Perspectives on Bacterial Flagellar Motor

A special issue of Biomolecules (ISSN 2218-273X). This special issue belongs to the section "Molecular Structure and Dynamics".

Deadline for manuscript submissions: closed (31 December 2020) | Viewed by 113581

Printed Edition Available!
A printed edition of this Special Issue is available here.

Special Issue Editors


E-Mail Website
Guest Editor
Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
Interests: bacterial flagella; bacterial motility; bacterial protein secretion; macromolecular assembly; energy transduction
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
Interests: bacterial flagella; muscle actomyosin; molecular motors; electron cryomicroscopy; macromolecular structures and dynamics

Special Issue Information

Dear Colleagues,

The bacterial flagellum is a supramolecular motility machinery that allows bacterial cells to migrate towards more favourable conditions and to escape from undesirable conditions in viscous liquid environments for their survival and/or infection. The bacterial flagellum also acts as a biosensor to detect changes in their environment to switch their life cycle from planktonic cells to the formation of a biofilm society. The flagellum consists of at least three parts: the basal body (reversible rotary motor), the hook (universal joint) and the filament (helical propeller). The bacterial flagellar motor composed of a rotor ring and a dozen stators is powered by an electrochemical-potential difference of specific ions across the cytoplasmic membrane and rotates in either the counterclockwise (CCW) or clockwise (CW) direction. A sensory signal transduction pathway regulates the switching between the CCW and CW states of the motor in response to environmental stimuli. Recently, it has been revealed that the bacterial flagellar motor induces structural remodeling of itself in response to changes in the environment to exert the motor function under different conditions.

The core structure of the bacterial flagellum is conserved among bacterial species. However, recent structural analyses of intact flagellar structures derived from various bacterial species by electron cryotomography and subtomogram averaging have shown that novel and divergent structures surround the core structure, suggesting that the flagellar motors have adapted to function in various environments of the habitat of bacteria.

This Special Issue of Biomolecules is dedicated to covering recent advances in our understanding of and perspectives on the flagellar motor derived from different bacterial species. Our aim is to compile a Special Issue describing recent advances in the structure and function of the bacterial flagellar motor.

Dr. Tohru Minamino
Prof. Keiichi Namba
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Biomolecules is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Bacterial flagellum
  • Flagellar assembly
  • Torque generation
  • Chemotaxis
  • Structural diversity

Published Papers (22 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

3 pages, 180 KiB  
Editorial
Recent Advances in the Bacterial Flagellar Motor Study
by Tohru Minamino and Keiichi Namba
Biomolecules 2021, 11(5), 741; https://doi.org/10.3390/biom11050741 - 17 May 2021
Viewed by 2260
Abstract
The bacterial flagellum is a supramolecular motility machine that allows bacterial cells to swim in liquid environments [...] Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)

Research

Jump to: Editorial, Review

20 pages, 4278 KiB  
Article
Fluctuations in Intracellular CheY-P Concentration Coordinate Reversals of Flagellar Motors in E. coli
by Yong-Suk Che, Takashi Sagawa, Yuichi Inoue, Hiroto Takahashi, Tatsuki Hamamoto, Akihiko Ishijima and Hajime Fukuoka
Biomolecules 2020, 10(11), 1544; https://doi.org/10.3390/biom10111544 - 12 Nov 2020
Cited by 8 | Viewed by 2477
Abstract
Signal transduction utilizing membrane-spanning receptors and cytoplasmic regulator proteins is a fundamental process for all living organisms, but quantitative studies of the behavior of signaling proteins, such as their diffusion within a cell, are limited. In this study, we show that fluctuations in [...] Read more.
Signal transduction utilizing membrane-spanning receptors and cytoplasmic regulator proteins is a fundamental process for all living organisms, but quantitative studies of the behavior of signaling proteins, such as their diffusion within a cell, are limited. In this study, we show that fluctuations in the concentration of the signaling molecule, phosphorylated CheY, constitute the basis of chemotaxis signaling. To analyze the propagation of the CheY-P signal quantitatively, we measured the coordination of directional switching between flagellar motors on the same cell. We analyzed the time lags of the switching of two motors in both CCW-to-CW and CW-to-CCW switching (∆τCCW-CW and ∆τCW-CCW). In wild-type cells, both time lags increased as a function of the relative distance of two motors from the polar receptor array. The apparent diffusion coefficient estimated for ∆τ values was ~9 µm2/s. The distance-dependency of ∆τCW-CCW disappeared upon loss of polar localization of the CheY-P phosphatase, CheZ. The distance-dependency of the response time for an instantaneously applied serine attractant signal also disappeared with the loss of polar localization of CheZ. These results were modeled by calculating the diffusion of CheY and CheY-P in cells in which phosphorylation and dephosphorylation occur in different subcellular regions. We conclude that diffusion of signaling molecules and their production and destruction through spontaneous activity of the receptor array generates fluctuations in CheY-P concentration over timescales of several hundred milliseconds. Signal fluctuation coordinates rotation among flagella and regulates steady-state run-and-tumble swimming of cells to facilitate efficient responses to environmental chemical signals. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Graphical abstract

11 pages, 1716 KiB  
Article
GFP Fusion to the N-Terminus of MotB Affects the Proton Channel Activity of the Bacterial Flagellar Motor in Salmonella
by Yusuke V. Morimoto, Keiichi Namba and Tohru Minamino
Biomolecules 2020, 10(9), 1255; https://doi.org/10.3390/biom10091255 - 29 Aug 2020
Cited by 3 | Viewed by 2752
Abstract
The bacterial flagellar motor converts the energy of proton flow through the MotA/MotB complex into mechanical works required for motor rotation. The rotational force is generated by electrostatic interactions between the stator protein MotA and the rotor protein FliG. The Arg-90 and Glu-98 [...] Read more.
The bacterial flagellar motor converts the energy of proton flow through the MotA/MotB complex into mechanical works required for motor rotation. The rotational force is generated by electrostatic interactions between the stator protein MotA and the rotor protein FliG. The Arg-90 and Glu-98 from MotA interact with Asp-289 and Arg-281 of FliG, respectively. An increase in the expression level of the wild-type MotA/MotB complex inhibits motility of the gfp-motBfliG(R281V) mutant but not the fliG(R281V) mutant, suggesting that the MotA/GFP-MotB complex cannot work together with wild-type MotA/MotB in the presence of the fliG(R281V) mutation. However, it remains unknown why. Here, we investigated the effect of the GFP fusion to MotB at its N-terminus on the MotA/MotB function. Over-expression of wild-type MotA/MotB significantly reduced the growth rate of the gfp-motBfliG(R281V) mutant. The over-expression of the MotA/GFP-MotB complex caused an excessive proton leakage through its proton channel, thereby inhibiting cell growth. These results suggest that the GFP tag on the MotB N-terminus affects well-regulated proton translocation through the MotA/MotB proton channel. Therefore, we propose that the N-terminal cytoplasmic tail of MotB couples the gating of the proton channel with the MotA–FliG interaction responsible for torque generation. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

9 pages, 911 KiB  
Article
Coupling Ion Specificity of the Flagellar Stator Proteins MotA1/MotB1 of Paenibacillus sp. TCA20
by Sakura Onoe, Myu Yoshida, Naoya Terahara and Yoshiyuki Sowa
Biomolecules 2020, 10(7), 1078; https://doi.org/10.3390/biom10071078 - 20 Jul 2020
Cited by 4 | Viewed by 2386
Abstract
The bacterial flagellar motor is a reversible rotary molecular nanomachine, which couples ion flux across the cytoplasmic membrane to torque generation. It comprises a rotor and multiple stator complexes, and each stator complex functions as an ion channel and determines the ion specificity [...] Read more.
The bacterial flagellar motor is a reversible rotary molecular nanomachine, which couples ion flux across the cytoplasmic membrane to torque generation. It comprises a rotor and multiple stator complexes, and each stator complex functions as an ion channel and determines the ion specificity of the motor. Although coupling ions for the motor rotation were presumed to be only monovalent cations, such as H+ and Na+, the stator complex MotA1/MotB1 of Paenibacillus sp. TCA20 (MotA1TCA/MotB1TCA) was reported to use divalent cations as coupling ions, such as Ca2+ and Mg2+. In this study, we initially aimed to measure the motor torque generated by MotA1TCA/MotB1TCA under the control of divalent cation motive force; however, we identified that the coupling ion of MotA1TCAMotB1TCA is very likely to be a monovalent ion. We engineered a series of functional chimeric stator proteins between MotB1TCA and Escherichia coli MotB. E. coli ΔmotAB cells expressing MotA1TCA and the chimeric MotB presented significant motility in the absence of divalent cations. Moreover, we confirmed that MotA1TCA/MotB1TCA in Bacillus subtilis ΔmotABΔmotPS cells generates torque without divalent cations. Based on two independent experimental results, we conclude that the MotA1TCA/MotB1TCA complex directly converts the energy released from monovalent cation flux to motor rotation. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

14 pages, 2204 KiB  
Article
A Novel Lysophosphatidic Acid Acyltransferase of Escherichia coli Produces Membrane Phospholipids with a cis-vaccenoyl Group and Is Related to Flagellar Formation
by Yosuke Toyotake, Masayoshi Nishiyama, Fumiaki Yokoyama, Takuya Ogawa, Jun Kawamoto and Tatsuo Kurihara
Biomolecules 2020, 10(5), 745; https://doi.org/10.3390/biom10050745 - 11 May 2020
Cited by 5 | Viewed by 2951
Abstract
Lysophosphatidic acid acyltransferase (LPAAT) introduces fatty acyl groups into the sn-2 position of membrane phospholipids (PLs). Various bacteria produce multiple LPAATs, whereas it is believed that Escherichia coli produces only one essential LPAAT homolog, PlsC—the deletion of which is lethal. However, we [...] Read more.
Lysophosphatidic acid acyltransferase (LPAAT) introduces fatty acyl groups into the sn-2 position of membrane phospholipids (PLs). Various bacteria produce multiple LPAATs, whereas it is believed that Escherichia coli produces only one essential LPAAT homolog, PlsC—the deletion of which is lethal. However, we found that E. coli possesses another LPAAT homolog named YihG. Here, we show that overexpression of YihG in E. coli carrying a temperature-sensitive mutation in plsC allowed its growth at non-permissive temperatures. Analysis of the fatty acyl composition of PLs from the yihG-deletion mutant (∆yihG) revealed that endogenous YihG introduces the cis-vaccenoyl group into the sn-2 position of PLs. Loss of YihG did not affect cell growth or morphology, but ∆yihG cells swam well in liquid medium in contrast to wild-type cells. Immunoblot analysis showed that FliC was highly expressed in ∆yihG cells, and this phenotype was suppressed by expression of recombinant YihG in ∆yihG cells. Transmission electron microscopy confirmed that the flagellar structure was observed only in ∆yihG cells. These results suggest that YihG has specific functions related to flagellar formation through modulation of the fatty acyl composition of membrane PLs. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

10 pages, 1166 KiB  
Article
The Homologous Components of Flagellar Type III Protein Apparatus Have Acquired a Novel Function to Control Twitching Motility in a Non-Flagellated Biocontrol Bacterium
by Alex M. Fulano, Danyu Shen, Miki Kinoshita, Shan-Ho Chou and Guoliang Qian
Biomolecules 2020, 10(5), 733; https://doi.org/10.3390/biom10050733 - 07 May 2020
Cited by 11 | Viewed by 2634
Abstract
The bacterial flagellum is one of the best-studied surface-attached appendages in bacteria. Flagellar assembly in vivo is promoted by its own protein export apparatus, a type III secretion system (T3SS) in pathogenic bacteria. Lysobacter enzymogenes OH11 is a non-flagellated soil bacterium that utilizes [...] Read more.
The bacterial flagellum is one of the best-studied surface-attached appendages in bacteria. Flagellar assembly in vivo is promoted by its own protein export apparatus, a type III secretion system (T3SS) in pathogenic bacteria. Lysobacter enzymogenes OH11 is a non-flagellated soil bacterium that utilizes type IV pilus (T4P)-driven twitching motility to prey upon nearby fungi for food. Interestingly, the strain OH11 encodes components homologous to the flagellar type III protein apparatus (FT3SS) on its genome, but it remains unknown whether this FT3SS-like system is functional. Here, we report that, despite the absence of flagella, the FT3SS homologous genes are responsible not only for the export of the heterologous flagellin in strain OH11 but also for twitching motility. Blocking the FT3SS-like system by in-frame deletion mutations in either flhB or fliI abolished the secretion of heterologous flagellin molecules into the culture medium, indicating that the FT3SS is functional in strain OH11. A deletion of flhA, flhB, fliI, or fliR inhibited T4P-driven twitching motility, whereas neither that of fliP nor fliQ did, suggesting that FlhA, FlhB, FliI, and FliR may obtain a novel function to modulate the twitching motility. The flagellar FliI ATPase was required for the secretion of the major pilus subunit, PilA, suggesting that FliI would have evolved to act as a PilB-like pilus ATPase. These observations lead to a plausible hypothesis that the non-flagellated L. enzymogenes OH11 could preserve FT3SS-like genes for acquiring a distinct function to regulate twitching motility associated with its predatory behavior. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

21 pages, 6067 KiB  
Article
MotP Subunit is Critical for Ion Selectivity and Evolution of a K+-Coupled Flagellar Motor
by Shun Naganawa and Masahiro Ito
Biomolecules 2020, 10(5), 691; https://doi.org/10.3390/biom10050691 - 29 Apr 2020
Cited by 6 | Viewed by 2984
Abstract
The bacterial flagellar motor is a sophisticated nanomachine embedded in the cell envelope. The flagellar motor is driven by an electrochemical gradient of cations such as H+, Na+, and K+ through ion channels in stator complexes embedded in [...] Read more.
The bacterial flagellar motor is a sophisticated nanomachine embedded in the cell envelope. The flagellar motor is driven by an electrochemical gradient of cations such as H+, Na+, and K+ through ion channels in stator complexes embedded in the cell membrane. The flagellum is believed to rotate as a result of electrostatic interaction forces between the stator and the rotor. In bacteria of the genus Bacillus and related species, the single transmembrane segment of MotB-type subunit protein (MotB and MotS) is critical for the selection of the H+ and Na+ coupling ions. Here, we constructed and characterized several hybrid stators combined with single Na+-coupled and dual Na+- and K+-coupled stator subunits, and we report that the MotP subunit is critical for the selection of K+. This result suggested that the K+ selectivity of the MotP/MotS complexes evolved from the single Na+-coupled stator MotP/MotS complexes. This finding will promote the understanding of the evolution of flagellar motors and the molecular mechanisms of coupling ion selectivity. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Graphical abstract

17 pages, 3130 KiB  
Article
A Factor Produced by Kaistia sp. 32K Accelerated the Motility of Methylobacterium sp. ME121
by Yoshiaki Usui, Yuu Wakabayashi, Tetsu Shimizu, Yuhei O. Tahara, Makoto Miyata, Akira Nakamura and Masahiro Ito
Biomolecules 2020, 10(4), 618; https://doi.org/10.3390/biom10040618 - 16 Apr 2020
Cited by 4 | Viewed by 3580
Abstract
Motile Methylobacterium sp. ME121 and non-motile Kaistia sp. 32K were isolated from the same soil sample. Interestingly, ME121 was significantly more motile in the coculture of ME121 and 32K than in the monoculture of ME121. This advanced motility of ME121 was also observed [...] Read more.
Motile Methylobacterium sp. ME121 and non-motile Kaistia sp. 32K were isolated from the same soil sample. Interestingly, ME121 was significantly more motile in the coculture of ME121 and 32K than in the monoculture of ME121. This advanced motility of ME121 was also observed in the 32K culture supernatant. A swimming acceleration factor, which we named the K factor, was identified in the 32K culture supernatant, purified, characterized as an extracellular polysaccharide (5–10 kDa), and precipitated with 70% ethanol. These results suggest the possibility that the K factor was directly or indirectly sensed by the flagellar stator, accelerating the flagellar rotation of ME121. To the best of our knowledge, no reports describing an acceleration in motility due to coculture with two or more types of bacteria have been published. We propose a mechanism by which the increase in rotational force of the ME121 flagellar motor is caused by the introduction of the additional stator into the motor by the K factor. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Graphical abstract

11 pages, 2152 KiB  
Article
Structural and Functional Comparison of Salmonella Flagellar Filaments Composed of FljB and FliC
by Tomoko Yamaguchi, Shoko Toma, Naoya Terahara, Tomoko Miyata, Masamichi Ashihara, Tohru Minamino, Keiichi Namba and Takayuki Kato
Biomolecules 2020, 10(2), 246; https://doi.org/10.3390/biom10020246 - 06 Feb 2020
Cited by 30 | Viewed by 5672
Abstract
The bacterial flagellum is a motility organelle consisting of a long helical filament as a propeller and a rotary motor that drives rapid filament rotation to produce thrust. Salmonella enterica serovar Typhimurium has two genes of flagellin, fljB and fliC, for flagellar [...] Read more.
The bacterial flagellum is a motility organelle consisting of a long helical filament as a propeller and a rotary motor that drives rapid filament rotation to produce thrust. Salmonella enterica serovar Typhimurium has two genes of flagellin, fljB and fliC, for flagellar filament formation and autonomously switches their expression at a frequency of 10−3–10−4 per cell per generation. We report here differences in their structures and motility functions under high-viscosity conditions. A Salmonella strain expressing FljB showed a higher motility than one expressing FliC under high viscosity. To examine the reasons for this motility difference, we carried out structural analyses of the FljB filament by electron cryomicroscopy and found that the structure was nearly identical to that of the FliC filament except for the position and orientation of the outermost domain D3 of flagellin. The density of domain D3 was much lower in FljB than FliC, suggesting that domain D3 of FljB is more flexible and mobile than that of FliC. These differences suggest that domain D3 plays an important role not only in changing antigenicity of the filament but also in optimizing motility function of the filament as a propeller under different conditions. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

14 pages, 2391 KiB  
Article
In Vitro Autonomous Construction of the Flagellar Axial Structure in Inverted Membrane Vesicles
by Hiroyuki Terashima, Chinatsu Tatsumi, Akihiro Kawamoto, Keiichi Namba, Tohru Minamino and Katsumi Imada
Biomolecules 2020, 10(1), 126; https://doi.org/10.3390/biom10010126 - 11 Jan 2020
Cited by 8 | Viewed by 4352
Abstract
The bacterial flagellum is a filamentous organelle extending from the cell surface. The axial structure of the flagellum consists of the rod, hook, junction, filament, and cap. The axial structure is formed by axial component proteins exported via a specific protein export apparatus [...] Read more.
The bacterial flagellum is a filamentous organelle extending from the cell surface. The axial structure of the flagellum consists of the rod, hook, junction, filament, and cap. The axial structure is formed by axial component proteins exported via a specific protein export apparatus in a well-regulated manner. Although previous studies have revealed the outline of the flagellar construction process, the mechanism of axial structure formation, including axial protein export, is still obscure due to difficulties in direct observation of protein export and assembly in vivo. We recently developed an in vitro flagellar protein transport assay system using inverted membrane vesicles (IMVs) and succeeded in reproducing the early stage of flagellar assembly. However, the late stage of the flagellar formation process remained to be examined in the IMVs. In this study, we showed that the filament-type proteins are transported into the IMVs to produce the filament on the hook inside the IMVs. Furthermore, we provide direct evidence that coordinated flagellar protein export and assembly can occur at the post-translational level. These results indicate that the ordered construction of the entire flagellar structure can be regulated by only the interactions between the protein export apparatus, the export substrate proteins, and their cognate chaperones. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

13 pages, 5308 KiB  
Article
Structure of Salmonella Flagellar Hook Reveals Intermolecular Domain Interactions for the Universal Joint Function
by Péter Horváth, Takayuki Kato, Tomoko Miyata and Keiichi Namba
Biomolecules 2019, 9(9), 462; https://doi.org/10.3390/biom9090462 - 09 Sep 2019
Cited by 16 | Viewed by 3481
Abstract
The bacterial flagellum is a motility organelle consisting of a rotary motor and a long helical filament as a propeller. The flagellar hook is a flexible universal joint that transmits motor torque to the filament in its various orientations that change dynamically between [...] Read more.
The bacterial flagellum is a motility organelle consisting of a rotary motor and a long helical filament as a propeller. The flagellar hook is a flexible universal joint that transmits motor torque to the filament in its various orientations that change dynamically between swimming and tumbling of the cell upon switching the motor rotation for chemotaxis. Although the structures of the hook and hook protein FlgE from different bacterial species have been studied, the structure of Salmonella hook, which has been studied most over the years, has not been solved at a high enough resolution to allow building an atomic model of entire FlgE for understanding the mechanisms of self-assembly, stability and the universal joint function. Here we report the structure of Salmonella polyhook at 4.1 Å resolution by electron cryomicroscopy and helical image analysis. The density map clearly revealed folding of the entire FlgE chain forming the three domains D0, D1 and D2 and allowed us to build an atomic model. The model includes domain Dc with a long β-hairpin structure that connects domains D0 and D1 and contributes to the structural stability of the hook while allowing the flexible bending of the hook as a molecular universal joint. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

11 pages, 3108 KiB  
Article
Architecture of the Bacterial Flagellar Distal Rod and Hook of Salmonella
by Yumiko Saijo-Hamano, Hideyuki Matsunami, Keiichi Namba and Katsumi Imada
Biomolecules 2019, 9(7), 260; https://doi.org/10.3390/biom9070260 - 07 Jul 2019
Cited by 13 | Viewed by 4281
Abstract
The bacterial flagellum is a large molecular complex composed of thousands of protein subunits for motility. The filamentous part of the flagellum, which is called the axial structure, consists of the filament, the hook, and the rods, with other minor components—the cap protein [...] Read more.
The bacterial flagellum is a large molecular complex composed of thousands of protein subunits for motility. The filamentous part of the flagellum, which is called the axial structure, consists of the filament, the hook, and the rods, with other minor components—the cap protein and the hook associated proteins. They share a common basic architecture of subunit arrangement, but each part shows quite distinct mechanical properties to achieve its specific function. The distal rod and the hook are helical assemblies of a single protein, FlgG and FlgE, respectively. They show a significant sequence similarity but have distinct mechanical characteristics. The rod is a rigid, straight cylinder, whereas the hook is a curved tube with high bending flexibility. Here, we report a structural model of the rod constructed by using the crystal structure of a core fragment of FlgG with a density map obtained previously by electron cryomicroscopy. Our structural model suggests that a segment called L-stretch plays a key role in achieving the distinct mechanical properties of the rod using a structurally similar component protein to that of the hook. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

Review

Jump to: Editorial, Research

18 pages, 1707 KiB  
Review
Protein Export via the Type III Secretion System of the Bacterial Flagellum
by Manuel Halte and Marc Erhardt
Biomolecules 2021, 11(2), 186; https://doi.org/10.3390/biom11020186 - 29 Jan 2021
Cited by 16 | Viewed by 3763
Abstract
The bacterial flagellum and the related virulence-associated injectisome system of pathogenic bacteria utilize a type III secretion system (T3SS) to export substrate proteins across the inner membrane in a proton motive force-dependent manner. The T3SS is composed of an export gate (FliPQR/FlhA/FlhB) located [...] Read more.
The bacterial flagellum and the related virulence-associated injectisome system of pathogenic bacteria utilize a type III secretion system (T3SS) to export substrate proteins across the inner membrane in a proton motive force-dependent manner. The T3SS is composed of an export gate (FliPQR/FlhA/FlhB) located in the flagellar basal body and an associated soluble ATPase complex in the cytoplasm (FliHIJ). Here, we summarise recent insights into the structure, assembly and protein secretion mechanisms of the T3SS with a focus on energy transduction and protein transport across the cytoplasmic membrane. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

14 pages, 3800 KiB  
Review
Construction and Loss of Bacterial Flagellar Filaments
by Xiang-Yu Zhuang and Chien-Jung Lo
Biomolecules 2020, 10(11), 1528; https://doi.org/10.3390/biom10111528 - 09 Nov 2020
Cited by 11 | Viewed by 3892
Abstract
The bacterial flagellar filament is an extracellular tubular protein structure that acts as a propeller for bacterial swimming motility. It is connected to the membrane-anchored rotary bacterial flagellar motor through a short hook. The bacterial flagellar filament consists of approximately 20,000 flagellins and [...] Read more.
The bacterial flagellar filament is an extracellular tubular protein structure that acts as a propeller for bacterial swimming motility. It is connected to the membrane-anchored rotary bacterial flagellar motor through a short hook. The bacterial flagellar filament consists of approximately 20,000 flagellins and can be several micrometers long. In this article, we reviewed the experimental works and models of flagellar filament construction and the recent findings of flagellar filament ejection during the cell cycle. The length-dependent decay of flagellar filament growth data supports the injection-diffusion model. The decay of flagellar growth rate is due to reduced transportation of long-distance diffusion and jamming. However, the filament is not a permeant structure. Several bacterial species actively abandon their flagella under starvation. Flagellum is disassembled when the rod is broken, resulting in an ejection of the filament with a partial rod and hook. The inner membrane component is then diffused on the membrane before further breakdown. These new findings open a new field of bacterial macro-molecule assembly, disassembly, and signal transduction. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

24 pages, 1511 KiB  
Review
Structural Conservation and Adaptation of the Bacterial Flagella Motor
by Brittany L. Carroll and Jun Liu
Biomolecules 2020, 10(11), 1492; https://doi.org/10.3390/biom10111492 - 29 Oct 2020
Cited by 23 | Viewed by 8887
Abstract
Many bacteria require flagella for the ability to move, survive, and cause infection. The flagellum is a complex nanomachine that has evolved to increase the fitness of each bacterium to diverse environments. Over several decades, molecular, biochemical, and structural insights into the flagella [...] Read more.
Many bacteria require flagella for the ability to move, survive, and cause infection. The flagellum is a complex nanomachine that has evolved to increase the fitness of each bacterium to diverse environments. Over several decades, molecular, biochemical, and structural insights into the flagella have led to a comprehensive understanding of the structure and function of this fascinating nanomachine. Notably, X-ray crystallography, cryo-electron microscopy (cryo-EM), and cryo-electron tomography (cryo-ET) have elucidated the flagella and their components to unprecedented resolution, gleaning insights into their structural conservation and adaptation. In this review, we focus on recent structural studies that have led to a mechanistic understanding of flagellar assembly, function, and evolution. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

26 pages, 5642 KiB  
Review
The Architectural Dynamics of the Bacterial Flagellar Motor Switch
by Shahid Khan
Biomolecules 2020, 10(6), 833; https://doi.org/10.3390/biom10060833 - 29 May 2020
Cited by 6 | Viewed by 3094
Abstract
The rotary bacterial flagellar motor is remarkable in biochemistry for its highly synchronized operation and amplification during switching of rotation sense. The motor is part of the flagellar basal body, a complex multi-protein assembly. Sensory and energy transduction depends on a core of [...] Read more.
The rotary bacterial flagellar motor is remarkable in biochemistry for its highly synchronized operation and amplification during switching of rotation sense. The motor is part of the flagellar basal body, a complex multi-protein assembly. Sensory and energy transduction depends on a core of six proteins that are adapted in different species to adjust torque and produce diverse switches. Motor response to chemotactic and environmental stimuli is driven by interactions of the core with small signal proteins. The initial protein interactions are propagated across a multi-subunit cytoplasmic ring to switch torque. Torque reversal triggers structural transitions in the flagellar filament to change motile behavior. Subtle variations in the core components invert or block switch operation. The mechanics of the flagellar switch have been studied with multiple approaches, from protein dynamics to single molecule and cell biophysics. The architecture, driven by recent advances in electron cryo-microscopy, is available for several species. Computational methods have correlated structure with genetic and biochemical databases. The design principles underlying the basis of switch ultra-sensitivity and its dependence on motor torque remain elusive, but tantalizing clues have emerged. This review aims to consolidate recent knowledge into a unified platform that can inspire new research strategies. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

14 pages, 2515 KiB  
Review
Living in a Foster Home: The Single Subpolar Flagellum Fla1 of Rhodobacter sphaeroides
by Laura Camarena and Georges Dreyfus
Biomolecules 2020, 10(5), 774; https://doi.org/10.3390/biom10050774 - 16 May 2020
Cited by 5 | Viewed by 3554
Abstract
Rhodobacter sphaeroides is an α-proteobacterium that has the particularity of having two functional flagellar systems used for swimming. Under the growth conditions commonly used in the laboratory, a single subpolar flagellum that traverses the cell membrane, is assembled on the surface. This flagellum [...] Read more.
Rhodobacter sphaeroides is an α-proteobacterium that has the particularity of having two functional flagellar systems used for swimming. Under the growth conditions commonly used in the laboratory, a single subpolar flagellum that traverses the cell membrane, is assembled on the surface. This flagellum has been named Fla1. Phylogenetic analyses have suggested that this flagellar genetic system was acquired from an ancient γ-proteobacterium. It has been shown that this flagellum has components homologous to those present in other γ-proteobacteria such as the H-ring characteristic of the Vibrio species. Other features of this flagellum such as a straight hook, and a prominent HAP region have been studied and the molecular basis underlying these features has been revealed. It has also been shown that FliL, and the protein MotF, mainly found in several species of the family Rhodobacteraceae, contribute to remodel the amphipathic region of MotB, known as the plug, in order to allow flagellar rotation. In the absence of the plug region of MotB, FliL and MotF are dispensable. In this review we have covered the most relevant aspects of the Fla1 flagellum of this remarkable photosynthetic bacterium. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

16 pages, 2327 KiB  
Review
Spirochete Flagella and Motility
by Shuichi Nakamura
Biomolecules 2020, 10(4), 550; https://doi.org/10.3390/biom10040550 - 04 Apr 2020
Cited by 30 | Viewed by 11111
Abstract
Spirochetes can be distinguished from other flagellated bacteria by their long, thin, spiral (or wavy) cell bodies and endoflagella that reside within the periplasmic space, designated as periplasmic flagella (PFs). Some members of the spirochetes are pathogenic, including the causative agents of syphilis, [...] Read more.
Spirochetes can be distinguished from other flagellated bacteria by their long, thin, spiral (or wavy) cell bodies and endoflagella that reside within the periplasmic space, designated as periplasmic flagella (PFs). Some members of the spirochetes are pathogenic, including the causative agents of syphilis, Lyme disease, swine dysentery, and leptospirosis. Furthermore, their unique morphologies have attracted attention of structural biologists; however, the underlying physics of viscoelasticity-dependent spirochetal motility is a longstanding mystery. Elucidating the molecular basis of spirochetal invasion and interaction with hosts, resulting in the appearance of symptoms or the generation of asymptomatic reservoirs, will lead to a deeper understanding of host–pathogen relationships and the development of antimicrobials. Moreover, the mechanism of propulsion in fluids or on surfaces by the rotation of PFs within the narrow periplasmic space could be a designing base for an autonomously driving micro-robot with high efficiency. This review describes diverse morphology and motility observed among the spirochetes and further summarizes the current knowledge on their mechanisms and relations to pathogenicity, mainly from the standpoint of experimental biophysics. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

17 pages, 4598 KiB  
Review
Regulation of the Single Polar Flagellar Biogenesis
by Seiji Kojima, Hiroyuki Terashima and Michio Homma
Biomolecules 2020, 10(4), 533; https://doi.org/10.3390/biom10040533 - 01 Apr 2020
Cited by 15 | Viewed by 4459
Abstract
Some bacterial species, such as the marine bacterium Vibrio alginolyticus, have a single polar flagellum that allows it to swim in liquid environments. Two regulators, FlhF and FlhG, function antagonistically to generate only one flagellum at the cell pole. FlhF, a signal recognition [...] Read more.
Some bacterial species, such as the marine bacterium Vibrio alginolyticus, have a single polar flagellum that allows it to swim in liquid environments. Two regulators, FlhF and FlhG, function antagonistically to generate only one flagellum at the cell pole. FlhF, a signal recognition particle (SRP)-type guanosine triphosphate (GTP)ase, works as a positive regulator for flagellar biogenesis and determines the location of flagellar assembly at the pole, whereas FlhG, a MinD-type ATPase, works as a negative regulator that inhibits flagellar formation. FlhF intrinsically localizes at the cell pole, and guanosine triphosphate (GTP) binding to FlhF is critical for its polar localization and flagellation. FlhG also localizes at the cell pole via the polar landmark protein HubP to directly inhibit FlhF function at the cell pole, and this localization depends on ATP binding to FlhG. However, the detailed regulatory mechanisms involved, played by FlhF and FlhG as the major factors, remain largely unknown. This article reviews recent studies that highlight the post-translational regulation mechanism that allows the synthesis of only a single flagellum at the cell pole. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

14 pages, 1068 KiB  
Review
Flagella and Swimming Behavior of Marine Magnetotactic Bacteria
by Wei-Jia Zhang and Long-Fei Wu
Biomolecules 2020, 10(3), 460; https://doi.org/10.3390/biom10030460 - 16 Mar 2020
Cited by 14 | Viewed by 4579
Abstract
Marine environments are generally characterized by low bulk concentrations of nutrients that are susceptible to steady or intermittent motion driven by currents and local turbulence. Marine bacteria have therefore developed strategies, such as very fast-swimming and the exploitation of multiple directional sensing–response systems [...] Read more.
Marine environments are generally characterized by low bulk concentrations of nutrients that are susceptible to steady or intermittent motion driven by currents and local turbulence. Marine bacteria have therefore developed strategies, such as very fast-swimming and the exploitation of multiple directional sensing–response systems in order to efficiently migrate towards favorable places in nutrient gradients. The magnetotactic bacteria (MTB) even utilize Earth’s magnetic field to facilitate downward swimming into the oxic–anoxic interface, which is the most favorable place for their persistence and proliferation, in chemically stratified sediments or water columns. To ensure the desired flagella-propelled motility, marine MTBs have evolved an exquisite flagellar apparatus, and an extremely high number (tens of thousands) of flagella can be found on a single entity, displaying a complex polar, axial, bounce, and photosensitive magnetotactic behavior. In this review, we describe gene clusters, the flagellar apparatus architecture, and the swimming behavior of marine unicellular and multicellular magnetotactic bacteria. The physiological significance and mechanisms that govern these motions are discussed. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

15 pages, 1562 KiB  
Review
Phylogenetic Distribution, Ultrastructure, and Function of Bacterial Flagellar Sheaths
by Joshua Chu, Jun Liu and Timothy R. Hoover
Biomolecules 2020, 10(3), 363; https://doi.org/10.3390/biom10030363 - 27 Feb 2020
Cited by 19 | Viewed by 4386
Abstract
A number of Gram-negative bacteria have a membrane surrounding their flagella, referred to as the flagellar sheath, which is continuous with the outer membrane. The flagellar sheath was initially described in Vibrio metschnikovii in the early 1950s as an extension of the outer [...] Read more.
A number of Gram-negative bacteria have a membrane surrounding their flagella, referred to as the flagellar sheath, which is continuous with the outer membrane. The flagellar sheath was initially described in Vibrio metschnikovii in the early 1950s as an extension of the outer cell wall layer that completely surrounded the flagellar filament. Subsequent studies identified other bacteria that possess flagellar sheaths, most of which are restricted to a few genera of the phylum Proteobacteria. Biochemical analysis of the flagellar sheaths from a few bacterial species revealed the presence of lipopolysaccharide, phospholipids, and outer membrane proteins in the sheath. Some proteins localize preferentially to the flagellar sheath, indicating mechanisms exist for protein partitioning to the sheath. Recent cryo-electron tomography studies have yielded high resolution images of the flagellar sheath and other structures closely associated with the sheath, which has generated insights and new hypotheses for how the flagellar sheath is synthesized. Various functions have been proposed for the flagellar sheath, including preventing disassociation of the flagellin subunits in the presence of gastric acid, avoiding activation of the host innate immune response by flagellin, activating the host immune response, adherence to host cells, and protecting the bacterium from bacteriophages. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Figure 1

23 pages, 2555 KiB  
Review
Flagella-Driven Motility of Bacteria
by Shuichi Nakamura and Tohru Minamino
Biomolecules 2019, 9(7), 279; https://doi.org/10.3390/biom9070279 - 14 Jul 2019
Cited by 188 | Viewed by 23171
Abstract
The bacterial flagellum is a helical filamentous organelle responsible for motility. In bacterial species possessing flagella at the cell exterior, the long helical flagellar filament acts as a molecular screw to generate thrust. Meanwhile, the flagella of spirochetes reside within the periplasmic space [...] Read more.
The bacterial flagellum is a helical filamentous organelle responsible for motility. In bacterial species possessing flagella at the cell exterior, the long helical flagellar filament acts as a molecular screw to generate thrust. Meanwhile, the flagella of spirochetes reside within the periplasmic space and not only act as a cytoskeleton to determine the helicity of the cell body, but also rotate or undulate the helical cell body for propulsion. Despite structural diversity of the flagella among bacterial species, flagellated bacteria share a common rotary nanomachine, namely the flagellar motor, which is located at the base of the filament. The flagellar motor is composed of a rotor ring complex and multiple transmembrane stator units and converts the ion flux through an ion channel of each stator unit into the mechanical work required for motor rotation. Intracellular chemotactic signaling pathways regulate the direction of flagella-driven motility in response to changes in the environments, allowing bacteria to migrate towards more desirable environments for their survival. Recent experimental and theoretical studies have been deepening our understanding of the molecular mechanisms of the flagellar motor. In this review article, we describe the current understanding of the structure and dynamics of the bacterial flagellum. Full article
(This article belongs to the Special Issue Perspectives on Bacterial Flagellar Motor)
Show Figures

Graphical abstract

Back to TopTop