100 results on '"Miake, J"'
Search Results
2. The impact of anatomical variation of pulmonary veins on pulmonary vein isolation procedure for paroxysmal atrial fibrillation patients
- Author
-
Kondo, K., Adachi, M., Miake, J., Yano, A., Ogura, K., Kato, M., and Shigemasa, C.
- Published
- 2011
3. Transcriptional activation of the anchoring protein SAP97 by heat shock factor (HSF)-1 stabilizes Kv1.5 channels in HL-1 cells
- Author
-
Ting, Y K, Morikawa, K, Kurata, Y, Li, P, Bahrudin, U, Mizuta, E, Kato, M, Miake, J, Yamamoto, Y, Yoshida, A, Murata, M, Inoue, T, Nakai, A, Shiota, G, Higaki, K, Nanba, E, Ninomiya, H, Shirayoshi, Y, and Hisatome, I
- Published
- 2011
- Full Text
- View/download PDF
4. Poster Session 3
- Author
-
Fabbri, G. M. T., primary, Baldasseroni, S., additional, Panuccio, D., additional, Zoni Berisso, M., additional, Scherillo, M., additional, Lucci, D., additional, Di Pasquale, G., additional, Mathieu, G., additional, Burazor, I., additional, Burazor, M., additional, Perisic, Z., additional, Atanaskovic, V., additional, Erakovic, V., additional, Stojkovic, A., additional, Vogtmann, T., additional, Schoebel, C., additional, Sogorski, S., additional, Sebert, M., additional, Schaarschmidt, J., additional, Fietze, I., additional, Baumann, G., additional, Penzel, T., additional, Mornos, C., additional, Ionac, A., additional, Cozma, D., additional, Dragulescu, D., additional, Mornos, A., additional, Petrescu, L., additional, Pescariu, L., additional, Brembilla-Perrot, B., additional, Khachab, H., additional, Lamberti, F., additional, Bellini, C., additional, Remoli, R., additional, Cogliandro, T., additional, Nardo, R., additional, Bellusci, F., additional, Mazzuca, V., additional, Gaspardone, A., additional, Aguinaga Arrascue, L. E., additional, Bravo, A., additional, Garcia Freire, P., additional, Gallardo, P., additional, Hasbani, E., additional, Quintana, R., additional, Dantur, J., additional, Inoue, K., additional, Ueoka, A., additional, Tsubakimoto, Y., additional, Sakatani, T., additional, Matsuo, A., additional, Fujita, H., additional, Kitamura, M., additional, Wegrzynowska, M., additional, Konduracka, E., additional, Pietrucha, A. Z., additional, Mroczek-Czernecka, D., additional, Paradowski, A., additional, Bzukala, I., additional, Nessler, J., additional, Igawa, O., additional, Adachi, M., additional, Atarashi, H., additional, Kusama, Y., additional, Kodani, E., additional, Okazaki, R., additional, Nakagomi, A., additional, Endoh, Y., additional, Baez-Escudero, J. L., additional, Dave, A. S., additional, Sasaridis, C. M., additional, Valderrabano, M., additional, Tilz, R., additional, Bai, R., additional, Di Biase, L., additional, Gallinghouse, G. J., additional, Gibson, D., additional, Pisapia, A., additional, Wazni, O., additional, Natale, A., additional, Arujuna, A., additional, Karim, R., additional, Rinaldi, A., additional, Cooklin, M., additional, Rhode, K., additional, Razavi, R., additional, O'neill, M., additional, Gill, J., additional, Kusa, S., additional, Komatsu, Y., additional, Kakita, K., additional, Takayama, K., additional, Taniguchi, H., additional, Otomo, K., additional, Iesaka, Y., additional, Ammar, S., additional, Reents, T., additional, Fichtner, S., additional, Wu, J., additional, Zhu, P., additional, Kolb, C., additional, Hessling, G., additional, Deisenhofer, I., additional, Gilbert, G., additional, Mohanty, P., additional, Cunningham, J., additional, Metz, T., additional, Horton, R., additional, Tao, S., additional, Yamauchi, Y., additional, Okada, H., additional, Maeda, S., additional, Obayashi, T., additional, Isobe, M., additional, Chan, J., additional, Johar, S., additional, Wong, T., additional, Markides, V., additional, Hussain, W., additional, Konstantinidou, M., additional, Wissner, E., additional, Fuernkranz, A., additional, Yoshiga, Y., additional, Metzner, A., additional, Kuck, K.- H., additional, Ouyang, F., additional, Kettering, K., additional, Gramley, F., additional, Mollnau, H., additional, Weiss, C., additional, Bardeleben, S., additional, Biasco, L., additional, Scaglione, M., additional, Caponi, D., additional, Di Donna, P., additional, Sergi, D., additional, Cerrato, N., additional, Blandino, A., additional, Gaita, F., additional, Fiala, M., additional, Wichterle, D., additional, Sknouril, L., additional, Bulkova, V., additional, Chovancik, J., additional, Nevralova, R., additional, Pindor, J., additional, Januska, J., additional, Choi, J. I., additional, Ban, J. E., additional, Yasutsugu, N., additional, Park, J. S., additional, Jung, J. S., additional, Lim, H. E., additional, Park, S. W., additional, Kim, Y. H., additional, Kuhne, M., additional, Reichlin, T., additional, Ammann, P., additional, Schaer, B., additional, Osswald, S., additional, Sticherling, C., additional, Ohe, M., additional, Goya, M., additional, Hiroshima, K., additional, Hayashi, K., additional, Makihara, Y., additional, Nagashima, M., additional, Fukunaga, M., additional, An, Y., additional, Dorwarth, U., additional, Schmidt, M., additional, Wankerl, M., additional, Krieg, J., additional, Straube, F., additional, Hoffmann, E., additional, Kathan, S., additional, Defaye, P., additional, Mbaye, A., additional, Cassagneau, R., additional, Gagniere, V., additional, Jacon, P., additional, Pokushalov, E., additional, Romanov, A., additional, Artemenko, S., additional, Shabanov, V., additional, Elesin, D., additional, Stenin, I., additional, Turov, A., additional, Losik, D., additional, Kondo, K., additional, Miake, J., additional, Yano, A., additional, Ogura, K., additional, Kato, M., additional, Shigemasa, C., additional, Sekiguchi, Y., additional, Tada, H., additional, Yoshida, K., additional, Naruse, Y., additional, Yamasaki, H., additional, Igarashi, M., additional, Machino, T., additional, Aonuma, K., additional, Chen, S., additional, Liu, S., additional, Chen, G., additional, Meng, W., additional, Zhang, F., additional, Yan, Y., additional, Sciarra, L., additional, Dottori, S., additional, Lanzillo, C., additional, De Ruvo, E., additional, De Luca, L., additional, Minati, M., additional, Lioy, E., additional, Calo', L., additional, Lin, J., additional, Nie, Z., additional, Zhu, M., additional, Wang, X., additional, Zhao, J., additional, Hu, W., additional, Tao, H., additional, Ge, J., additional, Johansson, B., additional, Houltz, B., additional, Edvardsson, N., additional, Schersten, H., additional, Karlsson, T., additional, Wandt, B., additional, Berglin, E., additional, Hoyt, R. H., additional, Jenson, B. P., additional, Trines, S. A. I. P., additional, Braun, J., additional, Tjon Joek Tjien, A., additional, Zeppenfeld, K., additional, Tavilla, G., additional, Klautz, R. J. M., additional, Schalij, M. J., additional, Krausova, R., additional, Cihak, R., additional, Peichl, P., additional, Kautzner, J., additional, Pirk, J., additional, Skalsky, I., additional, Maly, J., additional, Imai, K., additional, Sueda, T., additional, Orihashi, K., additional, Picarra, B. C., additional, Santos, A. R., additional, Dionisio, P., additional, Semedo, P., additional, Matos, R., additional, Leitao, M., additional, Banha, M., additional, Trinca, M., additional, Elder, D. H. J., additional, George, J., additional, Jain, R., additional, Lang, C. C., additional, Choy, A. M., additional, Konert, M., additional, Loescher, S., additional, Hartmann, A., additional, Aversa, E., additional, Chirife, R., additional, Sztyglic, E., additional, Mazzetti, H., additional, Mascheroni, O., additional, Tentori, M. C., additional, Pop, R. M., additional, Margulescu, A. D., additional, Dulgheru, R., additional, Enescu, O., additional, Siliste, C., additional, Vinereanu, D., additional, Menezes Junior, A., additional, Castro Carneiro, A. R., additional, De Oliveira, B. L., additional, Shah, A. N., additional, Kantharia, B., additional, De Lucia, R., additional, Soldati, E., additional, Segreti, L., additional, Di Cori, A., additional, Zucchelli, G., additional, Viani, S., additional, Paperini, L., additional, Bongiorni, M. G., additional, Kutarski, A., additional, Czajkowski, M., additional, Pietura, R., additional, Malecka, B., additional, Heintze, J., additional, Eckardt, L., additional, Bauer, A., additional, Meine, M., additional, Van Erven, L., additional, Bloch Thomsen, P. E., additional, Lopez Chicharro, M. P., additional, Merhi, O., additional, Soga, Y., additional, Andou, K., additional, Nobuyoshi, M., additional, Gonzalez-Mansilla, A., additional, Martin-Asenjo, R., additional, Unzue, L., additional, Torres, J., additional, Garralda, E., additional, Coma, R. R., additional, Rodriguez Garcia, J. E., additional, Yaegashi, T., additional, Furusho, H., additional, Kato, T., additional, Chikata, A., additional, Takashima, S., additional, Usui, S., additional, Takamura, M., additional, Kaneko, S., additional, Chudzik, M., additional, Mitkowski, P., additional, Przybylski, A., additional, Lewek, J., additional, Smukowski, T., additional, Maciag, A., additional, Castrejon Castrejon, S., additional, Perez-Silva, A., additional, Estrada, A., additional, Doiny, D., additional, Ortega, M., additional, Lopez-Sendon, J. L., additional, Merino, J. L., additional, O'mahony, C., additional, Coats, C., additional, Cardona, M., additional, Garcia, A., additional, Calcagnino, M., additional, Lachmann, R., additional, Hughes, D., additional, Elliott, P. M., additional, Conti, S., additional, Pruiti, G. P., additional, Puzzangara, E., additional, Romano, S. A., additional, Di Grazia, A., additional, Ussia, G. P., additional, Tamburino, C., additional, Calvi, V., additional, Radinovic, A., additional, Sala, S., additional, Latib, A., additional, Mussardo, M., additional, Sora, S., additional, Paglino, G., additional, Gullace, M., additional, Colombo, A., additional, Ohlow, M.- A. G., additional, Lauer, B., additional, Wagner, A., additional, Schreiber, M., additional, Buchter, B., additional, Farah, A., additional, Fuhrmann, J. T., additional, Geller, J. C., additional, Nascimento Cardoso, R. M., additional, Batista Sa, L. A., additional, Campos Filho, L. F. C., additional, Rodrigues, S. V., additional, Dutra, M. V. F., additional, Borges, T. R. S. A., additional, Portilho, D. R., additional, Deering, T., additional, Bernardes, A., additional, Veiga, A., additional, Gartenlaub, O., additional, Goncalves, A., additional, Jimenez, A., additional, Rousseauplasse, A., additional, Deharo, J. C., additional, Striekwold, H., additional, Gosselin, G., additional, Sitbon, H., additional, Martins, V., additional, Molon, G., additional, Ayala-Paredes, F., additional, Sancho-Tello, M. J., additional, Fazal, I. A., additional, Brady, S., additional, Cronin, J., additional, Mcnally, S., additional, Tynan, M., additional, Plummer, C. J., additional, Mccomb, J. M., additional, Val-Mejias, J. E., additional, Oliveira, R. M., additional, Costa, R., additional, Martinelli Filho, M., additional, Silva, K. R., additional, Menezes, L. M., additional, Tamaki, W. T., additional, Mathias, W., additional, Stolf, N. A. G., additional, Misawa, T., additional, Ohta, I., additional, Shishido, T., additional, Miyasita, T., additional, Miyamoto, T., additional, Nitobe, J., additional, Watanabe, T., additional, Kubota, I., additional, Thibault, B., additional, Ducharme, A., additional, Simpson, C., additional, Stuglin, C., additional, Gagne, C. E., additional, Williams, R., additional, Mcnicoll, S., additional, Silvetti, M. S., additional, Drago, F., additional, Penela, D., additional, Bijnens, B., additional, Doltra, A., additional, Silva, E., additional, Berruezo, A., additional, Mont, L., additional, Sitges, M., additional, Mcintosh, R., additional, Baumann, O., additional, Raju, P., additional, Gurunathan, S., additional, Furniss, S., additional, Patel, N., additional, Sulke, N., additional, Lloyd, G., additional, Mor, M., additional, Dror, S., additional, Tsadok, Y., additional, Bachner-Hinenzon, N., additional, Katz, A., additional, Liel-Cohen, N., additional, Etzion, Y., additional, Mlynarski, R., additional, Mlynarska, A., additional, Wilczek, J., additional, Sosnowski, M., additional, Sinha, A. M., additional, Sinha, D., additional, Noelker, G., additional, Brachmann, J., additional, Weidemann, F., additional, Ertl, G., additional, Jones, M., additional, Searle, N., additional, Cocker, M., additional, Ilsley, E., additional, Foley, P., additional, Khiani, R., additional, Nelson, K. E., additional, Turley, A. J., additional, Owens, W. A., additional, James, S. A., additional, Linker, N. J., additional, Velagic, V., additional, Cikes, M., additional, Pezo Nikolic, B., additional, Puljevic, D., additional, Separovic-Hanzevacki, J., additional, Lovric-Bencic, M., additional, Biocina, B., additional, Milicic, D., additional, Kawata, H., additional, Chen, L., additional, Phan, H., additional, Anand, K., additional, Feld, G., additional, Birgesdotter-Green, U., additional, Fernandez Lozano, I., additional, Mitroi, C., additional, Toquero Ramos, J., additional, Castro Urda, V., additional, Monivas Palomero, V., additional, Corona Figueroa, A., additional, Hernandez Reina, L., additional, Alonso Pulpon, L., additional, Gate-Martinet, A., additional, Da Costa, A., additional, Rouffiange, P., additional, Cerisier, A., additional, Bisch, L., additional, Romeyer-Bouchard, C., additional, Isaaz, K., additional, Morales, M.- A., additional, Bianchini, E., additional, Startari, U., additional, Faita, F., additional, Bombardini, T., additional, Gemignani, V., additional, Piacenti, M., additional, Adhya, S., additional, Kamdar, R. H., additional, Millar, L. M., additional, Burchardt, C., additional, Murgatroyd, F. D., additional, Klug, D., additional, Kouakam, C., additional, Guedon-Moreau, L., additional, Marquie, C., additional, Benard, S., additional, Kacet, S., additional, Cortez-Dias, N., additional, Carrilho-Ferreira, P., additional, Silva, D., additional, Goncalves, S., additional, Valente, M., additional, Marques, P., additional, Carpinteiro, L., additional, Sousa, J., additional, Keida, T., additional, Nishikido, T., additional, Fujita, M., additional, Chinen, T., additional, Kikuchi, T., additional, Nakamura, K., additional, Ohira, H., additional, Takami, M., additional, Anjo, D., additional, Meireles, A., additional, Gomes, C., additional, Roque, C., additional, Pinheiro Vieira, A., additional, Lagarto, V., additional, Reis, H., additional, Torres, S., additional, Ortega, D. F., additional, Barja, L. D., additional, Montes, J. P., additional, Logarzo, E., additional, Bonomini, P., additional, Mangani, N., additional, Paladino, C., additional, Chwyczko, T., additional, Smolis-Bak, E., additional, Sterlinski, M., additional, Pytkowski, M., additional, Firek, B., additional, Jankowska, A., additional, Szwed, H., additional, Nakajima, I., additional, Noda, T., additional, Okamura, H., additional, Satomi, K., additional, Aiba, T., additional, Shimizu, W., additional, Aihara, N., additional, Kamakura, S., additional, Brzozowski, W., additional, Tomaszewski, A., additional, Wysokinski, A., additional, Bertoldi, E. G., additional, Rohde, L. E., additional, Zimerman, L. I., additional, Pimentel, M., additional, Polanczyk, C. A., additional, Boriani, G., additional, Lunati, M., additional, Gasparini, M., additional, Landolina, M., additional, Lonardi, G., additional, Pecora, D., additional, Santini, M., additional, Valsecchi, S., additional, Rubinstein, B. J., additional, Wang, D. Y., additional, Cabreriza, S. E., additional, Richmond, M. E., additional, Rusanov, A., additional, Quinn, T. A., additional, Cheng, B., additional, Spotnitz, H. M., additional, Kristiansen, H. M., additional, Vollan, G., additional, Hovstad, T., additional, Keilegavlen, H., additional, Faerestrand, S., additional, Brigesdotter-Green, U., additional, Nawar, A. M. R., additional, Ragab, D. A. L. I. A., additional, Eluhsseiny, R. A. N. I. A., additional, Abdelaziz, A. H. M. E. D., additional, Nof, E., additional, Abu Shama, R., additional, Buber, J., additional, Kuperstein, R., additional, Feinberg, M. S., additional, Barlev, D., additional, Eldar, M., additional, Glikson, M., additional, Badran, H., additional, Samir, R., additional, Tawfik, M., additional, Amin, M., additional, Eldamnhoury, H., additional, Khaled, S., additional, Tolosana, J. M., additional, Martin, A. M., additional, Hernandez-Madrid, A., additional, Macias, A., additional, Fernandez-Lozano, I., additional, Osca, J., additional, Quesada, A., additional, Padeletti, L., additional, Botto, G. L., additional, De Santo, T., additional, Szwed, A., additional, Martinez, J. G., additional, Degand, B., additional, Villani, G. Q., additional, Leclercq, C., additional, Ritter, P., additional, Watanabe, I., additional, Nagashima, K., additional, Okumura, Y., additional, Kofune, M., additional, Ohkubo, K., additional, Nakai, T., additional, Hirayama, A., additional, Mikhaylov, E., additional, Vander, M., additional, Lebedev, D., additional, Zarse, M., additional, Suleimann, H., additional, Bogossian, H., additional, Stegelmeyer, J., additional, Ninios, I., additional, Karosienne, Z., additional, Kloppe, A., additional, Lemke, B., additional, John, S., additional, Gaspar, T., additional, Rolf, S., additional, Sommer, P., additional, Hindricks, G., additional, Piorkowski, C., additional, Fernandez-Armenta, J., additional, Mont, L. L., additional, Zeljko, H., additional, Andreu, D., additional, Herzcku, C., additional, Boussy, T., additional, Brugada, J., additional, Obayahi, T., additional, Hegrenes, J., additional, Lim, E., additional, Mediratta, V., additional, Bautista, R., additional, Teplitsky, L., additional, Van Huls Van Taxis, C. F. B., additional, Wijnmaalen, A. P., additional, Gawrysiak, M., additional, Schuijf, J. D., additional, Bax, J. J., additional, Huo, Y., additional, Richter, S., additional, Arya, A., additional, Bollmann, A., additional, Akca, F., additional, Bauernfeind, T., additional, Schwagten, B., additional, De Groot, N. M. S., additional, Jordaens, L., additional, Szili-Torok, T., additional, Miller, S., additional, Kastner, G., additional, Maury, P., additional, Della Bella, P., additional, Delacretaz, E., additional, Sacher, F., additional, Maccabelli, G., additional, Brenner, R., additional, Rollin, A., additional, Jais, P., additional, Vergara, P., additional, Trevisi, N., additional, Ricco, A., additional, Petracca, F., additional, Bisceglia, C., additional, Baratto, F., additional, Salguero Bodes, R., additional, Fontenla Cerezuela, A., additional, De Riva Silva, M., additional, Lopez Gil, M., additional, Mejia Martinez, E., additional, Jurado Roman, A., additional, Montero Alvarez, M., additional, Arribas Ynsaurriaga, F., additional, Baszko, A., additional, Krzyzanowski, K., additional, Bobkowski, W., additional, Surmacz, R., additional, Zinka, E., additional, Siwinska, A., additional, Szyszka, A., additional, Perez Silva, A., additional, Estrada Mucci, A., additional, Ortega Molina, M., additional, Lopez Sendon, J. L., additional, Merino Llorens, J. L., additional, Kaitani, K., additional, Hanazawa, K., additional, Izumi, C., additional, Nakagawa, Y., additional, Yamanaka, I., additional, Hirahara, T., additional, Sugawara, Y., additional, Suga, C., additional, Ako, J., additional, Momomura, S., additional, Galizio, N., additional, Gonzalez, J., additional, Robles, F., additional, Palazzo, A., additional, Favaloro, L., additional, Diez, M., additional, Guevara, E., additional, Fernandez, A., additional, Greenberg, S., additional, Epstein, A., additional, Goldman, D. S., additional, Sangli, C., additional, Keeney, J. A., additional, Lee, K., additional, Piers, S. R. D., additional, Van Rees, J. B., additional, Thijssen, J., additional, Borleffs, C. J. W., additional, Van Der Velde, E. T., additional, Leclercq, C. H., additional, Hero, M., additional, Mizobuchi, M., additional, Enjoji, Y., additional, Yazaki, Y., additional, Shibata, K., additional, Funatsu, A., additional, Kobayashi, T., additional, Nakamura, S., additional, Amit, G., additional, Pertzov, B., additional, Zahger, D., additional, Medesani, L., additional, Rana, R., additional, Albano, F., additional, Fraguas, H., additional, Pedersen, S. S., additional, Hoogwegt, M. T., additional, Theuns, D. A. M. J., additional, Van Den Broek, K. C., additional, Tekle, F. B., additional, Habibovic, M., additional, Alings, M., additional, Van Der Voort, P., additional, Denollet, J., additional, Vrazic, H., additional, Jilek, C., additional, Lesevic, H., additional, Tzeis, S., additional, Semmler, V., additional, Gold, M. R., additional, Burke, M. C., additional, Bardy, G. H., additional, Varma, N., additional, Pavri, B., additional, Stambler, B., additional, Michalski, J., additional, Investigators, T. R. U. S. T., additional, Safak, E., additional, Schmitz, D., additional, Konorza, T., additional, Wende, C., additional, Schirdewan, A., additional, Neuzner, J., additional, Simmers, T., additional, Erglis, A., additional, Gradaus, R., additional, Goetzke, J., additional, Coutrot, L., additional, Goehl, K., additional, Bazan Gelizo, V., additional, Grau, N., additional, Valles, E., additional, Felez, M., additional, Sanjuas, C., additional, Bruguera, J., additional, Marti-Almor, J., additional, Chu, S. Y., additional, Li, P. W., additional, Ding, W. H., additional, Schukro, C., additional, Leitner, L., additional, Siebermair, J., additional, Stix, G., additional, Pezawas, T., additional, Kastner, J., additional, Wolzt, M., additional, Schmidinger, H., additional, Behar, N. A. T. H. A. L. I. E., additional, Kervio, G., additional, Petit, B., additional, Maison-Balnche, P., additional, Bodi, S., additional, Mabo, P., additional, Foley, P. W. X., additional, Mutch, E., additional, Brashaw-Smith, J., additional, Ball, L., additional, Leyva, F., additional, Kim, D. H., additional, Lee, M. J., additional, Lee, W. S., additional, Park, S. D., additional, Shin, S. H., additional, Woo, S. I., additional, Kwan, J., additional, Park, K. S., additional, Munetsugu, Y., additional, Tanno, K., additional, Kikuchi, M., additional, Ito, H., additional, Miyoshi, F., additional, Kawamura, M., additional, Kobayashi, Y., additional, Man, S., additional, Algra, A. M., additional, Schreurs, C. A., additional, Van Der Wall, E. E., additional, Cannegieter, S. C., additional, Swenne, C. A., additional, Iitsuka, K., additional, Kondo, T., additional, Goebbert, K., additional, Karossiene, Z., additional, Goldman, D., additional, Kallen, B., additional, Kerpi, E., additional, Sardo, J., additional, Arsenos, P., additional, Gatzoulis, K., additional, Manis, G., additional, Dilaveris, P., additional, Tsiachris, D., additional, Mytas, D., additional, Asimakopoulos, S., additional, Stefanadis, C., additional, Sideris, S., additional, Kartsagoulis, E., additional, Barbosa, O., additional, Marocolo Junior, M., additional, Silva Cortes, R., additional, Moraes Brandolis, R. A., additional, Oliveira, L. F., additional, Pertili Rodrigues De Resende, L. A., additional, Vieira Da Silva, M. A., additional, Dias Da Silva, V. J., additional, Hegazy, R. A., additional, Sharaf, I. A., additional, Fadel, F., additional, Bazaraa, H., additional, Esam, R., additional, Deshko, M. S., additional, Snezhitsky, V. A., additional, Stempen, T. P., additional, Kuroki, K., additional, Igawa, M., additional, Kuga, K., additional, Ferreira Santos, L., additional, Dionisio, T., additional, Nunes, L., additional, Machado, J., additional, Castedo, S., additional, Henriques, C., additional, Matos, A., additional, Oliveira Santos, J., additional, Kraaier, K., additional, Olimulder, M. A. G. M., additional, Galjee, M. A., additional, Van Dessel, P. F. H. M., additional, Van Der Palen, J., additional, Wilde, A. A. M., additional, Scholten, M. F., additional, Chouchou, F., additional, Poupard, L., additional, Philippe, C., additional, Court-Fortune, I., additional, Barthelemy, J.- C., additional, Roche, F., additional, Dolgoshey, T. S., additional, Madekina, G. A., additional, Sugiura, S., additional, Fujii, E., additional, Senga, M., additional, Dohi, K., additional, Sugiura, E., additional, Nakamura, M., additional, Ito, M., additional, Eitel, C., additional, Mendell, J., additional, Lasseter, K., additional, Shi, M., additional, Urban, L., additional, Hatala, R., additional, Hlivak, P., additional, De Melis, M., additional, Garutti, C., additional, Corbucci, G., additional, Mlcochova, H., additional, Maxian, R., additional, Arbelo, E., additional, Dogac, A., additional, Luepkes, C., additional, Ploessnig, M., additional, Chronaki, C., additional, Hinterbuchner, L., additional, Guillen, A., additional, Bun, S. S., additional, Latcu, D. G., additional, Franceschi, F., additional, Prevot, S., additional, Koutbi, L., additional, Ricard, P., additional, Saoudi, N., additional, Nazari, N., additional, Alizadeh, A., additional, Sayah, S., additional, Hekmat, M., additional, Assadian, M., additional, Ahmadzadeh, A., additional, Wnuk, M., additional, Jedrzejczyk-Spaho, J., additional, Kruszelnicka, O., additional, Piwowarska, W., additional, Fedorowski, A., additional, Burri, P., additional, Juul-Moller, S., additional, Melander, O., additional, Mitro, P., additional, Murin, P., additional, Kirsch, P., additional, Habalova, V., additional, Slaba, E., additional, Matyasova, E., additional, Barlow, M. A., additional, Blake, R. J., additional, Rostoff, P., additional, Wojewodka Zak, E., additional, Froidevaux, L., additional, Sarasin, F. P., additional, Louis-Simonet, M., additional, Hugli, O., additional, Yersin, B., additional, Schlaepfer, J., additional, Mischler, C., additional, Pruvot, E., additional, Occhetta, E., additional, Frascarelli, F., additional, Burali, A., additional, and Dovellini, E., additional
- Published
- 2011
- Full Text
- View/download PDF
5. Long-term reliability of AAI mode pacing in patients with sinus node dysfunction and low Wenckebach block rate
- Author
-
Adachi, M., primary, Igawa, O., additional, Yano, A., additional, Miake, J., additional, Inoue, Y., additional, Ogura, K., additional, Kato, M., additional, Iitsuka, K., additional, and Hisatome, I., additional
- Published
- 2008
- Full Text
- View/download PDF
6. The small diverticulum in the right anterior wall of the left atrium
- Author
-
Igawa, O., primary, Miake, J., additional, and Adachi, M., additional
- Published
- 2007
- Full Text
- View/download PDF
7. Transcriptional activation of the anchoring protein SAP97 by heat shock factor (HSF)-1 stabilizes K(v) 1.5 channels in HL-1 cells.
- Author
-
Ting, YK, Morikawa, K, Kurata, Y, Li, P, Bahrudin, U, Mizuta, E, Kato, M, Miake, J, Yamamoto, Y, Yoshida, A, Murata, M, Inoue, T, Nakai, A, Shiota, G, Higaki, K, Nanba, E, Ninomiya, H, Shirayoshi, Y, Hisatome, I, and Ting, Y K
- Subjects
HEAT shock proteins ,MUSCLE cells ,GENE expression ,TRANSCRIPTION factors ,ACETONE ,SYNAPSES ,WESTERN immunoblotting ,POLYMERASE chain reaction ,SMALL interfering RNA - Abstract
Background and Purpose: The expression of voltage-dependent K(+) channels (K(v) ) 1.5 is regulated by members of the heat shock protein (Hsp) family. We examined whether the heat shock transcription factor 1 (HSF-1) and its inducer geranylgeranylacetone (GGA) could affect the expression of K(v) 1.5 channels and its anchoring protein, synapse associated protein 97 (SAP97).Experimental Approach: Transfected mouse atrial cardiomyocytes (HL-1 cells) and COS7 cells were subjected to luciferase reporter gene assay and whole-cell patch clamp. Protein and mRNA extracts were subjected to Western blot and quantitative real-time polymerase chain reaction.Key Results: Heat shock of HL-1 cells induced expression of Hsp70, HSF-1, SAP97 and K(v) 1.5 proteins. These effects were reproduced by wild-type HSF-1. Both heat shock and expression of HSF-1, but not the R71G mutant, increased the SAP97 mRNA level. Small interfering RNA (siRNA) against SAP97 abolished HSF-1-induced increase of K(v) 1.5 and SAP97 proteins. A luciferase reporter gene assay revealed that the SAP97 promoter region (from -919 to -740) that contains heat shock elements (HSEs) was required for this induction. Suppression of SIRT1 function either by nicotinamide or siRNA decreased the level of SAP97 mRNA. SIRT1 activation by resveratrol had opposing effects. A treatment of the cells with GGA increased the level of SAP97 mRNA, K(v) 1.5 proteins and I(Kur) current, which could be modified with either resveratrol or nicotinamide.Conclusions and Implications: HSF-1 induced transcription of SAP97 through SIRT1-dependent interaction with HSEs; the increase in SAP97 resulted in stabilization of K(v)1.5 channels. These effects were mimicked by GGA. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
8. QRS complex widening due to loss of left bundle branch capture: pitfall of para-Hisian pacing.
- Author
-
Adachi M, Igawa O, Miake J, Yano A, Inoue Y, Ogura K, Kato M, Iitsuka K, Hisatome I, Adachi, Masamitsu, Igawa, Osamu, Miake, Junichiro, Yano, Akio, Inoue, Yoshiaki, Ogura, Kazuyoshi, Kato, Masaru, Iitsuka, Kazuhiko, and Hisatome, Ichiro
- Abstract
During para-Hisian pacing, widening of the paced QRS complex usually indicates loss of His bundle capture. We describe a patient without any accessory pathways in whom widening of the paced QRS complex occurred due to loss of left bundle branch capture during para-Hisian pacing. After initial widening of the QRS complex, further widening was observed due to loss of His bundle capture. With the initial QRS widening, the stimulus-atrial interval and retrograde atrial activation sequence were almost unchanged, so the findings mimicked retrograde conduction over an accessory pathway. This may be a pitfall of the para-Hisian pacing technique. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
9. The small diverticulum in the right anterior wall of the left atrium.
- Author
-
Igawa O, Miake J, and Adachi M
- Published
- 2008
10. Mitochondrial Responses to Sublethal Doxorubicin in H9c2 Cardiomyocytes: The Role of Phosphorylated CaMKII.
- Author
-
Priyono AK, Miake J, Sawano T, Ichihara Y, Nagata K, Okamura A, Tomomori T, Takami A, Notsu T, Yamamoto K, and Imamura T
- Abstract
Background: Doxorubicin (Dox) is effective against different types of cancers, but it poses cardiotoxic side effects, frequently resulting in irreversible heart failure. However, the complexities surrounding this cardiotoxicity, especially at sublethal dosages, remain to be fully elucidated. We investigated early cellular disruptions in response to sublethal Dox, with a specific emphasis on the role of phosphorylated calcium/calmodulin-dependent protein kinase II (CaMKII) in initiating mitochondrial dysfunction., Methods: This study utilized the H9c2 cardiomyocyte model to identify a sublethal concentration of Dox and investigate its impact on mitochondrial health using markers such as mitochondrial membrane potential (MMP), mitophagy initiation, and mitochondrial calcium dynamics. We examined the roles of and interactions between CaMKII, dynamin-related protein 1 (Drp1), and the mitochondrial calcium uniporter (MCU) in Dox-induced mitochondrial disruption using specific inhibitors, such as KN-93, Mdivi-1, and Ru360, respectively., Results: Exposure to a sublethal dose of Dox reduced the MMP red-to-green fluorescence ratio in H9c2 cells by 40.6% compared with vehicle, and increased the proportion of cells undergoing mitophagy from negligible levels compared with vehicle to 62.2%. Mitochondrial calcium levels also increased by 8.7-fold compared with the vehicle group. Notably, the activation of CaMKII, particularly its phosphorylated form, was pivotal in driving these mitochondrial changes, as inhibition using KN-93 restored MMP and decreased mitophagy. However, inhibition of Drp1 and MCU functions had a limited impact on the observed mitochondrial disruptions., Conclusion: Sublethal administration of Dox is closely linked to CaMKII activation through phosphorylation, emphasizing its pivotal role in early mitochondrial disruption. These findings present a promising direction for developing therapeutic strategies that may alleviate the cardiotoxic effects of Dox, potentially increasing its clinical efficacy., Competing Interests: The authors declare no conflict of interest., (©2024 Tottori University Medical Press.)
- Published
- 2024
- Full Text
- View/download PDF
11. Inhibition of the uric acid efflux transporter ABCG2 enhances stimulating effect of soluble uric acid on IL-1β production in murine macrophage-like J774.1 cells.
- Author
-
Notsu T, Kurata Y, Ninomiya H, Taufiq F, Komatsu K, Miake J, Sawano T, Tsuneto M, Shirayoshi Y, and Hisatome I
- Subjects
- Mice, Animals, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Lipopolysaccharides pharmacology, Macrophages metabolism, Interleukin-1beta genetics, Interleukin-1beta metabolism, Interleukin-1beta pharmacology, RNA, Small Interfering pharmacology, RNA, Messenger pharmacology, Caspases pharmacology, Uric Acid pharmacology, Inflammasomes metabolism, Inflammasomes pharmacology
- Abstract
Soluble uric acid (UA) absorbed by cells through UA transporters (UATs) accumulates intracellularly, activates the NLRP3 inflammasome and thereby increases IL-1β secretion. ABCG2 transporter excludes intracellular UA. However, it remains unknown whether ABCG2 inhibition leads to intracellular accumulation of UA and increases IL-1β production. In this study, we examined whether genetic and pharmacological inhibition of ABCG2 could increase IL-1β production in mouse macrophage-like J774.1 cells especially under hyperuricemic conditions. We determined mRNA and protein levels of pro-IL-1β, mature IL-1β, caspase-1 and several UATs in culture supernatants and lysates of J774.1 cells with or without soluble UA pretreatment. Knockdown experiments using an shRNA against ABCG2 and pharmacological experiments with an ABCG2 inhibitor were conducted. Extracellularly applied soluble UA increased protein levels of pro-IL-1β, mature IL-1β and caspase-1 in the culture supernatant from lipopolysaccharide (LPS)-primed and monosodium urate crystal (MSU)-stimulated J774.1 cells. J774.1 cells expressed UATs of ABCG2, GLUT9 and MRP4, and shRNA knockdown of ABCG2 increased protein levels of pro-IL-1β and mature IL-1β in the culture supernatant. Soluble UA increased mRNA and protein levels of ABCG2 in J774.1 cells without either LPS or MSU treatment. An ABCG2 inhibitor, febuxostat, but not a urate reabsorption inhibitor, dotinurad, enhanced IL-1β production in cells pretreated with soluble UA. In conclusion, genetic and pharmacological inhibition of ABCG2 enhanced IL-1β production especially under hyperuricemic conditions by increasing intracellularly accumulated soluble UA that activates the NLRP3 inflammasome and pro-IL-1β transcription in macrophage-like J774.1 cells., (© 2023. The Author(s), under exclusive licence to The Japanese Society of Hypertension.)
- Published
- 2023
- Full Text
- View/download PDF
12. Epstein-Barr virus reactivation in peripheral B lymphocytes induces IgM-type thyrotropin receptor autoantibody production in patients with Graves' disease.
- Author
-
Nagata K, Hayashi K, Kumata K, Satoh Y, Osaki M, Nakayama Y, Kuwamoto S, Ichihara Y, Okura T, Matsuzawa K, Miake J, Fukata S, and Imamura T
- Subjects
- Animals, Swine, Humans, Herpesvirus 4, Human physiology, Long-Acting Thyroid Stimulator, Leukocytes, Mononuclear, Receptors, Thyrotropin, Immunoglobulin M, B-Lymphocytes, Thyrotropin, Autoantibodies, Immunoglobulins, Thyroid-Stimulating, Epstein-Barr Virus Infections, Graves Disease
- Abstract
Epstein-Barr virus (EBV) is a human herpes virus that latently infects B lymphocytes. When EBV is reactivated, host B cells differentiate into plasma cells and produce IgM-dominant antibodies as well as many progeny virions. The aims of the present study were to confirm the IgM dominance of thyrotropin-receptor antibodies (TRAbs) produced by EBV reactivation and investigate the roles of TRAb-IgM in Graves' disease. Peripheral blood mononuclear cells (PBMCs) containing TRAb-producing cells were stimulated for EBV reactivation, and TRAb-IgM and TRAb-IgG were measured by ELISA. TRAb-IgM were purified and TSH-binding inhibitory activities were assessed using a radio-receptor assay. Porcine thyroid follicular epithelial cells were cultured with TRAb-IgM and/or complements to measure the intracellular levels of cAMP and the amount of LDH released. TRAb-IgM/TRAb-IgG (the MG ratio) was examined in sequential serum samples of Graves' disease and compared among groups of thyroid function. The results obtained showed that IgM-dominant TRAb production was induced by EBV reactivation. TRAb-IgM did not inhibit TSH binding to TSH receptors and did not transduce hormone-producing signals. However, it destroyed thyroid follicular epithelial cells with complements. The MG ratio was significantly higher in samples of hyperthyroidism or hypothyroidism than in those with normal function or in healthy controls. A close relationship was observed between TRAb-IgM produced by EBV reactivation and the development and exacerbation of Graves' disease. The present results provide novel insights for the development of prophylaxis and therapeutics for Graves' disease.
- Published
- 2023
- Full Text
- View/download PDF
13. Impact of Hyper- and Hypo-Uricemia on Kidney Function.
- Author
-
Miake J, Hisatome I, Tomita K, Isoyama T, Sugihara S, Kuwabara M, Ogino K, and Ninomiya H
- Abstract
Uric acid (UA) forms monosodium urate (MSU) crystals to exert proinflammatory actions, thus causing gout arthritis, urolithiasis, kidney disease, and cardiovascular disease. UA is also one of the most potent antioxidants that suppresses oxidative stress. Hyper andhypouricemia are caused by genetic mutations or polymorphism. Hyperuricemia increases urinary UA concentration and is frequently associated with urolithiasis, which is augmented by low urinary pH. Renal hypouricemia (RHU) is associated with renal stones by increased level of urinary UA, which correlates with the impaired tubular reabsorption of UA. Hyperuricemia causes gout nephropathy, characterized by renal interstitium and tubular damage because MSU precipitates in the tubules. RHU is also frequently associated with tubular damage with elevated urinary beta2-microglobulin due to increased urinary UA concentration, which is related to impaired tubular UA reabsorption through URAT1. Hyperuricemia could induce renal arteriopathy and reduce renal blood flow, while increasing urinary albumin excretion, which is correlated with plasma xanthine oxidoreductase (XOR) activity. RHU is associated with exercise-induced kidney injury, since low levels of SUA could induce the vasoconstriction of the kidney and the enhanced urinary UA excretion could form intratubular precipitation. A U-shaped association of SUA with organ damage is observed in patients with kidney diseases related to impaired endothelial function. Under hyperuricemia, intracellular UA, MSU crystals, and XOR could reduce NO and activate several proinflammatory signals, impairing endothelial functions. Under hypouricemia, the genetic and pharmacological depletion of UA could impair the NO-dependent and independent endothelial functions, suggesting that RHU and secondary hypouricemia might be a risk factor for the loss of kidney functions. In order to protect kidney functions in hyperuricemic patients, the use of urate lowering agents could be recommended to target SUA below 6 mg/dL. In order to protect the kidney functions in RHU patients, hydration and urinary alkalization may be recommended, and in some cases an XOR inhibitor might be recommended in order to reduce oxidative stress.
- Published
- 2023
- Full Text
- View/download PDF
14. CORRIGENDUM: β-Adrenergic Blocker, Carvedilol, Abolishes Ameliorating Actions of Adipose-Derived Stem Cell Sheets on Cardiac Dysfunction and Remodeling After Myocardial Infarction.
- Author
-
Adachi M, Watanabe M, Kurata Y, Inoue Y, Notsu T, Yamamoto K, Horie H, Tanno S, Morita M, Miake J, Hamada T, Kuwabara M, Nakasone N, Ninomiya H, Tsuneto M, Shirayoshi Y, Yoshida A, Nishimura M, Yamamoto K, and Hisatome I
- Published
- 2023
- Full Text
- View/download PDF
15. [Gut microbiota and cardiovascular disease-insights into the potential and challenges of drug discovery].
- Author
-
Sawano T, Imamura T, and Miake J
- Subjects
- Humans, Ecosystem, Drug Discovery, Cardiovascular Diseases drug therapy, Cardiovascular Diseases etiology, Gastrointestinal Microbiome, Heart Failure
- Abstract
Cardiovascular disease is a major cause of death worldwide, with high prevalence and morbidity. Recent advances in technology have reported that abnormalities in the gut microbiota are associated with a variety of diseases, including cardiovascular diseases. The gut microbiota is a complex ecosystem that plays an important role in maintaining host health. It has been reported that the imbalance of gut microbiota causes changes in the production of substances derived from gut bacteria, such as short-chain fatty acids, trimethylamine-N-oxide, and lipopolysaccharide, and contributes to the development of cardiovascular diseases. In the drug discovery, it is a promising approach to prevention and therapy of the cardiovascular disease to focus on the relation between gut and heart, such as gut bacteria. However, there are challenges that must be overcome to convert this approach into effective therapy. In this review, we focus on cardiovascular diseases, particularly atherosclerotic disease, heart failure, and atrial fibrillation, and discuss the relationship between gut bacteria and substances derived from gut bacteria in cardiovascular disease. We also discuss the challenges and potential of drug discovery targeting the gut-heart relationship for the treatment and prevention of cardiovascular disease.
- Published
- 2023
- Full Text
- View/download PDF
16. JCS/JHRS 2020 Guideline on Pharmacotherapy of Cardiac Arrhythmias.
- Author
-
Ono K, Iwasaki YK, Akao M, Ikeda T, Ishii K, Inden Y, Kusano K, Kobayashi Y, Koretsune Y, Sasano T, Sumitomo N, Takahashi N, Niwano S, Hagiwara N, Hisatome I, Furukawa T, Honjo H, Maruyama T, Murakawa Y, Yasaka M, Watanabe E, Aiba T, Amino M, Itoh H, Ogawa H, Okumura Y, Aoki-Kamiya C, Kishihara J, Kodani E, Komatsu T, Sakamoto Y, Satomi K, Shiga T, Shinohara T, Suzuki A, Suzuki S, Sekiguchi Y, Nagase S, Hayami N, Harada M, Fujino T, Makiyama T, Maruyama M, Miake J, Muraji S, Murata H, Morita N, Yokoshiki H, Yoshioka K, Yodogawa K, Inoue H, Okumura K, Kimura T, Tsutsui H, and Shimizu W
- Published
- 2022
- Full Text
- View/download PDF
17. SHOX2 refines the identification of human sinoatrial nodal cell population in the in vitro cardiac differentiation.
- Author
-
Wakimizu T, Morikawa K, Fukumura K, Yuki T, Adachi T, Kurata Y, Miake J, Hisatome I, Tsuneto M, and Shirayoshi Y
- Abstract
Introduction: Dysfunction of the sinoatrial node (SAN) cells causes arrhythmias, and many patients require artificial cardiac pacemaker implantation. However, the mechanism of impaired SAN automaticity remains unknown, and the generation of human SAN cells in vitro may provide a platform for understanding the pathogenesis of SAN dysfunction. The short stature homeobox 2 ( SHOX2 ) and hyperpolarization-activated cyclic nucleotide-gated cation channel 4 ( HCN4 ) genes are specifically expressed in SAN cells and are important for SAN development and automaticity. In this study, we aimed to purify and characterize human SAN-like cells in vitro, using HCN4 and SHOX2 as SAN markers., Methods: We developed an HCN4-EGFP/SHOX2-mCherry dual reporter cell line derived from human induced pluripotent stem cells (hiPSCs), and HCN4 and SHOX2 gene expressions were visualized using the fluorescent proteins EGFP and mCherry, respectively. The dual reporter cell line was established using an HCN4-EGFP bacterial artificial chromosome-based semi-knock-in system and a CRISPR-Cas9-dependent knock-in system with a SHOX2-mCherry targeting vector. Flow cytometry, RT-PCR, and whole-cell patch-clamp analyses were performed to identify SAN-like cells., Results: Flow cytometry analysis and cell sorting isolated HCN4-EGFP single-positive (HCN4
+ /SHOX2- ) and HCN4-EGFP/SHOX2-mCherry double-positive (HCN4+ /SHOX2+ ) cells. RT-PCR analyses showed that SAN-related genes were enriched within the HCN4+ /SHOX2+ cells. Further, electrophysiological analyses showed that approximately 70% of the HCN4+ /SHOX2+ cells exhibited SAN-like electrophysiological characteristics, as defined by the action potential parameters of the maximum upstroke velocity and action potential duration., Conclusions: The HCN4-EGFP/SHOX2-mCherry dual reporter hiPSC system developed in this study enabled the enrichment of SAN-like cells within a mixed HCN4+ /SHOX2+ population of differentiating cardiac cells. This novel cell line is useful for the further enrichment of human SAN-like cells. It may contribute to regenerative medicine, for example, biological pacemakers, as well as testing for cardiotoxic and chronotropic actions of novel drug candidates., Competing Interests: None., (© 2022 The Japanese Society for Regenerative Medicine. Production and hosting by Elsevier B.V.)- Published
- 2022
- Full Text
- View/download PDF
18. Thrombin induces a temporal biphasic vascular response through the differential phosphorylation of endothelial nitric oxide synthase via protease-activated receptor-1 and protein kinase C.
- Author
-
Okamura A, Miake J, Tomomori T, Takami A, Sawano T, Kato M, Ogura K, Tsujimoto D, Kawatani S, Agung KP, Notsu T, Hisatome I, Yamamoto K, and Imamura T
- Subjects
- Animals, Human Umbilical Vein Endothelial Cells enzymology, Humans, Phosphorylation, Rats, Nitric Oxide Synthase Type III metabolism, Protein Kinase C metabolism, Receptor, PAR-1 metabolism, Thrombin metabolism, Thrombin pharmacology, Thrombin physiology, Vasodilation drug effects
- Abstract
Endothelial nitric oxide synthase (eNOS) is a critical regulatory enzyme that controls vascular tone via the production of nitric oxide. Although thrombin also modulates vascular tone predominantly via the activation of protease-activated receptors (PARs), the time course and mechanisms involved in how thrombin controls eNOS enzymatic activity are unknown. eNOS enzymatic activity is enhanced by the phosphorylation of eNOS-Ser1177 and reduced by the phosphorylation of eNOS-Thr495. In this study, we hypothesized that thrombin regulates vascular tone through the differential phosphorylation of eNOS. Using rat descending aorta, we show that thrombin modulates vascular tone in an eNOS-dependent manner via activated PAR-1. We also show that thrombin causes a temporal biphasic response. Protein kinase C (PKC) is associated with second phase of thrombin-induced response. Western blot analysis demonstrated thrombin phosphorylated eNOS-Ser1177 and eNOS-Thr495 in human umbilical vein endothelial cells. A PKC inhibitor suppressed the thrombin-induced phosphorylation of eNOS-Thr495, but not that of eNOS-Ser1177. Our results suggest that thrombin induces a temporal biphasic vascular response through the differential phosphorylation of eNOS via activated PAR-1. Thrombin causes transient vasorelaxation by the phosphorylation of eNOS-Ser1177, and subsequent attenuation of vasorelaxation by the phosphorylation of eNOS-Thr495 via PKC, leading to the modulation of vascular tone., Competing Interests: Declaration of competing interest The authors indicated no potential conflicts of interest., (Copyright © 2022 The Authors. Production and hosting by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. α1-Adrenergic receptor mediates adipose-derived stem cell sheet-induced protection against chronic heart failure after myocardial infarction in rats.
- Author
-
Horie H, Hisatome I, Kurata Y, Yamamoto Y, Notsu T, Adachi M, Li P, Kuwabara M, Sakaguchi T, Kinugasa Y, Miake J, Koba S, Tsuneto M, Shirayoshi Y, Ninomiya H, Ito S, Kitakaze M, Yamamoto K, Yoshikawa Y, and Nishimura M
- Subjects
- Animals, Human Umbilical Vein Endothelial Cells, Humans, Neovascularization, Physiologic, Rats, Rats, Inbred Lew, Stem Cells, Vascular Endothelial Growth Factor A, Heart Failure, Myocardial Infarction complications, Receptors, Adrenergic, alpha-1
- Abstract
Cell-based therapy using adipose-derived stem cells (ADSCs) has emerged as a novel therapeutic approach to treat heart failure after myocardial infarction (MI). The purpose of this study was to determine whether inhibition of α1-adrenergic receptors (α1-ARs) in ADSCs attenuates ADSC sheet-induced improvements in cardiac functions and inhibition of remodeling after MI. ADSCs were isolated from fat tissues of Lewis rats. In in vitro studies using cultured ADSCs, we determined the mRNA levels of vascular endothelial growth factor (VEGF)-A and α1-AR under normoxia or hypoxia and the effects of norepinephrine and an α1-blocker, doxazosin, on the mRNA levels of angiogenic factors. Hypoxia increased α1-AR and VEGF mRNA levels in ADSCs. Norepinephrine further increased VEGF mRNA expression under hypoxia; this effect was abolished by doxazosin. Tube formation of human umbilical vein endothelial cells was promoted by conditioned media of ADSCs treated with the α1 stimulant phenylephrine under hypoxia but not by those of ADSCs pretreated with phenylephrine plus doxazosin. In in vivo studies using rats with MI, transplanted ADSC sheets improved cardiac functions, facilitated neovascularization, and suppressed fibrosis after MI. These effects were abolished by doxazosin treatment. Pathway analysis from RNA sequencing data predicted significant upregulation of α1-AR mRNA expression in transplanted ADSC sheets and the involvement of α1-ARs in angiogenesis through VEGF. In conclusion, doxazosin abolished the beneficial effects of ADSC sheets on rat MI hearts as well as the enhancing effect of norepinephrine on VEGF expression in ADSCs, indicating that ADSC sheets promote angiogenesis and prevent cardiac dysfunction and remodeling after MI via their α1-ARs., (© 2021. The Author(s), under exclusive licence to The Japanese Society of Hypertension.)
- Published
- 2022
- Full Text
- View/download PDF
20. Esm1 and Stc1 as Angiogenic Factors Responsible for Protective Actions of Adipose-Derived Stem Cell Sheets on Chronic Heart Failure After Rat Myocardial Infarction.
- Author
-
Watanabe M, Horie H, Kurata Y, Inoue Y, Notsu T, Wakimizu T, Adachi M, Yamamoto K, Morikawa K, Kuwabara M, Sakaguchi T, Morisaki T, Miake J, Nishimura M, Tsuneto M, Shirayoshi Y, Ito S, Kitakaze M, Ninomiya H, Yamamoto K, and Hisatome I
- Subjects
- Angiogenesis Inducing Agents, Animals, Human Umbilical Vein Endothelial Cells, Humans, Rats, Stem Cell Transplantation, Adipose Tissue, Heart Failure therapy, Myocardial Infarction therapy
- Abstract
Background: Although adipose-derived stem cell (ADSC) sheets improve the cardiac function after myocardial infarction (MI), underlying mechanisms remain to be elucidated. The aim of this study was to determine the fate of transplanted ADSC sheets and candidate angiogenic factors released from ADSCs for their cardiac protective actions., Methods and results: MI was induced by ligation of the left anterior descending coronary artery. Sheets of transgenic (Tg)-ADSCs expressing green fluorescence protein (GFP) and luciferase or wild-type (WT)-ADSCs were transplanted 1 week after MI. Both WT- and Tg-ADSC sheets improved cardiac functions evaluated by echocardiography at 3 and 5 weeks after MI. Histological examination at 5 weeks after MI demonstrated that either sheet suppressed fibrosis and increased vasculogenesis. Luciferase signals from Tg-ADSC sheets were detected at 1 and 2 weeks, but not at 4 weeks, after transplantation. RNA sequencing of PKH (yellow-orange fluorescent dye with long aliphatic tails)-labeled Tg-ADSCs identified mRNAs of 4 molecules related to angiogenesis, including those of Esm1 and Stc1 that increased under hypoxia. Administration of Esm1 or Stc1 promoted tube formation by human umbilical vein endothelial cells., Conclusions: ADSC sheets improved cardiac contractile functions after MI by suppressing cardiac fibrosis and enhancing neovascularization. Transplanted ADSCs existed for >2 weeks on MI hearts and produced the angiogenic factors Esm1 and Stc1, which may improve cardiac functions after MI.
- Published
- 2021
- Full Text
- View/download PDF
21. Uric Acid as a Risk Factor for Chronic Kidney Disease and Cardiovascular Disease - Japanese Guideline on the Management of Asymptomatic Hyperuricemia.
- Author
-
Hisatome I, Li P, Miake J, Taufiq F, Mahati E, Maharani N, Utami SB, Kuwabara M, Bahrudin U, and Ninomiya H
- Subjects
- Allopurinol therapeutic use, Endothelial Cells, Febuxostat therapeutic use, Gout Suppressants therapeutic use, Humans, Japan, Practice Guidelines as Topic, Risk Factors, Cardiovascular Diseases complications, Cardiovascular Diseases drug therapy, Gout drug therapy, Hyperuricemia drug therapy, Renal Insufficiency, Chronic complications, Renal Insufficiency, Chronic drug therapy, Uric Acid blood
- Abstract
Serum uric acid (UA) is taken up by endothelial cells and reduces the level of nitric oxide (NO) by inhibiting its production and accelerating its degradation. Cytosolic and plasma xanthine oxidase (XO) generates superoxide and also decreases the NO level. Thus, hyperuricemia is associated with impaired endothelial function. Hyperuricemia is often associated with vascular diseases such as chronic kidney disease (CKD) and cardiovascular disease (CVD). It has long been debated whether hyperuricemia is causally related to the development of these diseases. The 2020 American College of Rheumatology Guideline for the Management of Gout (ACR2020) does not recommend pharmacological treatment of hyperuricemia in patients with CKD/CVD. In contrast, the Japanese Guideline on Management of Hyperuricemia and Gout (JGMHG), 3
rd edition, recommends pharmacological treatment of hyperuricemia in patients with CKD. In a FREED study on Japanese hyperuricemic patients with CVD, an XO inhibitor, febuxostat, improved the primary composite endpoint of cerebro-cardio-renovascular events, providing a rationale for the use of urate-lowering agents (ULAs). Since a CARES study on American gout patients with CVD treated with febuxostat revealed increased mortality, ACR2020 recommends switching to different ULAs. However, there was no difference in the mortality of Japanese patients between the febuxostat-treated group and the placebo or allopurinol-treated groups in either the FEATHER or FREED studies.- Published
- 2021
- Full Text
- View/download PDF
22. MicroRNA-494-3p inhibits formation of fast oxidative muscle fibres by targeting E1A-binding protein p300 in human-induced pluripotent stem cells.
- Author
-
Iwasaki H, Ichihara Y, Morino K, Lemecha M, Sugawara L, Sawano T, Miake J, Sakurai H, Nishi E, Maegawa H, and Imamura T
- Subjects
- 3' Untranslated Regions genetics, Animals, Cell Differentiation genetics, Cell Line, Cell Proliferation genetics, Down-Regulation genetics, Humans, Male, Mice, Mice, Inbred C57BL, Muscle Development genetics, MyoD Protein genetics, Myoblasts metabolism, E1A-Associated p300 Protein metabolism, Induced Pluripotent Stem Cells metabolism, MicroRNAs metabolism, Muscle Fibers, Skeletal metabolism, Muscle, Skeletal metabolism, Oxidative Stress genetics
- Abstract
MYOD-induced microRNA-494-3p expression inhibits fast oxidative myotube formation by downregulating myosin heavy chain 2 (MYH2) in human induced pluripotent stem cells (hiPSCs) during skeletal myogenesis. However, the molecular mechanisms regulating MYH2 expression via miR-494-3p remain unknown. Here, using bioinformatic analyses, we show that miR-494-3p potentially targets the transcript of the E1A-binding protein p300 at its 3'-untranslated region (UTR). Myogenesis in hiPSCs with the Tet/ON-myogenic differentiation 1 (MYOD1) gene (MyoD-hiPSCs) was induced by culturing them in doxycycline-supplemented differentiation medium for 7 days. p300 protein expression decreased after transient induction of miR-494-3p during myogenesis. miR-494-3p mimics decreased the levels of p300 and its downstream targets MYOD and MYH2 and myotube formation efficiency. p300 knockdown decreased myotube formation efficiency, MYH2 expression, and basal oxygen consumption rate. The binding of miR-494-3p to the wild type p300 3'-UTR, but not the mutated site, was confirmed using luciferase assay. Overexpression of p300 rescued the miR-494-3p mimic-induced phenotype in MyoD-hiPSCs. Moreover, miR-494-3p mimic reduced the levels of p300, MYOD, and MYH2 in skeletal muscles in mice. Thus, miR-494-3p might modulate MYH2 expression and fast oxidative myotube formation by directly regulating p300 levels during skeletal myogenesis in MyoD-hiPSCs and murine skeletal muscle tissues.
- Published
- 2021
- Full Text
- View/download PDF
23. A Novel Treatment for Arrhythmias via the Control of the Degradation of Ion Channel Proteins.
- Author
-
Miake J
- Abstract
Although there are many reports on the regulation of ion channel expression in transcription and translation, few drugs have been studied to influence post-translational modification of ion channel proteins. The Kv1.5 channel is a potassium ion channel expressed in atrial muscle, belongs to the voltage-gated K
+ channel superfamily, and forms an ultrarapid delayed rectifier potassium ion current. It is important to understand the fate of these channel proteins, as cardiac Kv1.5 mutations can cause arrhythmias. Disruption of quantitative and qualitative control mechanisms of channels leads to stagnation and degradation of intracellular channel proteins. As a result, ion channel proteins are not transported to the cell membrane and are involved in the development of atrial fibrillation. This review takes the Kv1.5 channel as an example and focuses on the degradation mechanism of ion channel proteins, and discusses its application to the treatment of arrhythmia by drugs that control the mechanism of ion channel protein degradation., Competing Interests: The author declares no conflict of interest., (©2020 Tottori University Medical Press.)- Published
- 2020
- Full Text
- View/download PDF
24. Evidence for Urate Uptake Through Monocarboxylate Transporter 9 Expressed in Mammalian Cells and Its Enhancement by Heat Shock.
- Author
-
Otani N, Kurata Y, Maharani N, Kuwabara M, Ikeda N, Notsu T, Li P, Miake J, Yoshida A, Sakaguchi H, Higaki K, Nakasone N, Tsuneto M, Shirayoshi Y, Ouchi M, Ninomiya H, Yamamoto K, Anzai N, and Hisatome I
- Abstract
Background: Monocarboxylate transporter 9 (MCT9), an orphan transporter member of the solute carrier family 16 (SLC16), possibly reabsorbs uric acid in the renal tubule and has been suggested by genome-wide association studies to be involved in the development of hyperuricemia and gout. In this study we investigated the mechanisms regulating the expression of human (h) MCT9, its degradation, and physiological functions. Methods and Results: hMCT9-FLAG was stably expressed in HEK293 cells and its degradation, intracellular localization, and urate uptake activities were assessed by pulse-chase analysis, immunofluorescence, and [
14 C]-urate uptake experiments, respectively. hMCT9-FLAG was localized on the plasma membrane as well as in the endoplasmic reticulum and Golgi apparatus. The proteasome inhibitors MG132 and lactacystine increased levels of hMCT9-FLAG protein expression with enhanced ubiquitination, prolonged their half-life, and decreased [14 C]-urate uptake. [14 C]-urate uptake was increased by both heat shock (HS) and the HS protein inducer geranylgeranylacetone (GGA). Both HS and GGA restored the [14 C]-urate uptake impaired by MG132. Conclusions: hMCT9 does transport urate and is degraded by a proteasome, inhibition of which reduces hMCT9 expression on the cell membrane and urate uptake. HS enhanced urate uptake through hMCT9., Competing Interests: I.H. reports receiving lecturer fees from Mochida Pharmaceutical Company, Sanwa Kagaku Kenkyusho Co. Ltd., Pfizer Co. Ltd., Teijin Pharma Co. Ltd., and Fuji Yakuhin Co. Ltd., and research grants from Mochida Pharmaceutical Company, Teijin Pharma Co. Ltd., Fuji Yakuhin Co. Ltd., and Sanwa Kagaku Kenkyusho Co. Ltd. I.H. is a member of Circulation Reports ’ Editorial Team. The other authors report no conflicts of interest., (Copyright © 2020, THE JAPANESE CIRCULATION SOCIETY.)- Published
- 2020
- Full Text
- View/download PDF
25. Pretreatment with cilnidipine attenuates hypoxia/reoxygenation injury in HL-1 cardiomyocytes through enhanced NO production and action potential shortening.
- Author
-
Minato H, Hisatome I, Kurata Y, Notsu T, Nakasone N, Ninomiya H, Hamada T, Tomomori T, Okamura A, Miake J, Tsuneto M, Shirayoshi Y, Endo R, Otsuki A, Okada F, and Inagaki Y
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Cell Survival drug effects, Gene Knockdown Techniques, Mice, Myocytes, Cardiac metabolism, Nitric Oxide Synthase Type III genetics, Nitric Oxide Synthase Type III metabolism, Phosphorylation drug effects, RNA, Small Interfering, Rats, Action Potentials drug effects, Calcium Channel Blockers pharmacology, Dihydropyridines pharmacology, Hypoxia metabolism, Myocytes, Cardiac drug effects, Nitric Oxide metabolism
- Abstract
Myocardial ischemia/reperfusion injury worsens in the absence of nitric oxide synthase (NOS). Cilnidipine, a Ca
2+ channel blocker, has been reported to activate endothelial NOS (eNOS) and increases nitric oxide (NO) in vascular endothelial cells. We examined whether pretreatment with cilnidipine could attenuate cardiac cell deaths including apoptosis caused by hypoxia/reoxygenation (H/R) injury. HL-1 mouse atrial myocytes as well as H9c2 rat ventricular cells were exposed to H/R, and cell viability was evaluated by an autoanalyzer and flow cytometry; eNOS expression, NO production, and electrophysiological properties were also evaluated by western blotting, colorimetry, and patch clamping, respectively, in the absence and presence of cilnidipine. Cilnidipine enhanced phosphorylation of eNOS and NO production in a concentration-dependent manner, which was abolished by siRNAs against eNOS or an Hsp90 inhibitor, geldanamycin. Pretreatment with cilnidipine attenuated cell deaths including apoptosis during H/R; this effect was reproduced by an NO donor and a xanthine oxidase inhibitor. The NOS inhibitor L-NAME abolished the protective action of cilnidipine. Pretreatment with cilnidipine also attenuated H9c2 cell death during H/R. Additional cilnidipine treatment during H/R did not significantly enhance its protective action. There was no significant difference in the protective effect of cilnidipine under normal and high Ca2+ conditions. Action potential duration (APD) of HL-1 cells was shortened by cilnidipine, with this shortening augmented after H/R. L-NAME attenuated the APD shortening caused by cilnidipine. These findings indicate that cilnidipine enhances NO production, shortens APD in part by L-type Ca2+ channel block, and thereby prevents HL-1 cell deaths during H/R.- Published
- 2020
- Full Text
- View/download PDF
26. Different Effects of Pulmonary Vein Isolation on Quality of Life Between Patients with Persistent and Paroxysmal Atrial Fibrillation.
- Author
-
Kato M, Miake J, Ogura K, Iitsuka K, Okamura A, Tomomori T, Tsujimoto D, Kato M, and Yamamoto K
- Subjects
- Aged, Atrial Fibrillation physiopathology, Female, Follow-Up Studies, Heart Rate, Humans, Male, Middle Aged, Natriuretic Peptide, Brain blood, Recurrence, Regression Analysis, Treatment Outcome, Atrial Fibrillation psychology, Atrial Fibrillation therapy, Catheter Ablation, Pulmonary Veins, Quality of Life
- Abstract
The effect of restoring sinus rhythm by pulmonary vein isolation (PVI) on the quality of life (QOL) of patients with persistent atrial fibrillation (PerAF) has not been adequately investigated. This study was performed to compare the changes in QOL after extended PVI between patients with PerAF and paroxysmal AF (PAF).Patients with PAF (n = 38) and PerAF (n = 22) who underwent their first PVI and developed no AF recurrence 6 months after PVI were enrolled. QOL surveys were performed at baseline and 6 months post-ablation using Short Form-36 surveys.The mental component summary score (MCS) (53.4 ± 10.2 to 56.5 ± 7.1, P = 0.019) and physical component summary score (PCS) (46.1 ± 10.6 to 48.5 ± 8.3, P = 0.015) improved after PVI in the PAF group. The PCS, but not the MCS, improved after PVI in the PerAF group (45.8 ± 7.9 to 51.5 ± 6.2, P < 0.001). Changes in the PCS were greater in the PerAF group than in the PAF group (8.6 ± 6.9 versus 2.8 ± 5.2, P = 0.009). Multivariate regression analysis demonstrated that a low baseline MCS and the type of AF (PAF) were independent predictors of an increased MCS and that a low baseline PCS and the type of AF (PerAF) were independent predictors of an increased PCS.The changes in QOL differed between PAF and PerAF after PVI. Although most patients with PerAF were asymptomatic before PVI, their improvement in physical QOL was greater than that in patients with PAF. Such beneficial effects on physical QOL are likely expected in patients with PerAF with a low PCS before PVI.
- Published
- 2019
- Full Text
- View/download PDF
27. Hyperuricemia as a Risk Factor for Atrial Fibrillation Due to Soluble and Crystalized Uric Acid.
- Author
-
Taufiq F, Li P, Miake J, and Hisatome I
- Abstract
Among the several independent risk factors for atrial fibrillation (AF), hyperuricemia has been widely accepted as associated with the incidence of paroxysmal or persistent AF, as well as with the risk of AF in patients undergoing cardiovascular surgery. The electrophysiological mechanism of AF involves electrical remodeling of the arrhythmogenic substrate and abnormal automaticity as trigger. Both electrical and structural remodeling mediated by oxidative stress derived from either xanthine oxidoreductase (XOR), soluble uric acid (UA) or monosodium urate (MSU) crystals might be plausible explanations for the association of AF with hyperuricemia. XOR generates reactive oxygen species (ROS) that lead to atrial structural remodeling via inflammation. Soluble UA accumulates intracellularly through UA transporters (UAT), shortening the atrial action potential via enhanced expression and activity of Kv1.5 channel proteins. Intracellular accumulation of soluble UA generates ROS in atrial myocytes via nicotinamide adenine dinucleotide phosphate oxidase, which phosphorylates ERK/Akt and heat shock factor 1 (HSF1), thereby increasing transcription and translation of Hsp70, which stabilizes Kv1.5. In macrophages, MSU activates the NLRP3 inflammasome and proteolytic processing mediated by caspase-1 with enhanced interleukin (IL)-1β and IL-18 secretion. Use of an XOR inhibitor, antioxidants, a UAT inhibitor such as a uricosuric agent, and an NLRP3 inflammasome inhibitor, might become a potential strategy to reduce the risk of hyperuricemia-induced AF, and control serum UA level., Competing Interests: I.H. has received lecture fees from Teijin Pharma Co. Ltd., Sanwa Kagaku Kenkyusho Co. Ltd., Feizer Co. Ltd., and Fuji Yakuhin Co. Ltd.; and research grants from Daiichi Sankyo Co. Ltd., Bayer Yakuhin, Ltd., Teijin Pharma Co. Ltd., Fuji Yakuhin Co. Ltd. and Sanwa Kagaku Kenkyusho Co. Ltd. I.H. is also a member of Circulation Reports’ Editorial Team. The other authors declare no conflicts of interest., (Copyright © 2019, THE JAPANESE CIRCULATION SOCIETY.)
- Published
- 2019
- Full Text
- View/download PDF
28. β-Adrenergic Blocker, Carvedilol, Abolishes Ameliorating Actions of Adipose-Derived Stem Cell Sheets on Cardiac Dysfunction and Remodeling After Myocardial Infarction.
- Author
-
Adachi M, Watanabe M, Kurata Y, Inoue Y, Notsu T, Yamamoto K, Horie H, Tanno S, Morita M, Miake J, Hamada T, Kuwabara M, Nakasone N, Ninomiya H, Tsuneto M, Shirayoshi Y, Yoshida A, Nishimura M, Yamamoto K, and Hisatome I
- Subjects
- Animals, Cell Hypoxia, Cells, Cultured, Disease Models, Animal, Fibrosis, Human Umbilical Vein Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells metabolism, Humans, Male, Mesenchymal Stem Cells metabolism, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Neovascularization, Physiologic drug effects, Phosphorylation, Rats, Inbred Lew, Receptors, Vascular Endothelial Growth Factor metabolism, Recovery of Function, Vascular Endothelial Growth Factor A metabolism, Ventricular Remodeling drug effects, Adrenergic beta-Antagonists pharmacology, Carvedilol pharmacology, Mesenchymal Stem Cell Transplantation, Mesenchymal Stem Cells drug effects, Myocardial Contraction drug effects, Myocardial Infarction surgery, Subcutaneous Fat cytology, Ventricular Function, Left drug effects
- Abstract
Background: Treatment of myocardial infarction (MI) includes inhibition of the sympathetic nervous system (SNS). Cell-based therapy using adipose-derived stem cells (ASCs) has emerged as a novel therapeutic approach to treat heart failure in MI. The purpose of this study was to determine whether a combination of ASC transplantation and SNS inhibition synergistically improves cardiac functions after MI.Methods and Results:ASCs were isolated from fat tissues of Lewis rats. In in vitro studies using cultured ASC cells, mRNA levels of angiogenic factors under normoxia or hypoxia, and the effects of norepinephrine and a β-blocker, carvedilol, on the mRNA levels were determined. Hypoxia increased vascular endothelial growth factor (VEGF) mRNA in ASCs. Norepinephrine further increased VEGF mRNA; this effect was unaffected by carvedilol. VEGF promoted VEGF receptor phosphorylation and tube formation of human umbilical vein endothelial cells, which were inhibited by carvedilol. In in vivo studies using a rat MI model, transplanted ASC sheets improved contractile functions of MI hearts; they also facilitated neovascularization and suppressed fibrosis after MI. These beneficial effects of ASC sheets were abolished by carvedilol. The effects of ASC sheets and carvedilol on MI heart functions were confirmed by Langendorff perfusion experiments using isolated hearts., Conclusions: ASC sheets prevented cardiac dysfunctions and remodeling after MI in a rat model via VEGF secretion. Inhibition of VEGF effects by carvedilol abolished their beneficial effects.
- Published
- 2019
- Full Text
- View/download PDF
29. Uric Acid-Induced Enhancements of Kv1.5 Protein Expression and Channel Activity via the Akt-HSF1-Hsp70 Pathway in HL-1 Atrial Myocytes.
- Author
-
Taufiq F, Maharani N, Li P, Kurata Y, Ikeda N, Kuwabara M, Otani N, Miake J, Hasegawa A, Tsuneto M, Shirayoshi Y, Ninomiya H, Saitoh T, Nakai A, Yamamoto K, and Hisatome I
- Subjects
- Animals, Cell Line, Kv1.5 Potassium Channel drug effects, Mice, Phosphorylation drug effects, Protein Biosynthesis, Transcription, Genetic, HSP70 Heat-Shock Proteins metabolism, Heat Shock Transcription Factors metabolism, Kv1.5 Potassium Channel metabolism, Myocytes, Cardiac metabolism, Proto-Oncogene Proteins c-akt metabolism, Uric Acid pharmacology
- Abstract
Background: Intracellular uric acid is known to increase the protein level and channel current of atrial Kv1.5; however, mechanisms of the uric acid-induced enhancement of Kv1.5 expression remain unclear. Methods and Results: The effects of uric acid on mRNA and protein levels of Kv1.5, as well as those of Akt, HSF1 and Hsp70, in HL-1 cardiomyocytes were studied by using qRT-PCR and Western blotting. The uptake of uric acid was measured using radio-labeled uric acid. The Kv1.5-mediated channel current was also measured by using patch clamp techniques. Uric acid up-taken by HL-1 cells significantly increased the level of Kv1.5 proteins in a concentration-dependent manner, with this increase abolished by an uric acid transporter inhibitor. Uric acid slowed degradation of Kv1.5 proteins without altering its mRNA level. Uric acid enhanced phosphorylation of Akt and HSF1, and thereby increased both transcription and translation of Hsp70; these effects were abolished by a PI3K inhibitor. Hsp70 knockdown abolished the uric acid-induced increases of Kv1.5 proteins and channel currents., Conclusions: Intracellular uric acid could stabilize Kv1.5 proteins through phosphorylation of Akt and HSF1 leading to enhanced expression of Hsp70.
- Published
- 2019
- Full Text
- View/download PDF
30. Pre-ablation levels of brain natriuretic peptide are independently associated with the recurrence of atrial fibrillation after radiofrequency catheter ablation in patients with nonvalvular atrial fibrillation.
- Author
-
Miake J, Kato M, Ogura K, Iitsuka K, Okamura A, Tomomori T, Tsujimoto D, Kato M, and Yamamoto K
- Subjects
- Aged, Atrial Fibrillation blood, Atrial Fibrillation physiopathology, Biomarkers blood, Endothelium, Vascular physiopathology, Enzyme-Linked Immunosorbent Assay, Female, Follow-Up Studies, Humans, Male, Middle Aged, Predictive Value of Tests, Preoperative Period, Prognosis, Recurrence, Retrospective Studies, Risk Factors, Vasodilation, Atrial Fibrillation surgery, Catheter Ablation methods, Natriuretic Peptide, Brain blood
- Abstract
Association between pre-ablation levels of biomarkers of cardiac and endothelial dysfunctions, CHADS2, CHA2DS2-VASc, and APPLE scores and the recurrence of atrial fibrillation (AF) after radiofrequency catheter ablation has not been fully studied. A total of 254 patients with nonvalvular AF were prospectively followed for AF recurrence after a single ablation procedure. During a two-year follow-up period, AF recurred in 65 (25.6%) patients. Patients with AF recurrence had significantly greater baseline ln brain natriuretic peptide (BNP) than those without AF recurrence (P < 0.01), whereas there were no significant differences in the levels of biomarkers of endothelial dysfunction and points of scoring systems. In the Cox regression analyses, the baseline ln BNP was significantly independently associated with AF recurrence (adjusted HR =1.286, 95% CI =1.000-1.655, P < 0.05). The baseline levels of ln BNP were significantly associated with rhythm at blood collection, age, sex, and left atrial diameter, and left ventricular ejection fraction (P < 0.05).The subgroup analysis showed a significant interaction on the risk of AF recurrence between ln BNP, sex difference, and rhythm at blood collection (P for interaction < 0.05). In conclusion, the results suggest that the pre-ablation levels of ln BNP are useful to evaluate the risk of AF recurrence after ablation therapy; however, there is a need to be careful while using BNP as a biomarker for the risk of AF recurrence by taking account of the effects of rhythm status at blood collection and sex difference.
- Published
- 2019
- Full Text
- View/download PDF
31. Inhibitory effects of class I antiarrhythmic agents on Na + and Ca 2+ currents of human iPS cell-derived cardiomyocytes.
- Author
-
Yonemizu S, Masuda K, Kurata Y, Notsu T, Higashi Y, Fukumura K, Li P, Ninomiya H, Miake J, Tsuneto M, Shirayoshi Y, and Hisatome I
- Abstract
Introduction: Human induced pluripotent stem cells (hiPSCs) harboring cardiac myosin heavy chain 6 promoter can differentiate into functional cardiomyocytes called "iCell cardiomyocytes" under blasticidin treatment condition. While iCell cardiomyocytes are expected to be used for predicting cardiotoxicity of drugs, their responses to antiarrhythmic agents remain to be elucidated. We first examined electrophysiological properties of iCell cardiomyocytes and mRNA levels of ion channels and Ca handling proteins, and then evaluated effects of class I antiarrhythmic agents on their Na
+ and Ca2+ currents., Methods: iCell cardiomyocytes were cultured for 8-14 days (38-44 days after inducing their differentiation), according to the manufacturer's protocol. We determined their action potentials (APs) and sarcolemmal ionic currents using whole-cell patch clamp techniques, and also mRNA levels of ion channels and Ca handling proteins by RT-PCR. Effects of three class I antiarrhythmic agents, pirmenol, pilsicainide and mexiletine, on Na+ channel current (INa ) and L-type Ca2+ channel current (ICaL ) were evaluated by the whole-cell patch clamp., Results: iCell cardiomyocytes revealed sinoatrial node-type (18%), atrial-type (18%) and ventricular-type (64%) spontaneous APs. The maximum peak amplitudes of INa , ICaL , and rapidly-activating delayed-rectifier K+ channel current were -62.7 ± 13.7, -8.1 ± 0.7, and 3.0 ± 1.0 pA/pF, respectively. The hyperpolarization-activated cation channel and inward-rectifier K+ channel currents were observed, whereas the T-type Ca2+ channel or slowly-activating delayed-rectifier K+ channel current was not detectable. mRNAs of Nav1.5, Cav1.2, Kir2.1, HCN4, KvLQT1, hERG and SERCA2 were detected, while that of HCN1, minK or MiRP was not. The class Ia antiarrhythmic agent pirmenol and class Ic agent pilsicainide blocked INa in a concentration-dependent manner with IC50 of 0.87 ± 0.37 and 0.88 ± 0.16 μM, respectively; the class Ib agent mexiletine revealed weak INa block with a higher IC50 of 30.0 ± 3.0 μM. Pirmenol, pilsicainide and mexiletine blocked ICaL with IC50 of 2.00 ± 0.39, 7.7 ± 2.5 and 5.0 ± 0.1 μM, respectively., Conclusions: In iCell cardiomyocytes, INa was blocked by the class Ia and Ic antiarrhythmic agents and ICaL was blocked by all the class I agents within the ranges of clinical concentrations, suggesting their cardiotoxicity.- Published
- 2019
- Full Text
- View/download PDF
32. Establishment of a Novel In Situ Rat Model for Direct Measuring of Intestinal Drug Absorption: Confirmation of Inhibitory Effects of Daijokito on the Absorption of Ranitidine.
- Author
-
Qian W, Hasegawa J, Yang J, Endo Y, and Miake J
- Abstract
Background: Daijokito (DJKT), a classical traditional Kampo and Chinese medicine, has been used to treat acute pancreatitis in China. In our previous study, DJKT was found to reduce the area under the plasma concentration-time curve (AUC) of ranitidine in humans. Therefore, we established a novel rat model to examine the direct absorption of ranitidine after daijokito administration., Methods: An in situ intestinal injection with portal vein sampling (IIPS) model was created to determine the rate of intestinal drug absorption. Rats were divided into two groups: the ranitidine group (R, n = 6) or the ranitidine and daijokito group (RD, n = 6). Blood was collected after intestinal injection of drugs. After the experiment, the concentrations of ranitidine were measured by LC/MS/MS analysis., Results: The concentrations of ranitidine increased linearly with time in both groups. Compared with the R group, the concentrations of ranitidine in RD group significantly decreased throughout the experiment., Conclusion: Co-administration of ranitidine with DJKT resulted in significant decreases in intestinal absorption in rats. The reduction of the systemic ranitidine concentration by co-administration of DJKT may be due, at least in part, to the inhibition of intestinal absorption of ranitidine.
- Published
- 2018
33. Pretreatment with an angiotensin II receptor blocker abolished ameliorating actions of adipose-derived stem cell sheets on cardiac dysfunction and remodeling after myocardial infarction.
- Author
-
Yamamoto K, Kurata Y, Inoue Y, Adachi M, Tsuneto M, Miake J, Ogino K, Ninomiya H, Yoshida A, Shirayoshi Y, Suyama Y, Yagi S, Nishimura M, Yamamoto K, and Hisatome I
- Abstract
Introduction: Cell sheets using myoblasts have been developed for the treatment of heart failure after myocardial infarction (MI) bridging to heart transplantation. Stem cells are supposed to be better than myoblasts as a source of cells, since they possess a potential to proliferate and differentiate into cardiomyocytes, and also have capacity to secrete angiogenic factors. Adipose-derived stem cells (ASCs) obtained from fat tissues are expected to be a new cell source for ASC sheet therapies. Administration of angiotensin II receptor blockers (ARBs) is a standard therapy for heart failure after MI. However, it is not known whether ARBs affect the cell sheet therapy. This study aimed to examine ameliorating effects of ASC sheets on heart failure and remodeling after MI, and how pretreatment with ARBs prior to the creation of MI and ASC sheet transplantation modifies the effects of ASC sheets., Methods: ASCs were isolated from fat tissues of wild-type rats, and ASC sheets were engineered on temperature-responsive dishes. In in vitro studies using cultured cells, mRNA levels of vascular endothelial growth factor (VEGF) in ASCs were determined by RT-PCR in the presence of angiotensin II and/or an ARB, irbesartan, under normoxia and hypoxia; mRNA and protein levels of angiotensin II receptor type 1a (AT1aR), type 1b (AT1bR) and type 2 (AT2R) were also determined by RT-PCR and western blotting. In in vivo studies using a rat MI model, effects of transplanted ASC sheets and/or irbesartan on cardiac functions and remodeling after MI were evaluated by echocardiography, histological analysis and molecular biological techniques., Results: In the in vitro studies, ASCs expressed higher levels of VEGF mRNA under hypoxia. They also expressed mRNA and protein of AT1aR but not AT1bR or AT2R. Under normoxia, angiotensin II increased the level of VEGF mRNA in ASCs, which was abolished by irbesartan. Under hypoxia, irbesartan reduced the level of VEGF mRNA in ASCs regardless of whether angiotensin II was present or not. In the in vivo studies, ASC sheets improved cardiac functions after MI, leading to decreased interstitial fibrosis and increased capillary density in border zones. These effects of ASC sheets were abolished by oral administration of irbesartan before MI and their transplantation., Conclusions: ASC sheets ameliorated cardiac dysfunctions and remodeling after MI via increasing VEGF expression, which was abolished by pretreatment with irbesartan before the creation of MI and transplantation.
- Published
- 2018
- Full Text
- View/download PDF
34. Protective Effects of Topiroxostat on an Ischemia-Reperfusion Model of Rat Hearts.
- Author
-
Tanno S, Yamamoto K, Kurata Y, Adachi M, Inoue Y, Otani N, Mishima M, Yamamoto Y, Kuwabara M, Ogino K, Miake J, Ninomiya H, Shirayoshi Y, Okada F, Yamamoto K, and Hisatome I
- Subjects
- Allopurinol pharmacology, Allopurinol therapeutic use, Animals, Arrhythmias, Cardiac drug therapy, Nitriles pharmacology, Protective Agents pharmacology, Protective Agents therapeutic use, Pyridines pharmacology, Rats, Reactive Oxygen Species metabolism, Thiobarbituric Acid Reactive Substances metabolism, Ventricular Dysfunction, Left prevention & control, Xanthine Dehydrogenase antagonists & inhibitors, Myocardial Reperfusion Injury drug therapy, Nitriles therapeutic use, Pyridines therapeutic use
- Abstract
Background: Ischemia/reperfusion (I/R) injury triggers cardiac dysfunctions via creating reactive oxygen species (ROS). Because xanthine oxidase (XO) is one of the major enzymes that generate ROS, inhibition of XO is expected to suppress ROS-induced I/R injury. However, it remains unclear whether XO inhibition really yields cardioprotection during I/R. The protective effects of the XO inhibitors, topiroxostat and allopurinol, on cardiac I/R injury were evaluated.Methods and Results:Using isolated rat hearts, ventricular functions, occurrence of arrhythmias, XO activities and thiobarbituric acid reactive substances (TBARS) productions and myocardial levels of adenine nucleotides before and after I/R, and cardiomyocyte death markers during reperfusion, were evaluated. Topiroxostat prevented left ventricular dysfunctions and facilitated recovery from arrhythmias during I/R. Allopurinol and the antioxidant, N-acetylcysteine (NAC), exhibited similar effects at higher concentrations. Topiroxostat inhibited myocardial XO activities and TBARS productions after I/R. I/R decreased myocardial levels of ATP, ADP and AMP, but increased that of xanthine. While topiroxostat, allopurinol or NAC did not change myocardial levels of ATP, ADP or AMP after I/R, all of the agents decreased the level of xanthine. They also decreased releases of CPK and LDH during reperfusion., Conclusions: Topiroxostat showed protective effects against I/R injury with higher potency than allopurinol or NAC. It dramatically inhibited XO activity and TBARS production, suggesting suppression of ROS generation.
- Published
- 2018
- Full Text
- View/download PDF
35. Cited4 is related to cardiogenic induction and maintenance of proliferation capacity of embryonic stem cell-derived cardiomyocytes during in vitro cardiogenesis.
- Author
-
Miake J, Notsu T, Higaki K, Hidaka K, Morisaki T, Yamamoto K, and Hisatome I
- Subjects
- Animals, Cell Separation, Cells, Cultured, Embryonic Stem Cells metabolism, Flow Cytometry, Gene Expression Regulation, Developmental, Mice, Myocytes, Cardiac metabolism, Transcription Factors genetics, Cell Proliferation, Embryonic Stem Cells cytology, Heart embryology, Myocytes, Cardiac cytology, Transcription Factors metabolism
- Abstract
Cardiac progenitor cells have a limited proliferative capacity. The CREB-binding protein/p300-interacting transactivator, with the Glu/Asp-rich carboxy-terminal domain (Cited) gene family, regulates gene transcription. Increased expression of the Cited4 gene in an adult mouse is associated with exercise-induced cardiomyocyte hypertrophy and proliferation. However, the expression patterns and functional roles of the Cited4 gene during cardiogenesis are largely unknown. Therefore, in the present study, we investigated the expression patterns and functional roles of the Cited4 gene during in vitro cardiogenesis. Using embryoid bodies formed from mouse embryonic stem cells, we evaluated the expression patterns of the Cited4 gene by quantitative reverse transcriptase-polymerase chain reaction. Cited4 gene expression levels increased and decreased during the early and late phases of cardiogenesis, respectively. Moreover, Cited4 gene levels were significantly high in the cardiac progenitor cell population. A functional assay of the Cited4 gene in cardiac progenitor cells using flow cytometry indicated that overexpression of the Cited4 gene significantly increased the cardiac progenitor cell population compared with the control and knockdown groups. A cell proliferation assay, with 5-ethynyl-2'-deoxyuridine incorporation and Ki67 expression during the late phase of cardiogenesis, indicated that the number of troponin T-positive embryonic stem cell-direived cardiomyocytes with proliferative capacity was significantly greater in the overexpression group than in the control and knockdown groups. Our study results suggest that the Cited4 gene is related to cardiac differentiation and maintenance of proliferation capacity of embryonic stem cell-derived cardiomyocytes during in vitro cardiogenesis. Therefore, manipulation of Cited4 gene expression may be of great interest for cardiac regeneration.
- Published
- 2017
- Full Text
- View/download PDF
36. Molecular mechanisms underlying the pilsicainide-induced stabilization of hERG proteins in transfected mammalian cells.
- Author
-
Onohara T, Hisatome I, Kurata Y, Li P, Notsu T, Morikawa K, Otani N, Yoshida A, Iitsuka K, Kato M, Miake J, Ninomiya H, Higaki K, Shirayoshi Y, Nishihara T, Itoh T, Nakamura Y, and Nishimura M
- Abstract
Background: Pilsicainide, classified as a relatively selective Na
+ channel blocker, also has an inhibitory action on the rapidly-activating delayed-rectifier K+ current ( IKr ) through human ether-a-go-go-related gene (hERG) channels. We studied the effects of chronic exposure to pilsicainide on the expression of wild-type (WT) hERG proteins and WT-hERG channel currents, as well as on the expression of mutant hERG proteins, in a heterologous expression system., Methods: HEK293 cells stably expressing WT or mutant hERG proteins were subjected to Western blotting, immunofluorescence microscopy and patch-clamp experiments., Results: Acute exposure to pilsicainide at 0.03-10 μM influenced neither the expression of WT-hERG proteins nor WT-hERG channel currents. Chronic treatment with 0.03-10 μM pilsicainide for 48 h, however, increased the expression of WT-hERG proteins and channel currents in a concentration-dependent manner. Chronic treatment with 3 μM pilsicainide for 48 h delayed degradation of WT-hERG proteins and increased the channels expressed on the plasma membrane. A cell membrane-impermeant pilsicainide derivative did not influence the expression of WT-hERG, indicating that pilsicainide stabilized the protein inside the cell. Pilsicainide did not influence phosphorylation of Akt (protein kinase B) or expression of heat shock protein families such as HSF-1, hsp70 and hsp90. E4031, a chemical chaperone for hERG, abolished the pilsicainide effect on hERG. Chronic treatment with pilsicainide could also increase the protein expression of trafficking-defective mutant hERG, G601S and R752W., Conclusions: Pilsicainide penetrates the plasma membrane, stabilizes WT-hERG proteins by acting as a chemical chaperone, and enhances WT-hERG channel currents. This mechanism could also be applicable to modulations of certain mutant-hERG proteins.- Published
- 2017
- Full Text
- View/download PDF
37. M3 Muscarinic Receptor Signaling Stabilizes a Novel Mutant Human Ether-a-Go-Go-Related Gene Channel Protein via Phosphorylation of Heat Shock Factor 1 in Transfected Cells.
- Author
-
Mahati E, Li P, Kurata Y, Maharani N, Ikeda N, Sakata S, Ogura K, Miake J, Aiba T, Shimizu W, Nakasone N, Ninomiya H, Higaki K, Yamamoto K, Nakai A, Shirayoshi Y, and Hisatome I
- Subjects
- Adolescent, DNA-Binding Proteins genetics, HEK293 Cells, Heat Shock Transcription Factors, Humans, Male, Phosphorylation genetics, Protein Stability, Receptor, Muscarinic M3 genetics, Transcription Factors genetics, Transfection, DNA-Binding Proteins metabolism, ERG1 Potassium Channel genetics, ERG1 Potassium Channel metabolism, Long QT Syndrome genetics, Long QT Syndrome metabolism, Mutation, Receptor, Muscarinic M3 metabolism, Signal Transduction, Transcription Factors metabolism
- Abstract
Background: Long QT syndrome 2 (LQT2) is caused by mutations in the human ether-a-go-go-related gene (hERG). Most of its mutations give rise to unstable hERG proteins degraded by the proteasome. Recently, carbachol was reported to stabilize the wild-type hERG-FLAG via activation of the muscarinic type 3 receptor (M3-mAChR). Its action on mutant hERG-FLAG, however, remains uninvestigated.Methods and Results:A novel mutant hERG-FLAG carried 2 mutations: an amino acid substitution G572S and an in-frame insertion D1037_V1038insGD. When expressed in HEK293 cells, this mutant hERG-FLAG was degraded by the proteasome and failed to be transported to the cell surface. Carbachol restored stability of the mutant hERG-FLAG and facilitated cell-surface expression. Carbachol activated PKC, augmented phosphorylation of heat shock factor 1 (HSF1) and enhanced expression of heat shock proteins (hsps), hsp70 and hsp90. Both a M3-mAChR antagonist, 4-DAMP, and a PKC inhibitor, bisindolylmaleimide, abolished carbachol-induced stabilization of the mutant hERG-FLAG., Conclusions: M3-mAChR activation leads to enhancement of hsp expression via PKC-dependent phosphorylation of HSF1, thereby stabilizing the mutant hERG-FLAG protein. Thus, M3-mAChR activators may have a therapeutic value for patients with LQT2. (Circ J 2016; 80: 2443-2452).
- Published
- 2016
- Full Text
- View/download PDF
38. Characterization of the novel mutant A78T-HERG from a long QT syndrome type 2 patient: Instability of the mutant protein and stabilization by heat shock factor 1.
- Author
-
Kondo T, Hisatome I, Yoshimura S, Mahati E, Notsu T, Li P, Iitsuka K, Kato M, Ogura K, Miake J, Aiba T, Shimizu W, Kurata Y, Sakata S, Nakasone N, Ninomiya H, Nakai A, Higaki K, Kawata Y, Shirayoshi Y, Yoshida A, and Yamamoto K
- Abstract
Background: The human ether-a-go-go-related gene (HERG) encodes the α-subunit of rapidly activating delayed-rectifier potassium channels. Mutations in this gene cause long QT syndrome type 2 (LQT2). In most cases, mutations reduce the stability of the channel protein, which can be restored by heat shock (HS)., Methods: We identified the novel mutant A78T-HERG in a patient with LQT2. The purpose of the current study was to characterize this mutant protein and test whether HS and heat shock factors (HSFs) could stabilize the mutant protein. A78T-HERG and wild-type HERG (WT-HERG) were expressed in HEK293 cells and analyzed by immunoblotting, immunoprecipitation, immunofluorescence, and whole-cell patch clamping., Results: When expressed in HEK293 cells, WT-HERG gave rise to immature and mature forms of the protein at 135 and 155 kDa, respectively. A78T-HERG gave rise only to the immature form, which was heavily ubiquitinated. The proteasome inhibitor MG132 increased the expression of immature A78T-HERG and increased both the immature and mature forms of WT-HERG. WT-HERG, but not A78T-HERG, was expressed on the plasma membrane. In whole-cell patch clamping experiments, depolarizing pulses evoked E4031-sensitive HERG channel currents in cells transfected with WT-HERG, but not in cells transfected with A78T-HERG. The A78V mutant, but not A78G mutant, remained in the immature form similarly to A78T. Maturation of the A78T-HERG protein was facilitated by HS, expression of HSF-1, or exposure to geranyl geranyl acetone., Conclusions: A78T-HERG was characterized by protein instability and reduced expression on the plasma membrane. The stability of the mutant was partially restored by HSF-1, indicating that HSF-1 is a target for the treatment for LQT2 caused by the A78T mutation in HERG.
- Published
- 2016
- Full Text
- View/download PDF
39. Impact of postprocedural antiarrhythmic drug therapy with bepridil on maintaining sinus rhythm after catheter ablation for persistent atrial fibrillation.
- Author
-
Kondo T, Miake J, Kato M, Ogura K, Iitsuka K, and Yamamoto K
- Subjects
- Amiodarone therapeutic use, Case-Control Studies, Female, Follow-Up Studies, Humans, Male, Middle Aged, Recurrence, Sodium Channel Blockers therapeutic use, Anti-Arrhythmia Agents therapeutic use, Atrial Fibrillation therapy, Bepridil therapeutic use, Catheter Ablation
- Abstract
Background: Although several studies have assessed the predictors of recurrent atrial fibrillation (AF) after catheter ablation for persistent AF, the impact of antiarrhythmic drug (AAD) therapy on maintaining sinus rhythm after catheter ablation for persistent AF has not been fully evaluated. This case-control study aimed to evaluate the effect of bepridil on maintaining sinus rhythm after catheter ablation for persistent AF., Methods and Results: We enrolled 122 consecutive patients (87 men; mean age: 62.3 years) who underwent catheter ablation for persistent AF and were administered AAD therapy after the initial procedure. Restoration of sinus rhythm was achieved in all of the patients by catheter ablation and cardioversion after the initial procedure. After a median 12-month follow up, 51 of 122 (41.8%) patients had recurrence of AF. In Cox proportional hazard regression analysis, postprocedural AAD therapy with bepridil was a significantly correlated factor with freedom from recurrent AF after the initial ablation procedure (hazard ratio 0.446, 95% confidence interval 0.236-0.842, p=0.012). In Kaplan-Meier analysis, AF-free survival was significantly better with bepridil compared with amiodarone (AMD) and sodium channel blocker (SCB) (log-rank test, bepridil vs AMD, p=0.012; bepridil vs SCB, p=0.018)., Conclusions: Bepridil reduced the recurrence of AF compared with AMD and SCB in patients who underwent catheter ablation for persistent AF., (Copyright © 2015 Japanese College of Cardiology. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
40. Electrophysiological properties of iPS cell-derived cardiomyocytes from a patient with long QT syndrome type 1 harboring the novel mutation M437V of KCNQ1.
- Author
-
Sogo T, Morikawa K, Kurata Y, Li P, Ichinose T, Yuasa S, Nozaki D, Miake J, Ninomiya H, Shimizu W, Fukuda K, Yamamoto K, Shirayoshi Y, and Hisatome I
- Abstract
Introduction: Long QT syndrome type 1 (LQT1) is caused by mutations in KCNQ1 coding slowly-activating delayed-rectifier K
+ channels. We identified the novel missense mutation M437V of KCNQ1 in a LQT1 patient. Here, we employed iPS cell (iPSC)-derived cardiomyocytes to investigate electrophysiological properties of the mutant channel and LQT1 cardiomyocytes., Methods: To generate iPSCs from the patient and a healthy subject, peripheral blood T cells were reprogrammed by Sendai virus vector encoding human OCT3/4, SOX2, KLF4, and c-MYC. Cardiomyocytes were prepared from iPSCs and human embryonic stem cells using a cytokine-based two-step differentiation method and were subjected to patch clamp experiments., Results: LQT1 iPSC-derived cardiomyocytes exhibited prolongation of action potential duration (APD), which was due to a reduction of the KCNQ1-mediated current IKs ; Na+ , Ca2+ and other K+ channel currents were comparable. When expressed in HEK293 and COS7 cells, the mutant KCNQ1 was normally expressed in the plasma membrane but generated smaller currents than the wild type. Isoproterenol significantly prolonged APDs of LQT1 cardiomyocytes, while shortening those of healthy ones. A mathematical model for IKs -reduced human ventricular myocytes reproduced APD prolongation and generation of early afterdepolarizations (EADs) under β-adrenergic stimulation., Conclusions: QT prolongation of the LQT1 patient with the mutation M437V of KCNQ1 was caused by IKs reduction, which may render the patient vulnerable to generation of EADs and arrhythmias.- Published
- 2016
- Full Text
- View/download PDF
41. Stabilization of Kv1.5 channel protein by the inotropic agent olprinone.
- Author
-
Endo R, Kurata Y, Notsu T, Li P, Morikawa K, Kondo T, Ogura K, Miake J, Yoshida A, Shirayoshi Y, Ninomiya H, Higaki K, Kuwabara M, Yamamoto K, Inagaki Y, and Hisatome I
- Subjects
- Animals, COS Cells, Chlorocebus aethiops, Dose-Response Relationship, Drug, Membrane Potentials drug effects, Membrane Potentials physiology, Protein Stability drug effects, Cardiotonic Agents metabolism, Cardiotonic Agents pharmacology, Imidazoles metabolism, Imidazoles pharmacology, Kv1.5 Potassium Channel metabolism, Pyridones metabolism, Pyridones pharmacology
- Abstract
Olprinone is an inotropic agent that inhibits phosphodiesterase (PDE) III and causes vasodilation. Olprinone has been shown to be less proarrhythmic and possibly affect expression of functional Kv1.5 channels that confer the ultra-rapid delayed-rectifier K+ channel current (IKur) responsible for action potential repolarization. To reveal involvement of Kv1.5 channels in the less arrhythmic effect of olprinone, we examined effects of the agent on the stability of Kv1.5 channel proteins expressed in COS7 cells. Olprinone at 30-1000 nM increased the protein level of Kv1.5 channels in a concentration-dependent manner. Chase experiments showed that olprinone delayed degradation of Kv1.5 channels. Olprinone increased the immunofluorescent signal of Kv1.5 channels in the endoplasmic reticulum (ER) and Golgi apparatus as well as on the cell surface. Kv1.5-mediated membrane currents, measured as 4-aminopyridine-sensitive currents, were increased by olprinone without changes in their activation kinetics. A protein transporter inhibitor, colchicine, abolished the olprinone-induced increase of Kv.1.5-mediated currents. The action of olprinone was inhibited by 4-aminopyridine, and was not mimicked by the application of 8-Bromo-cAMP. Taken together, we conclude that olprinone stabilizes Kv1.5 proteins at the ER through an action as a chemical chaperone, and thereby increases the density of Kv1.5 channels on the cell membrane. The enhancement of Kv1.5 currents could underlie less arrhythmogenicity of olprinone., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
42. Ultra-Rapid and Massive Thrombus Formation in Cardiac Chambers.
- Author
-
Okamura A, Miake J, Miyagi M, and Yamamoto K
- Subjects
- Adult, Echocardiography, Heart Atria, Heart Ventricles, Humans, Male, Photography, Thrombosis etiology, Thrombosis pathology, Ventricular Fibrillation complications
- Published
- 2015
- Full Text
- View/download PDF
43. Instability of KCNE1-D85N that causes long QT syndrome: stabilization by verapamil.
- Author
-
Sakata S, Kurata Y, Li P, Notsu T, Morikawa K, Miake J, Higaki K, Yamamoto Y, Yoshida A, Shirayoshi Y, Yamamoto K, Horie M, Ninomiya H, Kanzaki S, and Hisatome I
- Subjects
- Cells, Cultured, Humans, Calcium Channel Blockers pharmacology, Calcium Channel Blockers therapeutic use, Long QT Syndrome drug therapy, Long QT Syndrome genetics, Polymorphism, Genetic, Potassium Channels, Voltage-Gated drug effects, Potassium Channels, Voltage-Gated genetics, Verapamil pharmacology, Verapamil therapeutic use
- Abstract
Background: A KCNE1 polymorphism, D85N, causes long QT syndrome (LQTS) with a decrease in the slowly activating delayed-rectifier K(+) channel current (IKs ). We examined impacts of D85N polymorphism on KCNE1 protein stability and functions, and tested the ability of various drugs to modify them., Methods: KCNE1-D85N or the wild-type protein was coexpressed in COS7 cells with KCNQ1 to form K(+) channels. Expression, degradation, and intracellular localization of KCNE1 proteins, as well as the currents conferred by KCNQ1/KCNE1 complexes, were determined using immunoblots, immunofluorescence, and patch-clamp techniques., Results: The protein level of KCNE1-D85N was lower than that of the wild-type, in spite of the comparable levels of their mRNA. KCNE1-D85N was highly ubiquitinated and rapidly degraded as compared to the wild-type; a proteasome inhibitor, MG132, inhibited its degradation and increased its steady-state level. Both KCNE1-D85N and the wild-type proteins were co-immunoprecipitated with KCNQ1. Immunofluorescent signals of KCNE1-D85N accumulated in the endoplasmic reticulum and Golgi apparatus, with reduced levels on the cell membrane. Patch-clamp experiments demonstrated that the membrane current corresponding to IKs was much smaller in cells expressing KCNE1-D85N than in those expressing the wild-type. Verapamil (0.5-10 μM) increased the protein level of KCNE1-D85N, decreased its ubiquitination, slowed its degradation, and enhanced KCNQ1/KCNE1-D85N channel currents. Pretreatment with amiodarone abolished these effects of verapamil., Conclusion: KCNE1-D85N is less stable than the wild-type protein, and is rapidly degraded through the ubiquitin-proteasome system. Verapamil may be of a therapeutic value in LQTS patients via preventing degradation of KCNE1-D85N., (©2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
44. Hsp90 prevents interaction between CHIP and HERG proteins to facilitate maturation of wild-type and mutant HERG proteins.
- Author
-
Iwai C, Li P, Kurata Y, Hoshikawa Y, Morikawa K, Maharani N, Higaki K, Sasano T, Notsu T, Ishido Y, Miake J, Yamamoto Y, Shirayoshi Y, Ninomiya H, Nakai A, Murata S, Yoshida A, Yamamoto K, Hiraoka M, and Hisatome I
- Subjects
- Animals, Benzoquinones pharmacology, Cell Membrane enzymology, ERG1 Potassium Channel, Endoplasmic Reticulum enzymology, Ether-A-Go-Go Potassium Channels genetics, HEK293 Cells, HSP90 Heat-Shock Proteins antagonists & inhibitors, HSP90 Heat-Shock Proteins genetics, Humans, Lactams, Macrocyclic pharmacology, Long QT Syndrome enzymology, Long QT Syndrome genetics, Macrolides pharmacology, Membrane Potentials, Mice, Mutation, Missense, Myocytes, Cardiac drug effects, Protein Transport, Transfection, Ubiquitin-Protein Ligases genetics, Ubiquitination, Ether-A-Go-Go Potassium Channels metabolism, HSP90 Heat-Shock Proteins metabolism, Myocytes, Cardiac enzymology, Ubiquitin-Protein Ligases metabolism
- Abstract
Aims: We examined the role of Hsp90 in expression and maturation of wild-type (WT) and mutant ether-a-go-go related gene (HERG) proteins by using Hsp90 inhibitors, geldanamycin (GA) and radicicol, and Hsp90 overexpression., Methods and Results: The proteins were expressed in HEK293 cells or collected from HL-1 mouse cardiomyocytes, and analysed by western blotting, immunoprecipitation, immunofluorescence, and whole-cell patch-clamp techniques. GA and radicicol suppressed maturation of HERG-FLAG proteins and increased their immature forms. Co-expression of Hsp90 counteracted the effects of Hsp90 inhibitors and suppressed ubiquitination of HERG proteins. Overexpressed Hsp90 also inhibited the binding of endogenous C-terminus of Hsp70-interacting protein (CHIP) to HERG-FLAG proteins. Hsp90-induced increase of functional HERG proteins was verified by their increased expression on the cell surface and enhanced HERG channel currents. CHIP overexpression decreased both mature and immature forms of HERG-FLAG proteins in cells treated with GA. Hsp90 facilitated maturation of endogenous ERG proteins, whereas CHIP decreased both forms of ERG proteins in HL-1 cells. Mutant HERG proteins harbouring disease-causing missense mutations were mainly in the immature form and had a higher binding capacity to CHIP than the WT; Hsp90 overexpression suppressed this association. Overexpressed Hsp90 increased the mature form of HERG(1122fs/147) proteins, reduced its ubiquitinated form, increased its immunoreactivity in the endoplasmic reticulum and on the plasma membrane, and increased the mutant-mediated membrane current. CHIP overexpression decreased the immature form of HERG(1122fs/147) proteins., Conclusion: Enhancement of HERG protein expression through Hsp90 inhibition of CHIP binding might be a novel therapeutic strategy for long QT syndrome 2 caused by trafficking abnormalities of HERG proteins.
- Published
- 2013
- Full Text
- View/download PDF
45. Stabilization of Kv1.5 channel protein by bepridil through its action as a chemical chaperone.
- Author
-
Suzuki S, Kurata Y, Li P, Notsu T, Hasegawa A, Ikeda N, Kato M, Miake J, Sakata S, Shiota G, Yoshida A, Ninomiya H, Higaki K, Yamamoto K, Shirayoshi Y, and Hisatome I
- Subjects
- 4-Aminopyridine pharmacology, Animals, COS Cells, Chlorocebus aethiops, Endoplasmic Reticulum drug effects, Endoplasmic Reticulum metabolism, Golgi Apparatus drug effects, Golgi Apparatus metabolism, Potassium Channel Blockers pharmacology, Rats, Bepridil pharmacology, Kv1.5 Potassium Channel metabolism, Membrane Transport Modulators pharmacology
- Abstract
While bepridil has been reported to alter the stability of ion channel proteins, the precise mechanism of action remains unclear. We examined the effect of bepridil on the stability of Kv1.5 channel proteins expressed in COS7 cells. Bepridil at 0.3-30 μM increased the protein level of Kv1.5 channels in a concentration-dependent manner. Chase experiments showed that bepridil delayed the degradation process of Kv1.5 channel proteins in the same manner as a proteasomal inhibitor, MG132, did. Bepridil increased the immunofluorescent signal of Kv1.5 channel proteins in the endoplasmic reticulum (ER) and Golgi apparatus and on the cell surface. The cell fraction experiment also showed bepridil-induced increases in Kv1.5 in the ER, Golgi apparatus, and the cell membrane. Bepridil at a lower concentration of 1 μM had no effect on the proteasome activity in vitro. A blocker of the ultrarapid delayed-rectifier K(+) channel current, 4-aminopyridine (4AP), abolished bepridil-induced increases in Kv1.5. Kv1.5-medicated membrane currents measured as 4AP-sensitive currents were increased by bepridil. Taken together, we conclude that bepridil stabilizes Kv1.5 proteins at the ER through an action as a chemical chaperone, thereby increasing the density of Kv1.5 channels in the cell membrane., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
46. Electrophysiological properties of prion-positive cardiac progenitors derived from murine embryonic stem cells.
- Author
-
Fujii H, Ikeuchi Y, Kurata Y, Ikeda N, Bahrudin U, Li P, Nakayama Y, Endo R, Hasegawa A, Morikawa K, Miake J, Yoshida A, Hidaka K, Morisaki T, Ninomiya H, Shirayoshi Y, Yamamoto K, and Hisatome I
- Subjects
- Animals, Biomarkers metabolism, Cell Differentiation, Cell Line, Cell Lineage, Cell Separation methods, Coculture Techniques, Cyclic Nucleotide-Gated Cation Channels genetics, Cyclic Nucleotide-Gated Cation Channels metabolism, Flow Cytometry, Gene Expression Regulation, Developmental, Immunohistochemistry, Mice, Mice, 129 Strain, Myocardial Contraction, Patch-Clamp Techniques, Periodicity, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Transcription Factors genetics, Transcription Factors metabolism, Transfection, Action Potentials, Embryonic Stem Cells metabolism, Myocytes, Cardiac metabolism, Prions metabolism
- Abstract
Background: The prion protein (PrP) has been reported to serve as a surface maker for isolation of cardiomyogenic progenitors from murine embryonic stem (ES) cells. Although PrP-positive cells exhibited automaticity, their electrophysiological characteristics remain unresolved. The aim of the present study was therefore to investigate the electrophysiological properties of PrP-positive cells in comparison with those of HCN4p-or Nkx2.5-positive cells., Methods and Results: Differentiation of AB1, HCN5p-EGFP and hcgp7 ES cells into cardiac progenitors was induced by embryoid body (EB) formation. EBs were dissociated and cells expressing PrP, HCN4-EGFP and/or Nkx2.5-GFP were collected via flow cytometry. Sorted cells were subjected to reverse transcriptase-polymerase chain reaction, immunostaining and patch-clamp experiments. PrP-positive cells expressed mRNA of undifferentiation markers, first and second heart field markers, and cardiac-specific genes and ion channels, indicating their commitment to cardiomyogenic progenitors. PrP-positive cells with automaticity showed positive and negative chronotropic responses to isoproterenol and carbamylcholine, respectively. Hyperpolarization-activated cation current (I(f)) was barely detectable, whereas Na(+) and L-type Ca(2+) channel currents were frequently observed. Their spontaneous activity was slowed by inhibition of sarcoplasmic reticulum Ca(2+) uptake and release but not by blocking I(f). The maximum diastolic potential of their spontaneous firings was more depolarized than that of Nkx2.5-GFP-positive cells., Conclusions: PrP-positive cells contained cardiac progenitors that separated from the lineage of sinoatrial node cells. PrP can be used as a marker to enrich nascent cardiac progenitors.
- Published
- 2012
- Full Text
- View/download PDF
47. Impairment of ubiquitin-proteasome system by E334K cMyBPC modifies channel proteins, leading to electrophysiological dysfunction.
- Author
-
Bahrudin U, Morikawa K, Takeuchi A, Kurata Y, Miake J, Mizuta E, Adachi K, Higaki K, Yamamoto Y, Shirayoshi Y, Yoshida A, Kato M, Yamamoto K, Nanba E, Morisaki H, Morisaki T, Matsuoka S, Ninomiya H, and Hisatome I
- Subjects
- Amino Acid Substitution, Animals, Apoptosis, Arrhythmias, Cardiac physiopathology, Calcium metabolism, Cardiomyopathy, Hypertrophic complications, Cardiomyopathy, Hypertrophic pathology, Cell Line, Humans, Mutant Proteins genetics, Mutant Proteins metabolism, Rats, Carrier Proteins genetics, Carrier Proteins metabolism, Ion Channels metabolism, Mutation, Missense, Proteasome Endopeptidase Complex metabolism, Ubiquitin metabolism
- Abstract
Cardiac arrhythmogenesis is regulated by channel proteins whose protein levels are in turn regulated by the ubiquitin-proteasome system (UPS). We have previously reported on UPS impairment induced by E334K cardiac myosin-binding protein C (cMyBPC), which causes hypertrophic cardiomyopathy (HCM) accompanied by arrhythmia. We hypothesized that UPS impairment induced by E334K cMyBPC causes accumulation of cardiac channel proteins, leading to electrophysiological dysfunction. Wild-type or E334K cMyBPC was overexpressed in HL-1 cells and primary cultured neonatal rat cardiac myocytes. The expression of E334K cMyBPC suppressed cellular proteasome activities. The protein levels of K(v)1.5, Na(v)1.5, Hcn4, Ca(v)3.2, Ca(v)1.2, Serca, RyR2, and Ncx1 were significantly higher in cells expressing E334K cMyBPC than in wild type. They further increased in cells pretreated with MG132 and had longer protein decays. The channel proteins retained the correct localization. Cells expressing E334K cMyBPC exhibited higher Ca(2+) transients and longer action potential durations (APDs), accompanied by afterdepolarizations and higher apoptosis. Those augments of APD and Ca(2+) transients were recapitulated by a simulation model. Although a Ca(2+) antagonist, azelnidipine, neither protected E334K cMyBPC from degradation nor affected E334K cMyBPC incorporation into the sarcomere, it normalized the APD and Ca(2+) transients and partially reversed the levels of those proteins regulating apoptosis, thereby attenuating apoptosis. In conclusion, UPS impairment caused by E334K cMyBPC may modify the levels of channel proteins, leading to electrophysiological dysfunction. Therefore, UPS impairment due to a mutant cMyBPC may partly contribute to the observed clinical arrhythmias in HCM patients., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
48. Enhancing effects of salicylate on quinidine-induced block of human wild type and LQT3 related mutant cardiac Na+ channels.
- Author
-
Urashima T, Kurata Y, Miake J, Kato M, Ogura K, Yano A, Adachi M, Tanaka Y, Yamada K, Hamada T, Mizuta E, Kuwabara M, Kato M, Yamamoto Y, Ogino K, Yoshida A, Shirayoshi Y, and Hisatome I
- Subjects
- Animals, COS Cells, Chlorocebus aethiops, Heart drug effects, Humans, Membrane Potentials drug effects, Membrane Potentials genetics, Muscle, Skeletal drug effects, Muscle, Skeletal metabolism, Mutation, Myocardium metabolism, NAV1.5 Voltage-Gated Sodium Channel, Protein Binding, Quinidine metabolism, Quinidine pharmacology, Salicylates pharmacology, Sodium Channel Blockers pharmacology, Sodium Channels genetics, Sodium Channels metabolism
- Abstract
It is unknown whether salicylate enhances the action of antiarrhythmic agents on human Na+ channels with state dependency and tissue specificity. We therefore investigated effects of salicylate on quinidine-induced block of human cardiac and skeletal muscle Na+ channels. Human cardiac wild-type (hH1), LQT3-related mutant (ΔKPQ), and skeletal muscle (hSkM1) Na+ channel α subunits were expressed in COS7 cells. Effects of salicylate on quinidine-induced tonic and use-dependent block of Na+ channel currents were examined by the whole-cell patch-clamp technique. Salicylate enhanced the quinidine-induced tonic and use-dependent block of both hH1 and hSkM1 currents at a holding potential (HP) of -100 mV but not at -140 mV. Salicylate decreased the IC50 value for the quinidine-induced tonic block of hH1 at an HP of -100 mV, and produced a negative shift in the steady-state inactivation curve of hH1 in the presence of quinidine. According to the modulated receptor theory, it is probable that salicylate decreases the dissociation constant for quinidine binding to inactivated-state channels. Furthermore, salicylate significantly enhanced the quinidine-induced tonic and use-dependent block of the peak and steady-state ΔKPQ channel currents. The results suggest that salicylate enhances quinidine-induced block of Na+ channels via increasing the affinity of quinidine to inactivated state channels.
- Published
- 2011
- Full Text
- View/download PDF
49. Reciprocal control of hERG stability by Hsp70 and Hsc70 with implication for restoration of LQT2 mutant stability.
- Author
-
Li P, Ninomiya H, Kurata Y, Kato M, Miake J, Yamamoto Y, Igawa O, Nakai A, Higaki K, Toyoda F, Wu J, Horie M, Matsuura H, Yoshida A, Shirayoshi Y, Hiraoka M, and Hisatome I
- Subjects
- Action Potentials physiology, Animals, Cell Membrane metabolism, Cells, Cultured, Disease Models, Animal, Electrophysiologic Techniques, Cardiac, Endoplasmic Reticulum metabolism, Ether-A-Go-Go Potassium Channels pharmacology, HEK293 Cells, Heat-Shock Response physiology, Humans, Mice, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, RNA, Small Interfering pharmacology, Ether-A-Go-Go Potassium Channels genetics, Ether-A-Go-Go Potassium Channels metabolism, HSC70 Heat-Shock Proteins metabolism, HSP70 Heat-Shock Proteins metabolism, Long QT Syndrome genetics, Long QT Syndrome metabolism, Mutation, Missense genetics
- Abstract
Rationale: The human ether-a-go-go-related gene (hERG) encodes the α subunit of the potassium current I(Kr). It is highly expressed in cardiomyocytes and its mutations cause long QT syndrome type 2. Heat shock protein (Hsp)70 is known to promote maturation of hERG. Hsp70 and heat shock cognate (Hsc70) 70 has been suggested to play a similar function. However, Hsc70 has recently been reported to counteract Hsp70., Objective: We investigated whether Hsc70 counteracts Hsp70 in the control of wild-type and mutant hERG stability., Methods and Results: Coexpression of Hsp70 with hERG in HEK293 cells suppressed hERG ubiquitination and increased the levels of both immature and mature forms of hERG. Immunocytochemistry revealed increased levels of hERG in the endoplasmic reticulum and on the cell surface. Electrophysiological studies showed increased I(Kr). All these effects of Hsp70 were abolished by Hsc70 coexpression. Heat shock treatment of HL-1 mouse cardiomyocytes induced endogenous Hsp70, switched mouse ERG associated with Hsc70 to Hsp70, increased I(Kr), and shortened action potential duration. Channels with disease-causing missense mutations in intracellular domains had a higher binding capacity to Hsc70 than wild-type channels and channels with mutations in the pore region. Knockdown of Hsc70 by small interfering RNA or heat shock prevented degradation of mutant hERG proteins with mutations in intracellular domains., Conclusions: These results indicate reciprocal control of hERG stability by Hsp70 and Hsc70. Hsc70 is a potential target in the treatment of LQT2 resulting from missense hERG mutations.
- Published
- 2011
- Full Text
- View/download PDF
50. Novel effects of extracts from poisonous mushrooms on expression and function of the human ether-a-go-go-related gene channel.
- Author
-
Li P, Tanaka S, Ichiyanagi T, Ninomiya H, Ting Y, Utami SB, Aimi T, Shirayoshi Y, Miake J, and Hisatome I
- Subjects
- Action Potentials drug effects, Base Sequence, Cell Line, DNA Primers, HSP70 Heat-Shock Proteins genetics, Humans, Ion Channel Gating drug effects, Patch-Clamp Techniques, RNA, Small Interfering, Reverse Transcriptase Polymerase Chain Reaction, Agaricales, Ether-A-Go-Go Potassium Channels drug effects
- Abstract
Unlabelled: The human ether-a-go-go-related gene (hERG) encodes the α subunit of the potassium current I(Kr), which plays a pivotal role in cardiac action potential repolarization. Inherited mutations of this gene cause Long QT syndrome type 2. hERG expression is altered by several types of drugs as well as by temperature. Heat shock protein 70 (Hsp70) and Heat shock cognate protein 70 (Hsc70) have reciprocal effects on hERG proteins. We examined the effects of poisonous mushrooms on hERG protein expression and its channel function., Methods: We evaluated the effects of several types of poisonous mushrooms on the expression and function of wild-type hERG by Western blotting, reverse transcription polymerase chain reaction (PCR), and patch clamping in transfected HEK293 cells and mouse HL-1 cardiomyocytes., Results: Extracts of Gymnopilus junonius (junonius) increased expression of both hERG and Hsp70 in HEK293 cells with concomitant decrease in Hsc70, whereas extracts of Amanita ibotengutake (ibotengutake) decreased hERG proteins with increase in Hsc70. Knockdown of Hsp70 and Hsc70 by small interfering RNA abolished the effects of the two mushrooms on hERG, respectively. Certain fractions of junonius increased expression of hERG proteins. hERG currents were increased by extracts of junonius, resulting in shortening of action potential duration (APD). In contrast, hERG currents were decreased and APD was prolonged by extracts of ibotengutake., Conclusion: junonius enhanced the expression and function of hERG by increasing Hsp70 and decreasing Hsc70. Ibotengutake decreased hERG expression via increase in Hsc70. Constituents of junonius may have the potential for use in treatment of arrhythmia.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.