Network


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

Hotspot


Dive into the research topics where Yoh Wada is active.

Publication


Featured researches published by Yoh Wada.


Circulation Research | 2010

The (Pro)renin Receptor/ATP6AP2 is Essential for Vacuolar H+-ATPase Assembly in Murine Cardiomyocytes

Kenichiro Kinouchi; Atsuhiro Ichihara; Motoaki Sano; Ge-Hong Sun-Wada; Yoh Wada; Asako Kurauchi-Mito; Kanako Bokuda; Tatsuya Narita; Yoichi Oshima; Mariyo Sakoda; Yoshitaka Tamai; Hiromu Sato; Keiichi Fukuda; Hiroshi Itoh

Rationale: The (pro)renin receptor [(P)RR], encoded in ATP6AP2, plays a key role in the activation of local renin-angiotensin system (RAS). A truncated form of (P)RR, termed M8.9, was also found to be associated with the vacuolar H+-ATPase (V-ATPase), implicating a non–RAS-related function of ATP6AP2. Objective: We investigated the role of (P)RR/ATP6AP2 in murine cardiomyocytes. Methods and Results: Cardiomyocyte-specific ablation of Atp6ap2 resulted in lethal heart failure; the cardiomyocytes contained RAB7- and lysosomal-associated membrane protein 2 (LAMP2)-positive multivesicular vacuoles, especially in the perinuclear regions. The myofibrils and mitochondria remained at the cell periphery. Cardiomyocyte death was accompanied by numerous autophagic vacuoles that contained undigested cellular constituents, as a result of impaired autophagic degradation. Notably, ablation of Atp6ap2 selectively suppressed expression of the VO subunits of V-ATPase, resulting in deacidification of the intracellular vesicles. Furthermore, the inhibition of intracellular acidification by treatment with bafilomycin A1 or chloroquine reproduced the phenotype observed for the (P)RR/ATP6AP2-deficient cardiomyocytes. Conclusions: Genetic ablation of Atp6ap2 created a loss-of-function model for V-ATPase. The gene product of ATP6AP2 is considered to act as in 2 ways: (1) as (P)RR, exerting a RAS-related function; and (2) as the V-ATPase-associated protein, exerting a non–RAS-related function that is essential for cell survival.


Journal of Cell Science | 2006

The a3 isoform of V-ATPase regulates insulin secretion from pancreatic β-cells

Ge-Hong Sun-Wada; Takao Toyomura; Yoshiko Murata; Akitsugu Yamamoto; Masamitsu Futai; Yoh Wada

Vacuolar-type H+-ATPase (V-ATPase) is a multi-subunit enzyme that has important roles in the acidification of a variety of intracellular compartments and some extracellular milieus. Four isoforms for the membrane-intrinsic subunit (subunit a) of the V-ATPase have been identified in mammals, and they confer distinct cellular localizations and activities on the proton pump. We found that V-ATPase with the a3 isoform is highly expressed in pancreatic islets, and is localized to membranes of insulin-containing secretory granules in β-cells. oc/oc mice, which have a null mutation at the a3 locus, exhibited a reduced level of insulin in the blood, even with high glucose administration. However, islet lysates contained mature insulin, and the ratio of the amount of insulin to proinsulin in oc/oc islets was similar to that of wild-type islets, indicating that processing of insulin was normal even in the absence of the a3 function. The insulin contents of oc/oc islets were reduced slightly, but this was not significant enough to explain the reduced levels of the blood insulin. The secretion of insulin from isolated islets in response to glucose or depolarizing stimulation was impaired. These results suggest that the a3 isoform of V-ATPase has a regulatory function in the exocytosis of insulin secretion.


Journal of Cell Science | 2009

Direct recruitment of H+-ATPase from lysosomes for phagosomal acidification

Ge-Hong Sun-Wada; Hiroyuki Tabata; Nobuyuki Kawamura; Minako Aoyama; Yoh Wada

The nascent phagosome progressively establishes an acidic milieu by acquiring a proton pump, the vacuolar-type ATPase (V-ATPase). However, the origin of phagosomal V-ATPase remains poorly understood. We found that phagosomes were enriched with the V-ATPase a3 subunit, which also accumulated in late endosomes and lysosomes. We modified the mouse Tcirg1 locus encoding subunit a3, to express an a3-GFP fusion protein. Live-cell imaging and immunofluorescence microscopy revealed that nascent phagosomes received the a3-GFP from tubular structures extending from lysosomes located in the perinuclear region. Macrophages from a3-deficient mice exhibited impaired acidification of phagosomes and delayed digestion of bacteria. These results show that lysosomal V-ATPase is recruited directly to the phagosomes via tubular lysosomes to establish the acidic environment hostile to pathogens.


Molecular Cancer Research | 2011

The a3 Isoform Vacuolar Type H+-ATPase Promotes Distant Metastasis in the Mouse B16 Melanoma Cells

Toshihiko Nishisho; Kenji Hata; Masako Nakanishi; Yoshihiro Morita; Ge-Hong Sun-Wada; Yoh Wada; Natsuo Yasui; Toshiyuki Yoneda

Accumulating evidence indicates that the acidic microenvironments critically influence malignant behaviors of cancer including invasiveness, metastasis, and chemoresistance. Because the vacuolar-type H+-ATPase (V-ATPase) has been shown to cause extracellular acidification by pumping protons, we studied the role of V-ATPase in distant metastasis. Real-time PCR analysis revealed that the high-metastatic B16-F10 melanoma cells strongly expressed the a3 isoform V-ATPase compared to the low-metastatic B16 parental cells. Consistent with this, B16-F10 cells created acidic environments in lung metastases by acridine orange staining and strong a3 V-ATPase expression in bone metastases by immunohistochemistry. Immunocytochemical analysis showed B16-F10 cells expressed a3 V-ATPase not only in cytoplasm but also plasma membrane, whereas B16 parental cells exhibited its expression only in cytoplasm. Of note, knockdown of a3 V-ATPase suppressed invasiveness and migration with reduced MMP-2 and MMP-9 expression in B16-F10 cells and significantly decreased lung and bone metastases, despite that tumor growth was not altered. Importantly, administration of a specific V-ATPase a3 inhibitor FR167356 reduced bone metastasis of B16-F10 cells. These results suggest that a3 V-ATPase promotes distant metastasis of B16-F10 cells by creating acidic environments via proton secretion. Our results also suggest that inhibition of the development of cancer-associated acidic environments by suppressing a3 V-ATPase could be a novel therapeutic approach for the treatment of cancer metastasis. Mol Cancer Res; 9(7); 845–55. ©2011 AACR.


Gene | 2003

Diversity of mouse proton-translocating ATPase: presence of multiple isoforms of the C, d and G subunits

Ge-Hong Sun-Wada; Takao Yoshimizu; Yoko Imai-Senga; Yoh Wada; Masamitsu Futai

Vacuolar-type proton-translocating ATPases (V-ATPases), multimeric proton pumps, are involved in a wide variety of physiological processes. For their diverse functions, V-ATPases utilize a specific subunit isoform(s). Here, we reported the molecular cloning and characterization of three novel subunit isoforms, C2, d2 and G3, of mouse V-ATPase. These isoforms were expressed in a tissue-specific manner, in contrast to the ubiquitously expressed C1, d1 and G1 isoforms. C2 was expressed predominantly in lung and kidney, and d2 and G3 specifically in kidney. We introduced these isoforms into yeasts lacking the corresponding genes. Although the G3 and d2 did not rescue the vmaDelta phenotype, d1 and the two C isoforms functionally complemented the Deltavma6 and Deltavma5, respectively, indicating that they are bona fide subunits of V-ATPase.


Journal of Biological Chemistry | 2003

Mouse Proton Pump ATPase C Subunit Isoforms (C2-a and C2-b) Specifically Expressed in Kidney and Lung

Ge-Hong Sun-Wada; Yoshiko Murata; Miwako Namba; Akitsugu Yamamoto; Yoh Wada; Masamitsu Futai

The vacuolar-type H+-ATPases (V-ATPases) are multimeric proton pumps involved in a wide variety of physiological processes. We have identified two alternative splicing variants of C2 subunit isoforms: C2-a, a lungspecific isoform containing a 46-amino acid insertion, and C2-b, a kidney-specific isoform without the insert. Immunohistochemistry with isoform-specific antibodies revealed that V-ATPase with C2-a is localized specifically in lamellar bodies of type II alveolar cells, whereas the C2-b isoform is found in the plasma membranes of renal α and β intercalated cells. Immunoprecipitation combined with immunohistological analysis revealed that C2-b together with other kidney-specific isoforms was selectively assembled to form a unique proton pump in intercalated cells. Furthermore, a chimeric yeast V-ATPase with mouse the C2-a or C2-b isoform showed a lower Km(ATP) and lower proton transport activity than that with C1 or Vma5p (yeast C subunit). These results suggest that V-ATPases with the C2-a and C2-b isoform are involved in luminal acidification of lamellar bodies and regulation of the renal acid-base balance, respectively.


Nature Communications | 2012

Delivery of endosomes to lysosomes via microautophagy in the visceral endoderm of mouse embryos

Nobuyuki Kawamura; Ge-Hong Sun-Wada; Minako Aoyama; Akihiro Harada; Shunsuke Takasuga; Takehiko Sasaki; Yoh Wada

The differentiation and patterning of murine early embryos are sustained by the visceral endoderm, an epithelial layer of polarised cells that has critical roles in multiple signalling pathways and nutrient uptake. Both nutritional and signalling functions rely upon the endocytosis of various molecules from the cell surface via the endocytic pathway. However, endocytic membrane dynamics in this embryonic tissue remain poorly understood. Here we show that the functions of rab7, a small GTP-binding protein regulating the late endocytic pathway, are essential for embryonic patterning during gastrulation. The endosomes of visceral endoderm cells are delivered via a unique microautophagy-like process to the apical vacuole, a large compartment exhibiting lysosomal characteristics. Loss of rab7 function results in severe inhibition of this endocytic pathway. Our results indicate that the microautophagic process and flow of the endocytic membrane have essential roles in early embryonic development.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Critical roles of type III phosphatidylinositol phosphate kinase in murine embryonic visceral endoderm and adult intestine

Shunsuke Takasuga; Yasuo Horie; Junko Sasaki; Ge-Hong Sun-Wada; Nobuyuki Kawamura; Ryota Iizuka; Katsunori Mizuno; Satoshi Eguchi; Satoshi Kofuji; Hirotaka Kimura; Masakazu Yamazaki; Chihoko Horie; Eri Odanaga; Yoshiko Sato; Shinsuke Chida; Kenji Kontani; Akihiro Harada; Toshiaki Katada; Akira Suzuki; Yoh Wada; Hirohide Ohnishi; Takehiko Sasaki

The metabolism of membrane phosphoinositides is critical for a variety of cellular processes. Phosphatidylinositol-3,5-bisphosphate [PtdIns(3,5)P2] controls multiple steps of the intracellular membrane trafficking system in both yeast and mammalian cells. However, other than in neuronal tissues, little is known about the physiological functions of PtdIns(3,5)P2 in mammals. Here, we provide genetic evidence that type III phosphatidylinositol phosphate kinase (PIPKIII), which produces PtdIns(3,5)P2, is essential for the functions of polarized epithelial cells. PIPKIII-null mouse embryos die by embryonic day 8.5 because of a failure of the visceral endoderm to supply the epiblast with maternal nutrients. Similarly, although intestine-specific PIPKIII-deficient mice are born, they fail to thrive and eventually die of malnutrition. At the mechanistic level, we show that PIPKIII regulates the trafficking of proteins to a cell’s apical membrane domain. Importantly, mice with intestine-specific deletion of PIPKIII exhibit diarrhea and bloody stool, and their gut epithelial layers show inflammation and fibrosis, making our mutants an improved model for inflammatory bowel diseases. In summary, our data demonstrate that PIPKIII is required for the structural and functional integrity of two different types of polarized epithelial cells and suggest that PtdIns(3,5)P2 metabolism is an unexpected and critical link between membrane trafficking in intestinal epithelial cells and the pathogenesis of inflammatory bowel disease.


Archive | 2004

Handbook of ATPases : biochemistry, cell biology, pathophysiology

Masamitsu Futai; Yoh Wada; Jack H. Kaplan

Preface. List of Contributors. PART I: P--TYPE ATPASES. 1. Yeast plasma--membrane H+--ATPase: Model System for Studies of Structure, Function, Biogenesis and Regulation (S. Lecchi & C. Slayman). 2. Regulation of the Sarco(endo)plasmic Reticulum Ca2+--ATPase by Phospholamban and Sarcolipin (D. McLennan & E. Kranias). 3. Catalysis and Transport Mechanism of the Sarco--(Endo)Plasmic Reticulum Ca2+--ATPase (SERCA) (G. Inesi & C. Toyoshima). 4. The Na,K--ATPase: A Current Overview (J. Kaplan). 5. Copper--Transporting ATPases: Key Regulators of Intracellular Copper Concentration (R. Tsivkovskii, et al.). 6. Bacterial Transport ATPases for Monovalent, Divalent and Trivalent Soft Metal Ions (M. Wong, et al.). 7. Gastric H+,K+--ATPase (J. Shin, et al.). 8. Plasma Membrane Calcium Pumps (E. Carafoli, et al.). PART II: F--TYPE ATPASES. 9. Proton Translocating ATPases: Introducing Unique Enzymes Coupling Catalysis and Proton Translocation through Mechanical Rotation (M. Futai, et al.). 10. Rotation of F1--ATPase (E. Muneyuki & M. Yoshida). 11. Coordinating Catalysis and Rotation in the F1--ATPase (R. Nakamoto, et al.). 12. ATP Synthase Stalk Subunits b, eth and e: Structures and Functions in Energy Coupling (S. Dunn, et al.). PART III: V--TYPE ATPASES. 13. Structure, Mechanism and Regulation of the Yeast and Coated Vesicle V--ATPases (E. Shao & M. Forgac). 14. Role of the V--ATPase in the Cellular Physiology of the Yeast Saccharomyces cerevisiae (L. Graham, et al.). 15. Vacuolar--Type Proton ATPases: Subunit Isoforms and Tissue--Specific Functions (G. Sun--Wada, et al.). PART IV: CELL BIOLOGY AND PATHOPHYSIOLOGY OF ATPASES AND THEIR COMPARTMENTS. 16. Physiological Role of Na,K--ATPases Isoforms (J. Lingrel, et al.). 17. Renal V--ATPase: Physiology and Pathophysiology (D. Brown & V. Marshansky). 18. Lytic Function of Vacuole and Molecular Dissection of Autophagy in Yeast (Y. Ohsumi). Index.


Journal of Biological Chemistry | 2007

Rotational Catalysis of Escherichia coli ATP Synthase F1 Sector STOCHASTIC FLUCTUATION AND A KEY DOMAIN OF THE β SUBUNIT

Mayumi Nakanishi-Matsui; Sachiko Kashiwagi; Toshiharu Ubukata; Atsuko Iwamoto-Kihara; Yoh Wada; Masamitsu Futai

A complex of γ, ϵ, and c subunits rotates in ATP synthase (FoF1) coupled with proton transport. A gold bead connected to the γ subunit of the Escherichia coli F1 sector exhibited stochastic rotation, confirming a previous study (Nakanishi-Matsui, M., Kashiwagi, S., Hosokawa, H., Cipriano, D. J., Dunn, S. D., Wada, Y., and Futai, M. (2006) J. Biol. Chem. 281, 4126-4131). A similar approach was taken for mutations in the β subunit key region; consistent with its bulk phase ATPase activities, F1 with the Ser-174 to Phe substitution (βS174F) exhibited a slower single revolution time (time required for 360 degree revolution) and paused almost 10 times longer than the wild type at one of the three 120° positions during the stepped revolution. The pause positions were probably not at the “ATP waiting” dwell but at the “ATP hydrolysis/product release” dwell, since the ATP concentration used for the assay was ∼30-fold higher than the Km value for ATP. A βGly-149 to Ala substitution in the phosphate binding P-loop suppressed the defect of βS174F. The revertant (βG149A/βS174F) exhibited similar rotation to the wild type, except that it showed long pauses less frequently. Essentially the same results were obtained with the Ser-174 to Leu substitution and the corresponding revertant βG149A/βS174L. These results indicate that the domain between β-sheet 4 (βSer-174) and P-loop (βGly-149) is important to drive rotation.

Collaboration


Dive into the Yoh Wada's collaboration.

Top Co-Authors

Avatar

Ge-Hong Sun-Wada

Doshisha Women's College of Liberal Arts

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nobuyuki Kawamura

Doshisha Women's College of Liberal Arts

View shared research outputs
Top Co-Authors

Avatar

Hiroyuki Tabata

Doshisha Women's College of Liberal Arts

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