation of Streptococcus pneumoniae FtsZ assembly by divalent cations: paradoxical effects of Ca2+ on the nucleation and bundling of FtsZ polymers
.
FEBS J.
286
,
3629
3646
[PubMed]
105.
White
E.L.
,
Ross
L.J.
,
Reynolds
R.C.
,
Seitz
L.E.
,
Moore
G.D.
and
Borhani
D.W.
(
2000
)
Slow polymerization of Mycobacterium tuberculosis FtsZ
.
J. Bacteriol.
182
,
4028
4034
[PubMed]
106.
ur Rahman
M.
,
Li
Z.
,
Zhang
T.
,
Du
S.
,
Ma
X.
et al.
(
2020
)
Assembly properties of bacterial tubulin homolog FtsZ regulated by the positive regulator protein ZipA and ZapA from Pseudomonas aeruginosa
.
Sci. Rep.
10
,
21369
[PubMed]
107.
Jumper
J.
,
Evans
R.
,
Pritzel
A.
,
Green
T.
,
Figurnov
M.
,
Ronneberger
O.
et al.
(
2021
)
Highly accurate protein structure prediction with AlphaFold
.
Nature
596
,
583
589
[PubMed]
108.
Varadi
M.
,
Anyango
S.
,
Deshpande
M.
,
Nair
S.
,
Natassia
C.
,
Yordanova
G.
et al.
(
2022
)
AlphaFold Protein Structure Database: massively expanding the structural coverage of protein-sequence space with high-accuracy models
.
Nucleic Acids Res.
50
,
D439
D444
[PubMed]
109.
Mosyak
L.
(
2000
)
The bacterial cell-division protein ZipA and its interaction with an FtsZ fragment revealed by X-ray crystallography
.
EMBO J.
19
,
3179
3191
[PubMed]
110.
Sogues
A.
,
Martinez
M.
,
Gaday
Q.
,
Ben Assaya
M.
,
Graña
M.
,
Voegele
A.
et al.
(
2020
)
Essential dynamic interdependence of FtsZ and SepF for Z-ring and septum formation in Corynebacterium glutamicum
.
Nat. Commun.
11
,
1641
[PubMed]
111.
Szwedziak
P.
,
Wang
Q.
,
Freund
S.M.
and
Löwe
J.
(
2012
)
FtsA forms actin-like protofilaments
.
EMBO J.
31
,
2249
2260
[PubMed]
112.
Schumacher
M.A.
,
Huang
K.H.
,
Zeng
W.
and
Janakiraman
A.
(
2017
)
Structure of the z ring-associated protein, Zapd, bound to the c-terminal domain of the tubulin-like protein, Ftsz, suggests mechanism of z ring stabilization through Ftsz cross-linking
.
J. Biol. Chem.
292
,
3740
3750
[PubMed]
113.
Viola
M.G.
,
Perdikari
T.M.
,
Trebino
C.E.
,
Rahmani
N.
,
Mathews
K.L.
,
Pena
C.M.
et al.
(
2022
)
An enhancer sequence in the intrinsically disordered region of FtsZ promotes polymer-guided substrate processing by ClpXP protease
.
Protein Sci.
31
,
e4306
[PubMed]
114.
Buske
P.J.
and
Levin
P.A.
(
2013
)
A flexible C-terminal linker is required for proper FtsZ assembly in vitro and cytokinetic ring formation in vivo
.
Mol. Microbiol.
89
,
249
263
[PubMed]
115.
Gardner
K.A.J.A.
,
Moore
D.A.
and
Erickson
H.P.
(
2013
)
The C-terminal linker of Escherichia ColiFtsZ functions as an intrinsically disordered peptide
.
Mol. Microbiol.
89
,
264
275
[PubMed]
116.
Buske
P.J.
,
Mittal
A.
,
Pappu
R.V.
and
Levin
P.A.
(
2015
)
An intrinsically disordered linker plays a critical role in bacterial cell division
.
Semin. Cell Dev. Biol.
3
10
[PubMed]
117.
Cohan
M.C.
,
Eddelbuettel
A.M.P.
,
Levin
P.A.
and
Pappu
R.V.
(
2020
)
Dissecting the functional contributions of the intrinsically disordered C-terminal tail of Bacillus subtilis FtsZ
.
J. Mol. Biol.
432
,
3205
3221
[PubMed]
118.
Huecas
S.
,
Ramírez-Aportela
E.
,
Vergoñós
A.
,
Núñez-Ramírez
R.
,
Llorca
O.
,
Díaz
J.F.
et al.
(
2017
)
Self-organization of FtsZ polymers in solution reveals spacer role of the disordered C-terminal tail
.
Biophys. J.
113
,
1831
1844
[PubMed]
119.
Buske
P.J.
and
Levin
P.A.
(
2012
)
Extreme C terminus of bacterial cytoskeletal protein FtsZ plays fundamental role in assembly independent of modulatory proteins
.
J. Biol. Chem.
287
,
10945
10957
[PubMed]
120.
Blasios
V.
,
Bisson-Filho
A.W.
,
Castellen
P.
,
Nogueira
M.L.C.
,
Bettini
J.
,
Portugal
R.V.
et al.
(
2013
)
Genetic and biochemical characterization of the MinC-FtsZ interaction in Bacillus subtilis
.
PloS ONE
8
,
e60690
[PubMed]
121.
Hu
Z.
,
Mukherjee
A.
,
Pichoff
S.
and
Lutkenhaus
J.
(
1999
)
The MinC component of the division site selection system in Escherichia coli interacts with FtsZ to prevent polymerization
.
Proc. Natl. Acad. Sci. U.S.A.
96
,
14819
14824
[PubMed]
122.
Dajkovic
A.
,
Lan
G.
,
Sun
S.X.
,
Wirtz
D.
and
Lutkenhaus
J.
(
2008
)
MinC spatially controls bacterial cytokinesis by antagonizing the scaffolding function of FtsZ
.
Curr. Biol.
18
,
235
244
[PubMed]
123.
Schumacher
M.A.
and
Zeng
W.
(
2016
)
Structures of the nucleoid occlusion protein SlmA bound to DNA and the C-terminal domain of the cytoskeletal protein FtsZ
.
Proc. Natl. Acad. Sci. U. S. A.
113
,
4988
4993
[PubMed]
124.
Du
S.
and
Lutkenhaus
J.
(
2014
)
SlmA antagonism of FtsZ assembly employs a two-pronged mechanism like MinCD
.
PLos Genet.
10
,
e1004460
[PubMed]
125.
Hosek
T.
,
Bougault
C.M.
,
Lavergne
J.P.
,
Martinez
D.
,
Ayala
I.
,
Fenel
D.
et al.
(
2020
)
Structural features of the interaction of MapZ with FtsZ and membranes in Streptococcus pneumoniae
.
Sci. Rep.
10
,
4051
[PubMed]
126.
Wang
X.
,
Huang
J.
,
Mukherjee
A.
,
Cao
C.
and
Lutkenhaus
J.
(
1997
)
Analysis of the interaction of FtsZ with Itself, GTP, and FtsA
.
J. Bacteriol.
179
,
5551
5559
[PubMed]
127.
Yan
K.
,
Pearce
K.H.
and
Payne
D.J.
(
2000
)
A conserved residue at the extreme C-terminus of FtsZ is critical for the FtsA-FtsZ interaction in Staphylococcus aureus
.
Biochem. Biophys. Res. Commun.
270
,
387
392
[PubMed]
128.
Ma
X.
and
Margolin
W.
(
1999
)
Genetic and functional analyses of the conserved C-terminal core domain of Escherichia coli FtsZ
.
J. Bacteriol.
181
,
7531
7544
[PubMed]
129.
Levin
P.A.
,
Kurtser
I.G.
and
Grossman
A.D.
(
1999
)
Identification and characterization of a negative regulator of FtsZ ring formation in Bacillus subtilis
.
Proc. Natl. Acad. Sci. U.S.A.
96
,
9642
9647
[PubMed]
130.
Singh
J.K.
,
Makde
R.D.
,
Kumar
V.
and
Panda
D.
(
2007
)
A membrane protein, EzrA, regulates assembly dynamics of FtsZ by interacting with the C-terminal tail of FtSZ
.
Biochemistry
46
,
11013
11022
[PubMed]
131.
Król
E.
,
van Kessel
S.P.
,
van Bezouwen
L.S.
,
Kumar
N.
,
Boekema
E.J.
and
Scheffers
D.J.
(
2012
)
Bacillus subtilis SepF binds to the C-terminus of FtsZ
.
PloS ONE
7
,
e43293
[PubMed]
132.
Pazos
M.
,
Natale
P.
and
Vicente
M.
(
2013
)
A specific role for the ZipA protein in cell division: stabilization of the FtsZ protein
.
J. Biol. Chem.
288
,
3219
3226
[PubMed]
133.
Hale
C.A.
and
De Boer
P.A.J.
(
1997
)
Direct binding of FtsZ to ZipA, an essential component of the septal ring structure that mediates cell division in E. coli
.
Cell
88
,
175
185
[PubMed]
134.
Hale
C.A.
,
Rhee
A.C.
and
De Boer
P.A.J.
(
2000
)
ZipA-induced bundling of FtsZ polymers mediated by an interaction between C-terminal domains
.
J. Bacteriol.
182
,
5153
5166
[PubMed]
135.
Durand-Heredia
J.
,
Rivkin
E.
,
Fan
G.
,
Morales
J.
and
Janakiraman
A.
(
2012
)
Identification of ZapD as a cell division factor that promotes the assembly of FtsZ in Escherichia coli
.
J. Bacteriol.
194
,
3189
3198
[PubMed]
136.
Durand-Heredia
J.M.
,
Yu
H.H.
,
De Carlo
S.
,
Lesser
C.F.
and
Janakiraman
A.
(
2011
)
Identification and characterization of ZapC, a stabilizer of the FtsZ ring in Escherichia coli
.
J. Bacteriol.
193
,
1405
1413
[PubMed]
137.
Roll-Mecak
A.
(
2015
)
Intrinsically disordered tubulin tails: complex tuners of microtubule functions?
Sem. Cell Dev. Biol.
37
,
11
19
[PubMed]
138.
Prassanawar
S.S.
and
Panda
D.
(
2019
)
Tubulin heterogeneity regulates functions and dynamics of microtubules and plays a role in the development of drug resistance in cancer
.
Biochem. J.
476
,
1359
1376
[PubMed]
139.
Janke
C.
and
Magiera
M.M.
(
2020
)
The tubulin code and its role in controlling microtubule properties and functions
.
Nat. Rev. Mol. Cell Biol.
21
,
307
326
[PubMed]
140.
Wall
K.P.
,
Pagratis
M.
,
Armstrong
G.
,
Balsbaugh
J.L.
,
Verbeke
E.
,
Pearson
C.G.
et al.
(
2016
)
Molecular determinants of tubulin’s C-terminal tail conformational ensemble
.
ACS Chem. Biol.
11
,
2981
2990
[PubMed]
141.
Rothfield
L.
,
Justice
S.
and
García-Lara
J.
(
1999
)
Bacterial cell division
.
Annu. Rev. Genet.
33
,
423
448
[PubMed]
142.
Margolin
W.
(
2000
)
Themes and variations in prokaryotic cell division
.
FEMS Microbiol. Rev.
24
,
531
548
[PubMed]
143.
Singh
P.
,
Jindal
B.
,
Surolia
A.
and
Panda
D.
(
2012
)
A rhodanine derivative CCR-11 inhibits bacterial proliferation by inhibiting the assembly and GTPase activity of FtsZ
.
Biochemistry
51
,
5434
5442
[PubMed]
144.
Ojima
I.
,
Kumar
K.
,
Awasthi
D.
and
Vineberg
J.G.
(
2014
)
Drug discovery targeting cell division proteins, microtubules and FtsZ
.
Bioorg. Med. Chem.
22
,
5060
5077
[PubMed]
145.
Han
H.
,
Wang
Z.
,
Li
T.
,
Teng
D.
,
Mao
R.
,
Hao
Y.
et al.
(
2021
)
Recent Progress of Bacterial FtsZ Inhibitors with a Focus on Peptides
.
FEBS J.
288
,
1091
1106
[PubMed]
146.
Pushpakaran
A.
,
Battaje
R.R.
and
Panda
D.
(
2022
)
Vitamin K3 inhibits FtsZ assembly, disrupts the Z-ring in Streptococcus pneumoniae and displays anti-pneumococcal activity
.
Biochem. J.
479
,
1543
1558
[PubMed]
147.
Bhattacharya
A.
,
Jindal
B.
,
Singh
P.
,
Datta
A.
and
Panda
D.
(
2013
)
Plumbagin inhibits cytokinesis in Bacillus subtilis by inhibiting FtsZ assembly - a mechanistic study of its antibacterial activity
.
FEBS J.
280
,
4585
4599
[PubMed]
148.
Haydon
D.J.
,
Stokes
N.R.
,
Ure
R.
,
Galbraith
G.
,
Bennett
J.M.
,
Brown
D.R.
et al.
(
2008
)
An inhibitor of FtsZ with potent and selective anti-staphylococcal activity
.
Science
321
,
1673
1675
[PubMed]
149.
Andreu
J.M.
,
Schaffner-Barbero
C.
,
Huecas
S.
,
Alonso
D.
,
Lopez-Rodriguez
M.L.
,
Ruiz-Avila
L.B.
et al.
(
2010
)
The antibacterial cell division inhibitor PC190723 is an FtsZ polymer-stabilizing agent that induces filament assembly and condensation
.
J. Biol. Chem.
285
,
14239
14246
[PubMed]
150.
Kaul
M.
,
Zhang
Y.
,
Parhi
A.K.
,
Lavoie
E.J.
,
Tuske
S.
,
Arnold
E.
et al.
(
2013
)
Enterococcal and streptococcal resistance to PC190723 and related compounds: molecular insights from a FtsZ mutational analysis
.
Biochimie
95
,
1880
1887
[PubMed]
151.
Miguel
A.
,
Hsin
J.
,
Liu
T.
,
Tang
G.
,
Altman
R.B.
and
Huang
K.C.
(
2015
)
Variations in the binding pocket of an inhibitor of the bacterial division protein FtsZ across genotypes and species
.
PLoS Comput. Biol.
11
,
e1004117
[PubMed]
152.
Kaul
M.
,
Parhi
A.K.
,
Zhang
Y.
,
Lavoie
E.J.
,
Tuske
S.
,
Arnold
E.
et al.
(
2012
)
A bactericidal guanidinomethyl biaryl that alters the dynamics of bacterial FtsZ polymerization
.
J. Med. Chem.
55
,
10160
10176
[PubMed]
153.
Ruiz-Avila
L.B.
,
Huecas
S.
,
Artola
M.
,
Vergoñós
A.
,
Ramírez-Aportela
E.
,
Cercenado
E.
et al.
(
2013
)
Synthetic inhibitors of bacterial cell division targeting the GTP-binding site of FtsZ
.
ACS Chem. Biol.
8
,
2072
2083
[PubMed]
154.
Butler
M.S.
and
Paterson
D.L.
(
2020
)
Antibiotics in the clinical pipeline in October 2019
.
J. Antibiot.
,
73
329
364
155.
Stokes
N.R.
,
Baker
N.
,
Bennett
J.M.
,
Berry
J.
,
Collins
I.
,
Czaplewski
L.G.
et al.
(
2013
)
An improved small-molecule inhibitor of FtsZ with superior in vitro potency, drug-likeproperties, and in vivo efficacy
.
Antimicrob. Agents Chemother.
57
,
317
325
[PubMed]
156.
Kaul
M.
,
Mark
L.
,
Zhang
Y.
,
Parhi
A.K.
,
Lavoie
E.J.
and
Pilch
D.S.
(
2013
)
An FtsZ-targeting prodrug with oral antistaphylococcal efficacy in vivo
.
Antimicrob. Agents Chemother.
57
,
5860
5869
[PubMed]
157.
Kaul
M.
,
Mark
L.
,
Zhang
Y.
,
Parhi
A.K.
,
Lavoie
E.J.
and
Pilch
D.S.
(
2013
)
Pharmacokinetics and in vivo antistaphylococcal efficacy of TXY541, a 1-methylpiperidine-4-carboxamide prodrug of PC190723
.
Biochem. Pharmacol.
86
,
1699
1707
[PubMed]
158.
Kaul
M.
,
Mark
L.
,
Zhang
Y.
,
Parhi
A.K.
,
Lyu
Y.L.
,
Pawlak
J.
et al.
(
2015
)
TXA709, an FtsZ-targeting benzamide prodrug with improved pharmacokinetics and enhanced in vivo efficacy against methicillin-resistant Staphylococcus aureus
.
Antimicrob. Agents Chemother.
59
,
4845
4855
[PubMed]
159.
Lepak
A.J.
,
Parhi
A.
,
Madison
M.
,
Marchillo
K.
,
Vanhecker
J.
and
Andes
D.R.
(
2015
)
In vivo pharmacodynamic evaluation of an FtsZ inhibitor, TXA-709, and its active metabolite, TXA-707, in a murine neutropenic thigh infection model
.
Antimicrob. Agents Chemother.
59
,
6568
6574
[PubMed]
160.
TAXIS Pharmaceuticals Inc
.
161.
Kaul
M.
,
Mark
L.
,
Parhi
A.K.
,
LaVoie
E.J.
and
Pilch
D.S.
(
2016
)
Combining the FtsZ-targeting prodrug TXA709 and the cephalosporin cefdinir confers synergy and reduces the frequency of resistance in methicillin-resistant Staphylococcus aureus
.
Antimicrob. Agents Chemother.
60
,
4290
6
162.
Fujita
J.
,
Maeda
Y.
,
Mizohata
E.
,
Inoue
T.
,
Kaul
M.
,
Parhi
A.K.
et al.
(
2017
)
Structural flexibility of an inhibitor overcomes drug resistance mutations in Staphylococcus Aureus FtsZ
.
ACS Chem. Biol.
12
,
1947
1955
[PubMed]
163.
Rosado-Lugo
J.D.
,
Sun
Y.
,
Banerjee
A.
,
Cao
Y.
,
Datta
P.
,
Zhang
Y.
et al.
(
2022
)
Evaluation of 2,6-difluoro-3-(Oxazol-2-Ylmethoxy)benzamide chemotypes as gram-negative FtsZ inhibitors
.
J. Antibiot. (Tokyo)
75
,
385
395
[PubMed]
164.
Melander
R.J.
,
Zurawski
D.V.
and
Melander
C.
(
2018
)
Narrow-spectrum antibacterial agents
.
Med. Chem. Comm.
9
,
12
21
[PubMed]
165.
Ruan
H.
,
Sun
Q.
,
Zhang
W.
,
Liu
Y.
and
Lai
L.
(
2019
)
Targeting intrinsically disordered proteins at the edge of chaos
.
Drug Discov. Today
24
,
217
227
[PubMed]
166.
Petri
L.
,
Ábrányi-Balogh
P.
,
Vagrys
D.
,
Imre
T.
,
Varró
N.
,
Mándity
I.
et al.
(
2022
)
A covalent strategy to target intrinsically disordered proteins: discovery of novel tau aggregation inhibitors
.
Eur. J. Med. Chem.
231
,
114163
167.
Li
S.
,
Mori
M.
,
Yang
M.
,
Elfazazi
S.
,
Hortigüela
R.
,
Chan
P.
et al.
(
2023
)
Targeting the tubulin C-terminal tail by charged small molecules
.
Org. Biomol. Chem.
21
,
153
162
168.
Tan
C.M.
,
Therien
A.G.
,
Lu
J.
,
Lee
S.H.
,
Caron
A.
,
Gill
C.J.
et al.
(
2012
)
Restoring methicillin-resistant Staphylococcus aureus susceptibility to -lactam antibiotics
.
Sci. Transl. Med.
4
,
126ra35
126ra35
[PubMed]
169.
Artola
M.
,
Ruíz-Avila
L.B.
,
Ramírez-Aportela
E.
,
Martínez
R.F.
,
Araujo-Bazán
L.
,
Vázquez-Villa
H.
et al.
(
2017
)
The structural assembly switch of cell division protein FtsZ Probed with fluorescent allosteric inhibitors
.
Chem. Sci.
8
,
1525
1534
[PubMed]
170.
Sutherland
A.G.
,
Alvarez
J.
,
Ding
W.
,
Foreman
K.W.
,
Kenny
C.H.
,
Labthavikul
P.
et al.
(
2003
)
Structure-Based Design of Carboxybiphenylindole Inhibitors of the ZipA-FtsZ Interaction
.
Org. Biomol. Chem.
1
,
4138
4140
[PubMed]
171.
Tsao
D.H.H.
,
Sutherland
A.G.
,
Jennings
L.D.
,
Li
Y.
,
Rush
T.S.
,
Alvarez
J.C.
et al.
(
2006
)
Discovery of Novel Inhibitors of the ZipA/FtsZ Complex by NMR Fragment Screening Coupled with Structure-Based Design
.
Bioorganic Med. Chem.
14
,
7953
7961
172.
Paradis-Bleau
C.
,
Sanschagrin
F.
and
Levesque
R.C.
(
2005
)
Peptide Inhibitors of the Essential Cell Division Protein FtsA
.
Protein Eng. Des. Sel.
18
,
85
91
[PubMed]
173.
Sundararajan
K.
,
Vecchiarelli
A.
,
Mizuuchi
K.
and
Goley
E.D.
(
2018
)
Species- and C-Terminal Linker-Dependent Variations in the Dynamic Behavior of FtsZ on Membranes in Vitro
.
Mol. Microbiol.
110
,
47
63
[PubMed]
174.
Botella
H.
,
Yang
G.
,
Ouerfelli
O.
,
Ehrt
S.
,
Nathan
C.F.
and
Vaubourgeix
J.
(
2017
)
Distinct Spatiotemporal Dynamics of Peptidoglycan Synthesis between Mycobacterium Smegmatis and Mycobacterium Tuberculosis
.
MBio
8
,
e01183
e01217
[PubMed]
175.
Beuria
T.K.
,
Santra
M.K.
and
Panda
D.
(
2005
)
Sanguinarine blocks cytokinesis in bacteria by inhibiting FtsZ assembly a bundling
.
Biochemistry
44
,
16584
16593
[PubMed]
176.
Domadia
P.N.
,
Bhunia
A.
,
Sivaraman
J.
,
Swarup
S.
and
Dasgupta
D.
(
2008
)
Berberine targets assembly of Escherichia coli cell division protein FtsZ
.
Biochemistry
47
,
3225
3234
[PubMed]
177.
Sun
N.
,
Chan
F.Y.
,
Lu
Y.J.
,
Neves
M.A.C.
,
Lui
H.K.
,
Wang
Y.
et al.
(
2014
)
Rational design of berberine-based FtsZ inhibitors with broad-spectrum antibacterial activity
.
PloS ONE
9
,
e97514
178.
Rai
D.
,
Singh
J.K.
,
Roy
N.
and
Panda
D.
(
2008
)
Curcumin inhibits FtsZ assembly: an attractive mechanism for its antibacterial activity
.
Biochem. J.
410
,
147
155
[PubMed]
179.
Plaza
A.
,
Keffer
J.L.
,
Bifulco
G.
,
Lloyd
J.R.
and
Bewley
C.A.
(
2010
)
Chrysophaentins A-H, Antibacterial Bisdiarylbutene Macrocycles That Inhibit the Bacterial Cell Division Protein FtsZ
.
J. Am. Chem. Soc.
132
,
9069
9077
[PubMed]
180.
Keffer
J.L.
,
Huecas
S.
,
Hammill
J.T.
,
Wipf
P.
,
Andreu
J.M.
and
Bewley
C.A.
(
2013
)
Chrysophaentins Are Competitive Inhibitors of FtsZ and Inhibit Z-Ring Formation in Live Bacteria
.
Bioorganic Med. Chem.
21
,
5673
5678
181.
Keffer
J.L.
,
Hammill
J.T.
,
Lloyd
J.R.
,
Plaza
A.
,
Wipf
P.
and
Bewley
C.A.
(
2012
)
Geographic Variability and Anti-Staphylococcal Activity of the Chrysophaentins and Their Synthetic Fragments
.
Mar. Drugs
10
,
1103
1125
[PubMed]
182.
Negar
S.
,
Boyuan
W.
,
Yu
I.
,
Miho
M.
,
Sangkeun
S.
,
Libang
L.
et al.
(
2022
)
A Silent Operon of Photorhabdus Luminescens Encodes a Prodrug Mimic of GTP
.
MBio
13
,
e00700
e00722
[PubMed]
183.
Paradis-Bleau
C.
,
Beaumont
M.
,
Sanschagrin
F.
,
Voyer
N.
and
Levesque
R.C.
(
2007
)
Parallel Solid Synthesis of Inhibitors of the Essential Cell Division FtsZ Enzyme as a New Potential Class of Antibacterials
.
Bioorganic Med. Chem.
15
,
1330
1340
184.
Ito
H.
,
Ura
A.
,
Oyamada
Y.
,
Tanitame
A.
,
Yoshida
H.
,
Yamada
S.
et al.
(
2006
)
A 4-Aminofurazan Derivative - A189 - Inhibits Assembly of Bacterial Cell Division Protein FtsZ in Vitro and in Vivo
.
Microbiol. Immunol.
50
,
759
764
[PubMed]
185.
Margalit
D.N.
,
Romberg
L.
,
Mets
R.B.
,
Hebert
A.M.
,
Mitchison
T.J.
,
Kirschner
M.W.
et al.
(
2004
)
Targeting Cell Division: Small-Molecule Inhibitors of FtsZ GTPase Perturb Cytokinetic Ring Assembly and Induce Bacterial Lethality
.
Proc. Natl. Acad. Sci. U. S. A.
101
,
11821
11826
[PubMed]
186.
Zheng
Y.Y.
,
Du
R.L.
,
Cai
S.Y.
,
Liu
Z.H.
,
Fang
Z.Y.
,
Liu
T.
et al.
(
2018
)
Study of Benzofuroquinolinium Derivatives as a New Class of Potent Antibacterial Agent and the Mode of Inhibition Targeting FtsZ
.
Front. Microbiol.
9
[PubMed]
187.
Chan
F.Y.
,
Sun
N.
,
Neves
M.A.C.
,
Lam
P.C.H.
,
Chung
W.H.
,
Wong
L.K.
et al.
(
2013
)
Identification of a New Class of FtsZ Inhibitors by Structure-Based Design and in Vitro Screening
.
J. Chem. Inf. Model.
53
,
2131
2140
[PubMed]
188.
Fang
Z.
,
Zheng
S.
,
Chan
K.F.
,
Yuan
W.
,
Guo
Q.
,
Wu
W.
et al.
(
2019
)
Design, synthesis and antibacterial evaluation of 2,4-disubstituted-6-thiophenyl-pyrimidines
.
Eur. J. Med. Chem.
161
,
141
153
[PubMed]
189.
Artola
M.
,
Ruiz-Avila
L.B.
,
Vergonos
A.
,
Huecas
S.
,
Araujo-Bazán
L.
,
Martin-Fontecha
M.
et al.
(
2015
)
Effective GTP-replacing FtsZ Inhibitors and antibacterial mechanism of action
.
ACS Chem. Biol.
10
,
834
843
[PubMed]
190.
Domadia
P.
,
Swarup
S.
,
Bhunia
A.
,
Sivaraman
J.
and
Dasgupta
D.
(
2007
)
Inhibition of bacterial cell division protein FtsZ by cinnamaldehyde
.
Biochem. Pharmacol.
74
,
831
840
[PubMed]
191.
Duggirala
S.
,
Nankar
R.P.
,
Rajendran
S.
and
Doble
M.
(
2014
)
Phytochemicals as Inhibitors of bacterial cell division protein FtsZ: coumarins are promising candidates
.
Appl. Biochem. Biotechnol.
174
,
283
296
[PubMed]
192.
Chiang
C.C.
,
Cheng
M.J.
,
Peng
C.F.
,
Huang
H.Y.
and
Chen
I.S.
(
2010
)
A Novel Dimeric Coumarin Analog and Antimycobacterial Constituents from Fatoua Pilosa
.
Chem. Biodivers.
7
,
1728
1736
[PubMed]
193.
Matsui
T.
,
Lallo
S.
,
Nisa
K.
and
Morita
H.
(
2017
)
Filamenting Temperature-Sensitive Mutant Z Inhibitors from Glycyrrhiza Glabra and Their Inhibitory Mode of Action
.
Bioorganic Med. Chem. Lett.
27
,
1420
1424
194.
Hemaiswarya
S.
,
Soudaminikkutty
R.
,
Narasumani
M.L.
and
Doble
M.
(
2011
)
Phenylpropanoids inhibit protofilament formation of Escherichia coli cell division protein FtsZ
.
J. Med. Microbiol.
60
,
1317
1325
[PubMed]
195.
Lou
Z.
,
Wang
H.
,
Rao
S.
,
Sun
J.
,
Ma
C.
and
Li
J.
(
2012
)
P-coumaric acid kills bacteria through dual damage mechanisms
.
Food Control
25
,
550
554
196.
Panda
P.
,
Taviti
A.C.
,
Satpati
S.
,
Kar
M.M.
,
Dixit
A.
and
Beuria
T.K.
(
2015
)
Doxorubicin Inhibits E. Coli Division by Interacting at a Novel Site in FtsZ
.
Biochem. J.
471
,
335
346
[PubMed]
197.
Ray
S.
,
Jindal
B.
,
Kunal
K.
,
Surolia
A.
and
Panda
D.
(
2015
)
BT-Benzo-29 inhibits bacterial cell proliferation by perturbing FtsZ assembly
.
FEBS J.
282
,
4015
4033
[PubMed]
198.
Singh
D.
,
Bhattacharya
A.
,
Rai
A.
,
Dhaked
H.P.S.
,
Awasthi
D.
,
Ojima
I.
et al.
(
2014
)
SB-RA-2001 Inhibits bacterial proliferation by targeting FtsZ assembly
.
Biochemistry
53
,
2979
2992
[PubMed]
199.
Sun
N.
,
Lu
Y.J.
,
Chan
F.Y.
,
Du
R.L.
,
Zheng
Y.-Y.
et al.
(
2017
)
A thiazole orange derivative targeting the bacterial protein FtsZ shows potent antibacterial activity
.
Front. Microbiol.
8
,
855
[PubMed]
200.
Lin
H.-Y.J.
,
Battaje
R.R.
,
Tan
J.
,
Doddareddy
M.
,
Dhaked
H.P.
,
Srivastava
S.
et al.
(
2022
)
Discovery of 2′,6-Bis(4-Hydroxybenzyl)-2-Acetylcyclohexanone, a Novel FtsZ Inhibitor
.
Molecules
27
,
6993
[PubMed]
201.
Li
Y.
,
Sun
N.
,
Ser
H.L.
,
Long
W.
,
Li
Y.
,
Chen
C.
et al.
(
2020
)
Antibacterial activity evaluation and mode of action study of novel thiazole-quinolinium derivatives
.
RSC Adv.
10
,
15000
15014
[PubMed]
202.
Dhameliya
T.M.
,
Tiwari
R.
,
Patel
K.I.
,
Vagolu
S.K.
,
Panda
D.
,
Sriram
D.
et al.
(
2022
)
Bacterial FtsZ Inhibition by Benzo[d]Imidazole-2-Carboxamide Derivative with Anti-TB activity
.
Future Med. Chem.
14
,
1361
1373
[PubMed]
203.
Chai
W.C.
,
Whittall
J.J.
,
Polyak
S.W.
,
Foo
K.
,
Li
X.
,
Dutschke
C.J.
et al.
(
2022
)
Cinnamaldehyde derivatives act as antimicrobial agents against acinetobacter baumannii through the inhibition of cell division
.
Front. Microbiol.
967949
204.
Jaiswal
R.
,
Beuria
T.K.
,
Mohan
R.
,
Mahajan
S.K.
and
Panda
D.
(
2007
)
Totarol inhibits bacterial cytokinesis by perturbing the assembly dynamics of FtsZ
.
Biochemistry
46
,
4211
4220
[PubMed]
205.
Constantine
G.H.
,
Karchesy
J.J.
,
Franzblau
S.G.
and
LaFleur
L.E.
(
2001
)
(+)-Totarol from chamaecyparis nootkatensis and activity against Mycobacterium tuberculosis
.
Fitoterapia
72
,
572
574
[PubMed]
206.
Wang
J.
,
Galgoci
A.
,
Kodali
S.
,
Herath
K.B.
,
Jayasuriya
H.
,
Dorso
K.
et al.
(
2003
)
Discovery of a small molecule that inhibits cell division by blocking FtsZ, a novel therapeutic target of antibiotics
.
J. Biol. Chem.
278
,
44424
44428
[PubMed]
207.
Urgaonkar
S.
,
La Pierre
H.S.
,
Meir
I.
,
Lund
H.
,
RayChaudhuri
D.
and
Shaw
J.T.
(
2005
)
Synthesis of antimicrobial natural products targeting FtsZ: (±)-Dichamanetin and (±)-2‴-Hydroxy-5″- Benzylisouvarinol-B
.
Org. Lett.
7
,
5609
5612
[PubMed]
208.
Beuria
T.K.
,
Singh
P.
,
Surolia
A.
and
Panda
D.
(
2009
)
Promoting assembly and bundling of FtsZ as a strategy to inhibit bacterial cell division: a new approach for developing novel antibacterial drugs
.
Biochem. J.
423
,
61
69
[PubMed]
209.
Groundwater
P.W.
,
Narlawar
R.
,
Liao
V.W.Y.
,
Bhattacharya
A.
,
Srivastava
S.
,
Kunal
K.
et al.
(
2017
)
A carbocyclic curcumin inhibits proliferation of gram-positive bacteria by targeting FtsZ
.
Biochemistry
56
,
514
524
[PubMed]
210.
White
E.L.
,
William
J.S.
,
Larry
J.R.
,
Lainne
E.S.
and
Reynolds
C.R.
(
2002
)
2-alkoxycarbonylaminopyridines: inhibitors of mycobacterium tuberculosis FtsZ
.
J. Antimicrob. Chemother.
50
,
111
114

Author notes

*

Present address: National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab 160062, India

This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).