Network


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

Hotspot


Dive into the research topics where Kojiro Mukai is active.

Publication


Featured researches published by Kojiro Mukai.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Intracellular phosphatidylserine is essential for retrograde membrane traffic through endosomes

Yasunori Uchida; Junya Hasegawa; Daniel J.-F. Chinnapen; Takao Inoue; Seiji Okazaki; Ryuichi Kato; Soichi Wakatsuki; Ryo Misaki; Masato Koike; Yasuo Uchiyama; Shun-ichiro Iemura; Tohru Natsume; Ryusuke Kuwahara; Takatoshi Nakagawa; Kiyotaka Nishikawa; Kojiro Mukai; Eiji Miyoshi; Naoyuki Taniguchi; David Sheff; Wayne I. Lencer; Tomohiko Taguchi; Hiroyuki Arai

Phosphatidylserine (PS) is a relatively minor constituent of biological membranes. Despite its low abundance, PS in the plasma membrane (PM) plays key roles in various phenomena such as the coagulation cascade, clearance of apoptotic cells, and recruitment of signaling molecules. PS also localizes in endocytic organelles, but how this relates to its cellular functions remains unknown. Here we report that PS is essential for retrograde membrane traffic at recycling endosomes (REs). PS was most concentrated in REs among intracellular organelles, and evectin-2 (evt-2), a protein of previously unknown function, was targeted to REs by the binding of its pleckstrin homology (PH) domain to PS. X-ray analysis supported the specificity of the binding of PS to the PH domain. Depletion of evt-2 or masking of intracellular PS suppressed membrane traffic from REs to the Golgi. These findings uncover the molecular basis that controls the RE-to-Golgi transport and identify a unique PH domain that specifically recognizes PS but not polyphosphoinositides.


The EMBO Journal | 2015

Transport through recycling endosomes requires EHD1 recruitment by a phosphatidylserine translocase

Shoken Lee; Yasunori Uchida; Jiao Wang; Tatsuyuki Matsudaira; Takatoshi Nakagawa; Takuma Kishimoto; Kojiro Mukai; Takehiko Inaba; Toshihide Kobayashi; Robert S. Molday; Tomohiko Taguchi; Hiroyuki Arai

P4‐ATPases translocate aminophospholipids, such as phosphatidylserine (PS), to the cytosolic leaflet of membranes. PS is highly enriched in recycling endosomes (REs) and is essential for endosomal membrane traffic. Here, we show that PS flipping by an RE‐localized P4‐ATPase is required for the recruitment of the membrane fission protein EHD1. Depletion of ATP8A1 impaired the asymmetric transbilayer distribution of PS in REs, dissociated EHD1 from REs, and generated aberrant endosomal tubules that appear resistant to fission. EHD1 did not show membrane localization in cells defective in PS synthesis. ATP8A2, a tissue‐specific ATP8A1 paralogue, is associated with a neurodegenerative disease (CAMRQ). ATP8A2, but not the disease‐causative ATP8A2 mutant, rescued the endosomal defects in ATP8A1‐depleted cells. Primary neurons from Atp8a2−/− mice showed a reduced level of transferrin receptors at the cell surface compared to Atp8a2+/+ mice. These findings demonstrate the role of P4‐ATPase in membrane fission and give insight into the molecular basis of CAMRQ.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Nitro-fatty acids are formed in response to virus infection and are potent inhibitors of STING palmitoylation and signaling.

Anne Louise Hansen; Gregory J. Buchan; Michael Rühl; Kojiro Mukai; Sonia R. Salvatore; Emari Ogawa; Sidsel D. Andersen; Marie B. Iversen; Anne L. Thielke; Camilla Gunderstofte; Mona Motwani; Charlotte T. Møller; Andreas S. Jakobsen; Katherine A. Fitzgerald; Jessica Roos; Rongtuan Lin; Thorsten J. Maier; Raphaela Goldbach-Mansky; Cathrine Miner; Wei Qian; Jonathan J. Miner; Rachel E. Rigby; Jan Rehwinkel; Martin R. Jakobsen; Hiroyuki Arai; Tomohiko Taguchi; Francisco J. Schopfer; David Olagnier; Christian K. Holm

Significance Several chronic inflammatory conditions have recently been shown to depend on abnormally high activity of the signaling protein stimulator of IFN genes (STING). These conditions include examples from systemic lupus erythematosus, Aicardi–Goutiéres syndrome, and STING-associated vasculopathy with onset in infancy. The involvement of STING in these diseases points to an unmet demand to identify inhibitors of STING signaling, which could form the basis of anti-STING therapeutics. With this report, we identify distinct endogenously formed lipid species as potent inhibitors of STING signaling—and propose that these lipids could have pharmaceutical potential for treatment of STING-dependent inflammatory diseases. The adaptor molecule stimulator of IFN genes (STING) is central to production of type I IFNs in response to infection with DNA viruses and to presence of host DNA in the cytosol. Excessive release of type I IFNs through STING-dependent mechanisms has emerged as a central driver of several interferonopathies, including systemic lupus erythematosus (SLE), Aicardi–Goutières syndrome (AGS), and stimulator of IFN genes-associated vasculopathy with onset in infancy (SAVI). The involvement of STING in these diseases points to an unmet need for the development of agents that inhibit STING signaling. Here, we report that endogenously formed nitro-fatty acids can covalently modify STING by nitro-alkylation. These nitro-alkylations inhibit STING palmitoylation, STING signaling, and subsequently, the release of type I IFN in both human and murine cells. Furthermore, treatment with nitro-fatty acids was sufficient to inhibit production of type I IFN in fibroblasts derived from SAVI patients with a gain-of-function mutation in STING. In conclusion, we have identified nitro-fatty acids as endogenously formed inhibitors of STING signaling and propose for these lipids to be considered in the treatment of STING-dependent inflammatory diseases.


ACS Omega | 2017

Magnetic Separation of Autophagosomes from Mammalian Cells Using Magnetic–Plasmonic Hybrid Nanobeads

Mari Takahashi; Priyank Mohan; Kojiro Mukai; Yuichi Takeda; Takeo Matsumoto; Kazuaki Matsumura; Masahiro Takakura; Hiroyuki Arai; Tomohiko Taguchi; Shinya Maenosono

Developments in subcellular fractionation strategies have provided the means to analyze the protein and lipid composition of organelles by proteomics. Here, we developed ultrasmall magnetic–plasmonic hybrid nanobeads and applied them to the isolation of autophagosomes by applying a magnetic field. The beads were chemically synthesized and comprised an Ag/FeCo/Ag core/shell/shell structure with a mean diameter of 15 nm. The Ag core and the FeCo shell conferred imaging and magnetic separation capabilities, respectively. The nanobeads were transfected into mammalian cells by lipofection. Thirty minutes after lipofection, the nanobeads colocalized with Vps26 and subsequently with LC3. Cell lysates were prepared at the appropriate time points and were subjected to magnetic separation. The separated fraction contained LC3-II, transferrin receptor, and LAMP2, but not LC3-I, suggesting that autophagosomes engulfing endosomal origin had been isolated. The magnetic separation process was completed in less than 30 min, providing a rapid method for isolation of autophagosomes. The present organelle isolation technique using the hybrid nanobeads with imaging and magnetic separation capabilities is highly promising for isolation of other types of organelles such as endosomes and endosome-related organelles.


Biochemical and Biophysical Research Communications | 2018

The binding of TBK1 to STING requires exocytic membrane traffic from the ER

Emari Ogawa; Kojiro Mukai; Kota Saito; Hiroyuki Arai; Tomohiko Taguchi

Stimulator of interferon genes (STING) is essential for the type I interferon and pro-inflammatory responses against DNA pathogens. In response to the presence of cytosolic DNA, STING translocates from the endoplasmic reticulum (ER) to the Golgi, and activates TANK-binding kinase 1 (TBK1), a cytosolic kinase that is essential for the activation of STING-dependent downstream signalling. The organelles where TBK1 binds to STING remain unknown. Here we show that TBK1 binds to STING at the Golgi, not at the ER. Treatment with brefeldin A, an agent to block ER-to-Golgi traffic, or knockdown of Sar1, a small GTPase that regulates coat protein complex II (COP-II)-mediated ER-to-Golgi traffic, inhibited the binding of TBK1 to STING. Endogenous TBK1 was recruited to the Golgi when STING was transported to the Golgi, as shown by immunofluorescence microscopy. STING variants that constitutively induce the type I interferon response were found in patients with autoinflammatory diseases. Even these disease-causative STING variants could not bind to TBK1 when the STING variants were trapped in the ER. These results demonstrate that the Golgi is an organelle at which STING recruits and activates TBK1 for triggering the STING-dependent type I interferon response.


The Japanese Biochemical Society/The Molecular Biology Society of Japan | 2017

Endosomal phosphatidylserine is critical for the YAP signaling pathway

Tatsuyuki Matsudaira; Kojiro Mukai; Taishin Noguchi; Junya Hasegawa; Tomohisa Hatta; Shun-ichiro Iemura; Tohru Natsume; Norio Miyamura; Hiroshi Nishina; Jun Nakayama; Kentaro Semba; Takuya Tomita; Shigeo Murata; Hiroyuki Arai; Tomohiko Taguchi


The Japanese Biochemical Society/The Molecular Biology Society of Japan | 2017

Deficiency of polyunsaturated fatty acids (PUFAs) induces the G1/S cell cycle arrest

Yuki Ishino; Yuri Saito; Kojiro Mukai; Tomohiko Taguchi; Hiroyuki Arai


The Japanese Biochemical Society/The Molecular Biology Society of Japan | 2017

The mechanism underlying the activation of STING variants associated with an autoinflammatory disease SAVI

Emari Ogawa; Kojiro Mukai; Tomohiko Taguchi; Hiroyuki Arai


Nature Communications | 2017

Endosomal phosphatidylserine is critical for the YAP signalling pathway in proliferating cells

Tatsuyuki Matsudaira; Kojiro Mukai; Taishin Noguchi; Junya Hasegawa; Tomohisa Hatta; Shun-ichiro Iemura; Tohru Natsume; Norio Miyamura; Hiroshi Nishina; Jun Nakayama; Kentaro Semba; Takuya Tomita; Shigeo Murata; Hiroyuki Arai; Tomohiko Taguchi


The Molecular Biology Society of Japan | 2016

Activation of STING requires palmitoylation at the Golgi

Kojiro Mukai; Tomohiko Taguchi; Hiroyuki Arai

Collaboration


Dive into the Kojiro Mukai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shun-ichiro Iemura

Fukushima Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tohru Natsume

National Institute of Advanced Industrial Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Nishina

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge