42 results on '"Calvier L"'
Search Results
2. P310Cardiotrophin-1 is involved in cardiovascular and renal fibrosis and dysfunction
- Author
-
Lopez-Andres, N., Rousseau, A., Calvier, L., Akhtar, R., Labat, C., Cruickshank, K., Diez, J., Zannad, F., Lacolley, P., and Rossignol, P.
- Published
- 2012
3. P152Galectin-3 is a potential mediator of aldosterone effects in vascular remodeling
- Author
-
Calvier, L., Reboul, P., Martin-Fernandez, B., Lahera, V., Zannad, F., Cachofeiro, V., Lacolley, P., Rossignol, P., and Lopez-Andres, N.
- Published
- 2012
4. Chronic TGFβ1-Signaling in Pulmonary Arterial Hypertension Induces Sustained Canonical Smad3 Pathways in Vascular SMC
- Author
-
Calvier, L., primary, Chouvarine, P., additional, Legchenko, E., additional, Kokeny, G., additional, Mozes, M.M., additional, and Hansmann, G., additional
- Published
- 2019
- Full Text
- View/download PDF
5. P4.11: Galectin-3 is A Potential Mediator of Aldosterone Effects in Vascular Remodeling
- Author
-
Calvier, L., Reboul, P., Martin-Fernandez, B., Lahera, V., Zannad, F., Cachofeiro, V., Lacolley, P., Rossignol, P., and Lopez-Andres, N.
- Published
- 2011
- Full Text
- View/download PDF
6. PPARgamma Links BMP2 and TGFβ1 Pathways in Vascular Smooth Muscle Cells
- Author
-
Calvier, L., primary, Chouvarine, P., additional, Mozes, M.M., additional, and Hansmann, G., additional
- Published
- 2017
- Full Text
- View/download PDF
7. Galectin-3 and Aldosterone as Potential Tandem Biomarkers in Pulmonary Arterial Hypertension
- Author
-
Calvier, L., primary, Legchenko, E., additional, Grimm, L., additional, Sallmon, H., additional, Hatch, A., additional, Plouffe, B., additional, Schroeder, C., additional, Bauersachs, J., additional, Murthy, S., additional, and Hansmann, G., additional
- Published
- 2016
- Full Text
- View/download PDF
8. (339) - PPARgamma Links BMP2 and TGFβ1 Pathways in Vascular Smooth Muscle Cells
- Author
-
Calvier, L., Chouvarine, P., Mozes, M.M., and Hansmann, G.
- Published
- 2017
- Full Text
- View/download PDF
9. Poster session 2
- Author
-
Perez-Pomares, J. M., primary, Ruiz-Villalba, A., additional, Ziogas, A., additional, Segovia, J. C., additional, Ehrbar, M., additional, Munoz-Chapuli, R., additional, De La Rosa, A., additional, Dominguez, J. N., additional, Hove-Madsen, L., additional, Sankova, B., additional, Sedmera, D., additional, Franco, D., additional, Aranega Jimenez, A., additional, Babaeva, G., additional, Chizh, N., additional, Galchenko, S., additional, Sandomirsky, B., additional, Schwarzl, M., additional, Seiler, S., additional, Steendijk, P., additional, Huber, S., additional, Maechler, H., additional, Truschnig-Wilders, M., additional, Pieske, B., additional, Post, H., additional, Simrick, S., additional, Kreutzer, R., additional, Rao, C., additional, Terracciano, C. M., additional, Kirchhof, P., additional, Fabritz, L., additional, Brand, T., additional, Theveniau-Ruissy, M., additional, Parisot, P., additional, Francou, A., additional, Saint-Michel, E., additional, Mesbah, K., additional, Kelly, R. G., additional, Wu, H.-T., additional, Sie, S.-S., additional, Chen, C.-Y., additional, Kuan, T.-C., additional, Lin, C. S., additional, Ismailoglu, Z., additional, Guven, M., additional, Yakici, A., additional, Ata, Y., additional, Ozcan, S., additional, Yildirim, E., additional, Ongen, Z., additional, Miroshnikova, V., additional, Demina, E., additional, Rodygina, T., additional, Kurjanov, P., additional, Denisenko, A., additional, Schwarzman, A., additional, Rubanenko, A., additional, Shchukin, Y., additional, Germanov, A., additional, Goldbergova, M., additional, Parenica, J., additional, Lipkova, J., additional, Pavek, N., additional, Kala, P., additional, Poloczek, M., additional, Vasku, A., additional, Parenicova, I., additional, Spinar, J., additional, Gambacciani, C., additional, Chiavacci, E., additional, Evangelista, M., additional, Vesentini, N., additional, Kusmic, C., additional, Pitto, L., additional, Chernova, A., additional, Nikulina, S. U. Y., additional, Arvanitis, D. A., additional, Mourouzis, I., additional, Pantos, C., additional, Kranias, E. G., additional, Cokkinos, D. V., additional, Sanoudou, D., additional, Vladimirskaya, T. E., additional, Shved, I. A., additional, Kryvorot, S. G., additional, Schirmer, I. M., additional, Appukuttan, A., additional, Pott, L., additional, Jaquet, K., additional, Ladilov, Y., additional, Archer, C. R., additional, Bootman, M. D., additional, Roderick, H. L., additional, Fusco, A., additional, Sorriento, D., additional, Santulli, G., additional, Trimarco, B., additional, Iaccarino, G., additional, Hagenmueller, M., additional, Riffel, J., additional, Bernhold, E., additional, Katus, H. A., additional, Hardt, S. E., additional, Maqsood, A., additional, Zi, M., additional, Prehar, S., additional, Neyses, L., additional, Ray, S., additional, Oceandy, D., additional, Khatami, N., additional, Wadowski, P., additional, Wagh, V., additional, Hescheler, J., additional, Sachinidis, A., additional, Mohl, W., additional, Chaudhry, B., additional, Burns, D., additional, Henderson, D. J., additional, Bax, N. A. M., additional, Van Marion, M. H., additional, Shah, B., additional, Goumans, M. J., additional, Bouten, C. V. C., additional, Van Der Schaft, D. W. J., additional, Van Oorschot, A. A. M., additional, Maas, S., additional, Braun, J., additional, Van Tuyn, J., additional, De Vries, A. A. F., additional, Gittenberger-De Groot, A. C., additional, Bageghni, S., additional, Drinkhill, M. J., additional, Batten, T. F. C., additional, Ainscough, J. F. X., additional, Onate, B., additional, Vilahur, G., additional, Ferrer-Lorente, R., additional, Ybarra, J., additional, Diez-Caballero, A., additional, Ballesta-Lopez, C., additional, Moscatiello, F., additional, Herrero, J., additional, Badimon, L., additional, Martin-Rendon, E., additional, Clifford, D. M., additional, Fisher, S. A., additional, Brusnkill, S. J., additional, Doree, C., additional, Mathur, A., additional, Clarke, M., additional, Watt, S. M., additional, Hernandez-Vera, R., additional, Kavanagh, D., additional, Yemm, A. I., additional, Frampton, J., additional, Kalia, N., additional, Terajima, Y., additional, Shimizu, T., additional, Tsuruyama, S., additional, Ishii, H., additional, Sekine, H., additional, Hagiwara, N., additional, Okano, T., additional, Vrijsen, K. R., additional, Chamuleau, S. A. J., additional, Sluijter, J. P. G., additional, Doevendans, P. F. M., additional, Madonna, R., additional, Delli Pizzi, S., additional, Di Donato, L., additional, Mariotti, A., additional, Di Carlo, L., additional, D'ugo, E., additional, Teberino, M. A., additional, Merla, A., additional, T, A., additional, De Caterina, R., additional, Kolker, L., additional, Ali, N. N., additional, Maclellan, K., additional, Moore, M., additional, Wheeler, J., additional, Harding, S. E., additional, Fleck, R. A., additional, Rowlinson, J. M., additional, Kraenkel, N., additional, Ascione, R., additional, Madeddu, P., additional, O'sullivan, J. F., additional, Leblond, A. L., additional, Kelly, G., additional, Kumar, A. H. S., additional, Metharom, P., additional, Buneker, C. K., additional, Alizadeh-Vikali, N., additional, Hynes, B. G., additional, O'connor, R., additional, Caplice, N. M., additional, Noseda, M., additional, De Smith, A. J., additional, Leja, T., additional, Rao, P. H., additional, Al-Beidh, F., additional, Abreu Pavia, M. S., additional, Blakemore, A. I., additional, Schneider, M. D., additional, Stathopoulou, K., additional, Cuello, F., additional, Ehler, E., additional, Haworth, R. S., additional, Avkiran, M., additional, Morawietz, H., additional, Eickholt, C., additional, Langbein, H., additional, Brux, M., additional, Goettsch, C., additional, Goettsch, W., additional, Arsov, A., additional, Brunssen, C., additional, Mazilu, L., additional, Parepa, I. R., additional, Suceveanu, A. I., additional, Suceveanu, A. P., additional, De Man, F. S., additional, Guignabert, C., additional, Tu, L., additional, Handoko, M. L., additional, Schalij, I., additional, Fadel, E., additional, Postmus, P. E., additional, Vonk-Noordegraaf, A., additional, Humbert, M., additional, Eddahibi, S., additional, Del Giudice, C., additional, Anastasio, A., additional, Fazal, L., additional, Azibani, F., additional, Bihry, N., additional, Merval, R., additional, Polidano, E., additional, Samuel, J.-L., additional, Delcayre, C., additional, Zhang, Y., additional, Mi, Y. M., additional, Ren, L. L., additional, Cheng, Y. P., additional, Guo, R., additional, Liu, Y., additional, Jiang, Y. N., additional, Kokkinos, A. D., additional, Tretjakovs, P., additional, Jurka, A., additional, Bormane, I., additional, Mikelsone, I., additional, Reihmane, D., additional, Elksne, K., additional, Krievina, G., additional, Verbovenko, J., additional, Bahs, G., additional, Lopez-Andres, N., additional, Rousseau, A., additional, Calvier, L., additional, Akhtar, R., additional, Labat, C., additional, Cruickshank, K., additional, Diez, J., additional, Zannad, F., additional, Lacolley, P., additional, Rossignol, P., additional, Hamesch, K., additional, Subramanian, P., additional, Li, X., additional, Thiemann, A., additional, Heyll, K., additional, Dembowsky, K., additional, Chevalier, E., additional, Weber, C., additional, Schober, A., additional, Yang, L., additional, Kim, G., additional, Gardner, B., additional, Earley, J., additional, Hofmann-Bowman, M., additional, Cheng, C.-F., additional, Lian, W.-S., additional, Lin, H., additional, Jinjolia, N. J., additional, Abuladze, G. A., additional, Tvalchrelidze, S. H. T., additional, Khamnagadaev, I., additional, Shkolnikova, M., additional, Kokov, L., additional, Miklashevich, I., additional, Drozdov, I., additional, Ilyich, I., additional, Bingen, B. O., additional, Askar, S. F. A., additional, Ypey, D. L., additional, Van Der Laarse, A., additional, Schalij, M. J., additional, Pijnappels, D. A., additional, Roney, C. H., additional, Ng, F. S., additional, Chowdhury, R. A., additional, Chang, E. T. Y., additional, Patel, P. M., additional, Lyon, A. R., additional, Siggers, J. H., additional, Peters, N. S., additional, Obergrussberger, A., additional, Stoelzle, S., additional, Bruggemann, A., additional, Haarmann, C., additional, George, M., additional, Fertig, N., additional, Moreira, D., additional, Souza, A., additional, Valente, P., additional, Kornej, J., additional, Reihardt, C., additional, Kosiuk, J., additional, Arya, A., additional, Hindricks, G., additional, Adams, V., additional, Husser, D., additional, Bollmann, A., additional, Camelliti, P., additional, Dudhia, J., additional, Dias, P., additional, Cartledge, J., additional, Connolly, D. J., additional, Nobles, M., additional, Sebastian, S., additional, Tinker, A., additional, Opel, A., additional, Daimi, H., additional, Haj Khelil, A., additional, Be Chibani, J., additional, Barana, A., additional, Amoros, I., additional, Gonzalez De La Fuente, M., additional, Caballero, R., additional, Aranega, A., additional, Kelly, A., additional, Bernus, O., additional, Kemi, O. J., additional, Myles, R. C., additional, Ghouri, I. A., additional, Burton, F. L., additional, Smith, G. L., additional, Del Lungo, M., additional, Sartiani, L., additional, Spinelli, V., additional, Baruscotti, M., additional, Difrancesco, D., additional, Mugelli, A., additional, Cerbai, E., additional, Thomas, A. M., additional, Aziz, Q., additional, Khambra, T., additional, Addlestone, J. M. A., additional, Cartwright, E. J., additional, Wilkinson, R., additional, Song, W., additional, Marston, S., additional, Jacquet, A., additional, Mougenot, N. M., additional, Lipskaia, A. J., additional, Paalberends, E. R., additional, Stam, K., additional, Van Dijk, S. J., additional, Van Slegtenhorst, M., additional, Dos Remedios, C., additional, Ten Cate, F. J., additional, Michels, M., additional, Niessen, H. W. M., additional, Stienen, G. J. M., additional, Van Der Velden, J., additional, Read, M. I., additional, Andreianova, A. A., additional, Harrison, J. C., additional, Goulton, C. S., additional, Kerr, D. S., additional, Sammut, I. A., additional, Wallner, M., additional, Von Lewinski, D., additional, Kindsvater, D., additional, Saes, M., additional, Morano, I., additional, Muegge, A., additional, Buyandelger, B., additional, Kostin, S., additional, Gunkel, S., additional, Vouffo, J., additional, Ng, K., additional, Chen, J., additional, Eilers, M., additional, Isaacson, R., additional, Milting, H., additional, Knoell, R., additional, Cattin, M.-E., additional, Crocini, C., additional, Schlossarek, S., additional, Maron, S., additional, Hansen, A., additional, Eschenhagen, T., additional, Carrier, L., additional, Bonne, G., additional, Coppini, R., additional, Ferrantini, C., additional, Olivotto, I., additional, Belardinelli, L., additional, Poggesi, C., additional, Leung, M. C., additional, Messer, A. E., additional, Copeland, O., additional, Marston, S. B., additional, Mills, A. M., additional, Collins, T., additional, O'gara, P., additional, Thum, T., additional, Regalla, K., additional, Macleod, K. T., additional, Prodromakis, T., additional, Chaudhry, U., additional, Darzi, A., additional, Yacoub, M. H., additional, Athanasiou, T., additional, Bogdanova, A., additional, Makhro, A., additional, Hoydal, M., additional, Stolen, T. O., additional, Johnssen, A. B., additional, Alves, M., additional, Catalucci, D., additional, Condorelli, G., additional, Koch, L. G., additional, Britton, S. L., additional, Wisloff, U., additional, Bito, V., additional, Claus, P., additional, Vermeulen, K., additional, Huysmans, C., additional, Ventura-Clapier, R., additional, Sipido, K. R., additional, Seliuk, M. N., additional, Burlaka, A. P., additional, Sidorik, E. P., additional, Khaitovych, N. V., additional, Kozachok, M. M., additional, Potaskalova, V. S., additional, Driesen, R. B., additional, Galan, D. T., additional, De Paulis, D., additional, Arnoux, T., additional, Schaller, S., additional, Pruss, R. M., additional, Poitz, D. M., additional, Augstein, A., additional, Braun-Dullaeus, R. C., additional, Schmeisser, A., additional, Strasser, R. H., additional, Micova, P., additional, Balkova, P., additional, Hlavackova, M., additional, Zurmanova, J., additional, Kasparova, D., additional, Kolar, F., additional, Neckar, J., additional, Novak, F., additional, Novakova, O., additional, Pollard, S., additional, Babba, M., additional, Hussain, A., additional, James, R., additional, Maddock, H., additional, Alshehri, A. S., additional, Baxter, G. F., additional, Dietel, B., additional, Altendorf, R., additional, Daniel, W. G., additional, Kollmar, R., additional, Garlichs, C. D., additional, Sirohi, R., additional, Roberts, N., additional, Lawrence, D., additional, Sheikh, A., additional, Kolvekar, S., additional, Yap, J., additional, Arend, M., additional, Walkinshaw, G., additional, Hausenloy, D. J., additional, Yellon, D. M., additional, Posa, A., additional, Szabo, R., additional, Szalai, Z., additional, Szablics, P., additional, Berko, M. A., additional, Orban, K., additional, Murlasits, Z. S., additional, Balogh, L., additional, Varga, C., additional, Ku, H. C., additional, Su, M. J., additional, Chreih, R.-M., additional, Ginghina, C., additional, Deleanu, D., additional, Ferreira, A. L. B. J., additional, Belal, A., additional, Ali, M. A., additional, Fan, X., additional, Holt, A., additional, Campbell, R., additional, Schulz, R., additional, Bonanad, C., additional, Bodi, V., additional, Sanchis, J., additional, Morales, J. M., additional, Marrachelli, V., additional, Nunez, J., additional, Forteza, M. J., additional, Chaustre, F., additional, Gomez, C., additional, Chorro, F. J., additional, Csont, T., additional, Fekete, V., additional, Murlasits, Z., additional, Aypar, E., additional, Bencsik, P., additional, Sarkozy, M., additional, Varga, Z. V., additional, Ferdinandy, P., additional, Duerr, G. D., additional, Zoerlein, M., additional, Dewald, D., additional, Mesenholl, B., additional, Schneider, P., additional, Ghanem, A., additional, Rittling, S., additional, Welz, A., additional, Dewald, O., additional, Becker, E., additional, Peigney, C., additional, Bouleti, C., additional, Galaup, A., additional, Monnot, C., additional, Ghaleh, B., additional, Germain, S., additional, Timmermans, A., additional, Ginion, A., additional, De Meester, C., additional, Sakamoto, K., additional, Vanoverschelde, J.-L., additional, Horman, S., additional, Beauloye, C., additional, Bertrand, L., additional, Maroz-Vadalazhskaya, N., additional, Drozd, E., additional, Kukharenko, L., additional, Russkich, I., additional, Krachak, D., additional, Seljun, Y., additional, Ostrovski, Y., additional, Martin, A.-C., additional, Le Bonniec, B., additional, Lecompte, T., additional, Dizier, B., additional, Emmerich, J., additional, Fischer, A.-M., additional, Samama, C.-M., additional, Godier, A., additional, Mogensen, S., additional, Furchtbauer, E. M., additional, Aalkjaer, C., additional, Choong, W. L., additional, Jovanovic, A., additional, Khan, F., additional, Daniel, J. M., additional, Dutzmann, J. M., additional, Widmer-Teske, R., additional, Guenduez, D., additional, Sedding, D., additional, Castro, M. M., additional, Cena, J. J. C., additional, Cho, W. J. C., additional, Goobie, G. G., additional, Walsh, M. P. W., additional, Schulz, R. S., additional, Dutzmann, J., additional, Preissner, K. T., additional, Sones, W., additional, Kotlikoff, M., additional, Serizawa, K., additional, Yogo, K., additional, Aizawa, K., additional, Hirata, M., additional, Tashiro, Y., additional, Ishizuka, N., additional, Varela, A., additional, Katsiboulas, M., additional, Tousoulis, D., additional, Papaioannou, T. G., additional, Vaina, S., additional, Davos, C. H., additional, Piperi, C., additional, Stefanadis, C., additional, Basdra, E. K., additional, Papavassiliou, A. G., additional, Hermenegildo, C., additional, Lazaro-Franco, M., additional, Sobrino, A., additional, Bueno-Beti, C., additional, Martinez-Gil, N., additional, Walther, T., additional, Peiro, C., additional, Sanchez-Ferrer, C. F., additional, Novella, S., additional, Ciccarelli, M., additional, Franco, A., additional, Dorn, G. W., additional, Cseplo, P., additional, Torok, O., additional, Springo, Z. S., additional, Vamos, Z., additional, Kosa, D., additional, Hamar, J., additional, Koller, A., additional, Bubb, K. J., additional, Ahluwalia, A., additional, Stepien, E. L., additional, Gruca, A., additional, Grzybowska, J., additional, Goralska, J., additional, Dembinska-Kiec, A., additional, Stolinski, J., additional, Partyka, L., additional, Zhang, H., additional, Sweeney, D., additional, Thomas, G. N., additional, Fish, P. V., additional, Taggart, D. P., additional, Cioffi, S., additional, Bilio, M., additional, Martucciello, S., additional, Illingworth, E., additional, Caporali, A., additional, Shantikumar, S., additional, Marchetti, M., additional, Martelli, F., additional, Emanueli, C., additional, Meloni, M., additional, Al Haj Zen, A., additional, Sala-Newby, G., additional, Del Turco, S., additional, Saponaro, C., additional, Dario, B., additional, Sartini, S., additional, Menciassi, A., additional, Dario, P., additional, La Motta, C., additional, Basta, G., additional, Santiemma, V., additional, Bertone, C., additional, Rossi, F., additional, Michelon, E., additional, Bianco, M. J., additional, Castelli, A., additional, Shin, D. I., additional, Seung, K. B., additional, Seo, S. M., additional, Park, H. J., additional, Kim, P. J., additional, Baek, S. H., additional, Choi, Y. S., additional, Her, S. H., additional, Kim, D. B., additional, Lee, J. M., additional, Park, C. S., additional, Rocchiccioli, S., additional, Cecchettini, A., additional, Pelosi, G., additional, Citti, L., additional, Parodi, O., additional, Trivella, M. G., additional, Michel-Monigadon, D., additional, Burger, F., additional, Dunoyer-Geindre, S., additional, Pelli, G., additional, Cravatt, B., additional, Steffens, S., additional, Didangelos, A., additional, Mayr, U., additional, Yin, X., additional, Stegemann, C., additional, Shalhoub, J., additional, Davies, A. H., additional, Monaco, C., additional, Mayr, M., additional, Lypovetska, S., additional, Grytsenko, S., additional, Njerve, I. U., additional, Pettersen, A. A., additional, Opstad, T. B., additional, Bratseth, V., additional, Arnesen, H., additional, Seljeflot, I., additional, Dumitriu, I. E., additional, Baruah, P., additional, Antunes, R. F., additional, Kaski, J. C., additional, Trapero, I., additional, Benet, I., additional, Alguero, C., additional, Chaustre, F. J., additional, Mangold, A., additional, Puthenkalam, S., additional, Distelmaier, K., additional, Adlbrecht, C., additional, Lang, I. M., additional, Koizumi, T., additional, Inoue, I., additional, Komiyama, N., additional, Nishimura, S., additional, Korneeva, O. N., additional, Drapkina, O. M., additional, Fornai, L., additional, Angelini, A., additional, Kiss, A., additional, Giskes, F., additional, Eijkel, G., additional, Fedrigo, M., additional, Valente, M. L., additional, Thiene, G., additional, Heeren, R. M. A., additional, Padro, T., additional, Casani, L., additional, Suades, R., additional, Bertoni, B., additional, Carminati, R., additional, Carlini, V., additional, Pettinari, L., additional, Martinelli, C., additional, Gagliano, N., additional, Noppe, G., additional, Buchlin, P., additional, Marquet, N., additional, Baeyens, N., additional, Morel, N., additional, Baysa, A., additional, Sagave, J., additional, Dahl, C. P., additional, Gullestad, L., additional, Carpi, A., additional, Di Lisa, F., additional, Giorgio, M., additional, Vaage, J., additional, Valen, G., additional, Vafiadaki, E., additional, Papalouka, V., additional, Terzis, G., additional, Spengos, K., additional, Manta, P., additional, Gales, C., additional, Genet, G., additional, Dague, E., additional, Cazorla, O., additional, Payre, B., additional, Mias, C., additional, Ouille, A., additional, Lacampagne, A., additional, Pathak, A., additional, Senard, J. M., additional, Abonnenc, M., additional, Da Costa Martins, P., additional, Srivastava, S., additional, Gautel, M., additional, De Windt, L., additional, Comelli, L., additional, Lande, C., additional, Ucciferri, N., additional, Ikonen, L., additional, Vuorenpaa, H., additional, Kujala, K., additional, Sarkanen, J.-R., additional, Heinonen, T., additional, Ylikomi, T., additional, Aalto-Setala, K., additional, Capros, H., additional, Sprincean, N., additional, Usurelu, N., additional, Egorov, V., additional, Stratu, N., additional, Matchkov, V., additional, Bouzinova, E., additional, Moeller-Nielsen, N., additional, Wiborg, O., additional, Gutierrez, P. S., additional, Aparecida-Silva, R., additional, Borges, L. F., additional, Moreira, L. F. P., additional, Dias, R. R., additional, Kalil, J., additional, Stolf, N. A. G., additional, Zhou, W., additional, Suntharalingam, K., additional, Brand, N., additional, Vilar Compte, R., additional, Ying, L., additional, Bicknell, K., additional, Dannoura, A., additional, Dash, P., additional, Brooks, G., additional, Tsimafeyeu, I., additional, Tishova, Y., additional, Wynn, N., additional, Oyeyipo, I. P., additional, Olatunji, L. A., additional, Maegdefessel, L., additional, Azuma, J., additional, Toh, R., additional, Raaz, U., additional, Merk, D. R., additional, Deng, A., additional, Spin, J. M., additional, Tsao, P. S., additional, Tedeschi, L., additional, Taranta, M., additional, Naldi, I., additional, Grimaldi, S., additional, Cinti, C., additional, Bousquenaud, M., additional, Maskali, F., additional, Poussier, S., additional, Marie, P. Y., additional, Boutley, H., additional, Karcher, G., additional, Wagner, D. R., additional, Devaux, Y., additional, Torre, I., additional, Psilodimitrakopoulos, S., additional, Iruretagoiena, I., additional, Gonzalez-Tendero, A., additional, Artigas, D., additional, Loza-Alvarez, P., additional, Gratacos, E., additional, Amat-Roldan, I., additional, Murray, L., additional, Carberry, D. M., additional, Dunton, P., additional, Miles, M. J., additional, Suleiman, M.-S., additional, Kanesalingam, K., additional, Taylor, R., additional, Mc Collum, C. N., additional, Parniczky, A., additional, Solymar, M., additional, Porpaczy, A., additional, Miseta, A., additional, Lenkey, Z. S., additional, Szabados, S., additional, Cziraki, A., additional, Garai, J., additional, Myloslavska, I., additional, Menazza, S. M., additional, Canton, M. C., additional, Di Lisa, F. D. L., additional, Oliveira, S. H. V., additional, Morais, C. A. S., additional, Miranda, M. R., additional, Oliveira, T. T., additional, Lamego, M. R. A., additional, Lima, L. M., additional, Goncharova, N. S., additional, Naymushin, A. V., additional, Kazimli, A. V., additional, Moiseeva, O. M., additional, Carvalho, M. G., additional, Sabino, A. P., additional, Mota, A. P. L., additional, Sousa, M. O., additional, Niessner, A., additional, Richter, B., additional, Hohensinner, P. J., additional, Rychli, K., additional, Zorn, G., additional, Berger, R., additional, Moertl, D., additional, Pacher, R., additional, Wojta, J., additional, Huelsmann, M., additional, Kukharchik, G., additional, Nesterova, N., additional, Pavlova, A., additional, Gaykovaya, L., additional, Krapivka, N., additional, Konstantinova, I., additional, Sichinava, L., additional, Prapa, S., additional, Mccarthy, K. P., additional, Kilner, P. J., additional, Xu, X. Y., additional, Johnson, M. R., additional, Ho, S. Y., additional, Gatzoulis, M. A., additional, Stoupel, E. G., additional, Garcia, R., additional, Merino, D., additional, Montalvo, C., additional, Hurle, M. A., additional, Nistal, J. F., additional, Villar, A. V., additional, Perez-Moreno, A., additional, Gilabert, R., additional, and Ros, E., additional
- Published
- 2012
- Full Text
- View/download PDF
10. GALECTIN-3 IS A POTENTIAL MEDIATOR OF ALDOSTERONE EFFECTS IN VASCULAR REMODELING
- Author
-
Calvier, L., primary, Martin-Fernãndez, B., additional, Lahera, V., additional, Zannad, F., additional, Cachofeiro, V., additional, Lacolley, P., additional, Rossignol, P., additional, and Lopez-Andrés, N., additional
- Published
- 2011
- Full Text
- View/download PDF
11. CARDIOTROPHIN-1 IS A NEW KEY PLAYER IN CARDIAC AND VASCULAR FIBROSIS AND IN CARDIOVASCULAR DYSFUNCTION BEYOND HYPERTENSION
- Author
-
Lopez-Andres, N., primary, Calvier, L., additional, Inigo, C., additional, Labat, C., additional, Sloboda, N., additional, Fortuno, M. A., additional, Diez, J., additional, Zannad, F., additional, Lacolley, P., additional, and Rossignol, P., additional
- Published
- 2011
- Full Text
- View/download PDF
12. CARDIOTROPHIN-1 IS A DETERMINANT OF ARTERIAL STIFFNESS AND THICKNESS IN RODENTS: 3A.03
- Author
-
Lopez-Andres, N, primary, Calvier, L, additional, Fortuno, MA, additional, Labat, C, additional, Sloboda, N, additional, Simon, G, additional, Marie, PY, additional, Diez, J, additional, Zannad, F, additional, Lacolley, P, additional, and Rossignol, P, additional
- Published
- 2010
- Full Text
- View/download PDF
13. Galectin-3 is a potential mediator of aldosterone effects in vascular remodeling
- Author
-
Calvier, L., Reboul, P., Martin-Fernandez, B., Lahera, V., Zannad, F., Cachofeiro, V., Lacolley, P., Rossignol, P., and Lopez-Andres, N.
- Published
- 2011
- Full Text
- View/download PDF
14. Cardiotrophin-1 is a determinant of arterial stiffness and thickness in rodents
- Author
-
Lopez-Andres, N., Fortuno, M. A., Calvier, L., Labat, C., Sloboda, N., Marie, P. Y., Diez, J., Lacolley, P., Zannad, F., and Rossignol, P.
15. Safety of Anti-Reelin Therapeutic Approaches for Chronic Inflammatory Diseases.
- Author
-
Calvier L, Alexander A, Marckx AT, Kounnas MZ, Durakoglugil M, and Herz J
- Subjects
- Animals, Mice, Cell Adhesion Molecules, Neuronal metabolism, Endothelial Cells metabolism, Extracellular Matrix Proteins metabolism, Nerve Tissue Proteins metabolism, Serine Endopeptidases metabolism, Inflammation drug therapy, Reelin Protein antagonists & inhibitors, Encephalomyelitis, Autoimmune, Experimental drug therapy, Anti-Inflammatory Agents therapeutic use
- Abstract
Reelin, a large extracellular glycoprotein, plays critical roles in neuronal development and synaptic plasticity in the central nervous system (CNS). Recent studies have revealed non-neuronal functions of plasma Reelin in inflammation by promoting endothelial-leukocyte adhesion through its canonical pathway in endothelial cells (via ApoER2 acting on NF-κB), as well as in vascular tone regulation and thrombosis. In this study, we have investigated the safety and efficacy of selectively depleting plasma Reelin as a potential therapeutic strategy for chronic inflammatory diseases. We found that Reelin expression remains stable throughout adulthood and that peripheral anti-Reelin antibody treatment with CR-50 efficiently depletes plasma Reelin without affecting its levels or functionality within the CNS. Notably, this approach preserves essential neuronal functions and synaptic plasticity. Furthermore, in mice induced with experimental autoimmune encephalomyelitis (EAE), selective modulation of endothelial responses by anti-Reelin antibodies reduces pathological leukocyte infiltration without completely abolishing diapedesis. Finally, long-term Reelin depletion under metabolic stress induced by a Western diet did not negatively impact the heart, kidney, or liver, suggesting a favorable safety profile. These findings underscore the promising role of peripheral anti-Reelin therapeutic strategies for autoimmune diseases and conditions where endothelial function is compromised, offering a novel approach that may avoid the immunosuppressive side effects associated with conventional anti-inflammatory therapies.
- Published
- 2024
- Full Text
- View/download PDF
16. Circulating Reelin promotes inflammation and modulates disease activity in acute and long COVID-19 cases.
- Author
-
Calvier L, Drelich A, Hsu J, Tseng CT, Mina Y, Nath A, Kounnas MZ, and Herz J
- Subjects
- Mice, Animals, Humans, Intercellular Adhesion Molecule-1, Post-Acute COVID-19 Syndrome, SARS-CoV-2, Chemokine CCL4, Chemokine CXCL10, Interleukin-4, Inflammation, COVID-19
- Abstract
Thromboembolic complications and excessive inflammation are frequent in severe COVID-19, potentially leading to long COVID. In non-COVID studies, we and others demonstrated that circulating Reelin promotes leukocyte infiltration and thrombosis. Thus, we hypothesized that Reelin participates in endothelial dysfunction and hyperinflammation during COVID-19. We showed that Reelin was increased in COVID-19 patients and correlated with the disease activity. In the severe COVID-19 group, we observed a hyperinflammatory state, as judged by increased concentration of cytokines (IL-1α, IL-4, IL-6, IL-10 and IL-17A), chemokines (IP-10 and MIP-1β), and adhesion markers (E-selectin and ICAM-1). Reelin level was correlated with IL-1α, IL-4, IP-10, MIP-1β, and ICAM-1, suggesting a specific role for Reelin in COVID-19 progression. Furthermore, Reelin and all of the inflammatory markers aforementioned returned to normal in a long COVID cohort, showing that the hyperinflammatory state was resolved. Finally, we tested Reelin inhibition with the anti-Reelin antibody CR-50 in hACE2 transgenic mice infected with SARS-CoV-2. CR-50 prophylactic treatment decreased mortality and disease severity in this model. These results demonstrate a direct proinflammatory function for Reelin in COVID-19 and identify it as a drug target. This work opens translational clinical applications in severe SARS-CoV-2 infection and beyond in auto-inflammatory diseases., Competing Interests: LC, MK, and JHE are co-shareholders of Reelin Therapeutics, Inc. LC and JHE are co-inventors of a patent related to anti-Reelin strategies application Number: 15/763,047 and Publication Number: 20180273637. MK was employed by the company Reelin Therapeutics. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Calvier, Drelich, Hsu, Tseng, Mina, Nath, Kounnas and Herz.)
- Published
- 2023
- Full Text
- View/download PDF
17. Reelin through the years: From brain development to inflammation.
- Author
-
Alexander A, Herz J, and Calvier L
- Subjects
- Humans, Brain metabolism, Inflammation, LDL-Receptor Related Proteins metabolism, Nerve Tissue Proteins metabolism, Receptors, LDL metabolism, Serine Endopeptidases metabolism, Cell Adhesion Molecules, Neuronal metabolism, Extracellular Matrix Proteins metabolism, Reelin Protein
- Abstract
Reelin was originally identified as a regulator of neuronal migration and synaptic function, but its non-neuronal functions have received far less attention. Reelin participates in organ development and physiological functions in various tissues, but it is also dysregulated in some diseases. In the cardiovascular system, Reelin is abundant in the blood, where it contributes to platelet adhesion and coagulation, as well as vascular adhesion and permeability of leukocytes. It is a pro-inflammatory and pro-thrombotic factor with important implications for autoinflammatory and autoimmune diseases such as multiple sclerosis, Alzheimer's disease, arthritis, atherosclerosis, or cancer. Mechanistically, Reelin is a large secreted glycoprotein that binds to several membrane receptors, including ApoER2, VLDLR, integrins, and ephrins. Reelin signaling depends on the cell type but mostly involves phosphorylation of NF-κB, PI3K, AKT, or JAK/STAT. This review focuses on non-neuronal functions and the therapeutic potential of Reelin, while highlighting secretion, signaling, and functional similarities between cell types., Competing Interests: Declaration of interests L.C. and J.H. are co-shareholders of Reelin Therapeutics, Inc. and coinventors of a patent related to anti-Reelin strategies (application 15/763,047 and publication 20180273637)., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. The "6B" strategy: Build Back a Better Blood-Brain Barrier.
- Author
-
Calvier L, Alexander AE, and Herz J
- Abstract
Under pathological conditions like multiple sclerosis (MS), leukocytes infiltrate the central nervous system where they, in concert with activated microglia, promote inflammatory demyelination resulting in a broad spectrum of symptoms including paralysis. Therefore, all current therapeutic approaches to MS target the immune system, blocking inflammation and paralysis progression, but may compromise the immune system. In this focused review, we present an underestimated compartment, the blood-brain barrier, which is compromised during MS and becomes permeable to leukocytes infiltrating the central nervous system. This barrier has the potential to offer new therapeutic strategies and is easily accessible for drugs. We highlight this paradigm using the example of the therapeutic anti-Reelin strategy we have developed. Reelin is a plasma protein that regulates the expression of adhesion markers on the endothelial surface, thus promoting the infiltration of inflammatory cells and propagating inflammation. Building Back a Better Blood-Brain Barrier (the "6B" strategy) may have advantages compared to actual immunosuppressive drugs because it restores a physiological function rather than suppressing the immune system., Competing Interests: DECLARATION OF INTERESTS L.C. and J.H. are co-shareholders of Reelin Therapeutics Inc. and coinventors of a patent related to anti-Reelin strategies (Application Number: 15/763,047 and Publication Number: 20180273637).
- Published
- 2022
- Full Text
- View/download PDF
19. Editorial: New Advances in Cardiorenal Syndrome.
- Author
-
Calvier L, Kökény G, and Martinez-Martinez E
- Published
- 2022
- Full Text
- View/download PDF
20. Interplay of Low-Density Lipoprotein Receptors, LRPs, and Lipoproteins in Pulmonary Hypertension.
- Author
-
Calvier L, Herz J, and Hansmann G
- Abstract
The low-density lipoprotein receptor (LDLR) gene family includes LDLR, very LDLR, and LDL receptor-related proteins (LRPs) such as LRP1, LRP1b (aka LRP-DIT), LRP2 (aka megalin), LRP4, and LRP5/6, and LRP8 (aka ApoER2). LDLR family members constitute a class of closely related multifunctional, transmembrane receptors, with diverse functions, from embryonic development to cancer, lipid metabolism, and cardiovascular homeostasis. While LDLR family members have been studied extensively in the systemic circulation in the context of atherosclerosis, their roles in pulmonary arterial hypertension (PAH) are understudied and largely unknown. Endothelial dysfunction, tissue infiltration of monocytes, and proliferation of pulmonary artery smooth muscle cells are hallmarks of PAH, leading to vascular remodeling, obliteration, increased pulmonary vascular resistance, heart failure, and death. LDLR family members are entangled with the aforementioned detrimental processes by controlling many pathways that are dysregulated in PAH; these include lipid metabolism and oxidation, but also platelet-derived growth factor, transforming growth factor β1, Wnt, apolipoprotein E, bone morpohogenetic proteins, and peroxisome proliferator-activated receptor gamma. In this paper, we discuss the current knowledge on LDLR family members in PAH. We also review mechanisms and drugs discovered in biological contexts and diseases other than PAH that are likely very relevant in the hypertensive pulmonary vasculature and the future care of patients with PAH or other chronic, progressive, debilitating cardiovascular diseases., Competing Interests: Dr Herz was supported by grants from the National Heart, Lung, and Blood Institute (R37 HL063762), the National Institute on Aging (RF AG053391), the National Institute on Neurological Disorders and Stroke and National Institute on Aging (R01 NS093382), and BrightFocus (A2016396S); the Bluefield Project to Cure FTD; and a Harrington Scholar Innovator Award (2019). Dr Hansmann has received financial support from the German Research Foundation (HA4348/2-2 and HA4348/6-2 KFO311), the Federal Ministry of Education and Research (BMBF ViP+ program-03VP08053; BMBF 01KC2001B), and the European Pediatric Pulmonary Vascular Disease Network. Dr Calvier and Dr Herz are shareholders of Reelin Therapeutics and co-inventors of a patent related to anti-Reelin strategies (application number 15/763,047 and publication number 20180273637). Dr Hansmann has reported that he has no relationships relevant to the contents of this paper to disclose., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
21. PPARγ and TGFβ-Major Regulators of Metabolism, Inflammation, and Fibrosis in the Lungs and Kidneys.
- Author
-
Kökény G, Calvier L, and Hansmann G
- Subjects
- Animals, Humans, PPAR gamma agonists, Pioglitazone therapeutic use, Pulmonary Arterial Hypertension drug therapy, Pulmonary Arterial Hypertension metabolism, Pulmonary Fibrosis drug therapy, Renal Insufficiency drug therapy, Kidney metabolism, Lung metabolism, PPAR gamma metabolism, Pulmonary Fibrosis metabolism, Renal Insufficiency metabolism, Signal Transduction, Transforming Growth Factor beta metabolism
- Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a type II nuclear receptor, initially recognized in adipose tissue for its role in fatty acid storage and glucose metabolism. It promotes lipid uptake and adipogenesis by increasing insulin sensitivity and adiponectin release. Later, PPARγ was implicated in cardiac development and in critical conditions such as pulmonary arterial hypertension (PAH) and kidney failure. Recently, a cluster of different papers linked PPARγ signaling with another superfamily, the transforming growth factor beta (TGFβ), and its receptors, all of which play a major role in PAH and kidney failure. TGFβ is a multifunctional cytokine that drives inflammation, fibrosis, and cell differentiation while PPARγ activation reverses these adverse events in many models. Such opposite biological effects emphasize the delicate balance and complex crosstalk between PPARγ and TGFβ. Based on solid experimental and clinical evidence, the present review summarizes connections and their implications for PAH and kidney failure, highlighting the similarities and differences between lung and kidney mechanisms as well as discussing the therapeutic potential of PPARγ agonist pioglitazone.
- Published
- 2021
- Full Text
- View/download PDF
22. Reelin changes hippocampal learning in aging and Alzheimer's disease.
- Author
-
Marckx AT, Fritschle KE, Calvier L, and Herz J
- Subjects
- Age Factors, Animals, Disease Models, Animal, Maze Learning physiology, Mice, Mice, 129 Strain, Mice, Knockout, Mice, Transgenic, Reelin Protein deficiency, Aging physiology, Alzheimer Disease metabolism, Alzheimer Disease physiopathology, Cognitive Dysfunction metabolism, Cognitive Dysfunction physiopathology, Hippocampus metabolism, Hippocampus physiopathology, Reelin Protein metabolism, tau Proteins metabolism
- Abstract
The hippocampal formation (HF) is a neuroanatomical region essential for learning and memory. As one of the earliest regions to display the histopathological hallmarks of Alzheimer's disease (AD), determining the specific mechanisms of the HF's vulnerability is of capital importance. Reelin, a glycoprotein crucial in cortical lamination during embryonic neurogenesis, has an uncommon expression pattern within the HF and has been implicated in both learning and AD pathogenesis. We hypothesized that Reelin deficiency would expedite behavioral impairments which accompany normal aging. Additionally, we hypothesized that Reelin deficiency in the presence of mutated human microtubule associated protein tau (MAPT) would further impair hippocampal function. To test our hypothesis, we utilized cohorts of aged mice, aged mice with Reelin conditional knockout (RcKO), and adult mice with both RcKO and MAPT in the Barnes maze and Trace fear conditioning. Consistent with prior literature, increased age in wild-type mice was sufficient to reduce spatial searching in the Barnes maze. Increased age both exacerbated spatial impairments and altered context learning in RcKO mice. Lastly, adult mice with both RcKO and the MAPT transgene displayed both the lowest age-of-onset and most severe spatial learning deficits. In conclusion, Reelin deficiency when combined with AD risk-factors produced consistent impairments in spatial memory tasks. Furthermore, our results further implicate Reelin's importance in both HF homeostasis and AD pathogenesis., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Apolipoprotein E receptor 2 deficiency decreases endothelial adhesion of monocytes and protects against autoimmune encephalomyelitis.
- Author
-
Calvier L, Manouchehri N, Sacharidou A, Mineo C, Shaul PW, Hui DY, Kounnas MZ, Stüve O, and Herz J
- Subjects
- Animals, Cell Adhesion immunology, LDL-Receptor Related Proteins deficiency, Male, Mice, Mice, Knockout, Encephalomyelitis, Autoimmune, Experimental immunology, Endothelium, Vascular immunology, LDL-Receptor Related Proteins immunology, Monocytes immunology
- Abstract
Under normal conditions, the blood-brain barrier effectively regulates the passage of immune cells into the central nervous system (CNS). However, under pathological conditions such as multiple sclerosis (MS), leukocytes, especially monocytes, infiltrate the CNS where they promote inflammatory demyelination, resulting in paralysis. Therapies targeting the immune cells directly and preventing leukocyte infiltration exist for MS but may compromise the immune system. Here, we explore how apolipoprotein E receptor 2 (ApoER2) regulates vascular adhesion and infiltration of monocytes during inflammation. We induced experimental autoimmune encephalitis in ApoER2 knockout mice and in mice carrying a loss-of-function mutation in the ApoER2 cytoplasmic domain. In both models, paralysis and neuroinflammation were largely abolished as a result of greatly diminished monocyte adherence due to reduced expression of adhesion molecules on the endothelial surface. Our findings expand our mechanistic understanding of the vascular barrier, the regulation of inflammation and vascular permeability, and the therapeutic potential of ApoER2-targeted therapies., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
24. CD11c + CD88 + CD317 + myeloid cells are critical mediators of persistent CNS autoimmunity.
- Author
-
Manouchehri N, Hussain RZ, Cravens PD, Esaulova E, Artyomov MN, Edelson BT, Wu GF, Cross AH, Doelger R, Loof N, Eagar TN, Forsthuber TG, Calvier L, Herz J, and Stüve O
- Subjects
- Animals, Antigen Presentation, Cells, Cultured, Central Nervous System immunology, Central Nervous System metabolism, Female, Humans, Male, Mice, Microglia metabolism, Antigens, CD metabolism, Encephalomyelitis, Autoimmune, Experimental immunology, Integrin alpha4 metabolism, Myeloid Cells metabolism
- Abstract
Natalizumab, a humanized monoclonal antibody (mAb) against α4-integrin, reduces the number of dendritic cells (DC) in cerebral perivascular spaces in multiple sclerosis (MS). Selective deletion of α4-integrin in CD11c
+ cells should curtail their migration to the central nervous system (CNS) and ameliorate experimental autoimmune encephalomyelitis (EAE). We generated CD11c.Cre+/- ITGA4fl/fl C57BL/6 mice to selectively delete α4-integrin in CD11c+ cells. Active immunization and adoptive transfer EAE models were employed and compared with WT controls. Multiparameter flow cytometry was utilized to immunophenotype leukocyte subsets. Single-cell RNA sequencing was used to profile individual cells. α4-Integrin expression by CD11c+ cells was significantly reduced in primary and secondary lymphoid organs in CD11c.Cre+/- ITGA4fl/fl mice. In active EAE, a delayed disease onset was observed in CD11c.Cre+/- ITGA4fl/fl mice, during which CD11c+ CD88+ cells were sequestered in the blood. Upon clinical EAE onset, CD11c+ CD88+ cells appeared in the CNS and expressed CD317+ In adoptive transfer experiments, CD11c.Cre+/- ITGA4fl/fl mice had ameliorated clinical disease phenotype associated with significantly diminished numbers of CNS CD11c+ CD88+ CD317+ cells. In human cerebrospinal fluid from subjects with neuroinflammation, microglia-like cells display coincident expression of ITGAX (CD11c), C5AR1 (CD88), and BST2 (CD317). In mice, we show that only activated, but not naïve microglia expressed CD11c, CD88, and CD317. Finally, anti-CD317 treatment prior to clinical EAE substantially enhanced recovery in mice., Competing Interests: Competing interest statement: O.S. serves on the editorial boards of Therapeutic Advances in Neurological Disorders. He has served on data monitoring committees for Genentech-Roche, Pfizer, and TG Therapeutics without monetary compensation. O.S. has advised EMD Serono, Celgene, Genentech, TG Therapeutics, and Genzyme. He currently receives grant support from Sanofi Genzyme and EMD Serono. G.F.W. has received honoraria for consulting for Novartis and Genentech, and received research funding from Biogen, EMD Serono, and Roche. A.H.C. has received honoraria for consulting for Biogen, Celgene, EMD Serono, Genentech, Novartis, Roche, and TG Therapeutics; she has received compensation for speaking for Genentech., (Copyright © 2021 the Author(s). Published by PNAS.)- Published
- 2021
- Full Text
- View/download PDF
25. Reelin Depletion Protects Against Atherosclerosis by Decreasing Vascular Adhesion of Leukocytes.
- Author
-
Calvier L, Xian X, Lee RG, Sacharidou A, Mineo C, Shaul PW, Kounnas MZ, Tsai S, and Herz J
- Subjects
- Animals, Atherosclerosis genetics, Atherosclerosis immunology, Atherosclerosis metabolism, CX3C Chemokine Receptor 1 genetics, Cell Adhesion Molecules, Neuronal deficiency, Cell Adhesion Molecules, Neuronal genetics, Coculture Techniques, Disease Models, Animal, Endothelial Cells immunology, Endothelial Cells metabolism, Extracellular Matrix Proteins deficiency, Extracellular Matrix Proteins genetics, Female, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, LDL-Receptor Related Proteins metabolism, Leukocytes immunology, Leukocytes metabolism, Male, Mice, Transgenic, Nerve Tissue Proteins deficiency, Nerve Tissue Proteins genetics, Plaque, Atherosclerotic, Receptors, LDL deficiency, Receptors, LDL genetics, Reelin Protein, Serine Endopeptidases deficiency, Serine Endopeptidases genetics, Signal Transduction, U937 Cells, Antibodies, Neutralizing pharmacology, Atherosclerosis prevention & control, Cell Adhesion drug effects, Cell Adhesion Molecules, Neuronal antagonists & inhibitors, Endothelial Cells drug effects, Extracellular Matrix Proteins antagonists & inhibitors, Leukocyte Rolling drug effects, Leukocytes drug effects, Nerve Tissue Proteins antagonists & inhibitors, Oligonucleotides, Antisense pharmacology
- Abstract
[Figure: see text].
- Published
- 2021
- Full Text
- View/download PDF
26. Reelin depletion protects against autoimmune encephalomyelitis by decreasing vascular adhesion of leukocytes.
- Author
-
Calvier L, Demuth G, Manouchehri N, Wong C, Sacharidou A, Mineo C, Shaul PW, Monson NL, Kounnas MZ, Stüve O, and Herz J
- Subjects
- Animals, Central Nervous System, Humans, Leukocytes, Mice, Mice, Inbred C57BL, Reelin Protein, Encephalomyelitis, Encephalomyelitis, Autoimmune, Experimental, Multiple Sclerosis
- Abstract
Neuroinflammation as a result of immune cell recruitment into the central nervous system (CNS) is a key pathogenic mechanism of multiple sclerosis (MS). However, current anti-inflammatory interventions depleting immune cells or directly targeting their trafficking into the CNS can have serious side effects, highlighting a need for better immunomodulatory strategies. We detected increased Reelin concentrations in the serum of patients with MS, resulting in increased endothelial permeability to leukocytes through increased nuclear factor κB-mediated expression of vascular adhesion molecules. We thus investigated the prophylactic and therapeutic potential of Reelin immunodepletion in experimental autoimmune encephalomyelitis (EAE) and further validated the results in Reelin knockout mice. Removal of plasma Reelin by either approach protected against neuroinflammation and largely abolished the neurological consequences by reducing endothelial permeability and immune cell accumulation in the CNS. Our findings suggest Reelin depletion as a therapeutic approach with an inherent good safety margin for the treatment of MS and other diseases where leukocyte extravasation is a major driver of pathogenicity., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
27. PPARγ is a gatekeeper for extracellular matrix and vascular cell homeostasis: beneficial role in pulmonary hypertension and renal/cardiac/pulmonary fibrosis.
- Author
-
Kökény G, Calvier L, Legchenko E, Chouvarine P, Mózes MM, and Hansmann G
- Subjects
- Animals, Bone Morphogenetic Protein Receptors, Type II physiology, DNA Damage, Fibrosis, Homeostasis, Humans, PPAR gamma agonists, Transforming Growth Factor beta1 physiology, Extracellular Matrix physiology, Hypertension, Pulmonary etiology, Kidney pathology, Muscle, Smooth, Vascular cytology, Myocardium pathology, Myocytes, Smooth Muscle physiology, PPAR gamma physiology, Pulmonary Fibrosis etiology
- Abstract
Purpose of Review: Pulmonary arterial hypertension (PAH) is characterized by pulmonary arterial endothelial cell (PAEC) dysfunction and apoptosis, pulmonary arterial smooth muscle cell (PASMC) proliferation, inflammation, vasoconstriction, and metabolic disturbances that include disrupted bone morphogenetic protein receptor (BMPR2)-peroxisome proliferator-activated receptor gamma (PPARγ) axis and DNA damage. Activation of PPARγ improves many of these mechanisms, although erroneous reports on potential adverse effects of thiazolidinedione (TZD)-class PPARγ agonists reduced their clinical use in the past decade. Here, we review recent findings in heart, lung, and kidney research related to the pathobiology of vascular remodeling and tissue fibrosis, and also potential therapeutic effects of the PPARγ agonist pioglitazone., Recent Findings: Independent of its metabolic effects (improved insulin sensitivity and fatty acid handling), PPARγ activation rescues BMPR2 dysfunction, inhibits TGFβ/Smad3/CTGF and TGFβ/pSTAT3/pFoxO1 pathways, and induces the PPARγ/apoE axis, inhibiting vascular remodeling. PPARγ activation dampens mtDNA damage via PPARγ/UBR5/ATM pathway, improves function of endothelial progenitor cells (EPCs), and decrease renal fibrosis by repressing TGFβ/pSTAT3 and TGFβ/EGR1., Summary: Pharmacological PPARγ activation improves many hallmarks of PAH, including dysfunction of BMPR2-PPARγ axis, PAEC, PASMC, EPC, mitochondria/metabolism, and inflammation. Recent randomized controlled trials, including IRIS (Insulin Resistance Intervention After Stroke Trial), emphasize the beneficial effects of PPARγ agonists in PAH patients, leading to recent revival for clinical use.
- Published
- 2020
- Full Text
- View/download PDF
28. Activation of the Metabolic Master Regulator PPARγ: A Potential PIOneering Therapy for Pulmonary Arterial Hypertension.
- Author
-
Hansmann G, Calvier L, Risbano MG, and Chan SY
- Subjects
- Animals, Humans, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary metabolism, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular metabolism, Thiazolidinediones pharmacology, Familial Primary Pulmonary Hypertension drug therapy, PPAR gamma metabolism, Pulmonary Arterial Hypertension drug therapy
- Abstract
Translational research is essential to the development of reverse-remodeling strategies for the treatment of pulmonary vascular disease, pulmonary hypertension, and heart failure via mechanistic in vivo studies using animal models resembling human pulmonary arterial hypertension (PAH), cardiovascular remodeling, and progressive right heart failure. Since 2007, peroxisome proliferator-activated receptor γ (PPARγ) agonists have emerged as promising novel, antiproliferative, antiinflammatory, insulin-sensitizing, efficient medications for the treatment of PAH. However, early diabetes study results, their subsequent misinterpretations, errors in published review articles, and rumors regarding potential adverse effects in the literature have dampened enthusiasm for considering pharmacological PPARγ activation for the treatment of cardiovascular diseases, including PAH. Most recently, the thiazolidinedione class PPARγ agonist pioglitazone underwent a clinical revival, especially based on the IRIS (Insulin Resistance Intervention After Stroke) study, a randomized controlled trial in 3,876 patients without diabetes status post-transient ischemic attack/ischemic stroke who were clinically followed for 4.8 years. We discuss preclinical basic translational findings and randomized controlled trials related to the beneficial and adverse effects of PPARγ agonists of the thiazolidinedione class, with a particular focus on the last 5 years. The objective is a data-driven approach to set the preclinical and clinical study record straight. The convincing recent clinical trial data on the lack of significant toxicity in high-risk populations justify the timely conduct of clinical studies to achieve "repurposing" or "repositioning" of pioglitazone for the treatment of clinical PAH.
- Published
- 2020
- Full Text
- View/download PDF
29. The PPARγ agonist pioglitazone prevents TGF-β induced renal fibrosis by repressing EGR-1 and STAT3.
- Author
-
Németh Á, Mózes MM, Calvier L, Hansmann G, and Kökény G
- Subjects
- Animals, Early Growth Response Protein 1 metabolism, Fibrosis, Kidney Diseases chemically induced, Kidney Diseases metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Pioglitazone pharmacology, STAT3 Transcription Factor metabolism, Transforming Growth Factor beta toxicity, Early Growth Response Protein 1 antagonists & inhibitors, Kidney Diseases prevention & control, PPAR gamma agonists, Pioglitazone therapeutic use, STAT3 Transcription Factor antagonists & inhibitors, Transforming Growth Factor beta antagonists & inhibitors
- Abstract
Background: It has been proposed that peroxisome proliferator-activated receptor-γ (PPARγ) agonists might reduce renal fibrosis, however, several studies had contradictory results. Moreover, the possible interaction of TGF-β
1 , PPARγ, and transcription factors in renal fibrosis have not been investigated. We hypothesized that oral pioglitazone treatment would inhibit TGF-β-driven renal fibrosis and its progression, by modulating profibrotic transcription factors in TGF-β1 transgenic mice., Methods: Male C57Bl/6 J mice (control, CTL, n = 14) and TGF-β overexpressing transgenic mice (TGFβ, n = 14, having elevated plasma TGF-β1 level) were divided in two sets at 10 weeks of age. Mice in the first set were fed with regular rodent chow (CTL and TGFβ, n = 7/group). Mice in the second set were fed with chow containing pioglitazone (at a dose of 20 mg/kg/day, CTL + Pio and TGFβ+Pio, n = 7/group). After 5 weeks of treatment, blood pressure was assessed and urine samples were collected, and the kidneys were analyzed for histology, mRNA and protein expression., Results: TGF-β1 induced glomerulosclerosis and tubulointerstitial damage were significantly reduced by pioglitazone. Pioglitazone inhibited renal mRNA expression of all the profibrotic effectors: type-III collagen, TGF-β1 , CTGF and TIMP-1, and alike transcription factors cFos/cJun and protein expression of EGR-1, and STAT3 protein phosphorylation., Conclusions: Oral administration of PPARγ agonist pioglitazone significantly reduces TGF-β1 -driven renal fibrosis, via the attenuation of EGR-1, STAT3 and AP-1. This implies that PPARγ agonists might be effective in the treatment of chronic kidney disease patients.- Published
- 2019
- Full Text
- View/download PDF
30. Chronic TGF-β1 Signaling in Pulmonary Arterial Hypertension Induces Sustained Canonical Smad3 Pathways in Vascular Smooth Muscle Cells.
- Author
-
Calvier L, Chouvarine P, Legchenko E, Kokeny G, Mozes MM, and Hansmann G
- Subjects
- Animals, Humans, Male, Mice, Transgenic, Rats, Sprague-Dawley, Muscle, Smooth, Vascular pathology, Myocytes, Smooth Muscle metabolism, Pulmonary Arterial Hypertension metabolism, Signal Transduction, Smad3 Protein metabolism, Transforming Growth Factor beta1 metabolism
- Published
- 2019
- Full Text
- View/download PDF
31. LRP1 Deficiency in Vascular SMC Leads to Pulmonary Arterial Hypertension That Is Reversed by PPARγ Activation.
- Author
-
Calvier L, Boucher P, Herz J, and Hansmann G
- Subjects
- Animals, Cells, Cultured, Female, Humans, Low Density Lipoprotein Receptor-Related Protein-1 genetics, Male, Mice, Mice, Knockout, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular pathology, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle pathology, Pulmonary Arterial Hypertension genetics, Pulmonary Arterial Hypertension pathology, Random Allocation, Transforming Growth Factor beta1 pharmacology, Vascular Remodeling drug effects, Vascular Remodeling physiology, Low Density Lipoprotein Receptor-Related Protein-1 deficiency, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, PPAR gamma metabolism, Pulmonary Arterial Hypertension metabolism
- Abstract
Rationale: Arterial remodeling-a hallmark of many cardiovascular pathologies including pulmonary arterial hypertension (PAH)-is regulated by TGFβ1 (transforming growth factor-β1)-TGFβ receptors and the antagonistic, vasoprotective BMPR2 (bone morphogenetic protein receptor 2)-PPARγ (peroxisome proliferator-activated receptor-γ) axis. However, it is unclear which factors drive detrimental TGFβ1 pathways in the hypertensive pulmonary vasculature., Objective: We hypothesized that LRP1 (low-density lipoprotein receptor-related protein 1) expression is decreased in PAH, leading to enhancement (disinhibition) of TGFβ1 signals and that the PPARγ agonist pioglitazone can restore vascular homeostasis and prevent PAH resulting from LRP1 deletion in vascular smooth muscle cells (SMCs)., Methods and Results: Targeted deletion of LRP1 in vascular SMC (smLRP1
-/- ) in mice disinhibited TGFβ1-CTGF (connective tissue growth factor) signaling, leading to spontaneous PAH and distal pulmonary arterial muscularization as assessed by closed-chest cardiac catheterization and anti-αSMA staining. Pioglitazone inhibited the canonical TGFβ1-CTGF axis in human pulmonary artery SMC and smLRP1-/- main pulmonary artery (CTGF and NOX4) and reversed PAH in smLRP1-/- mice. TGFβ1 boosted pSmad3 in PASMC from smLRP1-/- mice versus controls. Pioglitazone-activated PPARγ binds to Smad3 in human pulmonary artery SMC (coimmunoprecipitation), thereby blocking its phosphorylation and overriding LRP1 deficiency. Finally, mRNA and protein expression of LRP1 was decreased in pulmonary plexiform lesions of patients with end-stage idiopathic PAH (laser capture microdissection, qPCR, and immunohistochemistry). Downregulation of LRP1 protein was also demonstrated in explanted PASMC from patients with PAH and accompanied by enhanced TGFβ1-pSmad3-CTGF signaling and increased TGFβ1-induced PASMC proliferation that was prevented by pioglitazone., Conclusions: Here, we identify LRP1 as an integrator of TGFβ1-mediated mechanisms that regulate vascular remodeling in mice and clinical PAH and PPARγ as a therapeutic target that controls canonical TGFβ1 pathways. Hence, pharmacologic PPARγ activation represents a promising new therapy for patients with PAH who lack the vasoprotective LRP1 in vascular SMC.- Published
- 2019
- Full Text
- View/download PDF
32. Galectin-3 pharmacological inhibition attenuates early renal damage in spontaneously hypertensive rats.
- Author
-
Martínez-Martínez E, Ibarrola J, Fernández-Celis A, Calvier L, Leroy C, Cachofeiro V, Rossignol P, and López-Andrés N
- Subjects
- Actins metabolism, Acute Kidney Injury, Acute-Phase Proteins urine, Albuminuria drug therapy, Animals, Blood Pressure, Cell Line, Chemokine CCL2 metabolism, Collagen Type I metabolism, Connective Tissue Growth Factor metabolism, Creatinine blood, Epithelial-Mesenchymal Transition drug effects, Fibronectins metabolism, Fibrosis, Hypertension complications, Kidney Diseases etiology, Kidney Diseases pathology, Lipocalin-2, Lipocalins blood, Lipocalins urine, Male, Organ Size, Osteopontin metabolism, Proto-Oncogene Proteins blood, Proto-Oncogene Proteins urine, Rats, Rats, Inbred SHR, Transforming Growth Factor beta metabolism, Up-Regulation, beta Catenin metabolism, Antigens, CD metabolism, Galectin 3 antagonists & inhibitors, Hypertension drug therapy, Kidney pathology, Kidney Diseases prevention & control, Pectins pharmacology
- Abstract
Background: The pharmacological blockade of galectin-3 (Gal-3), a β-galactoside-binding lectin, reduces renal impairment in acute kidney injury, hyperaldosteronism or nephropathy. We herein investigated the effects of pharmacological Gal-3 inhibition by modified citrus pectin (MCP) in renal damage in spontaneously hypertensive rats (SHRs)., Methods and Results: Gal-3 inhibition did not modify blood pressure levels in 30-week-old SHR. Kidney weight was higher in SHR, with no effect of MCP treatment (100 mg/kg/day in the drinking water). Plasma creatinine and albuminuria were slightly but significantly increased in SHR and reduced by MCP, as well as plasma and urinary neutrophil gelatinase-associated lipocalin. In kidney from SHR, Gal-3 was upregulated, as well as the fibrotic markers (collagen type I, TGF-β and connective tissue growth factor) and tubulointerstitial fibrosis. MCP treatment reduced Gal-3 levels and fibrosis. The epithelial-mesenchymal transition (EMT) molecules (fibronectin, α-smooth muscle actin and β-catenin) were modified in SHR and normalized by Gal-3 inhibition. The inflammatory mediators (monocyte chemoattractant protein-1, osteopontin, cd68, cd80, cd44 and cd45) were elevated in SHR and attenuated by MCP. Renal damage markers (neutrophil gelatinase-associated lipocalin and kidney injury molecule-1) were augmented in SHR and improved by MCP. In renal epithelial normal rat kidney-52E cells, Gal-3 treatment induced EMT markers, whereas Gal-3 silencing attenuated EMT., Conclusion: Gal-3 inhibition attenuated early renal damage in SHR as indicated by reduced albuminuria, improved renal function and decreased renal fibrosis, EMT and inflammation, independently of blood pressure levels. These data suggest that Gal-3 could be a potential therapeutic candidate for the prevention of early renal alterations in hypertension.
- Published
- 2018
- Full Text
- View/download PDF
33. Transforming Growth Factor β1- and Bone Morphogenetic Protein 2/PPARγ-regulated MicroRNAs in Pulmonary Arterial Hypertension.
- Author
-
Calvier L, Chouvarine P, Legchenko E, and Hansmann G
- Subjects
- Cells, Cultured, Humans, Hypertension, Pulmonary, MicroRNAs, PPAR gamma, Transforming Growth Factor beta, Bone Morphogenetic Protein 2, Transforming Growth Factor beta1
- Published
- 2017
- Full Text
- View/download PDF
34. PPARγ Links BMP2 and TGFβ1 Pathways in Vascular Smooth Muscle Cells, Regulating Cell Proliferation and Glucose Metabolism.
- Author
-
Calvier L, Chouvarine P, Legchenko E, Hoffmann N, Geldner J, Borchert P, Jonigk D, Mozes MM, and Hansmann G
- Subjects
- Animals, Cells, Cultured, Female, Male, Mice, Inbred C57BL, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Pulmonary Artery cytology, Pulmonary Artery metabolism, Bone Morphogenetic Protein 2 metabolism, Cell Proliferation, Glucose metabolism, Myocytes, Smooth Muscle cytology, PPAR gamma metabolism, Signal Transduction, Transforming Growth Factor beta1 metabolism
- Abstract
BMP2 and TGFβ1 are functional antagonists of pathological remodeling in the arteries, heart, and lung; however, the mechanisms in VSMCs, and their disturbance in pulmonary arterial hypertension (PAH), are unclear. We found a pro-proliferative TGFβ1-Stat3-FoxO1 axis in VSMCs, and PPARγ as inhibitory regulator of TGFβ1-Stat3-FoxO1 and TGFβ1-Smad3/4, by physically interacting with Stat3 and Smad3. TGFβ1 induces fibrosis-related genes and miR-130a/301b, suppressing PPARγ. Conversely, PPARγ inhibits TGFβ1-induced mitochondrial activation and VSMC proliferation, and regulates two glucose metabolism-related enzymes, platelet isoform of phosphofructokinase (PFKP, a PPARγ target, via miR-331-5p) and protein phosphatase 1 regulatory subunit 3G (PPP1R3G, a Smad3 target). PPARγ knockdown/deletion in VSMCs activates TGFβ1 signaling. The PPARγ agonist pioglitazone reverses PAH and inhibits the TGFβ1-Stat3-FoxO1 axis in TGFβ1-overexpressing mice. We identified PPARγ as a missing link between BMP2 and TGFβ1 pathways in VSMCs. PPARγ activation can be beneficial in TGFβ1-associated diseases, such as PAH, parenchymal lung diseases, and Marfan's syndrome., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
35. Galectin-3 Blockade Reduces Renal Fibrosis in Two Normotensive Experimental Models of Renal Damage.
- Author
-
Martinez-Martinez E, Ibarrola J, Calvier L, Fernandez-Celis A, Leroy C, Cachofeiro V, Rossignol P, and Lopez-Andres N
- Subjects
- Acute Kidney Injury pathology, Animals, Aortic Valve Stenosis complications, Diet, High-Fat adverse effects, Disease Models, Animal, Fibrosis, Galectin 3 physiology, Male, Obesity complications, Rats, Rats, Wistar, Real-Time Polymerase Chain Reaction, Acute Kidney Injury prevention & control, Galectin 3 antagonists & inhibitors, Kidney pathology, Pectins pharmacology
- Abstract
Background: Galectin-3 (Gal-3), a β-galactoside-binding lectin, is increased in kidney injury and its pharmacological blockade reduces renal damage in acute kidney injury, hyperaldosteronism or hypertensive nephropathy. We herein investigated the effects of pharmacological Gal-3 inhibition by modified citrus pectin (MCP) in early renal damage associated with obesity and aortic stenosis (AS)., Results: Gal-3 was upregulated in kidneys from high fat diet (HFD) rats and in animals with partial occlusion of ascending aorta (AS). Urinary and plasma neutrophil gelatinase-associated lipocalin (NGAL) and urinary albumin were enhanced in HFD and AS rats. In kidney from obese rats, fibrotic markers (collagen, TFG-β), epithelial-mesenchymal transition molecules (α-smooth muscle actin, E-cadherin), inflammatory mediator (osteopontin) and kidney injury marker (kidney injury molecule-1) were modified. In kidney from AS rats, fibrotic markers (collagen, CTGF), epithelial-mesenchymal transition molecules (fibronectin, α-smooth muscle actin, β-catenin, E-cadherin) and kidney injury markers (NGAL, kidney injury molecule-1) were altered. Histologic observations of obese and AS rat kidneys revealed tubulointerstitial fibrosis. The pharmacological inhibition of Gal-3 with MCP normalized renal Gal-3 levels as well as functional, histological and molecular alterations in obese and AS rats., Conclusions: In experimental models of mild kidney damage, the increase in renal Gal-3 expression paralleled with renal fibrosis, inflammation and damage, while these alterations were prevented by Gal-3 blockade. These data suggest that Gal-3 could be a new player in renal molecular, histological and functional alterations at early stages of kidney damage., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
36. Galectin-3 and aldosterone as potential tandem biomarkers in pulmonary arterial hypertension.
- Author
-
Calvier L, Legchenko E, Grimm L, Sallmon H, Hatch A, Plouffe BD, Schroeder C, Bauersachs J, Murthy SK, and Hansmann G
- Subjects
- Adult, Aged, Biomarkers blood, Blood Proteins, Case-Control Studies, Enzyme-Linked Immunosorbent Assay, Female, Galectins, Humans, Hypertension, Pulmonary diagnosis, Hypertension, Pulmonary physiopathology, Inflammation Mediators blood, Male, Middle Aged, Natriuretic Peptide, Brain blood, Peptide Fragments blood, Predictive Value of Tests, Prognosis, Severity of Illness Index, Up-Regulation, Aldosterone blood, Galectin 3 blood, Hypertension, Pulmonary blood
- Abstract
Background: Several studies have identified circulating biomarkers to be associated with the presence and severity of pulmonary arterial hypertension (PAH). Recent evidence supports a role for galectin-3 (Gal-3) and the mineralcorticoid aldosterone in left ventricular failure. However, studies on aldosterone together with Gal-3 in PAH are lacking., Objective: We investigated a novel Aldosterone-galectin-3 (Gal-3) tandem and several other potential PAH biomarkers and their association with the disease severity., Methods: A total of 57 patients, 41 with idiopathic PAH. (IPAH) and 16 with PAH associated with connective tissue disease (CTD), and 8 age-matched, non-relative controls were studied. Gal-3, aldosterone and other potential protein plasma concentrations were measured by single ELISA and multi-array MSD (Meso Scale Discovery) technology., Results: Gal-3 values were increased in both patients with IPAH (12.2±0.6 ng/mL; p<0.05) and with PAH-CTD (14.1±1.6 ng/mL; p<0.05) versus control (8.5±0.9 ng/mL), while aldosterone was significantly elevated in IPAH only (248.5±38.8 pg/mL vs control 71.9±18.2 pg/mL; p<0.05). In addition, aldosterone, Gal-3, and N-terminal pro-brain natriuretic peptide (NT-proBNP) values were all higher in patients in WHO functional class II-III versus PAH functional class I or controls. The vascular injury marker intercellular adhesion molecule 1 (ICAM-1) was increased in IPAH and PAH-CTD versus controls (559.5±18.2 pg/mL and 734.1±59.4 pg/mL vs controls 394.8±39.3 pg/mL, p<0.05, p<0.0001, respectively), whereas vascular cell adhesion molecule 1 (VCAM-1) and proinflammatory, anti-angiogenic interleukin-12 (IL-12) were elevated in PAH-CTD only (879.5±110.0 pg/mL and 391.2±70.3 pg/mL vs controls 489.8±44.6 pg/mL, p<0.01, and 102.1±15.2 pg/mL, p<0.01, respectively)., Conclusions: Heightened Gal-3 and aldosterone plasma concentrations in PAH patients indicate a role for Gal-3 signalling in the pathobiology of IPAH and PAH-CTD, and may serve as biomarkers for functional status and progression of disease., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/)
- Published
- 2016
- Full Text
- View/download PDF
37. Galectin-3 blockade inhibits cardiac inflammation and fibrosis in experimental hyperaldosteronism and hypertension.
- Author
-
Martínez-Martínez E, Calvier L, Fernández-Celis A, Rousseau E, Jurado-López R, Rossoni LV, Jaisser F, Zannad F, Rossignol P, Cachofeiro V, and López-Andrés N
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Fibroblasts drug effects, Fibroblasts pathology, Fibrosis etiology, Fibrosis pathology, Galectin 3 biosynthesis, Humans, Hyperaldosteronism drug therapy, Hyperaldosteronism metabolism, Hypertension drug therapy, Hypertension metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mineralocorticoid Receptor Antagonists therapeutic use, Myocarditis etiology, Myocarditis pathology, Rats, Rats, Inbred WKY, Rats, Wistar, Galectin 3 antagonists & inhibitors, Hyperaldosteronism complications, Hypertension complications, Myocarditis prevention & control, Spironolactone therapeutic use
- Abstract
Hypertensive cardiac remodeling is accompanied by molecular inflammation and fibrosis, 2 mechanisms that finally affect cardiac function. At cardiac level, aldosterone promotes inflammation and fibrosis, although the precise mechanisms are still unclear. Galectin-3 (Gal-3), a β-galactoside-binding lectin, is associated with inflammation and fibrosis in the cardiovascular system. We herein investigated whether Gal-3 inhibition could block aldosterone-induced cardiac inflammation and fibrosis and its potential role in cardiac damage associated with hypertension. Aldosterone-salt-treated rats presented hypertension, cardiac inflammation, and fibrosis that were prevented by the pharmacological inhibition of Gal-3 with modified citrus pectin. Cardiac inflammation and fibrosis presented in spontaneously hypertensive rats were prevented by modified citrus pectin treatment, whereas Gal-3 blockade did not modify blood pressure levels. In the absence of blood pressure modifications, Gal-3 knockout mice were resistant to aldosterone-induced cardiac inflammation. In human cardiac fibroblasts, aldosterone increased Gal-3 expression via its mineralocorticoid receptor. Gal-3 and aldosterone enhanced proinflammatory and profibrotic markers, as well as metalloproteinase activities in human cardiac fibroblasts, effects that were not observed in Gal-3-silenced cells treated with aldosterone. In experimental hyperaldosteronism, the increase in Gal-3 expression was associated with cardiac inflammation and fibrosis, alterations that were prevented by Gal-3 blockade independently of blood pressure levels. These data suggest that Gal-3 could be a new molecular mechanism linking cardiac inflammation and fibrosis in situations with high-aldosterone levels, such as hypertension., (© 2015 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
38. Interleukin-33/ST2 system attenuates aldosterone-induced adipogenesis and inflammation.
- Author
-
Martínez-Martínez E, Cachofeiro V, Rousseau E, Álvarez V, Calvier L, Fernández-Celis A, Leroy C, Miana M, Jurado-López R, Briones AM, Jaisser F, Zannad F, Rossignol P, and López-Andrés N
- Subjects
- 3T3-L1 Cells, Adipogenesis drug effects, Adipose Tissue drug effects, Animals, Diet, High-Fat, Inflammation metabolism, Interleukin-1 Receptor-Like 1 Protein, Male, Mice, Mice, Inbred C57BL, Obesity metabolism, Rats, Rats, Wistar, Adipogenesis physiology, Adipose Tissue metabolism, Aldosterone pharmacology, Interleukin-33 metabolism, Receptors, Interleukin metabolism
- Abstract
Interleukin-33 (IL-33) but not soluble ST2 (sST2) exerts anti-inflammatory and protective effects in several tissues. Aldosterone, a proinflammatory mediator which promotes adipogenesis, is elevated in obese patients. The aim of this study was to investigate the interactions between IL-33/ST2 system and Aldosterone in adipose tissue. Rats fed a high fat diet presented increased sST2 expression, diminished IL-33/sST2 ratio and enhanced levels of differentiation and inflammation in adipose tissue as compared to controls. A similar pattern was observed in adipose tissue from C57BL/6 Aldosterone-treated mice. In both animal models, Aldosterone was correlated with sST2. Treatment of 3T3-L1 adipocytes with IL-33 delayed adipocyte differentiation diminished lipid accumulation and decreased inflammation. Aldosterone decreased IL-33 and increased sST2 expressions in differentiated adipocytes. Aldosterone-induced adipocyte differentiation and inflammation were blocked by IL-33 treatment, but sST2 did not exert any effects. The crosstalk between IL-33/ST2 and Aldosterone could be relevant in the metabolic consequences of obesity., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
39. The impact of galectin-3 inhibition on aldosterone-induced cardiac and renal injuries.
- Author
-
Calvier L, Martinez-Martinez E, Miana M, Cachofeiro V, Rousseau E, Sádaba JR, Zannad F, Rossignol P, and López-Andrés N
- Subjects
- Acute Kidney Injury chemically induced, Acute Kidney Injury metabolism, Aldosterone toxicity, Animals, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Galectin 3 biosynthesis, Heart Failure chemically induced, Heart Failure metabolism, Immunohistochemistry, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mineralocorticoid Receptor Antagonists therapeutic use, Rats, Rats, Wistar, Acute Kidney Injury drug therapy, Galectin 3 antagonists & inhibitors, Heart Failure drug therapy, Spironolactone therapeutic use
- Abstract
Objectives: This study investigated whether galectin (Gal)-3 inhibition could block aldosterone-induced cardiac and renal fibrosis and improve cardiorenal dysfunction., Background: Aldosterone is involved in cardiac and renal fibrosis that is associated with the development of cardiorenal injury. However, the mechanisms of these interactions remain unclear. Gal-3, a β-galactoside-binding lectin, is increased in heart failure and kidney injury., Methods: Rats were treated with aldosterone-salt combined with spironolactone (a mineralocorticoid receptor antagonist) or modified citrus pectin (a Gal-3 inhibitor), for 3 weeks. Wild-type and Gal-3 knockout mice were treated with aldosterone for 3 weeks. Hemodynamic, cardiac, and renal parameters were analyzed., Results: Hypertensive aldosterone-salt-treated rats presented cardiac and renal hypertrophy (at morphometric, cellular, and molecular levels) and dysfunction. Cardiac and renal expressions of Gal-3 as well as levels of molecular markers attesting fibrosis were also augmented by aldosterone-salt treatment. Spironolactone or modified citrus pectin treatment reversed all of these effects. In wild-type mice, aldosterone did not alter blood pressure levels but increased cardiac and renal Gal-3 expression, fibrosis, and renal epithelial-mesenchymal transition. Gal-3 knockout mice were resistant to aldosterone effects., Conclusions: In experimental hyperaldosteronism, the increase in Gal-3 expression was associated with cardiac and renal fibrosis and dysfunction but was prevented by pharmacological inhibition (modified citrus pectin) or genetic disruption of Gal-3. These data suggest a key role for Gal-3 in cardiorenal remodeling and dysfunction induced by aldosterone. Gal-3 could be used as a new biotarget for specific pharmacological interventions., (Copyright © 2015 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
40. Galectin-3 mediates aldosterone-induced vascular fibrosis.
- Author
-
Calvier L, Miana M, Reboul P, Cachofeiro V, Martinez-Martinez E, de Boer RA, Poirier F, Lacolley P, Zannad F, Rossignol P, and López-Andrés N
- Subjects
- Animals, Blood Pressure, Cells, Cultured, Collagen Type I biosynthesis, Disease Models, Animal, Fibrosis, Galectin 3 antagonists & inhibitors, Galectin 3 deficiency, Galectin 3 genetics, Humans, Hypertension chemically induced, Hypertension genetics, Hypertension pathology, Hypertension physiopathology, Hypertension prevention & control, Inflammation chemically induced, Inflammation genetics, Inflammation pathology, Inflammation physiopathology, Inflammation prevention & control, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mineralocorticoid Receptor Antagonists pharmacology, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular pathology, Muscle, Smooth, Vascular physiopathology, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle pathology, RNA Interference, Rats, Rats, Wistar, Time Factors, Transfection, Up-Regulation, Vascular Stiffness, Aldosterone, Galectin 3 metabolism, Hypertension metabolism, Inflammation metabolism, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism
- Abstract
Objective: Aldosterone (Aldo) is involved in arterial stiffness and heart failure, but the mechanisms have remained unclear. Galectin-3 (Gal-3), a β-galactoside-binding lectin, plays an important role in inflammation, fibrosis, and heart failure. We investigated here whether Gal-3 is involved in Aldo-induced vascular fibrosis., Methods and Results: In rat vascular smooth muscle cells Gal-3 overexpression enhanced specifically collagen type I synthesis. Moreover Gal-3 inhibition by modified citrus pectin or small interfering RNA blocked Aldo-induced collagen type I synthesis. Rats were treated with Aldo-salt combined with spironolactone or modified citrus pectin for 3 weeks. Hypertensive Aldo-treated rats presented vascular hypertrophy, inflammation, fibrosis, and increased aortic Gal-3 expression. Spironolactone or modified citrus pectin treatment reversed all the above effects. Wild-type and Gal-3 knock-out mice were treated with Aldo for 6 hours or 3 weeks. Aldo increased aortic Gal-3 expression, inflammation, and collagen type I in wild-type mice at both the short- and the long-term, whereas no changes occurred in Gal-3 knock-out mice., Conclusions: Our data indicate that Gal-3 is required for inflammatory and fibrotic responses to Aldo in vascular smooth muscle cells in vitro and in vivo, suggesting a key role for Gal-3 in vascular fibrosis.
- Published
- 2013
- Full Text
- View/download PDF
41. Absence of cardiotrophin 1 is associated with decreased age-dependent arterial stiffness and increased longevity in mice.
- Author
-
López-Andrés N, Calvier L, Labat C, Fay R, Díez J, Benetos A, Zannad F, Lacolley P, and Rossignol P
- Subjects
- Animals, Apoptosis physiology, Carotid Intima-Media Thickness, Cells, Cultured, Cellular Senescence physiology, Cytokines metabolism, Fibrosis, Male, Mice, Mice, Knockout, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular pathology, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle pathology, Rats, Vascular Stiffness genetics, Carotid Arteries metabolism, Cytokines genetics, Longevity physiology, Vascular Stiffness physiology
- Abstract
Cardiotrophin 1 (CT-1), an interleukin 6 family member, promotes fibrosis and arterial stiffness. We hypothesized that the absence of CT-1 influences arterial fibrosis and stiffness, senescence, and life span. In senescent 29-month-old mice, vascular function was analyzed by echotracking device. Arterial histomorphology, senescence, metabolic, inflammatory, and oxidative stress parameters were measured by immunohistochemistry, reverse transcription polymerase chain reaction, Western blot, and ELISA. Survival rate of wild-type and CT-1-null mice was studied. Vascular smooth muscle cells were treated with CT-1 (10(-9) mol/L) for 15 days to analyze senescence. The wall stress-incremental elastic modulus curve of old CT-1-null mice was shifted rightward as compared with wild-type mice, indicating decreased arterial stiffness. Media thickness and wall fibrosis were lower in CT-1-null mice. CT-1-null mice showed decreased levels of inflammatory, apoptotic, and senescence pathways, whereas telomere-linked proteins, DNA repair proteins, and antioxidant enzyme activities were increased. CT-1-null mice displayed a 5-month increased median longevity compared with wild-type mice. In vascular smooth muscle cells, chronic CT-1 stimulation upregulated apoptotic and senescence markers and downregulated telomere-linked proteins. The absence of CT-1 is associated with decreased arterial fibrosis, stiffness, and senescence and increased longevity in mice likely through downregulating apoptotic, senescence, and inflammatory pathways. CT-1 may be a major regulator of arterial stiffness with a major impact on the aging process.
- Published
- 2013
- Full Text
- View/download PDF
42. Cardiotrophin 1 is involved in cardiac, vascular, and renal fibrosis and dysfunction.
- Author
-
López-Andrés N, Rousseau A, Akhtar R, Calvier L, Iñigo C, Labat C, Zhao X, Cruickshank K, Díez J, Zannad F, Lacolley P, and Rossignol P
- Subjects
- Animals, Blood Vessels drug effects, Cardiovascular System drug effects, Creatinine blood, Cytokines pharmacology, Fibrosis, Heart drug effects, Heart physiopathology, Heart Ventricles diagnostic imaging, Heart Ventricles drug effects, Kidney drug effects, Male, Models, Animal, Rats, Rats, Wistar, Stroke Volume drug effects, Stroke Volume physiology, Ultrasonography, Vascular Stiffness drug effects, Vascular Stiffness physiology, Blood Vessels pathology, Cardiovascular System physiopathology, Cytokines physiology, Kidney pathology, Kidney physiopathology, Myocardium pathology
- Abstract
Cardiotrophin 1 (CT-1), a cytokine belonging to the interleukin 6 family, is increased in hypertension and in heart failure. We aimed to study the precise role of CT-1 on cardiac, vascular, and renal function; morphology; and remodeling in early stages without hypertension. CT-1 (20 μg/kg per day) or vehicle was administrated to Wistar rats for 6 weeks. Cardiac and vascular functions were analyzed in vivo using M-mode echocardiography, Doppler, and echo tracking device and ex vivo using a scanning acoustic microscopy method. Cardiovascular and renal histomorphology were measured by immunohistochemistry, RT-PCR, and Western blot. Kidney functional properties were assessed by serum creatinine and neutrophile gelatinase-associated lipocalin and microalbuminuria/creatininuria ratio. Without alterations in blood pressure levels, CT-1 treatment increased left ventricular volumes, reduced fractional shortening and ejection fraction, and induced myocardial dilatation and myocardial fibrosis. In the carotid artery of CT-1-treated rats, the circumferential wall stress-incremental elastic modulus curve was shifted leftward, and the acoustic speed of sound in the aorta was augmented, indicating increased arterial stiffness. Vascular media thickness, collagen, and fibronectin content were increased by CT-1 treatment. CT-1-treated rats presented unaltered serum creatinine concentrations but increased urinary and serum neutrophile gelatinase-associated lipocalin and microalbuminuria/creatininuria ratio. This paralleled a glomerular and tubulointerstitial fibrosis accompanied by renal epithelial-mesenchymal transition. CT-1 is a new potent fibrotic agent in heart, vessels, and kidney able to induce cardiovascular-renal dysfunction independent from blood pressure. Thus, CT-1 could be a new target simultaneously integrating alterations of heart, vessels, and kidney in early stages of heart failure.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.