Network


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

Hotspot


Dive into the research topics where Muneaki Miyata is active.

Publication


Featured researches published by Muneaki Miyata.


Journal of Biological Chemistry | 2010

Involvement of the Interaction of Afadin with ZO-1 in the Formation of Tight Junctions in Madin-Darby Canine Kidney Cells

Takako Ooshio; Reiko Kobayashi; Wataru Ikeda; Muneaki Miyata; Yuri Fukumoto; Naomi Matsuzawa; Hisakazu Ogita; Yoshimi Takai

Tight junctions (TJs) and adherens junctions (AJs) are major junctional apparatuses in epithelial cells. Claudins and junctional adhesion molecules (JAMs) are major cell adhesion molecules (CAMs) at TJs, whereas cadherins and nectins are major CAMs at AJs. Claudins and JAMs are associated with ZO proteins, whereas cadherins are associated with β- and α-catenins, and nectins are associated with afadin. We previously showed that nectins first form cell-cell adhesions where the cadherin-catenin complex is recruited to form AJs, followed by the recruitment of the JAM-ZO and claudin-ZO complexes to the apical side of AJs to form TJs. It is not fully understood how TJ components are recruited to the apical side of AJs. We studied the roles of afadin and ZO-1 in the formation of TJs in Madin-Darby canine kidney (MDCK) cells. Before the formation of TJs, ZO-1 interacted with afadin through the two proline-rich regions of afadin and the SH3 domain of ZO-1. During and after the formation of TJs, ZO-1 dissociated from afadin and associated with JAM-A. Knockdown of afadin impaired the formation of both AJs and TJs in MDCK cells, whereas knockdown of ZO-1 impaired the formation of TJs, but not AJs. Re-expression of full-length afadin restored the formation of both AJs and TJs in afadin-knockdown MDCK cells, whereas re-expression of afadin-ΔPR1–2, which is incapable of binding to ZO-1, restored the formation of AJs, but not TJs. These results indicate that the transient interaction of afadin with ZO-1 is necessary for the formation of TJs in MDCK cells.


Circulation Research | 2010

Role of Afadin in Vascular Endothelial Growth Factor– and Sphingosine 1-Phosphate–Induced Angiogenesis

Hideto Tawa; Yoshiyuki Rikitake; Motonori Takahashi; Hisayuki Amano; Muneaki Miyata; Seimi Satomi-Kobayashi; Mitsuo Kinugasa; Yuichi Nagamatsu; Takashi Majima; Hisakazu Ogita; Jun Miyoshi; Ken-ichi Hirata; Yoshimi Takai

Rationale: Angiogenesis contributes to physiological and pathological conditions, including atherosclerosis. The Rap1 small G protein regulates vascular integrity and angiogenesis. However, little is known about the effectors of Rap1 involved in angiogenesis. It is not known whether afadin, an adherens junction protein that connects immunoglobulin-like adhesion molecule nectins to the actin cytoskeleton and binds activated Rap1, plays a role in angiogenesis. Objective: We investigated the role of endothelial afadin in angiogenesis and attempted to clarify the underlying molecular mechanism. Methods and Results: Treatment of human umbilical vein endothelial cells (HUVECs) with vascular endothelial growth factor (VEGF) and sphingosine 1-phosphate (S1P) induced the activation of Rap1. Activated Rap1 regulated intracellular localization of afadin. Knockdown of Rap1 or afadin by small interfering RNA inhibited the VEGF- and S1P-induced capillary-like network formation, migration, and proliferation, and increased the serum deprivation-induced apoptosis of HUVECs. Knockdown of Rap1 or afadin decreased the accumulation of adherens and tight junction proteins to the cell–cell contact sites. Rap1 regulated the interaction between afadin and phosphatidylinositol 3-kinase (PI3K), recruitment of the afadin–PI3K complex to the leading edge, and the activation of Akt, indicating the involvement of Rap1 and afadin in the PI3K–Akt signaling pathway. Binding of afadin to Rap1 regulated the activity of Rap1 in a positive-feedback manner. In vivo, conditional deletion of afadin in mouse vascular endothelium using a Cre-loxP system impaired the VEGF- and S1P-induced angiogenesis. Conclusions: These results demonstrate a novel molecular mechanism by which Rap1 and afadin regulate the VEGF- and S1P-induced angiogenesis.


Journal of Cell Science | 2009

Localization of nectin-free afadin at the leading edge and its involvement in directional cell movement induced by platelet-derived growth factor

Muneaki Miyata; Hisakazu Ogita; Hitomi Komura; Shinsuke Nakata; Ryoko Okamoto; Misa Ozaki; Takashi Majima; Naomi Matsuzawa; Satoshi Kawano; Akihiro Minami; Masumi Waseda; Naoyuki Fujita; Kiyohito Mizutani; Yoshiyuki Rikitake; Yoshimi Takai

Afadin is an actin-filament-binding protein that binds to nectin, an immunoglobulin-like cell-cell adhesion molecule, and plays an important role in the formation of adherens junctions. Here, we show that afadin, which did not bind to nectin and was localized at the leading edge of moving cells, has another role: enhancement of the directional, but not random, cell movement. When NIH3T3 cells were stimulated with platelet-derived growth factor (PDGF), afadin colocalized with PDGF receptor, αvβ3 integrin and nectin-like molecule-5 at the leading edge and facilitated the formation of leading-edge structures and directional cell movement in the direction of PDGF stimulation. However, these phenotypes were markedly perturbed by knockdown of afadin, and were dependent on the binding of afadin to active Rap1. Binding of Rap1 to afadin was necessary for the recruitment of afadin and the tyrosine phosphatase SHP-2 to the leading edge. SHP-2 was previously reported to tightly regulate the activation of PDGF receptor and its downstream signaling pathway for the formation of the leading edge. These results indicate that afadin has a novel role in PDGF-induced directional cell movement, presumably in cooperation with active Rap1 and SHP-2.


Journal of Biological Chemistry | 2009

Regulation by afadin of cyclical activation and inactivation of Rap1, Rac1, and RhoA small G proteins at leading edges of moving NIH3T3 cells.

Muneaki Miyata; Yoshiyuki Rikitake; Motonori Takahashi; Yuichi Nagamatsu; Yusuke Yamauchi; Hisakazu Ogita; Ken-ichi Hirata; Yoshimi Takai

Cyclical activation and inactivation of Rho family small G proteins, such as Rho, Rac, and Cdc42, are needed for moving cells to form leading edge structures in response to chemoattractants. However, the mechanisms underlying the dynamic regulation of their activities are not fully understood. We recently showed that another small G protein, Rap1, plays a crucial role in the platelet-derived growth factor (PDGF)-induced formation of leading edge structures and activation of Rac1 in NIH3T3 cells. We showed here that knockdown of afadin, an actin-binding protein, in NIH3T3 cells resulted in a failure to develop leading edge structures in association with an impairment of the activation of Rap1 and Rac1 and inactivation of RhoA in response to PDGF. Overexpression of a constitutively active mutant of Rap1 (Rap1-CA) and knockdown of SPA-1, a Rap1 GTPase-activating protein that was negatively regulated by afadin by virtue of binding to it, in afadin-knockdown NIH3T3 cells restored the formation of leading edge structures and the reduction of the PDGF-induced activation of Rac1 and inactivation of RhoA, suggesting that the inactivation of Rap1 by SPA-1 is responsible for inhibition of the formation of leading edge structures. The effect of Rap1-CA on the restoration of the formation of leading edge structures and RhoA inactivation was diminished by additional knockdown of ARAP1, a Rap-activated Rho GAP, which localized at the leading edges of moving NIH3T3 cells. These results indicate that afadin regulates the cyclical activation and inactivation of Rap1, Rac1, and RhoA through SPA-1 and ARAP1.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

FGD5 Mediates Proangiogenic Action of Vascular Endothelial Growth Factor in Human Vascular Endothelial Cells

Yusuke Kurogane; Muneaki Miyata; Yoshiki Kubo; Yuichi Nagamatsu; Ramendra K. Kundu; Akiyoshi Uemura; Tatsuro Ishida; Thomas Quertermous; Ken-ichi Hirata; Yoshiyuki Rikitake

Objective—Vascular endothelial growth factor (VEGF) exerts proangiogenic action and induces activation of a variety of proangiogenic signaling pathways, including the Rho family small G proteins. However, regulators of the Rho family small G proteins in vascular endothelial cells (ECs) are poorly understood. Here we attempted to clarify the expression, subcellular localization, downstream effectors, and proangiogenic role of FGD5, a member of the FGD family of guanine nucleotide exchange factors. Methods and Results—FGD5 was shown to be selectively expressed in cultured human vascular ECs. Immunofluorescence microscopy showed that the signal for FGD5 was observed at peripheral membrane ruffles and perinuclear regions in human umbilical vein ECs. Overexpression of FGD5 increased Cdc42 activity, whereas knockdown of FGD5 by small interfering RNAs inhibited the VEGF-induced activation of Cdc42 and extracellular signal–regulated kinase. VEGF-promoted capillary-like network formation, permeability, directional movement, and proliferation of human umbilical vein ECs and the reorientation of the Golgi complex during directional cell movement were attenuated by knockdown of FGD5. Conclusion—This study provides the first demonstration of expression, subcellular localization, and function of FGD5 in vascular ECs. The results suggest that FGD5 regulates proangiogenic action of VEGF in vascular ECs, including network formation, permeability, directional movement, and proliferation.


Circulation Research | 2012

Necl-5/Poliovirus Receptor Interacts With VEGFR2 and Regulates VEGF-Induced Angiogenesis

Mitsuo Kinugasa; Hisayuki Amano; Seimi Satomi-Kobayashi; Kazuhiko Nakayama; Muneaki Miyata; Yoshiki Kubo; Yuichi Nagamatsu; Yusuke Kurogane; Fumie Kureha; Shota Yamana; Ken-ichi Hirata; Jun Miyoshi; Yoshimi Takai; Yoshiyuki Rikitake

Rationale: Vascular endothelial growth factor (VEGF), a major proangiogenic agent, exerts its proangiogenic action by binding to VEGF receptor 2 (VEGFR2), the activity of which is regulated by direct interactions with other cell surface proteins, including integrin &agr;V&bgr;3. However, how the interaction between VEGFR2 and integrin &agr;V&bgr;3 is regulated is not clear. Objective: To investigate whether Necl-5/poliovirus receptor, an immunoglobulin-like molecule that is known to bind integrin &agr;V&bgr;3, regulates the interaction between VEGFR2 and integrin &agr;V&bgr;3, and to clarify the role of Necl-5 in the VEGF-induced angiogenesis. Methods and Results: Necl-5-knockout mice displayed no obvious defect in vascular development; however, recovery of blood flow after hindlimb ischemia and the VEGF-induced neovascularization in implanted Matrigel plugs were impaired in Necl-5-knockout mice. To clarify the mechanism of the regulation of angiogenesis by Necl-5, we investigated the roles of Necl-5 in the VEGF-induced angiogenic responses in vitro. Knockdown of Necl-5 by siRNAs in human umbilical vein endothelial cells (HUVECs) inhibited the VEGF-induced capillary-like network formation on Matrigel, migration, and proliferation, and conversely, enhanced apoptosis. Coimmunoprecipitation assays showed the interaction of Necl-5 with VEGFR2, and knockdown of Necl-5 prevented the VEGF-induced interaction of integrin &agr;V&bgr;3 with VEGFR2. Knockdown of Necl-5 suppressed the VEGFR2-mediated activation of downstream proangiogenic and survival signals, including Rap1, Akt, and endothelial nitric oxide synthase. Conclusions: These results demonstrate the critical role of Necl-5 in angiogenesis and suggest that Necl-5 may regulate the VEGF-induced angiogenesis by controlling the interaction of VEGFR2 with integrin &agr;v&bgr;3, and the VEGFR2-mediated Rap1-Akt signaling pathway.


Genes to Cells | 2010

Necl-5/PVR enhances PDGF-induced attraction of growing microtubules to the plasma membrane of the leading edge of moving NIH3T3 cells.

Akihiro Minami; Kiyohito Mizutani; Masazumi Waseda; Mihoko Kajita; Muneaki Miyata; Wataru Ikeda; Yoshimi Takai

Microtubules (MTs) search for and grow toward the leading edge of moving cells, followed by their stabilization at a specific structure at the rear site of the leading edge. This dynamic re‐orientation of MTs is critical to directional cell movement. We previously showed that Necl‐5/poliovirus receptor (PVR) interacts with platelet‐derived growth factor (PDGF) receptor and integrin αvβ3 at the leading edge of moving NIH3T3 cells, resulting in an enhancement of their directional movement. We studied here the role of Necl‐5 in the PDGF‐induced attraction of growing MTs to the leading edge of NIH3T3 cells. Necl‐5 enhanced the PDGF‐induced growth of MTs and attracted them near to the plasma membrane of the leading edge of NIH3T3 cells in an integrin αvβ3‐dependent manner. Furthermore, Necl‐5 enhanced the PDGF‐induced attraction of the plus‐end‐tracking proteins (+TIPs), including EB1, CLIP170, an intermediate chain subunit of cytoplasmic dynein, and p150Glued, a subunit of dynactin, near to the plasma membrane of the leading edge. Thus, Necl‐5 plays a role in the attraction of growing MTs to the plasma membrane of the leading edge of moving cells.


Genes to Cells | 2010

Interaction of integrin α6β4 with ErbB3 and implication in heregulin-induced ErbB3/ErbB2-mediated DNA synthesis

Satoshi Kawano; Kiyohito Mizutani; Muneaki Miyata; Wataru Ikeda; Yoshimi Takai

Integrin α6β4 is abundantly expressed in normal epithelial cells and forms hemidesmosomes, one of cell–extracellular matrix junctions. In many types of cancer cells, integrin α6β4 is up‐regulated, laminin, an integrin α6β4‐binding extracellular matrix protein, is cleaved, and hemidesmosomes are disrupted, eventually causing an enhancement of cancer cell movement and a facilitation of their invasion. It was previously shown that integrin α6β4 interacts with ErbB1 and ErbB2 and enhances cell proliferation and motility. Here we show that integrin α6β4 interacts with ErbB3 but not with ErbB1, ErbB2 or ErbB4, and enhances the heregulin‐induced, ErbB3/ErbB2 heterodimer‐mediated DNA synthesis, but not cell motility, in A549 cells.


Genes to Cells | 2014

s‐Afadin binds more preferentially to the cell adhesion molecules nectins than l‐afadin

Reiko Kobayashi; Souichi Kurita; Muneaki Miyata; Tomohiko Maruo; Kenji Mandai; Yoshiyuki Rikitake; Yoshimi Takai

l‐Afadin was originally purified from rat brain as an actin filament (F‐actin)‐binding protein that was homologous to the AF‐6 gene product. Concomitantly, s‐afadin that did not show an F‐actin‐binding capability was copurified with l‐afadin. Structurally, s‐afadin lacks the C‐terminal F‐actin‐binding domain but has two short sequences that were not present in l‐afadin. The properties and roles of l‐afadin have intensively been investigated, but those of s‐afadin have poorly been understood. We show here an additional difference in their biochemical properties other than binding to F‐actin between l‐afadin and s‐afadin. Both l‐afadin and s‐afadin bound to nectins, immunoglobulin‐like cell adhesion molecules, whereas s‐afadin more preferentially bound to nectins than l‐afadin. The PDZ domain of l‐afadin and s‐afadin was essential for their binding to nectin‐3. The dilute domain of l‐afadin negatively regulated its binding to nectin‐3, but the deletion of the C‐terminal F‐actin‐binding domain of l‐afadin did not increase the binding of l‐afadin to nectin‐3. These results indicate that the s‐afadin‐specific C‐terminal inserts may be involved in its preference of binding to nectin‐3 and raise the possibility that there are proteins other than nectins that more preferentially bind s‐afadin than l‐afadin.


The Journal of Comparative Neurology | 2017

Multiple roles of afadin in the ultrastructural morphogenesis of mouse hippocampal mossy fiber synapses: SAI et al.

Kousyoku Sai; Shujie Wang; Aika Kaito; Takeshi Fujiwara; Tomohiko Maruo; Yu Itoh; Muneaki Miyata; Shotaro Sakakibara; Naoyuki Miyazaki; Kazuyoshi Murata; Yuuki Yamaguchi; Tomohiro Haruta; Hideo Nishioka; Yuki Motojima; Miyuki Komura; Kazushi Kimura; Kenji Mandai; Yoshimi Takai; Akira Mizoguchi

A hippocampal mossy fiber synapse, which is implicated in learning and memory, has a complex structure in which mossy fiber boutons attach to the dendritic shaft by puncta adherentia junctions (PAJs) and wrap around a multiply‐branched spine, forming synaptic junctions. Here, we electron microscopically analyzed the ultrastructure of this synapse in afadin‐deficient mice. Transmission electron microscopy analysis revealed that typical PAJs with prominent symmetrical plasma membrane darkening undercoated with the thick filamentous cytoskeleton were observed in the control synapse, whereas in the afadin‐deficient synapse, atypical PAJs with the symmetrical plasma membrane darkening, which was much less in thickness and darkness than those of the control typical PAJs, were observed. Immunoelectron microscopy analysis revealed that nectin‐1, nectin‐3, and N‐cadherin were localized at the control typical PAJs, whereas nectin‐1 and nectin‐3 were localized at the afadin‐deficient atypical PAJs to extents lower than those in the control synapse and N‐cadherin was localized at their nonjunctional flanking regions. These results indicate that the atypical PAJs are formed by nectin‐1 and nectin‐3 independently of afadin and N‐cadherin and that the typical PAJs are formed by afadin and N‐cadherin cooperatively with nectin‐1 and nectin‐3. Serial block face‐scanning electron microscopy analysis revealed that the complexity of postsynaptic spines and mossy fiber boutons, the number of spine heads, the area of postsynaptic densities, and the density of synaptic vesicles docked to active zones were decreased in the afadin‐deficient synapse. These results indicate that afadin plays multiple roles in the complex ultrastructural morphogenesis of hippocampal mossy fiber synapses.

Collaboration


Dive into the Muneaki Miyata's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoshiyuki Rikitake

Kobe Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge