75 results on '"S. Rayego-Mateos"'
Search Results
2. P6276CCN2 deletion induces early aortic aneurysm development after Angiotensin II infusion in mice. New aneurysm generation experimental model
- Author
-
R. Goldschmeding, V. Esteban Vazquez, R. Rodrigues, R. Selgas Gutierrez, S. Rayego Mateos, L. Falke, M. Ruiz Ortega, and M. Orejudo Del Rio
- Subjects
Aortic aneurysm ,medicine.medical_specialty ,Aneurysm ,Experimental model ,business.industry ,Internal medicine ,medicine ,Cardiology ,Cardiology and Cardiovascular Medicine ,medicine.disease ,business ,Angiotensin II - Published
- 2017
3. Experimental models of CKD
- Author
-
R. Kanlaya, K. Sintiprungrat, V. Thongboonkerd, N. Torremade, R. Bindels, J. Hoenderop, E. Fernandez, A. Dusso, J. M. Valdivielso, T. Krueger, P. Boor, C. Schafer, R. Westenfeld, V. Brandenburg, G. Schlieper, W. Jahnen-Dechent, M. Ketteler, W. Jee, X. Li, B. Richards, J. Floege, J. G. Goncalves, D. Canale, A. C. de Braganca, M. H. M. Shimizu, R. M. A. Moyses, L. Andrade, A. C. Seguro, R. A. Volpini, S. Romoli, A. Migliorini, H.-J. Anders, O. Eskova, N. Neprintseva, N. Tchebotareva, I. Bobkova, L. Kozlovskaya, I. Simic, M. Tabatabaeifar, T. Wlodkowski, H. Denc, G. Mollet, C. Antignac, F. Schaefer, I. A. Ekaterina, L. Giardino, M. P. Rastaldi, L. Van den Heuvel, E. Levtchenko, C. Okina, T. Okamoto, M. Kamata, J. Murano, K. Kobayashi, K. Takeuchi, F. Kamata, T. Sakai, S. Naito, T. Aoyama, T. Sano, Y. Takeuchi, K. Kamata, D. Thomasova, H. A. Bruns, H. Liapis, T. Iwashita, H. Hasegawa, K. Takayanagi, T. Shimizu, J. Asakura, S. Okazaki, Y. Kogure, M. Hatano, H. Hara, M. Inamura, M. Iwanaga, T. Mitani, T. Mitarai, V. J. Savin, M. Sharma, C. Wei, J. Reiser, E. T. McCarthy, R. Sharma, J.-F. Gauchat, B. Eneman, K. Freson, C. Van Geet, D. E. Choi, J. Y. Jeong, Y. K. Chang, K.-R. Na, K. W. Lee, Y. T. Shin, H.-F. Ni, J.-F. Chen, M.-H. Zhang, M.-M. Pan, B.-C. Liu, S. S. Kim, T. Suzuki, M. Iyoda, K. Matsumoto, Y. Shindo-Hirai, Y. Kuno, Y. Wada, Y. Yamamoto, T. Shibata, T. Akizawa, J. M. Munoz-Felix, J. M. Lopez-Novoa, C. Martinez-Salgado, J. Ehling, J. Babickova, F. Gremse, F. Kiessling, T. Lammers, M. Lech, R. Gunthner, G. Lorenz, M. Ryu, R. Grobmayr, H. Susanti, K. S. Kobayashi, R. A. Flavell, S. Rayego-Mateos, J. Morgado, A. B. Sanz, S. Eguchi, J. Pato, G. Keri, J. Egido, A. Ortiz, M. Ruiz-Ortega, M. Leduc, L. Geerts, B. Grouix, F. Sarra-Bournet, A. Felton, L. Gervais, S. Abbott, J.-S. Duceppe, B. Zacharie, C. Penney, P. Laurin, L. Gagnon, M. G. Detsika, P. Duann, E. A. Lianos, K. I. Leong, C.-K. Chiang, C.-C. Yang, C.-T. Wu, L.-P. Chen, K.-Y. Hung, S.-H. Liu, F. F. Carvalho, V. P. Teixeira, W. S. Almeida, N. Schor, D. M. Small, N. C. Bennett, J. Coombes, D. W. Johnson, G. C. Gobe, N. Montero, A. Prada, M. Riera, M. Orfila, J. Pascual, E. Rodriguez, C. Barrios, G. Kokeny, K. Fazekas, L. Rosivall, M. M. Mozes, N. Hornigold, J. Hughes, A. Mooney, A. Benardeau, W. Riboulet, A. Vandjour, B. Jacobsen, C. Apfel, K. Conde-Knape, J.-F. Bienvenu, T. Tanaka, J. Yamaguchi, M. Nangaku, T. Niwa, D. Bolati, H. Shimizu, M. Yisireyili, F. Nishijima, A. Brocca, G. Virzi, M. de Cal, C. Ronco, G. Priante, E. Musacchio, C. Valvason, L. Sartori, A. Piccoli, B. Baggio, M. Perkuhn, M. Weibrecht, S. Zok, I. V. Martin, F. Schoth, T. Ostendorf, C. Kuhl, A. Karabaeva, A. Essaian, O. Beresneva, M. Parastaeva, I. Kayukov, A. Smirnov, I. Audzeyenka, M. Kasztan, A. Piwkowska, D. Rogacka, S. Angielski, M. Jankowski, C. L. Bockmeyer, K. Kokowicz, P. A. Agustian, S. Zell, J. Wittig, J. U. Becker, R. Nishizono, M. P. Venkatareddy, M. A. Chowdhury, S. Q. Wang, A. Fukuda, L. T. Wickman, Y. Yang, R. C. Wiggins, M. R. Fazio, V. Donato, S. Lucisano, V. Cernaro, R. Lupica, D. Trimboli, G. Montalto, C. Aloisi, A. T. Mazzeo, M. Buemi, O. Gawrys, K. H. Olszynski, M. Kuczeriszka, K. Gawarecka, E. Swiezewska, M. Chmielewski, M. Masnyk, J. Rafalowska, E. Kompanowska-Jezierska, W.-C. Lee, Y.-Y. Chau, L.-C. Lee, C.-H. Chiu, C.-T. Lee, J.-B. Chen, W.-K. Kim, and S. J. Shin
- Subjects
Transplantation ,medicine.medical_specialty ,Nephrology ,business.industry ,Medicine ,business ,Intensive care medicine - Published
- 2013
4. Hypertension
- Author
-
S. J. Shin, M.-Y. Rhee, C. Lim, C. Lavoz, R. Rodrigues-Di, S. Rayego-Mateos, A. Benito-Martin, R. Rodrigues-Diez, M. Alique, A. Ortiz, S. Mezzano, M. Ruiz-Ortega, J. Axelsson, A. Rippe, K. Sverrisson, B. Rippe, L. Calo, L. Dal Maso, E. Pagnin, P. Caielli, G. Spanos, R. Kalaitzidis, D. Karasavvidou, K. Pappas, O. Balafa, K. Siamopoulos, T.-C. Fang, T. J. F. Lee, E. Pappas, E. Ermeidi, A. Tatsioni, A. Blazquez-Medela, O. Garcia-Sanchez, Y. Quiros, F. J. Lopez-Hernandez, J. M. Lopez-Novoa, C. Martinez-Salgado, H.-Y. Wu, Y.-S. Peng, K.-Y. Hung, T.-J. Tsai, Y.-K. Tu, K.-L. Chien, T. Larsen, F. H. Mose, A. B. Hansen, E. B. Pedersen, Y. Quiroz, M. Rivero, K. Yaguas, B. Rodriguez-Iturbe, D. Xydakis, M. Sfakianaki, C. Petra, E. Maragaki, E. Antonaki, E. Krasoudaki, K. Kostakis, K. Stylianou, A. Papadogiannakis, Y. Sagliker, N. Paylar, A. Heidland, A. Keck, R. Erek, P. Kolasin, P. S Ozkaynak, H. S. Sagliker, I. Gokcay, E. Ritz, N. Koleganova, M.-L. Gross-Weissmann, G. Piecha, N. Reinecke, T. Marquez Cunha, E. M . S. Higa, I. Pfeferman Heilberg, J. A. Neder, J. L. Nishiura, W. Silva Almeida, N. Schor, E. Tapia, L. G. Sanchez-Lozada, M. Cristobal, V. Soto, F. Garci, a-Arroyo, F. Monroy-Sanchez, M. Madero, R. Johnson, S. M. Kim, S. H. Yang, Y. S. Kim, S. Karanovic, M. Fistrek, J. Kos, I. Pecin, V. Premuzic, M. Abramovic, V. Matijevic, D. Cvoriscec, A. Cvitkovic, M. Knezevic, M. Bitunjac, M. Laganovic, B. Jelakovic, F. Liu, M. Wu, P. Fu, S. Klok Matthesen, T. Guldager Lauridsen, H. Vase, P. Gjorup Holland, K. M. Nykjaer, S. Nielsen, E. Bjerregaard Pedersen, M. J. Montero, E. Vink, V. Willemien, V. Michiel, S. Wilko, V. Evert-Jan, P. Blankestijn, S. Zerbi, L. A. Pedrini, E. Zbroch, J. Malyszko, E. Koc-Zorawska, M. Mysliwiec, J. Quelhas-Santos, P. Serrao, I. Soares-Silva, L. Tang, B. Sampaio-Maia, G. Desir, M. Pestana, R. Elsurer, T. Demir, G. Celik, M. Yavas, O. Yavas, M. Murphy, G. Jacquillet, R. J. Unwin, H. Chichger, D. G. Shirley, A. Caraba, M. Andreea, S. Corina, R. Ioan, M. Nowicki, M. Bobik, A. Pawelec, J. Lacisz, A. Zapala, K. Bryc, C. Esposito, M. L. Scaramuzzi, A. Manini, M. Torreggiani, F. Beneventi, A. Spinillo, F. Grosjean, G. Fasoli, A. Dal Canton, C. Christos, S. Bernhard M.W., N. Martin, K. Jan, M. Claus, R. Leyla, B. Jan, K. Ulrich, H. Hermann, J. Menne, M. Pavicevic, S. Markovic, and Z. Igrutinovic
- Subjects
Transplantation ,Nephrology - Published
- 2012
5. Peritoneal dialysis
- Author
-
F.-X. Beck, C. Kuper, W. Neuhofer, R. Rodrigues-Diez, L. G. Stark Aroeira, J. A. Jimenez, S. Rayego-Mateos, A. Bajo Rubio, A. Ortiz, J. Egido, M. Lopez-Cabrera, R. Selgas, M. Ruiz-Ortega, A. Pedro Ventura, S. Olivia, L. Teixeira, V. Joana, F. Francisco, C. Maria Joao, C. Antonio, A. S. Rodrigues, A. Vychytil, J. Kerschbaum, K. Lhotta, F. Prischl, M. Wiesholzer, G. Kopriva-Altfahrt, V. Machold-Fabrizii, C. Schwarz, P. Balcke, R. Oberbauer, R. Kramar, P. Konig, M. Rudnicki, M. Habib, M. Betjes, M. Korte, E. Vidal, A. Edefonti, R. Chimenz, B. Gianoglio, G. Leozappa, S. Maringhini, F. Mencarelli, C. Pecoraro, F. Puteo, S. Testa, R. Cannavo, and E. Verrina
- Subjects
Transplantation ,Nephrology - Published
- 2012
6. [Connective tissue growth factor (CTGF): a key factor in the onset and progression of kidney damage]
- Author
-
E, Sánchez-López, R, Rodrigues Díez, J, Rodríguez Vita, S, Rayego Mateos, R R, Rodrigues Díez, E, Rodríguez García, C, Lavoz Barria, S, Mezzano, J, Egido, A, Ortiz, M, Ruiz-Ortega, and R, Selgas
- Subjects
Connective Tissue Growth Factor ,Disease Progression ,Humans ,Kidney Diseases - Abstract
Connective tissue growth factor (CTGF) is increased in several pathologies associated with fibrosis, including multiple renal diseases. CTGF is involved in biological processes such as cell cycle regulation, migration, adhesion and angiogenesis. Its expression is regulated by various factors involved in renal damage, such as transforming growth factor- , Angiotensin II, high concentrations of glucose and cellular stress. CTGF is involved in the initiation and progression of renal damage to be able to induce an inflammatory response and promote fibrosis, identified as a potential therapeutic target in the treatment of kidney diseases. In this paper we review the main actions of CTGF in renal disease, the intracellular action mechanisms and therapeutic strategies for its blocking.
- Published
- 2009
7. Mechanisms and targets of glomerular damage
- Author
-
S. Rayego-Mateos, R. Rodrigues-Diez, R. R. Rodrigues-Diez, C. Lavoz-Barria, M. Alique, S. Mas, J. Pato, G. Keri, J. Egido, A. Ortiz, M. Ruiz-Ortega, L. Ying, M. Tepel, E. Frank, T. Florian, T. Gregor, J. Boye, G. Maik, B. Teng, C. Gu, H. Haller, S. Sever, M. Schiffer, K. Worthmann, M. Leitges, O. Dittrich-Breiholz, M. Kracht, A. Peired, M. L. Angelotti, E. Ronconi, E. Lazzeri, A. Sisti, L. Lasagni, and P. Romagnani
- Subjects
Transplantation ,Pathology ,medicine.medical_specialty ,Nephrology ,business.industry ,Medicine ,business - Published
- 2012
8. Experimental pathology
- Author
-
D. X. Yi Chun, H. Alexandre, B. Edith, O. Nacera, P. Julie, J. Chantal, R. Eric, X. Zhang, Y. Jin, M. Miravete, R. Dissard, J. Klein, J. Gonzalez, C. Caubet, C. Pecher, B. Pipy, J.-L. Bascands, M. Mercier-Bonin, J. Schanstra, B. Buffin-Meyer, R. Claire, C. Rigothier, D. Richard, L. Sebastien, S. Moin, B. Chantal, C. Christian, R. Jean, M. Migliori, V. Cantaluppi, C. Mannari, D. Medica, L. Giovannini, V. Panichi, A. Goldwich, S. Alexander, G. Andre, K. Amann, A. Migliorini, C. Sagrinati, M. L. Angelotti, S. R. Mulay, E. Ronconi, A. Peired, P. Romagnani, H.-J. Anders, W. C. Chiang, C. F. Lai, W.-H. Peng, C. F. Wu, F.-C. Chang, Y.-T. Chen, S.-L. Lin, Y. M. Chen, K. D. Wu, K.-S. Lu, T. J. Tsai, O. Virgine, F. Qing Feng, S.-Y. Zhang, D. Dominique, A. Vincent, C. Marina, L. Philippe, G. Georges, A. Pawlak, D. Sahali, S. Matsumoto, H. Kiyomoto, A. Ichimura, T. Dan, T. Nakamichi, T. Tsujita, K. Akahori, S. Ito, T. Miyata, S. Xie, B. Zhang, W. Shi, Y. Yang, H. Nagasu, M. Satoh, K. Kidokoro, Y. Nishi, C. Ihoriya, H. Kadoya, T. Sasaki, N. Kashihara, C.-F. Wu, Y.-H. Chou, J. Duffield, C. Rocca, M. Gregorini, V. Corradetti, T. Valsania, G. Bedino, F. Bosio, E. F. Pattonieri, P. Esposito, V. Sepe, C. Libetta, T. Rampino, A. Dal Canton, H. Omori, N. Kawada, K. Inoue, Y. Ueda, R. Yamamoto, I. Matsui, J. Kaimori, Y. Takabatake, T. Moriyama, Y. Isaka, H. Rakugi, A. Wasilewska, K. Taranta-Janusz, W. Deebek, E. Kuroczycka-Saniutycz, A. S. Lee, J. E. Lee, Y. J. Jung, K. P. Kang, S. Lee, W. Kim, N. Arfian, N. Emoto, K. Yagi, K. Nakayama, A. B. Hartopo, D. A. Nugrahaningsih, M. Yanagisawa, K.-I. Hirata, J. M. Munoz-Felix, J. M. Lopez-Novoa, C. Martinez-Salgado, B. Oujo, M. Arevalo, C. Bernabeu, F. Perez-Barriocanal, K. Jesper, V. Nathalie, G. Pierre, M. Iyoda, T. Shibata, K. Matsumoto, Y. Shindo-Hirai, Y. Kuno, Y. Wada, T. Akizawa, I. Schwartz, D. Schwartz, C. Prot Bertoye, S. Terryn, J. Claver, W. B. Beghdadi, R. Monteiro, U. Blank, O. Devuyst, E. Daugas, K. Van Beneden, C. Geers, M. Pauwels, I. Mannaerts, C. Van den Branden, L. A. Van Grunsven, I. Seckin, M. Pekpak, M. Uzunalan, B. Uruluer, S. Kokturk, Z. Ozturk, H. Sonmez, E. Yaprak, Y. Furuno, M. Tsutsui, T. Morishita, H. Shimokawa, Y. Otsuji, N. Yanagihara, N. Kabashima, S. Ryota, K. Kanegae, T. Miyamoto, J. Nakamata, N. Ishimatsu, M. Tamura, T. Nakagawa, K. Ichikawa, M. Miyamoto, D. Takabayashi, H. Yamazaki, K. Kakeshita, T. Koike, S. Kagitani, F. Tomoda, T. Hamashima, Y. Ishii, H. Inoue, M. Sasahara, F. El Machhour, M. Kerroch, L. Mesnard, C. Chatziantoniou, J.-C. Dussaule, K. Inui, F. Sasai, Y. Maruta, H. Nishiwaki, E. Kawashima, Y. Inoue, A. Yoshimura, E. Musacchio, G. Priante, C. Valvason, L. Sartori, B. Baggio, J. H. Kim, O. Gross, R. Diana, D. H. Gry, B. Asimal, T. Johanna, S.-E. Imke, W. Lydia, M. Gerhard-Anton, D. Hassan, J. L. Cano, M. Griera, G. Olmos, P. Martin, M. A. Cortes, S. Lopez-Ongil, D. Rodriguez-Puyol, S. DE Frutos, M. Gonzalez, A. Luengo, M. Rodriguez-Puyol, L. Calleros, R. Lupica, A. Lacquaniti, V. Donato, R. Maggio, C. Mastroeni, S. Lucisano, V. Cernaro, M. R. Fazio, A. Quartarone, M. Buemi, M. Kacik, S. Goedicke, H. Eggert, J. D. Hoyer, S. Wurm, A. Steege, M. Banas, A. Kurtz, B. Banas, L. Lasagni, E. Lazzeri, S. Romoli, I. Schaefer, B. Teng, K. Worthmann, H. Haller, M. Schiffer, C. Prattichizzo, G. S. Netti, M. T. Rocchetti, L. Cormio, G. Carrieri, G. Stallone, G. Grandaliano, E. Ranieri, L. Gesualdo, A. Kucher, A. Smirnov, M. Parastayeva, O. Beresneva, I. Kayukov, I. Zubina, G. Ivanova, A. Abed, L. Schlekenbach, B. Foglia, B. Kwak, C. Chadjichristos, N. Queisser, N. Schupp, S. Brand, L. Himer, B. Szebeni, E. Sziksz, S. Saijo, E. Kis, A. Prokai, N. F. Banki, A. Fekete, T. Tulassay, A. Vannay, B. Hegner, T. Schaub, C. Lange, D. Dragun, B. M. Klinkhammer, K. Rafael, M. Monika, M. Anna, C. Van Roeyen, P. Boor, B. Eva Bettina, O. Simon, S. Esther, J. Floege, U. Kunter, D. Janke, J. Jankowski, M. Hayashi, I. Takamatsu, C. Horimai, T. Yoshida, G. Seno DI Marco, M. Koenig, C. Stock, S. Reiermann, S. Amler, G. Koehler, M. Fobker, F. Buck, H. Pavenstaedt, D. Lang, M. Brand, E. Plotnikov, M. Morosanova, I. Pevzner, L. Zorova, N. Pulkova, D. Zorov, M. Wornle, A. Ribeiro, F. Belling, M. Merkle, D. Nakazawa, S. Nishio, S. Shibasaki, U. Tomaru, I. Akihiro, I. Kobayashi, Y. Imanishi, M. Kurajoh, Y. Nagata, M. Yamagata, M. Emoto, T. Michigami, E. Ishimura, M. Inaba, C.-C. Wu, K.-C. Lu, J.-S. Chen, P. Chu, Y.-F. Lin, K. Eller, A. Schroll, A. Kirsch, J. Huber, G. Weiss, I. Theurl, A. R. Rosenkranz, A. Zawada, K. Rogacev, M. Achenbach, D. Fliser, G. Held, G. H. Heine, Y. Miyamoto, Y. Iwao, H. Watanabe, D. Kadowaki, Y. Ishima, V. T. G. Chuang, K. Sato, M. Otagiri, T. Maruyama, H. Iwatani, D. Honda, T. Noguchi, M. Tanaka, H. Tanaka, M. Fukagawa, J. Pircher, S. Koppel, H. Mannell, F. Krotz, G. M. Virzi, C. Bolin, D. Cruz, E. Scalzotto, M. De Cal, G. Vescovo, C. Ronco, R. Grobmayr, M. Lech, M. Ryu, Y. Aoshima, M. Mizobuchi, H. Ogata, C. Kumata, A. Nakazawa, F. Kondo, N. Ono, F. Koiwa, E. Kinugasa, W. Freisinger, N. Lale, A. Lampert, T. Ditting, S. Heinlein, R. E. Schmieder, R. Veelken, H. Nave, R. Perthel, M. Suntharalingam, S. Bode-Boger, G. Beutel, J. Kielstein, R. Rodrigues-Diez, S. Rayego-Mateos, C. Lavoz, L. G. Stark Aroeira, M. Orejudo, M. Alique, A. Ortiz, J. Egido, M. Ruiz-Ortega, W. Oskar, C. Rusan, J.-S. Padberg, A. Wiesinger, S. Reuter, A. Grabner, D. Kentrup, A. Lukasz, H. Oberleithner, H. Pavenstadt, P. Kumpers, H. U. Eberhardt, C. Skerka, Q. Chen, T. Hallstroem, A. Hartmann, M. J. Kemper, P. F. Zipfel, K. N'gome-Sendeyo, Q.-F. Fan, J. Toblli, G. Cao, J. F. Giani, F. P. Dominici, J. S. Kim, J. W. Yang, M. K. Kim, B. G. Han, and S. O. Choi
- Subjects
Transplantation ,Nephrology - Published
- 2011
9. Ccn2 Deletion Reduces Cardiac Dysfunction, Oxidative Markers, and Fibrosis Induced by Doxorubicin Administration in Mice.
- Author
-
Tejera-Muñoz A, Cortés M, Rodriguez-Rodriguez A, Tejedor-Santamaria L, Marchant V, Rayego-Mateos S, Gimeno-Longas MJ, Leask A, Nguyen TQ, Martín M, Tuñón J, Rodríguez I, Ruiz-Ortega M, and Rodrigues-Díez RR
- Subjects
- Animals, Mice, Gene Deletion, Male, Myocardium metabolism, Myocardium pathology, Mice, Inbred C57BL, Cardiotoxicity genetics, Cardiotoxicity metabolism, Doxorubicin adverse effects, Connective Tissue Growth Factor metabolism, Connective Tissue Growth Factor genetics, Fibrosis, Mice, Knockout, Oxidative Stress drug effects
- Abstract
Cellular Communication Network Factor 2 (CCN2) is a matricellular protein implicated in cell communication and microenvironmental signaling. Overexpression of CCN2 has been documented in various cardiovascular pathologies, wherein it may exert either deleterious or protective effects depending on the pathological context, thereby suggesting that its role in the cardiovascular system is not yet fully elucidated. In this study, we aimed to investigate the effects of Ccn2 gene deletion on the progression of acute cardiac injury induced by doxorubicin (DOX), a widely utilized chemotherapeutic agent. To this end, we employed conditional knockout (KO) mice for the Ccn2 gene (CCN2-KO), which were administered DOX and compared to DOX-treated wild-type (WT) control mice. Our findings demonstrated that the ablation of CCN2 ameliorated DOX-induced cardiac dysfunction, as evidenced by improvements in ejection fraction (EF) and fractional shortening (FS) of the left ventricle. Furthermore, DOX-treated CCN2-KO mice exhibited a significant reduction in the gene expression and activation of oxidative stress markers (Hmox1 and Nfe2l2/NRF2) relative to DOX-treated WT controls. Additionally, the deletion of Ccn2 markedly attenuated DOX-induced cardiac fibrosis. Collectively, these results suggest that CCN2 plays a pivotal role in the pathogenesis of DOX-mediated cardiotoxicity by modulating oxidative stress and fibrotic pathways. These findings provide a novel avenue for future investigations to explore the therapeutic potential of targeting CCN2 in the prevention of DOX-induced cardiac dysfunction.
- Published
- 2024
- Full Text
- View/download PDF
10. Colony stimulating factor-1 receptor drives glomerular parietal epithelial cell activation in focal segmental glomerulosclerosis.
- Author
-
Cruzado JM, Manonelles A, Rayego-Mateos S, Doladé N, Amaya-Garrido A, Varela C, Guiteras R, Mosquera JL, Jung M, Codina S, Martínez-Valenzuela L, Draibe J, Couceiro C, Vigués F, Madrid Á, Florian MC, Ruíz-Ortega M, and Sola A
- Subjects
- Animals, Humans, Mice, Cell Proliferation drug effects, Epithelial Cells metabolism, Epithelial Cells pathology, Epithelial Cells drug effects, Receptor, Macrophage Colony-Stimulating Factor metabolism, Receptor, Macrophage Colony-Stimulating Factor genetics, Kidney Glomerulus pathology, Kidney Glomerulus metabolism, Male, Disease Models, Animal, Cells, Cultured, Female, Up-Regulation, Cell Movement drug effects, MAP Kinase Signaling System drug effects, Signal Transduction, Mice, Inbred C57BL, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor, Glomerulosclerosis, Focal Segmental pathology, Glomerulosclerosis, Focal Segmental metabolism, Glomerulosclerosis, Focal Segmental genetics, Podocytes metabolism, Podocytes pathology, Macrophage Colony-Stimulating Factor metabolism, Macrophage Colony-Stimulating Factor genetics, Hyaluronan Receptors metabolism, Hyaluronan Receptors genetics
- Abstract
Parietal epithelial cells (PECs) are kidney progenitor cells with similarities to a bone marrow stem cell niche. In focal segmental glomerulosclerosis (FSGS) PECs become activated and contribute to extracellular matrix deposition. Colony stimulating factor-1 (CSF-1), a hematopoietic growth factor, acts via its specific receptor, CSF-1R, and has been implicated in several glomerular diseases, although its role on PEC activation is unknown. Here, we found that CSF-1R was upregulated in PECs and podocytes in biopsies from patients with FSGS. Through in vitro studies, PECs were found to constitutively express CSF-1R. Incubation with CSF-1 induced CSF-1R upregulation and significant transcriptional regulation of genes involved in pathways associated with PEC activation. Specifically, CSF-1/CSF-1R activated the ERK1/2 signaling pathway and upregulated CD44 in PECs, while both ERK and CSF-1R inhibitors reduced CD44 expression. Functional studies showed that CSF-1 induced PEC proliferation and migration, while reducing the differentiation of PECs into podocytes. These results were validated in the Adriamycin-induced FSGS experimental mouse model. Importantly, treatment with either the CSF-1R-specific inhibitor GW2580 or Ki20227 provided a robust therapeutic effect. Thus, we provide evidence of the role of the CSF-1/CSF-1R pathway in PEC activation in FSGS, paving the way for future clinical studies investigating the therapeutic effect of CSF-1R inhibitors on patients with FSGS., (Copyright © 2024 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. BET Protein Inhibitor JQ1 Ameliorates Experimental Peritoneal Damage by Inhibition of Inflammation and Oxidative Stress.
- Author
-
Marchant V, Trionfetti F, Tejedor-Santamaria L, Rayego-Mateos S, Rotili D, Bontempi G, Domenici A, Menè P, Mai A, Martín-Cleary C, Ortiz A, Ramos AM, Strippoli R, and Ruiz-Ortega M
- Abstract
Peritoneal dialysis (PD) is a current replacement therapy for end-stage kidney diseases (ESKDs). However, long-term exposure to PD fluids may lead to damage of the peritoneal membrane (PM) through mechanisms involving the activation of the inflammatory response and mesothelial-to-mesenchymal transition (MMT), leading to filtration failure. Peritoneal damage depends on a complex interaction among external stimuli, intrinsic properties of the PM, and subsequent activities of the local innate-adaptive immune system. Epigenetic drugs targeting bromodomain and extra-terminal domain (BET) proteins have shown beneficial effects on different experimental preclinical diseases, mainly by inhibiting proliferative and inflammatory responses. However the effect of BET inhibition on peritoneal damage has not been studied. To this aim, we have evaluated the effects of treatment with the BET inhibitor JQ1 in a mouse model of peritoneal damage induced by chlorhexidine gluconate (CHX). We found that JQ1 ameliorated the CHX-induced PM thickness and inflammatory cell infiltration. Moreover, JQ1 decreased gene overexpression of proinflammatory and profibrotic markers, together with an inhibition of the nuclear factor-κB (NF-κB) pathway. Additionally, JQ1 blocked the activation of nuclear factor erythroid 2-related factor 2 (NRF2) and restored changes in the mRNA expression levels of NADPH oxidases (NOX1 and NOX4) and NRF2/target antioxidant response genes. To corroborate the in vivo findings, we evaluated the effects of the BET inhibitor JQ1 on PD patients' effluent-derived primary mesothelial cells and on the MeT-5A cell line. JQ1 inhibited tumor necrosis factor-α (TNF-α)-induced proinflammatory gene upregulation and restored MMT phenotype changes, together with the downmodulation of oxidative stress. Taken together, these results suggest that BET inhibitors may be a potential therapeutic option to ameliorate peritoneal damage.
- Published
- 2023
- Full Text
- View/download PDF
12. Regulated necrosis role in inflammation and repair in acute kidney injury.
- Author
-
Guerrero-Mauvecin J, Villar-Gómez N, Rayego-Mateos S, Ramos AM, Ruiz-Ortega M, Ortiz A, and Sanz AB
- Subjects
- Humans, Mice, Animals, Apoptosis, Necrosis, Inflammation complications, Acute Kidney Injury metabolism, Renal Insufficiency, Chronic metabolism
- Abstract
Acute kidney injury (AKI) frequently occurs in patients with chronic kidney disease (CKD) and in turn, may cause or accelerate CKD. Therapeutic options in AKI are limited and mostly relate to replacement of kidney function until the kidneys recover spontaneously. Furthermore, there is no treatment that prevents the AKI-to-CKD transition. Regulated necrosis has recently emerged as key player in kidney injury. Specifically, there is functional evidence for a role of necroptosis, ferroptosis or pyroptosis in AKI and the AKI-to-CKD progression. Regulated necrosis may be proinflammatory and immunogenic, triggering subsequent waves of regulated necrosis. In a paradigmatic murine nephrotoxic AKI model, a first wave of ferroptosis was followed by recruitment of inflammatory cytokines such as TWEAK that, in turn, triggered a secondary wave of necroptosis which led to persistent kidney injury and decreased kidney function. A correct understanding of the specific forms of regulated necrosis, their timing and intracellular molecular pathways may help design novel therapeutic strategies to prevent or treat AKI at different stages of the condition, thus improving patient survival and the AKI-to-CKD transition. We now review key regulated necrosis pathways and their role in AKI and the AKI-to-CKD transition both at the time of the initial insult and during the repair phase following AKI., Competing Interests: AO has received grants from Sanofi and consultancy or speaker fees or travel support from Adviccene, Alexion, Astellas, AstraZeneca, Amicus, Amgen, Boehringer Ingelheim, Fresenius Medical Care, GSK, Bayer, Sanofi-Genzyme, Menarini, Mundipharma, Kyowa Kirin, Lilly, Freeline, Idorsia, Chiesi, Otsuka, Novo-Nordisk, Sysmex and Vifor Fresenius Medical Care Renal Pharma and Spafarma and is Director of the Catedra UAM-Astrazeneca of chronic kidney disease and electrolytes. He has stock in Telara Farma. 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. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2023 Guerrero-Mauvecin, Villar-Gómez, Rayego-Mateos, Ramos, Ruiz-Ortega, Ortiz and Sanz.)
- Published
- 2023
- Full Text
- View/download PDF
13. STING1 deficiency ameliorates immune-mediated crescentic glomerulonephritis in mice.
- Author
-
García-Giménez J, Córdoba-David G, Rayego-Mateos S, Cannata-Ortiz P, Carrasco S, Ruiz-Ortega M, Fernandez-Fernandez B, Ortiz A, and Ramos AM
- Subjects
- Animals, Humans, Mice, Acute Disease, Cytokines metabolism, Kidney immunology, Kidney metabolism, Kidney pathology, Kidney Glomerulus pathology, Membrane Proteins genetics, Membrane Proteins metabolism, Glomerulonephritis genetics, Glomerulonephritis immunology, Glomerulonephritis metabolism, Glomerulonephritis pathology, Nephritis
- Abstract
Rapidly progressive/crescentic glomerulonephritis (RPGN/CGN) involves the formation of glomerular crescents by maladaptive differentiation of parietal epithelial cells that leads to rapid loss of renal function. The molecular mechanisms of crescent formation are poorly understood. Therefore, new insights into molecular mechanisms could identify alternative therapeutic targets for RPGN/CGN. Analysis of kidney biopsies from patients with RPGN revealed increased interstitial, glomerular, and tubular expression of STING1, an accessory protein of the c-GAS-dependent DNA-sensing pathway, which was also observed in murine nephrotoxic nephritis induced by an anti-GBM antibody. STING1 was expressed by key cell types involved in RPGN and crescent formation such as glomerular parietal epithelial cells, and tubular cells as well as by inflammation accessory cells. In functional in vivo studies, Sting1
-/- mice with nephrotoxic nephritis had lower kidney cytokine expression, milder kidney infiltration by innate and adaptive immune cells, and decreased disease severity. Pharmacological STING1 inhibition mirrored these findings. Direct STING1 agonism in parietal and tubular cells activated the NF-κB-dependent cytokine response and the interferon-induced genes (ISGs) program. These responses were also triggered in a STING1-dependent manner by the pro-inflammatory cytokine TWEAK. These results identify STING1 activation as a pathological mechanism in RPGN/CGN and TWEAK as an activator of STING1. Pharmacological strategies targeting STING1, or upstream regulators may therefore be potential alternatives to treat RPGN. © 2023 The Pathological Society of Great Britain and Ireland., (© 2023 The Pathological Society of Great Britain and Ireland.)- Published
- 2023
- Full Text
- View/download PDF
14. Intravascular hemolysis triggers NAFLD characterized by a deregulation of lipid metabolism and lipophagy blockade.
- Author
-
Rayego-Mateos S, Morgado-Pascual JL, García-Caballero C, Lazaro I, Sala-Vila A, Opazo-Rios L, Mas-Fontao S, Egido J, Ruiz-Ortega M, and Moreno JA
- Subjects
- Animals, Mice, Proprotein Convertase 9 metabolism, Lipid Metabolism, Hemolysis, Liver pathology, Hepatocytes pathology, Fatty Acids metabolism, Autophagy, Heme metabolism, Mice, Inbred C57BL, Non-alcoholic Fatty Liver Disease pathology
- Abstract
Intravascular hemolysis is a common feature of different clinical entities, including sickle cell disease and malaria. Chronic hemolytic disorders are associated with hepatic damage; however, it is unknown whether heme disturbs lipid metabolism and promotes liver steatosis, thereby favoring the progression to nonalcoholic fatty liver disease (NAFLD). Using an experimental model of acute intravascular hemolysis, we report here the presence of liver injury in association with microvesicular lipid droplet deposition. Hemolysis promoted serum hyperlipidemia and altered intrahepatic triglyceride fatty acid composition, with increments in oleic, palmitoleic, and palmitic acids. These findings were related to augmented expression of transporters involved in fatty acid uptake (CD36 and MSR1) and deregulation of LDL transport, as demonstrated by decreased levels of LDL receptor and increased PCSK9 expression. Hemolysis also upregulated hepatic enzymes associated with cholesterol biosynthesis (SREBP2, HMGC1, LCAT, SOAT1) and transcription factors regulating lipid metabolism (SREBP1). Increased LC3II/LC3I ratio and p62/SQSTM1 protein levels were reported in mice with intravascular hemolysis and hepatocytes stimulated with heme, indicating a blockade of lipophagy. In cultured hepatocytes, cell pretreatment with the autophagy inductor rapamycin diminished heme-mediated toxicity and accumulation of lipid droplets. In conclusion, intravascular hemolysis enhances liver damage by exacerbating lipid accumulation and blocking the lipophagy pathway, thereby promoting NAFLD. These new findings have a high translational potential as a novel NAFLD-promoting mechanism in individuals suffering from severe hemolysis episodes. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland., (© 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.)
- Published
- 2023
- Full Text
- View/download PDF
15. B- and T-lymphocyte attenuator could be a new player in accelerated atherosclerosis associated with chronic kidney disease.
- Author
-
Dolade N, Rayego-Mateos S, Garcia-Carrasco A, Guerin M, Martín-Ventura JL, Ruiz-Ortega M, Tharaux PL, and Valdivielso JM
- Subjects
- Animals, Humans, Mice, Rats, Down-Regulation, Macrophages, Atherosclerosis metabolism, Plaque, Atherosclerotic, Receptors, Immunologic
- Abstract
Background: In chronic kidney disease (CKD), cardiovascular morbi-mortality is higher than in general population. Atherosclerotic cardiovascular disease is accelerated in CKD, but specific CKD-related risk factors for atherosclerosis are unknown., Methods: CKD patients from the NEFRONA study were used. We performed mRNA array from blood of patients free from atheroma plaque at baseline, with (n=10) and without (n=10) de novo atherosclerotic plaque development 2 years later. Selected mRNA candidates were validated in a bigger sample (n=148). Validated candidates were investigated in vivo in an experimental model of CKD-accelerated atherosclerosis, and in vitro in murine macrophages., Results: mRNA array analysis showed 92 up-regulated and 67 down-regulated mRNAs in samples from CKD patients with de novo plaque development. The functional analysis pointed to a paramount role of the immune response. The validation in a bigger sample confirmed that B- and T-lymphocyte co-inhibitory molecule (BTLA) down-regulation was associated with de novo plaque presence after 2 years. However, BTLA down-regulation was not found to be associated with atherosclerotic progression in patients with plaque already present at baseline. In a model of CKD-accelerated atherosclerosis, mRNA and protein expression levels of BTLA were significantly decreased in blood samples and atheroma plaques. Plaques from animals with CKD were bigger, had more infiltration of inflammatory cells, higher expression of IL6 and IL17 and less presence of collagen than plaques from control animals. Incubation of macrophages with rat uremic serum decreased BTLA expression., Conclusions: BTLA could be a potential biomarker or therapeutic target for atherosclerosis incidence in CKD patients., (© 2023 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2023
- Full Text
- View/download PDF
16. CCN2 Activates RIPK3, NLRP3 Inflammasome, and NRF2/Oxidative Pathways Linked to Kidney Inflammation.
- Author
-
Rayego-Mateos S, Marquez-Exposito L, Basantes P, Tejedor-Santamaria L, Sanz AB, Nguyen TQ, Goldschmeding R, Ortiz A, and Ruiz-Ortega M
- Abstract
Inflammation is a key characteristic of both acute and chronic kidney diseases. Preclinical data suggest the involvement of the NLRP3/Inflammasome, receptor-interacting protein kinase-3 (RIPK3), and NRF2/oxidative pathways in the regulation of kidney inflammation. Cellular communication network factor 2 (CCN2, also called CTGF in the past) is an established fibrotic biomarker and a well-known mediator of kidney damage. CCN2 was shown to be involved in kidney damage through the regulation of proinflammatory and profibrotic responses. However, to date, the potential role of the NLRP3/RIPK3/NRF2 pathways in CCN2 actions has not been evaluated. In experimental acute kidney injury induced with folic acid in mice, CCN2 deficiency diminished renal inflammatory cell infiltration (monocytes/macrophages and T lymphocytes) as well as the upregulation of proinflammatory genes and the activation of NLRP3/Inflammasome-related components and specific cytokine products, such as IL-1β. Moreover, the NRF2/oxidative pathway was deregulated. Systemic administration of CCN2 to C57BL/6 mice induced kidney immune cell infiltration and activated the NLRP3 pathway. RIPK3 deficiency diminished the CCN2-induced renal upregulation of proinflammatory mediators and prevented NLRP3 modulation. These data suggest that CCN2 plays a fundamental role in sterile inflammation and acute kidney injury by modulating the RIKP3/NLRP3/NRF2 inflammatory pathways.
- Published
- 2023
- Full Text
- View/download PDF
17. BET Protein Inhibitor JQ1 Modulates Mitochondrial Dysfunction and Oxidative Stress Induced by Chronic Kidney Disease.
- Author
-
Rayego-Mateos S, Basantes P, Morgado-Pascual JL, Brazal Prieto B, Suarez-Alvarez B, Ortiz A, Lopez-Larrea C, and Ruiz-Ortega M
- Abstract
Among the mechanisms involved in the progression of kidney disease, mitochondrial dysfunction has special relevance. Epigenetic drugs such as inhibitors of extra-terminal domain proteins (iBET) have shown beneficial effects in experimental kidney disease, mainly by inhibiting proliferative and inflammatory responses. The impact of iBET on mitochondrial damage was explored in in vitro studies in renal cells stimulated with TGF-β1 and in vivo in murine unilateral ureteral obstruction (UUO) model of progressive kidney damage. In vitro, JQ1 pretreatment prevented the TGF-β1-induced downregulation of components of the oxidative phosphorylation chain (OXPHOS), such as cytochrome C and CV-ATP5a in human proximal tubular cells. In addition, JQ1 also prevented the altered mitochondrial dynamics by avoiding the increase in the DRP-1 fission factor. In UUO model, renal gene expression levels of cytochrome C and CV-ATP5a as well as protein levels of cytochrome C were reduced These changes were prevented by JQ1 administration. In addition, JQ1 decreased protein levels of the DRP1 fission protein and increased the OPA-1 fusion protein, restoring mitochondrial dynamics. Mitochondria also participate in the maintenance of redox balance. JQ1 restored the gene expression of antioxidant proteins, such as Catalase and Heme oxygenase 1 in TGF-β1-stimulated human proximal tubular cells and in murine obstructed kidneys. Indeed, in tubular cells, JQ1 decreased ROS production induced by stimulation with TGF-β1, as evaluated by MitoSOXTM. iBETs, such as JQ1, improve mitochondrial dynamics, functionality, and oxidative stress in kidney disease.
- Published
- 2023
- Full Text
- View/download PDF
18. Mitochondrial Dysfunction in the Cardio-Renal Axis.
- Author
-
Mendez-Barbero N, Oller J, Sanz AB, Ramos AM, Ortiz A, Ruiz-Ortega M, and Rayego-Mateos S
- Subjects
- Humans, Kidney metabolism, Heart, Mitochondria, Renal Insufficiency, Chronic metabolism, Cardio-Renal Syndrome
- Abstract
Cardiovascular disease (CVD) frequently complicates chronic kidney disease (CKD). The risk of all-cause mortality increases from 20% to 500% in patients who suffer both conditions; this is referred to as the so-called cardio-renal syndrome (CRS). Preclinical studies have described the key role of mitochondrial dysfunction in cardiovascular and renal diseases, suggesting that maintaining mitochondrial homeostasis is a promising therapeutic strategy for CRS. In this review, we explore the malfunction of mitochondrial homeostasis (mitochondrial biogenesis, dynamics, oxidative stress, and mitophagy) and how it contributes to the development and progression of the main vascular pathologies that could be affected by kidney injury and vice versa, and how this knowledge may guide the development of novel therapeutic strategies in CRS.
- Published
- 2023
- Full Text
- View/download PDF
19. Indoxyl sulfate impairs erythropoiesis at BFU-E stage in chronic kidney disease.
- Author
-
Hamza E, Vallejo-Mudarra M, Ouled-Haddou H, García-Caballero C, Guerrero-Hue M, Santier L, Rayego-Mateos S, Larabi IA, Alvarez JC, Garçon L, Massy ZA, Choukroun G, Moreno JA, Metzinger L, and Meuth VM
- Subjects
- Mice, Animals, Humans, Erythropoiesis physiology, Indican metabolism, Indican pharmacology, Erythroid Precursor Cells metabolism, Erythropoietin, Anemia metabolism, Renal Insufficiency, Chronic metabolism
- Abstract
Chronic kidney disease (CKD) is a global health condition characterized by a progressive deterioration of kidney function. It is associated with high serum levels of uremic toxins (UT), such as Indoxyl Sulfate (IS), which may participate in the genesis of several uremic complications. Anemia is one of the major complications in CKD patients that contribute to cardiovascular disease, increase morbi-mortality, and is associated with a deterioration of kidney failure in these patients. Our study aimed to characterize the impact of IS on CKD-related erythropoiesis. Using cellular and pre-clinical models, we studied cellular and molecular effects of IS on the growth and differentiation of erythroid cells. First, we examined the effect of clinically relevant concentrations of IS (up to 250 μM) in the UT7/EPO cell line. IS at 250 μM increased apoptosis of UT7/EPO cells at 48 h compared to the control condition. We confirmed this apoptotic effect of IS in erythropoiesis in human primary CD34
+ cells during the later stages of erythropoiesis. Then, in IS-treated human primary CD34+ cells and in a (5/6 Nx) mice model, a blockage at the burst-forming unit-erythroid (BFU-E) stage of erythropoiesis was also observed. Finally, IS deregulates a number of erythropoietic related genes such as GATA-1, Erythropoietin-Receptor (EPO-R), and β-globin. Our findings suggest that IS could affect cell viability and differentiation of erythroid progenitors by altering erythropoiesis and contributing to the development of anemia in CKD., Competing Interests: Declaration of Competing Interest ZAM reports grants for CKD-REIN and other research projects from Amgen, Baxter, Fresenius Medical Care, GlaxoSmithKline, Merck Sharp and Dohme-Chibret, Sanofi-Genzyme, Lilly, Otsuka and the French government, as well as fees and grants to charities from GlaxoSmithKline, Astra Zeneca, Boehringer Ingelheim, and Bayer; these sources of funding are not necessarily related to the content of the present manuscript. GC reports grants from Amgen, Vifor Pharma, GSK and Astellas (honorarium for consultancy and conferences). Other authors had nothing to disclose., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
20. Vitamin D, Cellular Senescence and Chronic Kidney Diseases: What Is Missing in the Equation?
- Author
-
Martinelli RP, Rayego-Mateos S, Alique M, Márquez-Expósito L, Tejedor-Santamaria L, Ortiz A, González-Parra E, and Ruiz-Ortega M
- Subjects
- Humans, Vitamin D, Kidney, Vitamins therapeutic use, Cellular Senescence, Renal Insufficiency, Chronic complications, Kidney Diseases etiology, Vitamin D Deficiency epidemiology
- Abstract
As life expectancy increases in many countries, the prevalence of age-related diseases also rises. Among these conditions, chronic kidney disease is predicted to become the second cause of death in some countries before the end of the century. An important problem with kidney diseases is the lack of biomarkers to detect early damage or to predict the progression to renal failure. In addition, current treatments only retard kidney disease progression, and better tools are needed. Preclinical research has shown the involvement of the activation of cellular senescence-related mechanisms in natural aging and kidney injury. Intensive research is searching for novel treatments for kidney diseases as well as for anti-aging therapies. In this sense, many experimental shreds of evidence support that treatment with vitamin D or its analogs can exert pleiotropic protective effects in kidney injury. Moreover, vitamin D deficiency has been described in patients with kidney diseases. Here, we review recent evidence about the relationship between vitamin D and kidney diseases, explaining the underlying mechanisms of the effect of vitamin D actions, with particular attention to the modulation of cellular senescence mechanisms.
- Published
- 2023
- Full Text
- View/download PDF
21. Targeting inflammation to treat diabetic kidney disease: the road to 2030.
- Author
-
Rayego-Mateos S, Rodrigues-Diez RR, Fernandez-Fernandez B, Mora-Fernández C, Marchant V, Donate-Correa J, Navarro-González JF, Ortiz A, and Ruiz-Ortega M
- Subjects
- Humans, Inflammation drug therapy, Inflammation complications, Diabetic Nephropathies drug therapy, Diabetic Nephropathies etiology, Diabetic Nephropathies pathology, Diabetes Mellitus, Type 2 complications, Sodium-Glucose Transporter 2 Inhibitors therapeutic use, Renal Insufficiency, Chronic complications
- Abstract
Diabetic kidney disease (DKD) is one of the fastest growing causes of chronic kidney disease and associated morbidity and mortality. Preclinical research has demonstrated the involvement of inflammation in its pathogenesis and in the progression of kidney damage, supporting clinical trials designed to explore anti-inflammatory strategies. However, the recent success of sodium-glucose cotransporter-2 inhibitors and the nonsteroidal mineralocorticoid receptor antagonist finerenone has changed both guidelines and standard of care, rendering obsolete older studies directly targeting inflammatory mediators and the clinical development was discontinued for most anti-inflammatory drugs undergoing clinical trials for DKD in 2016. Given the contribution of inflammation to the pathogenesis of DKD, we review the impact on kidney inflammation of the current standard of care, therapies undergoing clinical trials, or repositioned drugs for DKD. Moreover, we review recent advances in the molecular regulation of inflammation in DKD and discuss potential novel therapeutic strategies with clinical relevance. Finally, we provide a road map for future research aimed at integrating the growing knowledge on inflammation and DKD into clinical practice to foster improvement of patient outcomes., (Copyright © 2022 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
22. Type IV Collagen and SOX9 Are Molecular Targets of BET Inhibition in Experimental Glomerulosclerosis.
- Author
-
Morgado-Pascual JL, Suarez-Alvarez B, Marchant V, Basantes P, Tharaux PL, Ortiz A, Lopez-Larrea C, Ruiz-Ortega M, and Rayego-Mateos S
- Subjects
- Animals, Mice, Cell Cycle Proteins metabolism, Collagen Type IV genetics, Collagen Type IV metabolism, Gene Expression Regulation, Nuclear Proteins genetics, SOX9 Transcription Factor genetics, SOX9 Transcription Factor metabolism, Transcription Factors metabolism, Glomerulonephritis, Kidney Diseases
- Abstract
Progressive glomerulonephritis (GN) is characterized by an excessive accumulation of extracellular (ECM) proteins, mainly type IV collagen (COLIV), in the glomerulus leading to glomerulosclerosis. The current therapeutic approach to GN is suboptimal. Epigenetic drugs could be novel therapeutic options for human disease. Among these drugs, bromodomain and extra-terminal domain (BET) inhibitors (iBETs) have shown beneficial effects in experimental kidney disease and fibrotic disorders. Sex-determining region Y-box 9 (SOX9) is a transcription factor involved in regulating proliferation, migration, and regeneration, but its role in kidney fibrosis is still unclear. We investigated whether iBETs could regulate ECM accumulation in experimental GN and evaluated the role of SOX9 in this process. For this purpose, we tested the iBET JQ1 in mice with anti-glomerular basement membrane nephritis induced by nephrotoxic serum (NTS). In NTS-injected mice, JQ1 treatment reduced glomerular ECM deposition, mainly by inhibiting glomerular COLIV accumulation and Col4a3 gene overexpression. Moreover, chromatin immunoprecipitation assays demonstrated that JQ1 inhibited the recruitment and binding of BRD4 to the Col4a3 promoter and reduced its transcription. Active SOX9 was found in the nuclei of glomerular cells of NTS-injured kidneys, mainly in COLIV-stained regions. JQ1 treatment blocked SOX9 nuclear translocation in injured kidneys. Moreover, in vitro JQ1 blocked TGF-β1-induced SOX9 activation and ECM production in cultured mesangial cells. Additionally, SOX9 gene silencing inhibited ECM production, including COLIV production. Our results demonstrated that JQ1 inhibited SOX9/COLIV, to reduce experimental glomerulosclerosis, supporting further research of iBET as a potential therapeutic option in progressive glomerulosclerosis.
- Published
- 2022
- Full Text
- View/download PDF
23. Role of Toll-like receptor 4 in intravascular hemolysis-mediated injury.
- Author
-
Vázquez-Carballo C, Herencia C, Guerrero-Hue M, García-Caballero C, Rayego-Mateos S, Morgado-Pascual JL, Opazo-Rios L, González-Guerrero C, Vallejo-Mudarra M, Cortegano I, Gaspar ML, de Andrés B, Egido J, and Moreno JA
- Subjects
- Animals, Disease Models, Animal, Heme metabolism, Inflammation, Mice, Mice, Knockout, NF-kappa B metabolism, Phenylhydrazines pharmacology, Sulfonamides, Hemolysis, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 metabolism
- Abstract
Massive intravascular hemolysis is a common characteristic of several pathologies. It is associated with the release of large quantities of heme into the circulation, promoting injury in vulnerable organs, mainly kidney, liver, and spleen. Heme activates Toll-like receptor 4 (TLR4), a key regulator of the inflammatory response; however, the role of TLR4 in hemolysis and whether inhibition of this receptor may protect from heme-mediated injury are unknown. We induced intravascular hemolysis by injection of phenylhydrazine in wildtype and Tlr4-knockout mice. In this model, we analyzed physiological parameters, histological damage, inflammation and cell death in kidney, liver, and spleen. We also evaluated whether heme-mediated-inflammatory effects were prevented by TLR4 inhibition with the compound TAK-242, both in vivo and in vitro. Induction of massive hemolysis elicited acute kidney injury characterized by loss of renal function, morphological alterations of the tubular epithelium, cell death, and inflammation. These pathological effects were significantly ameliorated in the TLR4-deficient mice and in wildtype mice treated with TAK-242. In vitro studies showed that TAK-242 pretreatment reduced heme-mediated inflammation by inhibiting the TLR4/NF-κB (nuclear factor kappa B) axis. However, analysis in liver and spleen indicated that TLR4 deficiency did not protect against the toxic accumulation of heme in these organs. In conclusion, TLR4 is a key molecule involved in the renal inflammatory response triggered by massive intravascular hemolysis. TLR4 inhibition may be a potential therapeutic approach to prevent renal damage in patients suffering from hemolysis. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland., (© 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.)
- Published
- 2022
- Full Text
- View/download PDF
24. The Increase in FGF23 Induced by Calcium Is Partially Dependent on Vitamin D Signaling.
- Author
-
Rayego-Mateos S, Doladé N, García-Carrasco A, Diaz-Tocados JM, Ibarz M, and Valdivielso JM
- Subjects
- Animals, Calcium, Dietary administration & dosage, Mice, Parathyroid Hormone metabolism, Receptors, Calcitriol metabolism, Calcium metabolism, Calcium pharmacology, Fibroblast Growth Factor-23 metabolism, Hypercalcemia metabolism, Vitamin D metabolism
- Abstract
Background: Increased FGF23 levels are an early pathological feature in chronic kidney disease (CKD), causing increased cardiovascular risk. The regulation of FGF23 expression is complex and not completely understood. Thus, Ca
2+ has been shown to induce an increase in FGF23 expression, but whether that increase is mediated by simultaneous changes in parathyroid hormone (PTH) and/or vitamin D is not fully known., Methods: Osteoblast-like cells (OLCs) from vitamin D receptor (VDR)+/+ and VDR-/- mice were incubated with Ca2+ for 18 h. Experimental hypercalcemia was induced by calcium gluconate injection in thyro-parathyroidectomized (T-PTX) VDR+/+ and VDR-/- mice with constant PTH infusion., Results: Inorganic Ca2+ induced an increase in FGF23 gene and protein expression in osteoblast-like cells (OLCs), but the increase was blunted in cells lacking VDR. In T-PTX VDR+/+ and VDR-/- mice with constant PTH levels, hypercalcemia induced an increase in FGF23 levels, but to a lower extent in animals lacking VDR. Similar results were observed in FGF23 expression in bone. Renal and bone 1α-hydroxylase expression was also modulated., Conclusions: Our study demonstrates that Ca2+ can increase FGF23 levels independently of vitamin D and PTH, but part of the physiological increase in FGF23 induced by Ca2+ is mediated by vitamin D signaling.- Published
- 2022
- Full Text
- View/download PDF
25. CCN2 (Cellular Communication Network Factor 2) Deletion Alters Vascular Integrity and Function Predisposing to Aneurysm Formation.
- Author
-
Rodrigues-Díez RR, Tejera-Muñoz A, Esteban V, Steffensen LB, Rodrigues-Díez R, Orejudo M, Rayego-Mateos S, Falke LL, Cannata-Ortiz P, Ortiz A, Egido J, Mallat Z, Briones AM, Bajo MA, Goldschmeding R, and Ruiz-Ortega M
- Subjects
- Angiotensin II pharmacology, Animals, Aorta drug effects, Aortic Aneurysm genetics, Connective Tissue Growth Factor genetics, Disease Models, Animal, Mice, Mice, Knockout, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle metabolism, Signal Transduction drug effects, Up-Regulation drug effects, Aorta metabolism, Aortic Aneurysm metabolism, Connective Tissue Growth Factor metabolism
- Abstract
Background: CCN2 (cellular communication network factor 2) is a matricellular protein involved in cell communication and microenvironmental signaling responses. CCN2 is known to be overexpressed in several cardiovascular diseases, but its role is not completely understood., Methods: Here, CCN2 involvement in aortic wall homeostasis and response to vascular injury was investigated in inducible <i>Ccn2</i>-deficient mice, with induction of vascular damage by infusion of Ang II (angiotensin II; 15 days), which is known to upregulate CCN2 expression in the aorta., Results: Ang II infusion in CCN2-silenced mice lead to 60% mortality within 10 days due to rapid development and rupture of aortic aneurysms, as evidenced by magnetic resonance imaging, echography, and histological examination. <i>Ccn2</i> deletion decreased systolic blood pressure and caused aortic structural and functional changes, including elastin layer disruption, smooth muscle cell alterations, augmented distensibility, and increased metalloproteinase activity, which were aggravated by Ang II administration. Gene ontology analysis of RNA sequencing data identified aldosterone biosynthesis as one of the most enriched terms in CCN2-deficient aortas. Consistently, treatment with the mineralocorticoid receptor antagonist spironolactone before and during Ang II infusion reduced aneurysm formation and mortality, underscoring the importance of the aldosterone pathway in Ang II-induced aorta pathology., Conclusions: CCN2 is critically involved in the functional and structural homeostasis of the aorta and in maintenance of its integrity under Ang II-induced stress, at least, in part, by disruption of the aldosterone pathway. Thus, this study opens new avenues to future studies in disorders associated to vascular pathologies.
- Published
- 2022
- Full Text
- View/download PDF
26. CCN2 Binds to Tubular Epithelial Cells in the Kidney.
- Author
-
Rayego-Mateos S, Morgado-Pascual JL, Lavoz C, Rodrigues-Díez RR, Márquez-Expósito L, Tejera-Muñoz A, Tejedor-Santamaría L, Rubio-Soto I, Marchant V, and Ruiz-Ortega M
- Subjects
- Animals, Epithelial Cells metabolism, ErbB Receptors metabolism, Fibrosis, Kidney metabolism, Mice, Connective Tissue Growth Factor metabolism, Kidney Diseases metabolism
- Abstract
Cellular communication network-2 (CCN2), also called connective tissue growth factor (CTGF), is considered a fibrotic biomarker and has been suggested as a potential therapeutic target for kidney pathologies. CCN2 is a matricellular protein with four distinct structural modules that can exert a dual function as a matricellular protein and as a growth factor. Previous experiments using surface plasmon resonance and cultured renal cells have demonstrated that the C-terminal module of CCN2 (CCN2(IV)) interacts with the epidermal growth factor receptor (EGFR). Moreover, CCN2(IV) activates proinflammatory and profibrotic responses in the mouse kidney. The aim of this paper was to locate the in vivo cellular CCN2/EGFR binding sites in the kidney. To this aim, the C-terminal module CCN2(IV) was labeled with a fluorophore (Cy5), and two different administration routes were employed. Both intraperitoneal and direct intra-renal injection of Cy5-CCN2(IV) in mice demonstrated that CCN2(IV) preferentially binds to the tubular epithelial cells, while no signal was detected in glomeruli. Moreover, co-localization of Cy5-CCN2(IV) binding and activated EGFR was found in tubules. In cultured tubular epithelial cells, live-cell confocal microscopy experiments showed that EGFR gene silencing blocked Cy5-CCN2(IV) binding to tubuloepithelial cells. These data clearly show the existence of CCN2/EGFR binding sites in the kidney, mainly in tubular epithelial cells. In conclusion, these studies show that circulating CCN2(IV) can directly bind and activate tubular cells, supporting the role of CCN2 as a growth factor involved in kidney damage progression.
- Published
- 2022
- Full Text
- View/download PDF
27. Oxidative Stress and Cellular Senescence Are Involved in the Aging Kidney.
- Author
-
Marquez-Exposito L, Tejedor-Santamaria L, Valentijn FA, Tejera-Muñoz A, Rayego-Mateos S, Marchant V, Rodrigues-Diez RR, Rubio-Soto I, Knoppert SN, Ortiz A, Ramos AM, Goldschmeding R, and Ruiz-Ortega M
- Abstract
Chronic kidney disease (CKD) can be considered as a clinical model for premature aging. However, non-invasive biomarkers to detect early kidney damage and the onset of a senescent phenotype are lacking. Most of the preclinical senescence studies in aging have been done in very old mice. Furthermore, the precise characterization and over-time development of age-related senescence in the kidney remain unclear. To address these limitations, the age-related activation of cellular senescence-associated mechanisms and their correlation with early structural changes in the kidney were investigated in 3- to 18-month-old C57BL6 mice. Inflammatory cell infiltration was observed by 12 months, whereas tubular damage and collagen accumulation occurred later. Early activation of cellular-senescence-associated mechanisms was found in 12-month-old mice, characterized by activation of the DNA-damage-response (DDR), mainly in tubular cells; activation of the antioxidant NRF2 pathway; and klotho downregulation. However, induction of tubular-cell-cycle-arrest (CCA) and overexpression of renal senescent-associated secretory phenotype (SASP) components was only found in 18-month-old mice. In aging mice, both inflammation and oxidative stress (marked by elevated lipid peroxidation and NRF2 inactivation) remained increased. These findings support the hypothesis that prolonged DDR and CCA, loss of nephroprotective factors (klotho), and dysfunctional redox regulatory mechanisms (NRF2/antioxidant defense) can be early drivers of age-related kidney-damage progression.
- Published
- 2022
- Full Text
- View/download PDF
28. Molecular Mechanisms of Kidney Injury and Repair.
- Author
-
Rayego-Mateos S, Marquez-Expósito L, Rodrigues-Diez R, Sanz AB, Guiteras R, Doladé N, Rubio-Soto I, Manonelles A, Codina S, Ortiz A, Cruzado JM, Ruiz-Ortega M, and Sola A
- Subjects
- Animals, Biomarkers metabolism, Disease Progression, Humans, Regeneration, Senescence-Associated Secretory Phenotype, Acute Kidney Injury metabolism, Macrophages metabolism, Renal Insufficiency, Chronic metabolism
- Abstract
Chronic kidney disease (CKD) will become the fifth global cause of death by 2040, thus emphasizing the need to better understand the molecular mechanisms of damage and regeneration in the kidney. CKD predisposes to acute kidney injury (AKI) which, in turn, promotes CKD progression. This implies that CKD or the AKI-to-CKD transition are associated with dysfunctional kidney repair mechanisms. Current therapeutic options slow CKD progression but fail to treat or accelerate recovery from AKI and are unable to promote kidney regeneration. Unraveling the cellular and molecular mechanisms involved in kidney injury and repair, including the failure of this process, may provide novel biomarkers and therapeutic tools. We now review the contribution of different molecular and cellular events to the AKI-to-CKD transition, focusing on the role of macrophages in kidney injury, the different forms of regulated cell death and necroinflammation, cellular senescence and the senescence-associated secretory phenotype (SAPS), polyploidization, and podocyte injury and activation of parietal epithelial cells. Next, we discuss key contributors to repair of kidney injury and opportunities for their therapeutic manipulation, with a focus on resident renal progenitor cells, stem cells and their reparative secretome, certain macrophage subphenotypes within the M2 phenotype and senescent cell clearance.
- Published
- 2022
- Full Text
- View/download PDF
29. Epigenetic Modulation of Gremlin-1/NOTCH Pathway in Experimental Crescentic Immune-Mediated Glomerulonephritis.
- Author
-
Tejedor-Santamaria L, Morgado-Pascual JL, Marquez-Exposito L, Suarez-Alvarez B, Rodrigues-Diez RR, Tejera-Muñoz A, Marchant V, Mezzano S, Lopez-Larrea C, Sola A, Fernandez-Juarez GM, Ortiz A, Rayego-Mateos S, and Ruiz-Ortega M
- Abstract
Crescentic glomerulonephritis is a devastating autoimmune disease that without early and properly treatment may rapidly progress to end-stage renal disease and death. Current immunosuppressive treatment provides limited efficacy and an important burden of adverse events. Epigenetic drugs are a source of novel therapeutic tools. Among them, bromodomain and extraterminal domain (BET) inhibitors (iBETs) block the interaction between bromodomains and acetylated proteins, including histones and transcription factors. iBETs have demonstrated protective effects on malignancy, inflammatory disorders and experimental kidney disease. Recently, Gremlin-1 was proposed as a urinary biomarker of disease progression in human anti-neutrophil cytoplasmic antibody (ANCA)-associated crescentic glomerulonephritis. We have now evaluated whether iBETs could regulate Gremlin-1 in experimental anti-glomerular basement membrane nephritis induced by nephrotoxic serum (NTS) in mice, a model resembling human crescentic glomerulonephritis. In NTS-injected mice, the iBET JQ1 inhibited renal Gremlin-1 overexpression and diminished glomerular damage, restoring podocyte numbers. Chromatin immunoprecipitation assay demonstrated BRD4 enrichment of the Grem-1 gene promoter in injured kidneys, consistent with Gremlin-1 epigenetic regulation. Moreover, JQ1 blocked BRD4 binding and inhibited Grem-1 gene transcription. The beneficial effect of iBETs was also mediated by modulation of NOTCH pathway. JQ1 inhibited the gene expression of the NOTCH effectors Hes-1 and Hey-1 in NTS-injured kidneys. Our results further support the role for epigenetic drugs, such as iBETs, in the treatment of rapidly progressive crescentic glomerulonephritis.
- Published
- 2022
- Full Text
- View/download PDF
30. CCN2 Increases TGF-β Receptor Type II Expression in Vascular Smooth Muscle Cells: Essential Role of CCN2 in the TGF-β Pathway Regulation.
- Author
-
Tejera-Muñoz A, Marquez-Exposito L, Tejedor-Santamaría L, Rayego-Mateos S, Orejudo M, Suarez-Álvarez B, López-Larrea C, Ruíz-Ortega M, and Rodrigues-Díez RR
- Subjects
- Animals, Aorta cytology, ErbB Receptors metabolism, Male, Mice, Inbred C57BL, Models, Biological, Phosphorylation, RNA, Messenger genetics, RNA, Messenger metabolism, Smad Proteins metabolism, Mice, Connective Tissue Growth Factor metabolism, Muscle, Smooth, Vascular cytology, Myocytes, Smooth Muscle metabolism, Receptor, Transforming Growth Factor-beta Type II metabolism, Signal Transduction, Transforming Growth Factor beta metabolism
- Abstract
The cellular communication network factor 2 (CCN2/CTGF) has been traditionally described as a mediator of the fibrotic responses induced by other factors including the transforming growth factor β (TGF-β). However, several studies have defined a direct role of CCN2 acting as a growth factor inducing oxidative and proinflammatory responses. The presence of CCN2 and TGF-β together in the cellular context has been described as a requisite to induce a persistent fibrotic response, but the precise mechanisms implicated in this relation are not described yet. Considering the main role of TGF-β receptors (TβR) in the TGF-β pathway activation, our aim was to investigate the effects of CCN2 in the regulation of TβRI and TβRII levels in vascular smooth muscle cells (VSMCs). While no differences were observed in TβRI levels, an increase in TβRII expression at both gene and protein level were found 48 h after stimulation with the C-terminal fragment of CCN2 (CCN2(IV)). Cell pretreatment with a TβRI inhibitor did not modify TβRII increment induced by CCN2(VI), demonstrating a TGF-β-independent response. Secondly, CCN2(IV) rapidly activated the SMAD pathway in VSMCs, this being crucial in the upregulation of TβRII since the preincubation with an SMAD3 inhibitor prevented it. Similarly, pretreatment with the epidermal growth factor receptor (EGFR) inhibitor erlotinib abolished TβRII upregulation, indicating the participation of this receptor in the observed responses. Our findings suggest a direct role of CCN2 maintaining the TGF-β pathway activation by increasing TβRII expression in an EGFR-SMAD dependent manner activation.
- Published
- 2021
- Full Text
- View/download PDF
31. Interplay between extracellular matrix components and cellular and molecular mechanisms in kidney fibrosis.
- Author
-
Rayego-Mateos S, Campillo S, Rodrigues-Diez RR, Tejera-Muñoz A, Marquez-Exposito L, Goldschmeding R, Rodríguez-Puyol D, Calleros L, and Ruiz-Ortega M
- Subjects
- Animals, Biomarkers metabolism, Cell Physiological Phenomena, Fibroblasts metabolism, Fibroblasts pathology, Fibrosis, Humans, Mice, Renal Insufficiency, Chronic diagnosis, Extracellular Matrix metabolism, Kidney metabolism, Kidney pathology, Renal Insufficiency, Chronic metabolism, Renal Insufficiency, Chronic pathology
- Abstract
Chronic kidney disease (CKD) is characterized by pathological accumulation of extracellular matrix (ECM) proteins in renal structures. Tubulointerstitial fibrosis is observed in glomerular diseases as well as in the regeneration failure of acute kidney injury (AKI). Therefore, finding antifibrotic therapies comprises an intensive research field in Nephrology. Nowadays, ECM is not only considered as a cellular scaffold, but also exerts important cellular functions. In this review, we describe the cellular and molecular mechanisms involved in kidney fibrosis, paying particular attention to ECM components, profibrotic factors and cell-matrix interactions. In response to kidney damage, activation of glomerular and/or tubular cells may induce aberrant phenotypes characterized by overproduction of proinflammatory and profibrotic factors, and thus contribute to CKD progression. Among ECM components, matricellular proteins can regulate cell-ECM interactions, as well as cellular phenotype changes. Regarding kidney fibrosis, one of the most studied matricellular proteins is cellular communication network-2 (CCN2), also called connective tissue growth factor (CTGF), currently considered as a fibrotic marker and a potential therapeutic target. Integrins connect the ECM proteins to the actin cytoskeleton and several downstream signaling pathways that enable cells to respond to external stimuli in a coordinated manner and maintain optimal tissue stiffness. In kidney fibrosis, there is an increase in ECM deposition, lower ECM degradation and ECM proteins cross-linking, leading to an alteration in the tissue mechanical properties and their responses to injurious stimuli. A better understanding of these complex cellular and molecular events could help us to improve the antifibrotic therapies for CKD., (© 2021 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2021
- Full Text
- View/download PDF
32. Role of Macrophages and Related Cytokines in Kidney Disease.
- Author
-
Cantero-Navarro E, Rayego-Mateos S, Orejudo M, Tejedor-Santamaria L, Tejera-Muñoz A, Sanz AB, Marquez-Exposito L, Marchant V, Santos-Sanchez L, Egido J, Ortiz A, Bellon T, Rodrigues-Diez RR, and Ruiz-Ortega M
- Abstract
Inflammation is a key characteristic of kidney disease, but this immune response is two-faced. In the acute phase of kidney injury, there is an activation of the immune cells to fight against the insult, contributing to kidney repair and regeneration. However, in chronic kidney diseases (CKD), immune cells that infiltrate the kidney play a deleterious role, actively participating in disease progression, and contributing to nephron loss and fibrosis. Importantly, CKD is a chronic inflammatory disease. In early CKD stages, patients present sub-clinical inflammation, activation of immune circulating cells and therefore, anti-inflammatory strategies have been proposed as a common therapeutic target for renal diseases. Recent studies have highlighted the plasticity of immune cells and the complexity of their functions. Among immune cells, monocytes/macrophages play an important role in all steps of kidney injury. However, the phenotype characterization between human and mice immune cells showed different markers; therefore the extrapolation of experimental studies in mice could not reflect human renal diseases. Here we will review the current information about the characteristics of different macrophage phenotypes, mainly focused on macrophage-related cytokines, with special attention to the chemokine CCL18, and its murine functional homolog CCL8, and the macrophage marker CD163, and their role in kidney pathology., Competing Interests: The 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 © 2021 Cantero-Navarro, Rayego-Mateos, Orejudo, Tejedor-Santamaria, Tejera-Muñoz, Sanz, Marquez-Exposito, Marchant, Santos-Sanchez, Egido, Ortiz, Bellon, Rodrigues-Diez and Ruiz-Ortega.)
- Published
- 2021
- Full Text
- View/download PDF
33. Bisphenol A Modulates Autophagy and Exacerbates Chronic Kidney Damage in Mice.
- Author
-
Priego AR, Parra EG, Mas S, Morgado-Pascual JL, Ruiz-Ortega M, and Rayego-Mateos S
- Subjects
- Animals, Antioxidants pharmacology, Apoptosis drug effects, Autophagy drug effects, Benzhydryl Compounds metabolism, Cell Line, Female, Humans, Kidney metabolism, Kidney pathology, Kidney Diseases metabolism, Kidney Diseases physiopathology, Kidney Tubules drug effects, Male, Mice, Mice, Inbred C57BL, Oxidative Stress drug effects, Phenols metabolism, Renal Insufficiency, Chronic physiopathology, Benzhydryl Compounds adverse effects, Benzhydryl Compounds pharmacology, Phenols adverse effects, Phenols pharmacology, Renal Insufficiency, Chronic metabolism
- Abstract
Background: Bisphenol A (BPA) is a ubiquitous environmental toxin that accumulates in chronic kidney disease (CKD). Our aim was to explore the effect of chronic exposition of BPA in healthy and injured kidney investigating potential mechanisms involved., Methods: In C57Bl/6 mice, administration of BPA (120 mg/kg/day, i.p for 5 days/week) was done for 2 and 5 weeks. To study BPA effect on CKD, a model of subtotal nephrectomy (SNX) combined with BPA administration for 5 weeks was employed. In vitro studies were done in human proximal tubular epithelial cells (HK-2 line)., Results: Chronic BPA administration to healthy mice induces inflammatory infiltration in the kidney, tubular injury and renal fibrosis (assessed by increased collagen deposition). Moreover, in SNX mice BPA exposure exacerbates renal lesions, including overexpression of the tubular damage biomarker Hepatitis A virus cellular receptor 1 ( Havcr-1 /KIM-1). BPA upregulated several proinflammatory genes and increased the antioxidant response [Nuclear factor erythroid 2-related factor 2 ( Nrf2 ), Heme Oxygenase-1 ( Ho-1 ) and NAD(P)H dehydrogenase quinone 1 ( Nqo-1 )] both in healthy and SNX mice. The autophagy process was modulated by BPA, through elevated autophagy-related gene 5 ( Atg5), autophagy-related gene 7 ( Atg7), Microtubule-associated proteins 1A/1B light chain 3B (Map1lc3b/Lc3b) and Beclin-1 gene levels and blockaded the autophagosome maturation and flux (p62 levels). This autophagy deregulation was confirmed in vitro., Conclusions: BPA deregulates autophagy flux and redox protective mechanisms, suggesting a potential mechanism of BPA deleterious effects in the kidney.
- Published
- 2021
- Full Text
- View/download PDF
34. Acute Kidney Injury is Aggravated in Aged Mice by the Exacerbation of Proinflammatory Processes.
- Author
-
Marquez-Exposito L, Tejedor-Santamaria L, Santos-Sanchez L, Valentijn FA, Cantero-Navarro E, Rayego-Mateos S, Rodrigues-Diez RR, Tejera-Muñoz A, Marchant V, Sanz AB, Ortiz A, Goldschmeding R, and Ruiz-Ortega M
- Abstract
Acute kidney injury (AKI) is more frequent in elderly patients. Mechanisms contributing to AKI (tubular cell death, inflammatory cell infiltration, impaired mitochondrial function, and prolonged cell-cycle arrest) have been linked to cellular senescence, a process implicated in regeneration failure and progression to fibrosis. However, the molecular and pathological basis of the age-related increase in AKI incidence is not completely understood. To explore these mechanisms, experimental AKI was induced by folic acid (FA) administration in young (3-months-old) and old (1-year-old) mice, and kidneys were evaluated in the early phase of AKI, at 48 h. Tubular damage score, KIM-1 expression, the recruitment of infiltrating immune cells (mainly neutrophils and macrophages) and proinflammatory gene expression were higher in AKI kidneys of old than of young mice. Tubular cell death in FA-AKI involves several pathways, such as regulated necrosis and apoptosis. Ferroptosis and necroptosis cell-death pathways were upregulated in old AKI kidneys. In contrast, caspase-3 activation was only found in young but not in old mice. Moreover, the antiapoptotic factor BCL-xL was significantly overexpressed in old, injured kidneys, suggesting an age-related apoptosis suppression. AKI kidneys displayed evidence of cellular senescence, such as increased levels of cyclin dependent kinase inhibitors p16ink4a and p21cip1, and of the DNA damage response marker γH2AX. Furthermore, p21cip1 mRNA expression and nuclear staining for p21cip1 and γH2AX were higher in old than in young FA-AKI mice, as well as the expression of senescence-associated secretory phenotype (SASP) components ( Il-6, Tgfb1 , Ctgf, and Serpine1 ). Interestingly, some infiltrating immune cells were p21 or γH2AX positive, suggesting that molecular senescence in the immune cells ("immunosenescence") are involved in the increased severity of AKI in old mice. In contrast, expression of renal protective factors was dramatically downregulated in old AKI mice, including the antiaging factor Klotho and the mitochondrial biogenesis driver PGC-1α. In conclusion, aging resulted in more severe AKI after the exposure to toxic compounds. This increased toxicity may be related to magnification of proinflammatory-related pathways in older mice, including a switch to a proinflammatory cell death (necroptosis) instead of apoptosis, and overactivation of cellular senescence of resident renal cells and infiltrating inflammatory cells., Competing Interests: The 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 © 2021 Marquez-Exposito, Tejedor-Santamaria, Santos-Sanchez, Valentijn, Cantero-Navarro, Rayego-Mateos, Rodrigues-Diez, Tejera-Muñoz, Marchant, Sanz, Ortiz, Goldschmeding and Ruiz-Ortega.)
- Published
- 2021
- Full Text
- View/download PDF
35. Non-Coding RNAs in Kidney Diseases: The Long and Short of Them.
- Author
-
Moreno JA, Hamza E, Guerrero-Hue M, Rayego-Mateos S, García-Caballero C, Vallejo-Mudarra M, Metzinger L, and Metzinger-Le Meuth V
- Subjects
- Animals, Humans, RNA, Messenger genetics, Renal Insufficiency, Chronic genetics, Renal Insufficiency, Chronic metabolism, Gene Expression Regulation, RNA, Long Noncoding genetics, RNA, Messenger metabolism, Renal Insufficiency, Chronic pathology
- Abstract
Recent progress in genomic research has highlighted the genome to be much more transcribed than expected. The formerly so-called junk DNA encodes a miscellaneous group of largely unknown RNA transcripts, which contain the long non-coding RNAs (lncRNAs) family. lncRNAs are instrumental in gene regulation. Moreover, understanding their biological roles in the physiopathology of many diseases, including renal, is a new challenge. lncRNAs regulate the effects of microRNAs (miRNA) on mRNA expression. Understanding the complex crosstalk between lncRNA-miRNA-mRNA is one of the main challenges of modern molecular biology. This review aims to summarize the role of lncRNA on kidney diseases, the molecular mechanisms involved, and their function as emerging prognostic biomarkers for both acute and chronic kidney diseases. Finally, we will also outline new therapeutic opportunities to diminish renal injury by targeting lncRNA with antisense oligonucleotides.
- Published
- 2021
- Full Text
- View/download PDF
36. Renin-angiotensin system and inflammation update.
- Author
-
Cantero-Navarro E, Fernández-Fernández B, Ramos AM, Rayego-Mateos S, Rodrigues-Diez RR, Sánchez-Niño MD, Sanz AB, Ruiz-Ortega M, and Ortiz A
- Subjects
- Angiotensin I genetics, Angiotensin II genetics, Angiotensin-Converting Enzyme 2 genetics, Angiotensin-Converting Enzyme 2 immunology, Animals, Autoimmunity, Blood Pressure genetics, Blood Pressure immunology, Gene Expression Regulation, Humans, Inflammation genetics, Inflammation pathology, Kidney cytology, Kidney immunology, Klotho Proteins genetics, Klotho Proteins immunology, Peptide Fragments genetics, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha genetics, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha immunology, Receptor, Angiotensin, Type 1 genetics, Receptor, Angiotensin, Type 1 immunology, Receptor, Angiotensin, Type 2 genetics, Receptor, Angiotensin, Type 2 immunology, Renin-Angiotensin System genetics, Signal Transduction, T-Lymphocytes cytology, T-Lymphocytes immunology, Water-Electrolyte Balance genetics, Angiotensin I immunology, Angiotensin II immunology, Inflammation immunology, Peptide Fragments immunology, Renin-Angiotensin System immunology, Water-Electrolyte Balance immunology
- Abstract
The most classical view of the renin-angiotensin system (RAS) emphasizes its role as an endocrine regulator of sodium balance and blood pressure. However, it has long become clear that the RAS has pleiotropic actions that contribute to organ damage, including modulation of inflammation. Angiotensin II (Ang II) activates angiotensin type 1 receptors (AT1R) to promote an inflammatory response and organ damage. This represents the pathophysiological basis for the successful use of RAS blockers to prevent and treat kidney and heart disease. However, other RAS components could have a built-in capacity to brake proinflammatory responses. Angiotensin type 2 receptor (AT2R) activation can oppose AT1R actions, such as vasodilatation, but its involvement in modulation of inflammation has not been conclusively proven. Angiotensin-converting enzyme 2 (ACE2) can process Ang II to generate angiotensin-(1-7) (Ang-(1-7)), that activates the Mas receptor to exert predominantly anti-inflammatory responses depending on the context. We now review recent advances in the understanding of the interaction of the RAS with inflammation. Specific topics in which novel information became available recently include intracellular angiotensin receptors; AT1R posttranslational modifications by tissue transglutaminase (TG2) and anti-AT1R autoimmunity; RAS modulation of lymphoid vessels and T lymphocyte responses, especially of Th17 and Treg responses; interactions with toll-like receptors (TLRs), programmed necrosis, and regulation of epigenetic modulators (e.g. microRNAs and bromodomain and extraterminal domain (BET) proteins). We additionally discuss an often overlooked effect of the RAS on inflammation which is the downregulation of anti-inflammatory factors such as klotho, peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α), transient receptor potential ankyrin 1 (TRPA1), SNF-related serine/threonine-protein kinase (SNRK), serine/threonine-protein phosphatase 6 catalytic subunit (Ppp6C) and n-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP). Both transcription factors, such as nuclear factor κB (NF-κB), and epigenetic regulators, such as miRNAs are involved in downmodulation of anti-inflammatory responses. A detailed analysis of pathways and targets for downmodulation of anti-inflammatory responses constitutes a novel frontier in RAS research., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
37. [Interleukin-17A: Possible mediator and therapeutic target in hypertension].
- Author
-
Rodrigues-Diez RR, Tejera-Muñoz A, Orejudo M, Marquez-Exposito L, Santos L, Rayego-Mateos S, Cantero-Navarro E, Tejedor-Santamaria L, Marchant V, Ortiz A, Egido J, Mezzano S, Selgas R, Navarro-González JF, Valdivielso JM, Lavoz C, and Ruiz-Ortega M
- Subjects
- Animals, Humans, Mice, Hypertension drug therapy, Hypertension etiology, Interleukin-17 antagonists & inhibitors, Interleukin-17 physiology
- Abstract
Interleukin-17A (IL-17A) is a proinflammatory cytokine produced by cells of the immune system, predominantly Th17 lymphocytes and γδ lymphocytes. In this paper, we review the role of IL-17A in the pathogenesis of hypertension and target organ damage. Studies in mice have shown that IL-17A increases blood pressure, probably by acting on multiple levels. Furthermore, IL-17A plasma concentrations are already elevated in patients with mild or moderate hypertension. Preclinical studies on arterial hypertension have detected IL-17A-producing cells in target organs such as the heart, vessels and kidneys. Patients with hypertensive nephrosclerosis show kidney infiltration by Th17 lymphocytes and γδ lymphocytes that express IL-17A. In addition, in experimental models of hypertension, blocking IL-17A by genetic strategies, or using neutralising antibodies, lowers blood pressure by acting on the vascular wall and tubule sodium transport and reduces damage to target organs. As a whole, the data presented in this review suggest that IL-17A participates in the regulation of blood pressure and in the genesis and maintenance of arterial hypertension, and may constitute a therapeutic target in the future., (Copyright © 2021 Sociedad Española de Nefrología. Published by Elsevier España, S.L.U. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
38. Toll-Like Receptors in Acute Kidney Injury.
- Author
-
Vázquez-Carballo C, Guerrero-Hue M, García-Caballero C, Rayego-Mateos S, Opazo-Ríos L, Morgado-Pascual JL, Herencia-Bellido C, Vallejo-Mudarra M, Cortegano I, Gaspar ML, de Andrés B, Egido J, and Moreno JA
- Subjects
- Animals, Disease Progression, Humans, Kidney metabolism, Kidney pathology, Renal Insufficiency, Chronic metabolism, Renal Insufficiency, Chronic therapy, Risk Factors, Toll-Like Receptor 4 metabolism, Acute Kidney Injury metabolism, Acute Kidney Injury therapy, Renal Replacement Therapy methods, Toll-Like Receptors metabolism
- Abstract
Acute kidney injury (AKI) is an important health problem, affecting 13.3 million individuals/year. It is associated with increased mortality, mainly in low- and middle-income countries, where renal replacement therapy is limited. Moreover, survivors show adverse long-term outcomes, including increased risk of developing recurrent AKI bouts, cardiovascular events, and chronic kidney disease. However, there are no specific treatments to decrease the adverse consequences of AKI. Epidemiological and preclinical studies show the pathological role of inflammation in AKI, not only at the acute phase but also in the progression to chronic kidney disease. Toll-like receptors (TLRs) are key regulators of the inflammatory response and have been associated to many cellular processes activated during AKI. For that reason, a number of anti-inflammatory agents targeting TLRs have been analyzed in preclinical studies to decrease renal damage during AKI. In this review, we updated recent knowledge about the role of TLRs, mainly TLR4, in the initiation and development of AKI as well as novel compounds targeting these molecules to diminish kidney injury associated to this pathological condition.
- Published
- 2021
- Full Text
- View/download PDF
39. Deletion of delta-like 1 homologue accelerates renal inflammation by modulating the Th17 immune response.
- Author
-
Marquez-Exposito L, Rodrigues-Diez RR, Rayego-Mateos S, Fierro-Fernandez M, Rodrigues-Diez R, Orejudo M, Santos-Sanchez L, Blanco EM, Laborda J, Mezzano S, Lamas S, Lavoz C, and Ruiz-Ortega M
- Subjects
- Animals, Calcium-Binding Proteins immunology, Cell Cycle Proteins genetics, Cell Cycle Proteins immunology, Inflammation genetics, Inflammation immunology, Inflammation pathology, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins immunology, Kidney pathology, Kidney Diseases genetics, Kidney Diseases pathology, Mice, Th17 Cells pathology, Ureteral Obstruction genetics, Ureteral Obstruction immunology, Ureteral Obstruction pathology, Calcium-Binding Proteins deficiency, Gene Deletion, Immunity, Cellular, Kidney immunology, Kidney Diseases immunology, Th17 Cells immunology
- Abstract
Preclinical studies have demonstrated that activation of the NOTCH pathway plays a key role in the pathogenesis of kidney damage. There is currently no information on the role of the Delta-like homologue 1 (DLK1), a NOTCH inhibitor, in the regulation of renal damage. Here, we investigated the contribution of DLK1 to experimental renal damage and the underlying molecular mechanisms. Using a Dlk1-null mouse model in the experimental renal damage of unilateral ureteral obstruction, we found activation of NOTCH, as shown by increased nuclear translocation of the NOTCH1 intracellular domain, and upregulation of Dlk2/hey-1 expression compared to wild-type (WT) littermates. NOTCH1 over-activation in Dlk1-null injured kidneys was associated with a higher inflammatory response, characterized by infiltration of inflammatory cells, mainly CD4/IL17A + lymphocytes, and activation of the Th17 immune response. Furthermore, pharmacological NOTCH blockade inhibited the transcription factors controlling Th17 differentiation and gene expression of the Th17 effector cytokine IL-17A and other related-inflammatory factors, linked to a diminution of inflammation in the injured kidneys. We propose that the non-canonical NOTCH ligand DLK1 acts as a NOTCH antagonist in renal injury regulating the Th17-mediated inflammatory response., (© 2020 Federation of American Societies for Experimental Biology.)
- Published
- 2021
- Full Text
- View/download PDF
40. Protective Role of Nrf2 in Renal Disease.
- Author
-
Guerrero-Hue M, Rayego-Mateos S, Vázquez-Carballo C, Palomino-Antolín A, García-Caballero C, Opazo-Rios L, Morgado-Pascual JL, Herencia C, Mas S, Ortiz A, Rubio-Navarro A, Egea J, Villalba JM, Egido J, and Moreno JA
- Abstract
Chronic kidney disease (CKD) is one of the fastest-growing causes of death and is predicted to become by 2040 the fifth global cause of death. CKD is characterized by increased oxidative stress and chronic inflammation. However, therapies to slow or prevent CKD progression remain an unmet need. Nrf2 (nuclear factor erythroid 2-related factor 2) is a transcription factor that plays a key role in protection against oxidative stress and regulation of the inflammatory response. Consequently, the use of compounds targeting Nrf2 has generated growing interest for nephrologists. Pre-clinical and clinical studies have demonstrated that Nrf2-inducing strategies prevent CKD progression and protect from acute kidney injury (AKI). In this article, we review current knowledge on the protective mechanisms mediated by Nrf2 against kidney injury, novel therapeutic strategies to induce Nrf2 activation, and the status of ongoing clinical trials targeting Nrf2 in renal diseases.
- Published
- 2020
- Full Text
- View/download PDF
41. Statins: Could an old friend help in the fight against COVID-19?
- Author
-
Rodrigues-Diez RR, Tejera-Muñoz A, Marquez-Exposito L, Rayego-Mateos S, Santos Sanchez L, Marchant V, Tejedor Santamaria L, Ramos AM, Ortiz A, Egido J, and Ruiz-Ortega M
- Subjects
- Animals, Betacoronavirus isolation & purification, COVID-19, Coronavirus Infections immunology, Coronavirus Infections virology, Cytokines immunology, Drug Repositioning, Humans, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Pandemics, Pneumonia, Viral immunology, Pneumonia, Viral virology, SARS-CoV-2, Virus Internalization drug effects, COVID-19 Drug Treatment, Coronavirus Infections drug therapy, Hydroxymethylglutaryl-CoA Reductase Inhibitors administration & dosage, Pneumonia, Viral drug therapy
- Abstract
The COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has overwhelmed healthcare systems requiring the rapid development of treatments, at least, to reduce COVID-19 severity. Drug repurposing offers a fast track. Here, we discuss the potential beneficial effects of statins in COVID-19 patients based on evidence that they may target virus receptors, replication, degradation, and downstream responses in infected cells, addressing both basic research and epidemiological information. Briefly, statins could modulate virus entry, acting on the SARS-CoV-2 receptors, ACE2 and CD147, and/or lipid rafts engagement. Statins, by inducing autophagy activation, could regulate virus replication or degradation, exerting protective effects. The well-known anti-inflammatory properties of statins, by blocking several molecular mechanisms, including NF-κB and NLRP3 inflammasomes, could limit the "cytokine storm" in severe COVID-19 patients which is linked to fatal outcome. Finally, statin moderation of coagulation response activation may also contribute to improving COVID-19 outcomes. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc., (© 2020 The British Pharmacological Society.)
- Published
- 2020
- Full Text
- View/download PDF
42. IL-17A as a Potential Therapeutic Target for Patients on Peritoneal Dialysis.
- Author
-
Marchant V, Tejera-Muñoz A, Marquez-Expósito L, Rayego-Mateos S, Rodrigues-Diez RR, Tejedor L, Santos-Sanchez L, Egido J, Ortiz A, Valdivielso JM, Fraser DJ, López-Cabrera M, Selgas R, and Ruiz-Ortega M
- Subjects
- Dialysis Solutions chemistry, Fibrosis pathology, Humans, Neovascularization, Pathologic genetics, Neovascularization, Pathologic pathology, Peritoneum metabolism, Peritoneum pathology, Renal Insufficiency, Chronic pathology, Renal Insufficiency, Chronic therapy, Fibrosis genetics, Interleukin-17 genetics, Peritoneal Dialysis, Renal Insufficiency, Chronic genetics
- Abstract
Chronic kidney disease (CKD) is a health problem reaching epidemic proportions. There is no cure for CKD, and patients may progress to end-stage renal disease (ESRD). Peritoneal dialysis (PD) is a current replacement therapy option for ESRD patients until renal transplantation can be achieved. One important problem in long-term PD patients is peritoneal membrane failure. The mechanisms involved in peritoneal damage include activation of the inflammatory and immune responses, associated with submesothelial immune infiltrates, angiogenesis, loss of the mesothelial layer due to cell death and mesothelial to mesenchymal transition, and collagen accumulation in the submesothelial compact zone. These processes lead to fibrosis and loss of peritoneal membrane function. Peritoneal inflammation and membrane failure are strongly associated with additional problems in PD patients, mainly with a very high risk of cardiovascular disease. Among the inflammatory mediators involved in peritoneal damage, cytokine IL-17A has recently been proposed as a potential therapeutic target for chronic inflammatory diseases, including CKD. Although IL-17A is the hallmark cytokine of Th17 immune cells, many other cells can also produce or secrete IL-17A. In the peritoneum of PD patients, IL-17A-secreting cells comprise Th17 cells, γδ T cells, mast cells, and neutrophils. Experimental studies demonstrated that IL-17A blockade ameliorated peritoneal damage caused by exposure to PD fluids. This article provides a comprehensive review of recent advances on the role of IL-17A in peritoneal membrane injury during PD and other PD-associated complications.
- Published
- 2020
- Full Text
- View/download PDF
43. TRAF3 Modulation: Novel Mechanism for the Anti-inflammatory Effects of the Vitamin D Receptor Agonist Paricalcitol in Renal Disease.
- Author
-
Rayego-Mateos S, Morgado-Pascual JL, Valdivielso JM, Sanz AB, Bosch-Panadero E, Rodrigues-Díez RR, Egido J, Ortiz A, González-Parra E, and Ruiz-Ortega M
- Subjects
- Animals, Cells, Cultured, Cytokine TWEAK pharmacology, Female, Humans, Male, Mice, Mice, Inbred C57BL, NF-kappa B antagonists & inhibitors, NF-kappa B physiology, Receptors, Calcitriol physiology, Signal Transduction drug effects, TNF Receptor-Associated Factor 3 analysis, Anti-Inflammatory Agents pharmacology, Ergocalciferols pharmacology, Kidney Failure, Chronic drug therapy, Receptors, Calcitriol agonists, TNF Receptor-Associated Factor 3 physiology
- Abstract
Background: CKD leads to vitamin D deficiency. Treatment with vitamin D receptor agonists (VDRAs) may have nephroprotective and anti-inflammatory actions, but their mechanisms of action are poorly understood., Methods: Modulation of the noncanonical NF- κ B2 pathway and its component TNF receptor-associated factor 3 (TRAF3) by the VDRA paricalcitol was studied in PBMCs from patients with ESKD, cytokine-stimulated cells, and preclinical kidney injury models., Results: In PBMCs isolated from patients with ESKD, TRAF3 protein levels were lower than in healthy controls. This finding was associated with evidence of noncanonical NF- κ B2 activation and a proinflammatory state. However, PBMCs from patients with ESKD treated with paricalcitol did not exhibit these features. Experiments in cultured cells confirmed the link between TRAF3 and NF- κ B2/inflammation. Decreased TRAF3 ubiquitination in K48-linked chains and cIAP1-TRAF3 interaction mediated the mechanisms of paricalcitol action.TRAF3 overexpression by CRISPR/Cas9 technology mimicked VDRA's effects. In a preclinical model of kidney injury, paricalcitol inhibited renal NF- κ B2 activation and decreased renal inflammation. In VDR knockout mice with renal injury, paricalcitol prevented TRAF3 downregulation and NF- κ B2-dependent gene upregulation, suggesting a VDR-independent anti-inflammatory effect of paricalcitol., Conclusions: These data suggest the anti-inflammatory actions of paricalcitol depend on TRAF3 modulation and subsequent inhibition of the noncanonical NF- κ B2 pathway, identifying a novel mechanism for VDRA's effects. Circulating TRAF3 levels could be a biomarker of renal damage associated with the inflammatory state., (Copyright © 2020 by the American Society of Nephrology.)
- Published
- 2020
- Full Text
- View/download PDF
44. New therapeutic targets in chronic kidney disease progression and renal fibrosis.
- Author
-
Rayego-Mateos S and Valdivielso JM
- Subjects
- Animals, Disease Progression, Fibrosis pathology, Fibrosis therapy, Gastrointestinal Microbiome, Humans, Inflammation physiopathology, Oxidative Stress, Renal Insufficiency, Chronic physiopathology, Inflammation therapy, Molecular Targeted Therapy, Renal Insufficiency, Chronic therapy
- Abstract
Introduction: The current therapeutic armamentarium to prevent chronic kidney disease (CKD) progression is limited to the control of blood pressure and in diabetic patients, the strict control of glucose levels. Current research is primarily focused on the reduction of inflammation and fibrosis at different levels., Areas Covered: This article examines the latest progress in this field and places an emphasis on inflammation, oxidative stress, and fibrosis. New therapeutic targets are described and evidence from experimental and clinical studies is summarized. We performed a search in Medline for articles published over the last 10 years., Expert Opinion: The search for therapeutic targets of renal inflammation is hindered by an incomplete understanding of the pathophysiology. The determination of the specific inducers of inflammation in the kidney is an area of heightened potential. Prevention of the progression of renal fibrosis by blocking TGF-β signaling has been unsuccessful, but the investigation of signaling pathways involved in late stages of fibrosis progression could yield improved results. Preventive strategies such as the modification of microbiota-inducers of uremic toxins involved in CKD progression is a promising field because of the interaction between the gut microbiota and the renal system.
- Published
- 2020
- Full Text
- View/download PDF
45. Pathogenic Pathways and Therapeutic Approaches Targeting Inflammation in Diabetic Nephropathy.
- Author
-
Rayego-Mateos S, Morgado-Pascual JL, Opazo-Ríos L, Guerrero-Hue M, García-Caballero C, Vázquez-Carballo C, Mas S, Sanz AB, Herencia C, Mezzano S, Gómez-Guerrero C, Moreno JA, and Egido J
- Subjects
- Animals, Diabetic Nephropathies drug therapy, Diabetic Nephropathies immunology, Humans, Anti-Inflammatory Agents therapeutic use, Diabetic Nephropathies metabolism, Hypoglycemic Agents therapeutic use, Immunosuppressive Agents therapeutic use
- Abstract
Diabetic nephropathy (DN) is associated with an increased morbidity and mortality, resulting in elevated cost for public health systems. DN is the main cause of chronic kidney disease (CKD) and its incidence increases the number of patients that develop the end-stage renal disease (ESRD). There are growing epidemiological and preclinical evidence about the close relationship between inflammatory response and the occurrence and progression of DN. Several anti-inflammatory strategies targeting specific inflammatory mediators (cell adhesion molecules, chemokines and cytokines) and intracellular signaling pathways have shown beneficial effects in experimental models of DN, decreasing proteinuria and renal lesions. A number of inflammatory molecules have been shown useful to identify diabetic patients at high risk of developing renal complications. In this review, we focus on the key role of inflammation in the genesis and progression of DN, with a special interest in effector molecules and activated intracellular pathways leading to renal damage, as well as a comprehensive update of new therapeutic strategies targeting inflammation to prevent and/or retard renal injury.
- Published
- 2020
- Full Text
- View/download PDF
46. Targeting the progression of chronic kidney disease.
- Author
-
Ruiz-Ortega M, Rayego-Mateos S, Lamas S, Ortiz A, and Rodrigues-Diez RR
- Subjects
- Disease Progression, Humans, Renal Insufficiency, Chronic pathology, Renal Insufficiency, Chronic complications, Renal Insufficiency, Chronic therapy
- Abstract
Chronic kidney disease (CKD) is a devastating condition that is reaching epidemic levels owing to the increasing prevalence of diabetes mellitus, hypertension and obesity, as well as ageing of the population. Regardless of the underlying aetiology, CKD is slowly progressive and leads to irreversible nephron loss, end-stage renal disease and/or premature death. Factors that contribute to CKD progression include parenchymal cell loss, chronic inflammation, fibrosis and reduced regenerative capacity of the kidney. Current therapies have limited effectiveness and only delay disease progression, underscoring the need to develop novel therapeutic approaches to either stop or reverse progression. Preclinical studies have identified several approaches that reduce fibrosis in experimental models, including targeting cytokines, transcription factors, developmental and signalling pathways and epigenetic modulators, particularly microRNAs. Some of these nephroprotective strategies are now being tested in clinical trials. Lessons learned from the failure of clinical studies of transforming growth factor β1 (TGFβ1) blockade underscore the need for alternative approaches to CKD therapy, as strategies that target a single pathogenic process may result in unexpected negative effects on simultaneously occurring processes. Additional promising avenues include preventing tubular cell injury and anti-fibrotic therapies that target activated myofibroblasts, the main collagen-producing cells.
- Published
- 2020
- Full Text
- View/download PDF
47. Special Issue "Diabetic Nephropathy: Diagnosis, Prevention and Treatment".
- Author
-
Ruiz-Ortega M, Rodrigues-Diez RR, Lavoz C, and Rayego-Mateos S
- Abstract
Diabetic nephropathy (DN) is the main cause of end-stage renal disease. DN is a complex disease mediated by genetic and environmental factors, and many cellular and molecular mechanisms are involved in renal damage in diabetes. There are no biomarkers that reflect the severity of the underlying renal histopathological changes and can effectively predict the progression of renal damage and stratify the risk of DN among individuals with diabetes mellitus. Current therapeutic strategies are based on the strict control of glucose and blood pressure levels and, although there are new anti-diabetic drugs, these treatments only retard renal damage progression, being necessary novel therapies. In this Special Issue, there are several comprehensive reviews and interesting original papers covering all these topics, which would be of interest to the growing number of readers of the Journal of Clinical Medicine .
- Published
- 2020
- Full Text
- View/download PDF
48. Could IL-17A Be a Novel Therapeutic Target in Diabetic Nephropathy?
- Author
-
Lavoz C, Rayego-Mateos S, Orejudo M, Opazo-Ríos L, Marchant V, Marquez-Exposito L, Tejera-Muñoz A, Navarro-González JF, Droguett A, Ortiz A, Egido J, Mezzano S, Rodrigues-Diez RR, and Ruiz-Ortega M
- Abstract
Chronic kidney disease has become a major medical issue in recent years due to its high prevalence worldwide, its association with premature mortality, and its social and economic implications. A number of patients gradually progress to end-stage renal disease (ESRD), requiring then dialysis and kidney transplantation. Currently, approximately 40% of patients with diabetes develop kidney disease, making it the most prevalent cause of ESRD. Thus, more effective therapies for diabetic nephropathy are needed. In preclinical studies of diabetes, anti-inflammatory therapeutic strategies have been used to protect the kidneys. Recent evidence supports that immune cells play an active role in the pathogenesis of diabetic nephropathy. Th17 immune cells and their effector cytokine IL-17A have recently emerged as promising targets in several clinical conditions, including renal diseases. Here, we review current knowledge regarding the involvement of Th17/IL-17A in the genesis of diabetic renal injury, as well as the rationale behind targeting IL-17A as an additional therapy in patients with diabetic nephropathy., Competing Interests: The authors declare no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
49. Molecular Regulation of Notch Signaling by Gremlin.
- Author
-
Marquez-Exposito L, Cantero-Navarro E, R Rodrigues-Diez R, Orejudo M, Tejera-Muñoz A, Tejedor L, Rayego-Mateos S, Rández-Carbayo J, Santos-Sanchez L, Mezzano S, Lavoz C, and Ruiz-Ortega M
- Subjects
- Animals, Humans, Kidney metabolism, Kidney pathology, Transforming Growth Factor beta metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism, Intercellular Signaling Peptides and Proteins metabolism, Receptors, Notch metabolism, Signal Transduction
- Abstract
Gremlin is a member of the TGF-β superfamily that can act as a BMP antagonist, and recently, has been described as a ligand of the vascular endothelial growth factor receptor 2 (VEGFR2). Gremlin shares properties with the Notch signaling pathway. Both participate in embryonic development and are reactivated in pathological conditions. Gremlin is emerging as a potential therapeutic target and biomarker of renal diseases. Here we review the role of the Gremlin-VEGFR2 axis in renal damage and downstream signaling mechanisms, such as Notch pathway.
- Published
- 2020
- Full Text
- View/download PDF
50. Bromodomain and Extraterminal Proteins as Novel Epigenetic Targets for Renal Diseases.
- Author
-
Morgado-Pascual JL, Rayego-Mateos S, Tejedor L, Suarez-Alvarez B, and Ruiz-Ortega M
- Abstract
Epigenetic mechanisms, especially DNA methylation and histone modifications, are dynamic processes that regulate the gene expression transcriptional program in normal and diseased states. The bromodomain and extraterminal (BET) protein family (BRD2, BRD3, BRD4, and BRDT) are epigenetic readers that, via bromodomains, regulate gene transcription by binding to acetylated lysine residues on histones and master transcriptional factors. Experimental data have demonstrated the involvement of some BET proteins in many pathological conditions, including tumor development, infections, autoimmunity, and inflammation. Selective bromodomain inhibitors are epigenetic drugs that block the interaction between BET proteins and acetylated proteins, thus exerting beneficial effects. Recent data have described the beneficial effect of BET inhibition on experimental renal diseases. Emerging evidence underscores the importance of environmental modifications in the origin of pathological features in chronic kidney diseases (CKD). Several cellular processes such as oxidation, metabolic disorders, cytokines, inflammation, or accumulated uremic toxins may induce epigenetic modifications that regulate key processes involved in renal damage and in other pathological conditions observed in CKD patients. Here, we review how targeting bromodomains in BET proteins may regulate essential processes involved in renal diseases and in associated complications found in CKD patients, such as cardiovascular damage, highlighting the potential of epigenetic therapeutic strategies against BET proteins for CKD treatment and associated risks., (Copyright © 2019 Morgado-Pascual, Rayego-Mateos, Tejedor, Suarez-Alvarez and Ruiz-Ortega.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.