Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Miki Kinoshita is active.

Publication


Featured researches published by Miki Kinoshita.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Structural insight into the regulatory mechanisms of interactions of the flagellar type III chaperone FliT with its binding partners

Katsumi Imada; Tohru Minamino; Miki Kinoshita; Yukio Furukawa; Keiichi Namba

For self-assembly of the bacterial flagellum, most of the flagellar component proteins synthesized in the cytoplasm are exported by the flagellar type III export apparatus to the growing, distal end. Flagellar protein export is highly organized and well controlled in every step of the flagellar assembly process. Flagellar-specific chaperones not only facilitate the export of their cognate proteins, as well as prevent their premature aggregation in the cytoplasm, but also play a role in fine-tuning flagellar gene expression to be coupled with the flagellar assembly process. FliT is a flagellar-specific chaperone responsible for the export of the filament-capping protein FliD and for negative control of flagellar gene expression by binding to the FlhDC complex. Here we report the crystal structure of Salmonella FliT at 3.2-Å resolution. The structural and biochemical analyses clearly reveal that the C-terminal segment of FliT regulates its interactions with the FlhDC complex, FliI ATPase, and FliJ (subunits of the export apparatus), and that its conformational change is responsible for the switch in its binding partners during flagellar protein export.


PLOS Biology | 2011

Structural insight into the rotational switching mechanism of the bacterial flagellar motor.

Tohru Minamino; Katsumi Imada; Miki Kinoshita; Shuichi Nakamura; Yusuke V. Morimoto; Keiichi Namba

The bacterial flagellar motor can rotate either clockwise (CW) or counterclockwise (CCW). Three flagellar proteins, FliG, FliM, and FliN, are required for rapid switching between the CW and CCW directions. Switching is achieved by a conformational change in FliG induced by the binding of a chemotaxis signaling protein, phospho-CheY, to FliM and FliN. FliG consists of three domains, FliG(N), FliG(M), and FliG(C), and forms a ring on the cytoplasmic face of the MS ring of the flagellar basal body. Crystal structures have been reported for the FliG(MC) domains of Thermotoga maritima, which consist of the FliG(M) and FliG(C) domains and a helix E that connects these two domains, and full-length FliG of Aquifex aeolicus. However, the basis for the switching mechanism is based only on previously obtained genetic data and is hence rather indirect. We characterized a CW-biased mutant (fliG(ΔPAA)) of Salmonella enterica by direct observation of rotation of a single motor at high temporal and spatial resolution. We also determined the crystal structure of the FliG(MC) domains of an equivalent deletion mutant variant of T. maritima (fliG(ΔPEV)). The FliG(ΔPAA) motor produced torque at wild-type levels under a wide range of external load conditions. The wild-type motors rotated exclusively in the CCW direction under our experimental conditions, whereas the mutant motors rotated only in the CW direction. This result suggests that wild-type FliG is more stable in the CCW state than in the CW state, whereas FliG(ΔPAA) is more stable in the CW state than in the CCW state. The structure of the TM-FliG(MC)(ΔPEV) revealed that extremely CW-biased rotation was caused by a conformational change in helix E. Although the arrangement of FliG(C) relative to FliG(M) in a single molecule was different among the three crystals, a conserved FliG(M)-FliG(C) unit was observed in all three of them. We suggest that the conserved FliG(M)-FliG(C) unit is the basic functional element in the rotor ring and that the PAA deletion induces a conformational change in a hinge-loop between FliG(M) and helix E to achieve the CW state of the FliG ring. We also propose a novel model for the arrangement of FliG subunits within the motor. The model is in agreement with the previous mutational and cross-linking experiments and explains the cooperative switching mechanism of the flagellar motor.


Molecular Microbiology | 2012

Interaction of a bacterial flagellar chaperone FlgN with FlhA is required for efficient export of its cognate substrates

Tohru Minamino; Miki Kinoshita; Noritaka Hara; Shiori Takeuchi; Akira Hida; Satomi Koya; Helen Glenwright; Katsumi Imada; Phillip D. Aldridge; Keiichi Namba

FlgN chaperone acts as a bodyguard to protect its cognate substrates, FlgK and FlgL, from proteolysis in the cytoplasm. Docking of the FlgN–FlgK complex with the FliI ATPase of the flagellar type III export apparatus is key to the protein export process. However, a ΔfliH‐fliI flhB(P28T) mutant forms some flagella even in the absence of FliH and FliI, raising the question of how FlgN promotes the export of its cognate substrates. Here, we report that the interaction of FlgN with an integral membrane export protein, FlhA, is directly involved in efficient protein export. A ΔfliH‐fliI flhB(P28T) ΔflgN mutant caused extragenic suppressor mutations in the C‐terminal domain of FlhA (FlhAC). Pull‐down assays using GST affinity chromatography showed an interaction between FlgN and FlhAC. The FlgN–FlgK complex bound to FlhAC and FliJ to form the FlgN–FlgK–FliJ–FlhAC complex. The FlgN–FlhAC interaction was enhanced by FlgK but not by FliJ. FlgN120 missing the last 20 residues still bound to FlgK and FliJ but not to FlhAC. A highly conserved Tyr‐122 residue was required for the interaction with FlhAC. These results suggest that FlgN efficiently transfers FlgK/L subunits to FlhAC to promote their export.


Molecular Microbiology | 2013

Interactions of bacterial flagellar chaperone–substrate complexes with FlhA contribute to co-ordinating assembly of the flagellar filament

Miki Kinoshita; Noritaka Hara; Katsumi Imada; Keiichi Namba; Tohru Minamino

Assembly of the bacterial flagellar filament is strictly sequential; the junction proteins, FlgK and FlgL, are assembled at the distal end of the hook prior to the FliD cap, which supports assembly of as many as 30 000 FliC molecules into the filament. Export of these proteins requires assistance of flagellar chaperones: FlgN for FlgK and FlgL, FliT for FliD and FliS for FliC. The C‐terminal cytoplasmic domain of FlhA (FlhAC), a membrane component of the export apparatus, provides a binding‐site for these chaperone–substrate complexes but it remains unknown how it co‐ordinates flagellar protein export. Here, we report that the highly conserved hydrophobic dimple of FlhAC is involved in the export of FlgK, FlgL, FliD and FliC but not in proteins responsible for the structure and assembly of the hook, and that the binding affinity of FlhAC for the FlgN/FlgK complex is slightly higher than that for the FliT/FliD complex and about 14‐fold higher than that for the FliS/FliC complex, leading to the proposal that the different binding affinities of FlhAC for these chaperone/substrate complexes may confer an advantage for the efficient formation of the junction and cap structures at the tip of the hook prior to filament formation.


Molecular Microbiology | 2012

Interaction between FliI ATPase and a flagellar chaperone FliT during bacterial flagellar protein export.

Tohru Minamino; Miki Kinoshita; Katsumi Imada; Keiichi Namba

FliT is a flagellar type III export chaperone specific for the filament‐capping protein FliD. The FliT/FliD complex binds to the FliI ATPase of the flagellar export apparatus. The C‐terminal α4 helix of FliT controls its interaction with FliI but it remains unknown how it does so. Here, we analysed the FliI–FliT interaction by pull‐down assays using GST affinity chromatography. FliT94, missing the C‐terminal α4 helix, bound to the extreme N‐terminal region of FliI (FliIEN) with high affinity and to the C‐terminal ATPase domain (FliICAT) with low affinity. The C‐terminal α4 helix of FliT suppressed the interaction with FliIEN. FliH and FliT94 bound to a common binding site on FliIEN and hence FliH induced the release of FliI from FliT94 in an ATP‐independent manner. FliD increased the binding affinity of FliICAT for FliT. These results raise a possible hypothesis that the FliH/FliI complex binds to the FliT/FliD complex through FliICAT to escort it from the cytoplasm to the export gate made up of six integral membrane proteins and that, upon dissociation of FliD from FliT, FliT94 may bind to FliIEN and then FliI may transfer from FliT94 to FliH by the direct competition of FliT94 and FliH for FliIEN.


PLOS Pathogens | 2010

Crystal structure of Legionella DotD: insights into the relationship between type IVB and type II/III secretion systems.

Noboru Nakano; Tomoko Kubori; Miki Kinoshita; Katsumi Imada; Hiroki Nagai

The Dot/Icm type IVB secretion system (T4BSS) is a pivotal determinant of Legionella pneumophila pathogenesis. L. pneumophila translocate more than 100 effector proteins into host cytoplasm using Dot/Icm T4BSS, modulating host cellular functions to establish a replicative niche within host cells. The T4BSS core complex spanning the inner and outer membranes is thought to be made up of at least five proteins: DotC, DotD, DotF, DotG and DotH. DotH is the outer membrane protein; its targeting depends on lipoproteins DotC and DotD. However, the core complex structure and assembly mechanism are still unknown. Here, we report the crystal structure of DotD at 2.0 Å resolution. The structure of DotD is distinct from that of VirB7, the outer membrane lipoprotein of the type IVA secretion system. In contrast, the C-terminal domain of DotD is remarkably similar to the N-terminal subdomain of secretins, the integral outer membrane proteins that form substrate conduits for the type II and the type III secretion systems (T2SS and T3SS). A short β-segment in the otherwise disordered N-terminal region, located on the hydrophobic cleft of the C-terminal domain, is essential for outer membrane targeting of DotH and Dot/Icm T4BSS core complex formation. These findings uncover an intriguing link between T4BSS and T2SS/T3SS.


Scientific Reports | 2015

The bacterial flagellar protein export apparatus processively transports flagellar proteins even with extremely infrequent ATP hydrolysis

Tohru Minamino; Yusuke V. Morimoto; Miki Kinoshita; Phillip D. Aldridge; Keiichi Namba

For self-assembly of the bacterial flagellum, a specific protein export apparatus utilizes ATP and proton motive force (PMF) as the energy source to transport component proteins to the distal growing end. The export apparatus consists of a transmembrane PMF-driven export gate and a cytoplasmic ATPase complex composed of FliH, FliI and FliJ. The FliI6FliJ complex is structurally similar to the α3β3γ complex of FOF1-ATPase. FliJ allows the gate to efficiently utilize PMF to drive flagellar protein export but it remains unknown how. Here, we report the role of ATP hydrolysis by the FliI6FliJ complex. The export apparatus processively transported flagellar proteins to grow flagella even with extremely infrequent or no ATP hydrolysis by FliI mutation (E211D and E211Q, respectively). This indicates that the rate of ATP hydrolysis is not at all coupled with the export rate. Deletion of FliI residues 401 to 410 resulted in no flagellar formation although this FliI deletion mutant retained 40% of the ATPase activity, suggesting uncoupling between ATP hydrolysis and activation of the gate. We propose that infrequent ATP hydrolysis by the FliI6FliJ ring is sufficient for gate activation, allowing processive translocation of export substrates for efficient flagellar assembly.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Insight into the flagella type III export revealed by the complex structure of the type III ATPase and its regulator

Katsumi Imada; Tohru Minamino; Yumiko Uchida; Miki Kinoshita; Keiichi Namba

Significance The flagellar basal body contains a type III protein export machinery to construct the flagellar axial structure. ATP hydrolysis by FliI facilitates the flagellar protein export, and the ATPase activity is regulated by FliH. In this study, the structure of the homodimer of a FliH fragment (FliHC) complexed with FliI has been solved at 3.0-Å resolution. FliHC2 shows a marked structural similarity to the peripheral stalk of the A/V-type ATPases, and the proposed FliHC2–FliI hexamer model resembles in situ electron cryotomographic images. These results suggest that FliH2 functions as a peripheral stalk of the type III ATPase complex and that the flagellar export system and F/A/V-type ATPases share a similar functional mechanism and close evolutionary relationship. FliI and FliJ form the FliI6FliJ ATPase complex of the bacterial flagellar export apparatus, a member of the type III secretion system. The FliI6FliJ complex is structurally similar to the α3β3γ complex of F1-ATPase. The FliH homodimer binds to FliI to connect the ATPase complex to the flagellar base, but the details are unknown. Here we report the structure of the homodimer of a C-terminal fragment of FliH (FliHC2) in complex with FliI. FliHC2 shows an unusually asymmetric homodimeric structure that markedly resembles the peripheral stalk of the A/V-type ATPases. The FliHC2–FliI hexamer model reveals that the C-terminal domains of the FliI ATPase face the cell membrane in a way similar to the F/A/V-type ATPases. We discuss the mechanism of flagellar ATPase complex formation and a common origin shared by the type III secretion system and the F/A/V-type ATPases.


PLOS ONE | 2013

Glycine insertion makes yellow fluorescent protein sensitive to hydrostatic pressure.

Tomonobu M. Watanabe; Katsumi Imada; Keiko Yoshizawa; Masayoshi Nishiyama; Chiaki Kato; Fumiyoshi Abe; Takamitsu Morikawa; Miki Kinoshita; Hideaki Fujita; Toshio Yanagida

Fluorescent protein-based indicators for intracellular environment conditions such as pH and ion concentrations are commonly used to study the status and dynamics of living cells. Despite being an important factor in many biological processes, the development of an indicator for the physicochemical state of water, such as pressure, viscosity and temperature, however, has been neglected. We here found a novel mutation that dramatically enhances the pressure dependency of the yellow fluorescent protein (YFP) by inserting several glycines into it. The crystal structure of the mutant showed that the tyrosine near the chromophore flipped toward the outside of the β-can structure, resulting in the entry of a few water molecules near the chromophore. In response to changes in hydrostatic pressure, a spectrum shift and an intensity change of the fluorescence were observed. By measuring the fluorescence of the YFP mutant, we succeeded in measuring the intracellular pressure change in living cell. This study shows a new strategy of design to engineer fluorescent protein indicators to sense hydrostatic pressure.


MicrobiologyOpen | 2016

FliH and FliI ensure efficient energy coupling of flagellar type III protein export in Salmonella

Tohru Minamino; Miki Kinoshita; Yumi Inoue; Yusuke V. Morimoto; Kunio Ihara; Satomi Koya; Noritaka Hara; Noriko Nishioka; Seiji Kojima; Michio Homma; Keiichi Namba

For construction of the bacterial flagellum, flagellar proteins are exported via its specific export apparatus from the cytoplasm to the distal end of the growing flagellar structure. The flagellar export apparatus consists of a transmembrane (TM) export gate complex and a cytoplasmic ATPase complex consisting of FliH, FliI, and FliJ. FlhA is a TM export gate protein and plays important roles in energy coupling of protein translocation. However, the energy coupling mechanism remains unknown. Here, we performed a cross‐complementation assay to measure robustness of the energy transduction system of the export apparatus against genetic perturbations. Vibrio FlhA restored motility of a Salmonella ΔflhA mutant but not that of a ΔfliH‐fliI flhB(P28T) ΔflhA mutant. The flgM mutations significantly increased flagellar gene expression levels, allowing Vibrio FlhA to exert its export activity in the ΔfliH‐fliI flhB(P28T) ΔflhA mutant. Pull‐down assays revealed that the binding affinities of Vibrio FlhA for FliJ and the FlgN–FlgK chaperone–substrate complex were much lower than those of Salmonella FlhA. These suggest that Vibrio FlhA requires the support of FliH and FliI to efficiently and properly interact with FliJ and the FlgN–FlgK complex. We propose that FliH and FliI ensure robust and efficient energy coupling of protein export during flagellar assembly.

Collaboration


Dive into the Miki Kinoshita's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Satomi Koya

Doshisha Women's College of Liberal Arts

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge