61 results on '"Moritz OL"'
Search Results
2. Guidelines for the use and interpretation of assays for monitoring autophagy (4th edition)
- Author
-
Klionsky, DJ, Abdel-Aziz, AK, Abdelfatah, S, Abdellatif, M, Abdoli, A, Abel, S, Abeliovich, H, Abildgaard, MH, Abudu, YP, Acevedo-Arozena, A, Adamopoulos, IE, Adeli, K, Adolph, TE, Adornetto, A, Aflaki, E, Agam, G, Agarwal, A, Aggarwal, BB, Agnello, M, Agostinis, P, Agrewala, JN, Agrotis, A, Aguilar, PV, Ahmad, ST, Ahmed, ZM, Ahumada-Castro, U, Aits, S, Aizawa, S, Akkoc, Y, Akoumianaki, T, Akpinar, HA, Al-Abd, AM, Al-Akra, L, Al-Gharaibeh, A, Alaoui-Jamali, MA, Alberti, S, Alcocer-Gómez, E, Alessandri, C, Ali, M, Alim Al-Bari, MA, Aliwaini, S, Alizadeh, J, Almacellas, E, Almasan, A, Alonso, A, Alonso, GD, Altan-Bonnet, N, Altieri, DC, Álvarez, ÉMC, Alves, S, Alves da Costa, C, Alzaharna, MM, Amadio, M, Amantini, C, Amaral, C, Ambrosio, S, Amer, AO, Ammanathan, V, An, Z, Andersen, SU, Andrabi, SA, Andrade-Silva, M, Andres, AM, Angelini, S, Ann, D, Anozie, UC, Ansari, MY, Antas, P, Antebi, A, Antón, Z, Anwar, T, Apetoh, L, Apostolova, N, Araki, T, Araki, Y, Arasaki, K, Araújo, WL, Araya, J, Arden, C, Arévalo, M-A, Arguelles, S, Arias, E, Arikkath, J, Arimoto, H, Ariosa, AR, Armstrong-James, D, Arnauné-Pelloquin, L, Aroca, A, Arroyo, DS, Arsov, I, Artero, R, Asaro, DML, Aschner, M, Ashrafizadeh, M, Ashur-Fabian, O, Atanasov, AG, Au, AK, Auberger, P, Auner, HW, Aurelian, L, Autelli, R, Avagliano, L, Ávalos, Y, Aveic, S, Aveleira, CA, Avin-Wittenberg, T, Aydin, Y, Ayton, S, Ayyadevara, S, Azzopardi, M, Baba, M, Backer, JM, Backues, SK, Bae, D-H, Bae, O-N, Bae, SH, Baehrecke, EH, Baek, A, Baek, S-H, Baek, SH, Bagetta, G, Bagniewska-Zadworna, A, Bai, H, Bai, J, Bai, X, Bai, Y, Bairagi, N, Baksi, S, Balbi, T, Baldari, CT, Balduini, W, Ballabio, A, Ballester, M, Balazadeh, S, Balzan, R, Bandopadhyay, R, Banerjee, S, Bánréti, Á, Bao, Y, Baptista, MS, Baracca, A, Barbati, C, Bargiela, A, Barilà, D, Barlow, PG, Barmada, SJ, Barreiro, E, Barreto, GE, Bartek, J, Bartel, B, Bartolome, A, Barve, GR, Basagoudanavar, SH, Bassham, DC, Bast, RC, Basu, A, Batoko, H, Batten, I, Baulieu, EE, Baumgarner, BL, Bayry, J, Beale, R, Beau, I, Beaumatin, F, Bechara, LRG, Beck, GR, Beers, MF, Begun, J, Behrends, C, Behrens, GMN, Bei, R, Bejarano, E, Bel, S, Behl, C, Belaid, A, Belgareh-Touzé, N, Bellarosa, C, Belleudi, F, Belló Pérez, M, Bello-Morales, R, Beltran, JSDO, Beltran, S, Benbrook, DM, Bendorius, M, Benitez, BA, Benito-Cuesta, I, Bensalem, J, Berchtold, MW, Berezowska, S, Bergamaschi, D, Bergami, M, Bergmann, A, Berliocchi, L, Berlioz-Torrent, C, Bernard, A, Berthoux, L, Besirli, CG, Besteiro, S, Betin, VM, Beyaert, R, Bezbradica, JS, Bhaskar, K, Bhatia-Kissova, I, Bhattacharya, R, Bhattacharya, S, Bhattacharyya, S, Bhuiyan, MS, Bhutia, SK, Bi, L, Bi, X, Biden, TJ, Bijian, K, Billes, VA, Binart, N, Bincoletto, C, Birgisdottir, AB, Bjorkoy, G, Blanco, G, Blas-Garcia, A, Blasiak, J, Blomgran, R, Blomgren, K, Blum, JS, Boada-Romero, E, Boban, M, Boesze-Battaglia, K, Boeuf, P, Boland, B, Bomont, P, Bonaldo, P, Bonam, SR, Bonfili, L, Bonifacino, JS, Boone, BA, Bootman, MD, Bordi, M, Borner, C, Bornhauser, BC, Borthakur, G, Bosch, J, Bose, S, Botana, LM, Botas, J, Boulanger, CM, Boulton, ME, Bourdenx, M, Bourgeois, B, Bourke, NM, Bousquet, G, Boya, P, Bozhkov, PV, Bozi, LHM, Bozkurt, TO, Brackney, DE, Brandts, CH, Braun, RJ, Braus, GH, Bravo-Sagua, R, Bravo-San Pedro, JM, Brest, P, Bringer, M-A, Briones-Herrera, A, Broaddus, VC, Brodersen, P, Brodsky, JL, Brody, SL, Bronson, PG, Bronstein, JM, Brown, CN, Brown, RE, Brum, PC, Brumell, JH, Brunetti-Pierri, N, Bruno, D, Bryson-Richardson, RJ, Bucci, C, Buchrieser, C, Bueno, M, Buitrago-Molina, LE, Buraschi, S, Buch, S, Buchan, JR, Buckingham, EM, Budak, H, Budini, M, Bultynck, G, Burada, F, Burgoyne, JR, Burón, MI, Bustos, V, Büttner, S, Butturini, E, Byrd, A, Cabas, I, Cabrera-Benitez, S, Cadwell, K, Cai, J, Cai, L, Cai, Q, Cairó, M, Calbet, JA, Caldwell, GA, Caldwell, KA, Call, JA, Calvani, R, Calvo, AC, Calvo-Rubio Barrera, M, Camara, NO, Camonis, JH, Camougrand, N, Campanella, M, Campbell, EM, Campbell-Valois, F-X, Campello, S, Campesi, I, Campos, JC, Camuzard, O, Cancino, J, Candido de Almeida, D, Canesi, L, Caniggia, I, Canonico, B, Cantí, C, Cao, B, Caraglia, M, Caramés, B, Carchman, EH, Cardenal-Muñoz, E, Cardenas, C, Cardenas, L, Cardoso, SM, Carew, JS, Carle, GF, Carleton, G, Carloni, S, Carmona-Gutierrez, D, Carneiro, LA, Carnevali, O, Carosi, JM, Carra, S, Carrier, A, Carrier, L, Carroll, B, Carter, AB, Carvalho, AN, Casanova, M, Casas, C, Casas, J, Cassioli, C, Castillo, EF, Castillo, K, Castillo-Lluva, S, Castoldi, F, Castori, M, Castro, AF, Castro-Caldas, M, Castro-Hernandez, J, Castro-Obregon, S, Catz, SD, Cavadas, C, Cavaliere, F, Cavallini, G, Cavinato, M, Cayuela, ML, Cebollada Rica, P, Cecarini, V, Cecconi, F, Cechowska-Pasko, M, Cenci, S, Ceperuelo-Mallafré, V, Cerqueira, JJ, Cerutti, JM, Cervia, D, Cetintas, VB, Cetrullo, S, Chae, H-J, Chagin, AS, Chai, C-Y, Chakrabarti, G, Chakrabarti, O, Chakraborty, T, Chami, M, Chamilos, G, Chan, DW, Chan, EYW, Chan, ED, Chan, HYE, Chan, HH, Chan, H, Chan, MTV, Chan, YS, Chandra, PK, Chang, C-P, Chang, C, Chang, H-C, Chang, K, Chao, J, Chapman, T, Charlet-Berguerand, N, Chatterjee, S, Chaube, SK, Chaudhary, A, Chauhan, S, Chaum, E, Checler, F, Cheetham, ME, Chen, C-S, Chen, G-C, Chen, J-F, Chen, LL, Chen, L, Chen, M, Chen, M-K, Chen, N, Chen, Q, Chen, R-H, Chen, S, Chen, W, Chen, X-M, Chen, X-W, Chen, X, Chen, Y, Chen, Y-G, Chen, Y-J, Chen, Y-Q, Chen, ZS, Chen, Z, Chen, Z-H, Chen, ZJ, Cheng, H, Cheng, J, Cheng, S-Y, Cheng, W, Cheng, X, Cheng, X-T, Cheng, Y, Cheng, Z, Cheong, H, Cheong, JK, Chernyak, BV, Cherry, S, Cheung, CFR, Cheung, CHA, Cheung, K-H, Chevet, E, Chi, RJ, Chiang, AKS, Chiaradonna, F, Chiarelli, R, Chiariello, M, Chica, N, Chiocca, S, Chiong, M, Chiou, S-H, Chiramel, AI, Chiurchiù, V, Cho, D-H, Choe, S-K, Choi, AMK, Choi, ME, Choudhury, KR, Chow, NS, Chu, CT, Chua, JP, Chua, JJE, Chung, H, Chung, KP, Chung, S, Chung, S-H, Chung, Y-L, Cianfanelli, V, Ciechomska, IA, Cifuentes, M, Cinque, L, Cirak, S, Cirone, M, Clague, MJ, Clarke, R, Clementi, E, Coccia, EM, Codogno, P, Cohen, E, Cohen, MM, Colasanti, T, Colasuonno, F, Colbert, RA, Colell, A, Čolić, M, Coll, NS, Collins, MO, Colombo, MI, Colón-Ramos, DA, Combaret, L, Comincini, S, Cominetti, MR, Consiglio, A, Conte, A, Conti, F, Contu, VR, Cookson, MR, Coombs, KM, Coppens, I, Corasaniti, MT, Corkery, DP, Cordes, N, Cortese, K, Costa, MDC, Costantino, S, Costelli, P, Coto-Montes, A, Crack, PJ, Crespo, JL, Criollo, A, Crippa, V, Cristofani, R, Csizmadia, T, Cuadrado, A, Cui, B, Cui, J, Cui, Y, Culetto, E, Cumino, AC, Cybulsky, AV, Czaja, MJ, Czuczwar, SJ, D'Adamo, S, D'Amelio, M, D'Arcangelo, D, D'Lugos, AC, D'Orazi, G, da Silva, JA, Dafsari, HS, Dagda, RK, Dagdas, Y, Daglia, M, Dai, X, Dai, Y, Dal Col, J, Dalhaimer, P, Dalla Valle, L, Dallenga, T, Dalmasso, G, Damme, M, Dando, I, Dantuma, NP, Darling, AL, Das, H, Dasarathy, S, Dasari, SK, Dash, S, Daumke, O, Dauphinee, AN, Davies, JS, Dávila, VA, Davis, RJ, Davis, T, Dayalan Naidu, S, De Amicis, F, De Bosscher, K, De Felice, F, De Franceschi, L, De Leonibus, C, de Mattos Barbosa, MG, De Meyer, GRY, De Milito, A, De Nunzio, C, De Palma, C, De Santi, M, De Virgilio, C, De Zio, D, Debnath, J, DeBosch, BJ, Decuypere, J-P, Deehan, MA, Deflorian, G, DeGregori, J, Dehay, B, Del Rio, G, Delaney, JR, Delbridge, LMD, Delorme-Axford, E, Delpino, MV, Demarchi, F, Dembitz, V, Demers, ND, Deng, H, Deng, Z, Dengjel, J, Dent, P, Denton, D, DePamphilis, ML, Der, CJ, Deretic, V, Descoteaux, A, Devis, L, Devkota, S, Devuyst, O, Dewson, G, Dharmasivam, M, Dhiman, R, di Bernardo, D, Di Cristina, M, Di Domenico, F, Di Fazio, P, Di Fonzo, A, Di Guardo, G, Di Guglielmo, GM, Di Leo, L, Di Malta, C, Di Nardo, A, Di Rienzo, M, Di Sano, F, Diallinas, G, Diao, J, Diaz-Araya, G, Díaz-Laviada, I, Dickinson, JM, Diederich, M, Dieudé, M, Dikic, I, Ding, S, Ding, W-X, Dini, L, Dinić, J, Dinic, M, Dinkova-Kostova, AT, Dionne, MS, Distler, JHW, Diwan, A, Dixon, IMC, Djavaheri-Mergny, M, Dobrinski, I, Dobrovinskaya, O, Dobrowolski, R, Dobson, RCJ, Đokić, J, Dokmeci Emre, S, Donadelli, M, Dong, B, Dong, X, Dong, Z, Dorn Ii, GW, Dotsch, V, Dou, H, Dou, J, Dowaidar, M, Dridi, S, Drucker, L, Du, A, Du, C, Du, G, Du, H-N, Du, L-L, du Toit, A, Duan, S-B, Duan, X, Duarte, SP, Dubrovska, A, Dunlop, EA, Dupont, N, Durán, RV, Dwarakanath, BS, Dyshlovoy, SA, Ebrahimi-Fakhari, D, Eckhart, L, Edelstein, CL, Efferth, T, Eftekharpour, E, Eichinger, L, Eid, N, Eisenberg, T, Eissa, NT, Eissa, S, Ejarque, M, El Andaloussi, A, El-Hage, N, El-Naggar, S, Eleuteri, AM, El-Shafey, ES, Elgendy, M, Eliopoulos, AG, Elizalde, MM, Elks, PM, Elsasser, H-P, Elsherbiny, ES, Emerling, BM, Emre, NCT, Eng, CH, Engedal, N, Engelbrecht, A-M, Engelsen, AST, Enserink, JM, Escalante, R, Esclatine, A, Escobar-Henriques, M, Eskelinen, E-L, Espert, L, Eusebio, M-O, Fabrias, G, Fabrizi, C, Facchiano, A, Facchiano, F, Fadeel, B, Fader, C, Faesen, AC, Fairlie, WD, Falcó, A, Falkenburger, BH, Fan, D, Fan, J, Fan, Y, Fang, EF, Fang, Y, Fanto, M, Farfel-Becker, T, Faure, M, Fazeli, G, Fedele, AO, Feldman, AM, Feng, D, Feng, J, Feng, L, Feng, Y, Feng, W, Fenz Araujo, T, Ferguson, TA, Fernández, ÁF, Fernandez-Checa, JC, Fernández-Veledo, S, Fernie, AR, Ferrante, AW, Ferraresi, A, Ferrari, MF, Ferreira, JCB, Ferro-Novick, S, Figueras, A, Filadi, R, Filigheddu, N, Filippi-Chiela, E, Filomeni, G, Fimia, GM, Fineschi, V, Finetti, F, Finkbeiner, S, Fisher, EA, Fisher, PB, Flamigni, F, Fliesler, SJ, Flo, TH, Florance, I, Florey, O, Florio, T, Fodor, E, Follo, C, Fon, EA, Forlino, A, Fornai, F, Fortini, P, Fracassi, A, Fraldi, A, Franco, B, Franco, R, Franconi, F, Frankel, LB, Friedman, SL, Fröhlich, LF, Frühbeck, G, Fuentes, JM, Fujiki, Y, Fujita, N, Fujiwara, Y, Fukuda, M, Fulda, S, Furic, L, Furuya, N, Fusco, C, Gack, MU, Gaffke, L, Galadari, S, Galasso, A, Galindo, MF, Gallolu Kankanamalage, S, Galluzzi, L, Galy, V, Gammoh, N, Gan, B, Ganley, IG, Gao, F, Gao, H, Gao, M, Gao, P, Gao, S-J, Gao, W, Gao, X, Garcera, A, Garcia, MN, Garcia, VE, García-Del Portillo, F, Garcia-Escudero, V, Garcia-Garcia, A, Garcia-Macia, M, García-Moreno, D, Garcia-Ruiz, C, García-Sanz, P, Garg, AD, Gargini, R, Garofalo, T, Garry, RF, Gassen, NC, Gatica, D, Ge, L, Ge, W, Geiss-Friedlander, R, Gelfi, C, Genschik, P, Gentle, IE, Gerbino, V, Gerhardt, C, Germain, K, Germain, M, Gewirtz, DA, Ghasemipour Afshar, E, Ghavami, S, Ghigo, A, Ghosh, M, Giamas, G, Giampietri, C, Giatromanolaki, A, Gibson, GE, Gibson, SB, Ginet, V, Giniger, E, Giorgi, C, Girao, H, Girardin, SE, Giridharan, M, Giuliano, S, Giulivi, C, Giuriato, S, Giustiniani, J, Gluschko, A, Goder, V, Goginashvili, A, Golab, J, Goldstone, DC, Golebiewska, A, Gomes, LR, Gomez, R, Gómez-Sánchez, R, Gomez-Puerto, MC, Gomez-Sintes, R, Gong, Q, Goni, FM, González-Gallego, J, Gonzalez-Hernandez, T, Gonzalez-Polo, RA, Gonzalez-Reyes, JA, González-Rodríguez, P, Goping, IS, Gorbatyuk, MS, Gorbunov, NV, Görgülü, K, Gorojod, RM, Gorski, SM, Goruppi, S, Gotor, C, Gottlieb, RA, Gozes, I, Gozuacik, D, Graef, M, Gräler, MH, Granatiero, V, Grasso, D, Gray, JP, Green, DR, Greenhough, A, Gregory, SL, Griffin, EF, Grinstaff, MW, Gros, F, Grose, C, Gross, AS, Gruber, F, Grumati, P, Grune, T, Gu, X, Guan, J-L, Guardia, CM, Guda, K, Guerra, F, Guerri, C, Guha, P, Guillén, C, Gujar, S, Gukovskaya, A, Gukovsky, I, Gunst, J, Günther, A, Guntur, AR, Guo, C, Guo, H, Guo, L-W, Guo, M, Gupta, P, Gupta, SK, Gupta, S, Gupta, VB, Gupta, V, Gustafsson, AB, Gutterman, DD, H B, R, Haapasalo, A, Haber, JE, Hać, A, Hadano, S, Hafrén, AJ, Haidar, M, Hall, BS, Halldén, G, Hamacher-Brady, A, Hamann, A, Hamasaki, M, Han, W, Hansen, M, Hanson, PI, Hao, Z, Harada, M, Harhaji-Trajkovic, L, Hariharan, N, Haroon, N, Harris, J, Hasegawa, T, Hasima Nagoor, N, Haspel, JA, Haucke, V, Hawkins, WD, Hay, BA, Haynes, CM, Hayrabedyan, SB, Hays, TS, He, C, He, Q, He, R-R, He, Y-W, He, Y-Y, Heakal, Y, Heberle, AM, Hejtmancik, JF, Helgason, GV, Henkel, V, Herb, M, Hergovich, A, Herman-Antosiewicz, A, Hernández, A, Hernandez, C, Hernandez-Diaz, S, Hernandez-Gea, V, Herpin, A, Herreros, J, Hervás, JH, Hesselson, D, Hetz, C, Heussler, VT, Higuchi, Y, Hilfiker, S, Hill, JA, Hlavacek, WS, Ho, EA, Ho, IHT, Ho, PW-L, Ho, S-L, Ho, WY, Hobbs, GA, Hochstrasser, M, Hoet, PHM, Hofius, D, Hofman, P, Höhn, A, Holmberg, CI, Hombrebueno, JR, Yi-Ren Hong, C-WH, Hooper, LV, Hoppe, T, Horos, R, Hoshida, Y, Hsin, I-L, Hsu, H-Y, Hu, B, Hu, D, Hu, L-F, Hu, MC, Hu, R, Hu, W, Hu, Y-C, Hu, Z-W, Hua, F, Hua, J, Hua, Y, Huan, C, Huang, C, Huang, H, Huang, K, Huang, MLH, Huang, R, Huang, S, Huang, T, Huang, X, Huang, YJ, Huber, TB, Hubert, V, Hubner, CA, Hughes, SM, Hughes, WE, Humbert, M, Hummer, G, Hurley, JH, Hussain, S, Hussey, PJ, Hutabarat, M, Hwang, H-Y, Hwang, S, Ieni, A, Ikeda, F, Imagawa, Y, Imai, Y, Imbriano, C, Imoto, M, Inman, DM, Inoki, K, Iovanna, J, Iozzo, RV, Ippolito, G, Irazoqui, JE, Iribarren, P, Ishaq, M, Ishikawa, M, Ishimwe, N, Isidoro, C, Ismail, N, Issazadeh-Navikas, S, Itakura, E, Ito, D, Ivankovic, D, Ivanova, S, Iyer, AKV, Izquierdo, JM, Izumi, M, Jäättelä, M, Jabir, MS, Jackson, WT, Jacobo-Herrera, N, Jacomin, A-C, Jacquin, E, Jadiya, P, Jaeschke, H, Jagannath, C, Jakobi, AJ, Jakobsson, J, Janji, B, Jansen-Dürr, P, Jansson, PJ, Jantsch, J, Januszewski, S, Jassey, A, Jean, S, Jeltsch-David, H, Jendelova, P, Jenny, A, Jensen, TE, Jessen, N, Jewell, JL, Ji, J, Jia, L, Jia, R, Jiang, L, Jiang, Q, Jiang, R, Jiang, T, Jiang, X, Jiang, Y, Jimenez-Sanchez, M, Jin, E-J, Jin, F, Jin, H, Jin, L, Jin, M, Jin, S, Jo, E-K, Joffre, C, Johansen, T, Johnson, GVW, Johnston, SA, Jokitalo, E, Jolly, MK, Joosten, LAB, Jordan, J, Joseph, B, Ju, D, Ju, J-S, Ju, J, Juárez, E, Judith, D, Juhász, G, Jun, Y, Jung, CH, Jung, S-C, Jung, YK, Jungbluth, H, Jungverdorben, J, Just, S, Kaarniranta, K, Kaasik, A, Kabuta, T, Kaganovich, D, Kahana, A, Kain, R, Kajimura, S, Kalamvoki, M, Kalia, M, Kalinowski, DS, Kaludercic, N, Kalvari, I, Kaminska, J, Kaminskyy, VO, Kanamori, H, Kanasaki, K, Kang, C, Kang, R, Kang, SS, Kaniyappan, S, Kanki, T, Kanneganti, T-D, Kanthasamy, AG, Kanthasamy, A, Kantorow, M, Kapuy, O, Karamouzis, MV, Karim, MR, Karmakar, P, Katare, RG, Kato, M, Kaufmann, SHE, Kauppinen, A, Kaushal, GP, Kaushik, S, Kawasaki, K, Kazan, K, Ke, P-Y, Keating, DJ, Keber, U, Kehrl, JH, Keller, KE, Keller, CW, Kemper, JK, Kenific, CM, Kepp, O, Kermorgant, S, Kern, A, Ketteler, R, Keulers, TG, Khalfin, B, Khalil, H, Khambu, B, Khan, SY, Khandelwal, VKM, Khandia, R, Kho, W, Khobrekar, NV, Khuansuwan, S, Khundadze, M, Killackey, SA, Kim, D, Kim, DR, Kim, D-H, Kim, D-E, Kim, EY, Kim, E-K, Kim, H-R, Kim, H-S, Hyung-Ryong Kim, Kim, JH, Kim, JK, Kim, J-H, Kim, J, Kim, KI, Kim, PK, Kim, S-J, Kimball, SR, Kimchi, A, Kimmelman, AC, Kimura, T, King, MA, Kinghorn, KJ, Kinsey, CG, Kirkin, V, Kirshenbaum, LA, Kiselev, SL, Kishi, S, Kitamoto, K, Kitaoka, Y, Kitazato, K, Kitsis, RN, Kittler, JT, Kjaerulff, O, Klein, PS, Klopstock, T, Klucken, J, Knævelsrud, H, Knorr, RL, Ko, BCB, Ko, F, Ko, J-L, Kobayashi, H, Kobayashi, S, Koch, I, Koch, JC, Koenig, U, Kögel, D, Koh, YH, Koike, M, Kohlwein, SD, Kocaturk, NM, Komatsu, M, König, J, Kono, T, Kopp, BT, Korcsmaros, T, Korkmaz, G, Korolchuk, VI, Korsnes, MS, Koskela, A, Kota, J, Kotake, Y, Kotler, ML, Kou, Y, Koukourakis, MI, Koustas, E, Kovacs, AL, Kovács, T, Koya, D, Kozako, T, Kraft, C, Krainc, D, Krämer, H, Krasnodembskaya, AD, Kretz-Remy, C, Kroemer, G, Ktistakis, NT, Kuchitsu, K, Kuenen, S, Kuerschner, L, Kukar, T, Kumar, A, Kumar, D, Kumar, S, Kume, S, Kumsta, C, Kundu, CN, Kundu, M, Kunnumakkara, AB, Kurgan, L, Kutateladze, TG, Kutlu, O, Kwak, S, Kwon, HJ, Kwon, TK, Kwon, YT, Kyrmizi, I, La Spada, A, Labonté, P, Ladoire, S, Laface, I, Lafont, F, Lagace, DC, Lahiri, V, Lai, Z, Laird, AS, Lakkaraju, A, Lamark, T, Lan, S-H, Landajuela, A, Lane, DJR, Lane, JD, Lang, CH, Lange, C, Langel, Ü, Langer, R, Lapaquette, P, Laporte, J, LaRusso, NF, Lastres-Becker, I, Lau, WCY, Laurie, GW, Lavandero, S, Law, BYK, Law, HK-W, Layfield, R, Le, W, Le Stunff, H, Leary, AY, Lebrun, J-J, Leck, LYW, Leduc-Gaudet, J-P, Lee, C, Lee, C-P, Lee, D-H, Lee, EB, Lee, EF, Lee, GM, Lee, H-J, Lee, HK, Lee, JM, Lee, JS, Lee, J-A, Lee, J-Y, Lee, JH, Lee, M, Lee, MG, Lee, MJ, Lee, M-S, Lee, SY, Lee, S-J, Lee, SB, Lee, WH, Lee, Y-R, Lee, Y-H, Lee, Y, Lefebvre, C, Legouis, R, Lei, YL, Lei, Y, Leikin, S, Leitinger, G, Lemus, L, Leng, S, Lenoir, O, Lenz, G, Lenz, HJ, Lenzi, P, León, Y, Leopoldino, AM, Leschczyk, C, Leskelä, S, Letellier, E, Leung, C-T, Leung, PS, Leventhal, JS, Levine, B, Lewis, PA, Ley, K, Li, B, Li, D-Q, Li, J, Li, K, Li, L, Li, M, Li, P-L, Li, M-Q, Li, Q, Li, S, Li, T, Li, W, Li, X, Li, Y-P, Li, Y, Li, Z, Lian, J, Liang, C, Liang, Q, Liang, W, Liang, Y, Liao, G, Liao, L, Liao, M, Liao, Y-F, Librizzi, M, Lie, PPY, Lilly, MA, Lim, HJ, Lima, TRR, Limana, F, Lin, C, Lin, C-W, Lin, D-S, Lin, F-C, Lin, JD, Lin, KM, Lin, K-H, Lin, L-T, Lin, P-H, Lin, Q, Lin, S, Lin, S-J, Lin, W, Lin, X, Lin, Y-X, Lin, Y-S, Linden, R, Lindner, P, Ling, S-C, Lingor, P, Linnemann, AK, Liou, Y-C, Lipinski, MM, Lipovšek, S, Lira, VA, Lisiak, N, Liton, PB, Liu, C, Liu, C-H, Liu, C-F, Liu, CH, Liu, F, Liu, H, Liu, H-S, Liu, H-F, Liu, J, Liu, L, Liu, M, Liu, Q, Liu, W, Liu, X-H, Liu, X, Liu, Y, Livingston, JA, Lizard, G, Lizcano, JM, Ljubojevic-Holzer, S, LLeonart, ME, Llobet-Navàs, D, Llorente, A, Lo, CH, Lobato-Márquez, D, Long, Q, Long, YC, Loos, B, Loos, JA, López, MG, López-Doménech, G, López-Guerrero, JA, López-Jiménez, AT, López-Pérez, Ó, López-Valero, I, Lorenowicz, MJ, Lorente, M, Lorincz, P, Lossi, L, Lotersztajn, S, Lovat, PE, Lovell, JF, Lovy, A, Lőw, P, Lu, G, Lu, H, Lu, J-H, Lu, J-J, Lu, M, Lu, S, Luciani, A, Lucocq, JM, Ludovico, P, Luftig, MA, Luhr, M, Luis-Ravelo, D, Lum, JJ, Luna-Dulcey, L, Lund, AH, Lund, VK, Lünemann, JD, Lüningschrör, P, Luo, H, Luo, R, Luo, S, Luo, Z, Luparello, C, Lüscher, B, Luu, L, Lyakhovich, A, Lyamzaev, KG, Lystad, AH, Lytvynchuk, L, Ma, AC, Ma, C, Ma, M, Ma, N-F, Ma, Q-H, Ma, X, Ma, Y, Ma, Z, MacDougald, OA, Macian, F, MacIntosh, GC, MacKeigan, JP, Macleod, KF, Maday, S, Madeo, F, Madesh, M, Madl, T, Madrigal-Matute, J, Maeda, A, Maejima, Y, Magarinos, M, Mahavadi, P, Maiani, E, Maiese, K, Maiti, P, Maiuri, MC, Majello, B, Major, MB, Makareeva, E, Malik, F, Mallilankaraman, K, Malorni, W, Maloyan, A, Mammadova, N, Man, GCW, Manai, F, Mancias, JD, Mandelkow, E-M, Mandell, MA, Manfredi, AA, Manjili, MH, Manjithaya, R, Manque, P, Manshian, BB, Manzano, R, Manzoni, C, Mao, K, Marchese, C, Marchetti, S, Marconi, AM, Marcucci, F, Mardente, S, Mareninova, OA, Margeta, M, Mari, M, Marinelli, S, Marinelli, O, Mariño, G, Mariotto, S, Marshall, RS, Marten, MR, Martens, S, Martin, APJ, Martin, KR, Martin, S, Martín-Segura, A, Martín-Acebes, MA, Martin-Burriel, I, Martin-Rincon, M, Martin-Sanz, P, Martina, JA, Martinet, W, Martinez, A, Martinez, J, Martinez Velazquez, M, Martinez-Lopez, N, Martinez-Vicente, M, Martins, DO, Martins, JO, Martins, WK, Martins-Marques, T, Marzetti, E, Masaldan, S, Masclaux-Daubresse, C, Mashek, DG, Massa, V, Massieu, L, Masson, GR, Masuelli, L, Masyuk, AI, Masyuk, TV, Matarrese, P, Matheu, A, Matoba, S, Matsuzaki, S, Mattar, P, Matte, A, Mattoscio, D, Mauriz, JL, Mauthe, M, Mauvezin, C, Maverakis, E, Maycotte, P, Mayer, J, Mazzoccoli, G, Mazzoni, C, Mazzulli, JR, McCarty, N, McDonald, C, McGill, MR, McKenna, SL, McLaughlin, B, McLoughlin, F, McNiven, MA, McWilliams, TG, Mechta-Grigoriou, F, Medeiros, TC, Medina, DL, Megeney, LA, Megyeri, K, Mehrpour, M, Mehta, JL, Meijer, AJ, Meijer, AH, Mejlvang, J, Meléndez, A, Melk, A, Memisoglu, G, Mendes, AF, Meng, D, Meng, F, Meng, T, Menna-Barreto, R, Menon, MB, Mercer, C, Mercier, AE, Mergny, J-L, Merighi, A, Merkley, SD, Merla, G, Meske, V, Mestre, AC, Metur, SP, Meyer, C, Meyer, H, Mi, W, Mialet-Perez, J, Miao, J, Micale, L, Miki, Y, Milan, E, Milczarek, M, Miller, DL, Miller, SI, Miller, S, Millward, SW, Milosevic, I, Minina, EA, Mirzaei, H, Mirzaei, HR, Mirzaei, M, Mishra, A, Mishra, N, Mishra, PK, Misirkic Marjanovic, M, Misasi, R, Misra, A, Misso, G, Mitchell, C, Mitou, G, Miura, T, Miyamoto, S, Miyazaki, M, Miyazaki, T, Miyazawa, K, Mizushima, N, Mogensen, TH, Mograbi, B, Mohammadinejad, R, Mohamud, Y, Mohanty, A, Mohapatra, S, Möhlmann, T, Mohmmed, A, Moles, A, Moley, KH, Molinari, M, Mollace, V, Møller, AB, Mollereau, B, Mollinedo, F, Montagna, C, Monteiro, MJ, Montella, A, Montes, LR, Montico, B, Mony, VK, Monzio Compagnoni, G, Moore, MN, Moosavi, MA, Mora, AL, Mora, M, Morales-Alamo, D, Moratalla, R, Moreira, PI, Morelli, E, Moreno, S, Moreno-Blas, D, Moresi, V, Morga, B, Morgan, AH, Morin, F, Morishita, H, Moritz, OL, Moriyama, M, Moriyasu, Y, Morleo, M, Morselli, E, Moruno-Manchon, JF, Moscat, J, Mostowy, S, Motori, E, Moura, AF, Moustaid-Moussa, N, Mrakovcic, M, Muciño-Hernández, G, Mukherjee, A, Mukhopadhyay, S, Mulcahy Levy, JM, Mulero, V, Muller, S, Münch, C, Munjal, A, Munoz-Canoves, P, Muñoz-Galdeano, T, Münz, C, Murakawa, T, Muratori, C, Murphy, BM, Murphy, JP, Murthy, A, Myöhänen, TT, Mysorekar, IU, Mytych, J, Nabavi, SM, Nabissi, M, Nagy, P, Nah, J, Nahimana, A, Nakagawa, I, Nakamura, K, Nakatogawa, H, Nandi, SS, Nanjundan, M, Nanni, M, Napolitano, G, Nardacci, R, Narita, M, Nassif, M, Nathan, I, Natsumeda, M, Naude, RJ, Naumann, C, Naveiras, O, Navid, F, Nawrocki, ST, Nazarko, TY, Nazio, F, Negoita, F, Neill, T, Neisch, AL, Neri, LM, Netea, MG, Neubert, P, Neufeld, TP, Neumann, D, Neutzner, A, Newton, PT, Ney, PA, Nezis, IP, Ng, CCW, Ng, TB, Nguyen, HTT, Nguyen, LT, Ni, H-M, Ní Cheallaigh, C, Ni, Z, Nicolao, MC, Nicoli, F, Nieto-Diaz, M, Nilsson, P, Ning, S, Niranjan, R, Nishimune, H, Niso-Santano, M, Nixon, RA, Nobili, A, Nobrega, C, Noda, T, Nogueira-Recalde, U, Nolan, TM, Nombela, I, Novak, I, Novoa, B, Nozawa, T, Nukina, N, Nussbaum-Krammer, C, Nylandsted, J, O'Donovan, TR, O'Leary, SM, O'Rourke, EJ, O'Sullivan, MP, O'Sullivan, TE, Oddo, S, Oehme, I, Ogawa, M, Ogier-Denis, E, Ogmundsdottir, MH, Ogretmen, B, Oh, GT, Oh, S-H, Oh, YJ, Ohama, T, Ohashi, Y, Ohmuraya, M, Oikonomou, V, Ojha, R, Okamoto, K, Okazawa, H, Oku, M, Oliván, S, Oliveira, JMA, Ollmann, M, Olzmann, JA, Omari, S, Omary, MB, Önal, G, Ondrej, M, Ong, S-B, Ong, S-G, Onnis, A, Orellana, JA, Orellana-Muñoz, S, Ortega-Villaizan, MDM, Ortiz-Gonzalez, XR, Ortona, E, Osiewacz, HD, Osman, A-HK, Osta, R, Otegui, MS, Otsu, K, Ott, C, Ottobrini, L, Ou, J-HJ, Outeiro, TF, Oynebraten, I, Ozturk, M, Pagès, G, Pahari, S, Pajares, M, Pajvani, UB, Pal, R, Paladino, S, Pallet, N, Palmieri, M, Palmisano, G, Palumbo, C, Pampaloni, F, Pan, L, Pan, Q, Pan, W, Pan, X, Panasyuk, G, Pandey, R, Pandey, UB, Pandya, V, Paneni, F, Pang, SY, Panzarini, E, Papademetrio, DL, Papaleo, E, Papinski, D, Papp, D, Park, EC, Park, HT, Park, J-M, Park, J-I, Park, JT, Park, J, Park, SC, Park, S-Y, Parola, AH, Parys, JB, Pasquier, A, Pasquier, B, Passos, JF, Pastore, N, Patel, HH, Patschan, D, Pattingre, S, Pedraza-Alva, G, Pedraza-Chaverri, J, Pedrozo, Z, Pei, G, Pei, J, Peled-Zehavi, H, Pellegrini, JM, Pelletier, J, Peñalva, MA, Peng, D, Peng, Y, Penna, F, Pennuto, M, Pentimalli, F, Pereira, CM, Pereira, GJS, Pereira, LC, Pereira de Almeida, L, Perera, ND, Pérez-Lara, Á, Perez-Oliva, AB, Pérez-Pérez, ME, Periyasamy, P, Perl, A, Perrotta, C, Perrotta, I, Pestell, RG, Petersen, M, Petrache, I, Petrovski, G, Pfirrmann, T, Pfister, AS, Philips, JA, Pi, H, Picca, A, Pickrell, AM, Picot, S, Pierantoni, GM, Pierdominici, M, Pierre, P, Pierrefite-Carle, V, Pierzynowska, K, Pietrocola, F, Pietruczuk, M, Pignata, C, Pimentel-Muiños, FX, Pinar, M, Pinheiro, RO, Pinkas-Kramarski, R, Pinton, P, Pircs, K, Piya, S, Pizzo, P, Plantinga, TS, Platta, HW, Plaza-Zabala, A, Plomann, M, Plotnikov, EY, Plun-Favreau, H, Pluta, R, Pocock, R, Pöggeler, S, Pohl, C, Poirot, M, Poletti, A, Ponpuak, M, Popelka, H, Popova, B, Porta, H, Porte Alcon, S, Portilla-Fernandez, E, Post, M, Potts, MB, Poulton, J, Powers, T, Prahlad, V, Prajsnar, TK, Praticò, D, Prencipe, R, Priault, M, Proikas-Cezanne, T, Promponas, VJ, Proud, CG, Puertollano, R, Puglielli, L, Pulinilkunnil, T, Puri, D, Puri, R, Puyal, J, Qi, X, Qi, Y, Qian, W, Qiang, L, Qiu, Y, Quadrilatero, J, Quarleri, J, Raben, N, Rabinowich, H, Ragona, D, Ragusa, MJ, Rahimi, N, Rahmati, M, Raia, V, Raimundo, N, Rajasekaran, N-S, Ramachandra Rao, S, Rami, A, Ramírez-Pardo, I, Ramsden, DB, Randow, F, Rangarajan, PN, Ranieri, D, Rao, H, Rao, L, Rao, R, Rathore, S, Ratnayaka, JA, Ratovitski, EA, Ravanan, P, Ravegnini, G, Ray, SK, Razani, B, Rebecca, V, Reggiori, F, Régnier-Vigouroux, A, Reichert, AS, Reigada, D, Reiling, JH, Rein, T, Reipert, S, Rekha, RS, Ren, H, Ren, J, Ren, W, Renault, T, Renga, G, Reue, K, Rewitz, K, Ribeiro de Andrade Ramos, B, Riazuddin, SA, Ribeiro-Rodrigues, TM, Ricci, J-E, Ricci, R, Riccio, V, Richardson, DR, Rikihisa, Y, Risbud, MV, Risueño, RM, Ritis, K, Rizza, S, Rizzuto, R, Roberts, HC, Roberts, LD, Robinson, KJ, Roccheri, MC, Rocchi, S, Rodney, GG, Rodrigues, T, Rodrigues Silva, VR, Rodriguez, A, Rodriguez-Barrueco, R, Rodriguez-Henche, N, Rodriguez-Rocha, H, Roelofs, J, Rogers, RS, Rogov, VV, Rojo, AI, Rolka, K, Romanello, V, Romani, L, Romano, A, Romano, PS, Romeo-Guitart, D, Romero, LC, Romero, M, Roney, JC, Rongo, C, Roperto, S, Rosenfeldt, MT, Rosenstiel, P, Rosenwald, AG, Roth, KA, Roth, L, Roth, S, Rouschop, KMA, Roussel, BD, Roux, S, Rovere-Querini, P, Roy, A, Rozieres, A, Ruano, D, Rubinsztein, DC, Rubtsova, MP, Ruckdeschel, K, Ruckenstuhl, C, Rudolf, E, Rudolf, R, Ruggieri, A, Ruparelia, AA, Rusmini, P, Russell, RR, Russo, GL, Russo, M, Russo, R, Ryabaya, OO, Ryan, KM, Ryu, K-Y, Sabater-Arcis, M, Sachdev, U, Sacher, M, Sachse, C, Sadhu, A, Sadoshima, J, Safren, N, Saftig, P, Sagona, AP, Sahay, G, Sahebkar, A, Sahin, M, Sahin, O, Sahni, S, Saito, N, Saito, S, Saito, T, Sakai, R, Sakai, Y, Sakamaki, J-I, Saksela, K, Salazar, G, Salazar-Degracia, A, Salekdeh, GH, Saluja, AK, Sampaio-Marques, B, Sanchez, MC, Sanchez-Alcazar, JA, Sanchez-Vera, V, Sancho-Shimizu, V, Sanderson, JT, Sandri, M, Santaguida, S, Santambrogio, L, Santana, MM, Santoni, G, Sanz, A, Sanz, P, Saran, S, Sardiello, M, Sargeant, TJ, Sarin, A, Sarkar, C, Sarkar, S, Sarrias, M-R, Sarmah, DT, Sarparanta, J, Sathyanarayan, A, Sathyanarayanan, R, Scaglione, KM, Scatozza, F, Schaefer, L, Schafer, ZT, Schaible, UE, Schapira, AHV, Scharl, M, Schatzl, HM, Schein, CH, Scheper, W, Scheuring, D, Schiaffino, MV, Schiappacassi, M, Schindl, R, Schlattner, U, Schmidt, O, Schmitt, R, Schmidt, SD, Schmitz, I, Schmukler, E, Schneider, A, Schneider, BE, Schober, R, Schoijet, AC, Schott, MB, Schramm, M, Schröder, B, Schuh, K, Schüller, C, Schulze, RJ, Schürmanns, L, Schwamborn, JC, Schwarten, M, Scialo, F, Sciarretta, S, Scott, MJ, Scotto, KW, Scovassi, AI, Scrima, A, Scrivo, A, Sebastian, D, Sebti, S, Sedej, S, Segatori, L, Segev, N, Seglen, PO, Seiliez, I, Seki, E, Selleck, SB, Sellke, FW, Selsby, JT, Sendtner, M, Senturk, S, Seranova, E, Sergi, C, Serra-Moreno, R, Sesaki, H, Settembre, C, Setty, SRG, Sgarbi, G, Sha, O, Shacka, JJ, Shah, JA, Shang, D, Shao, C, Shao, F, Sharbati, S, Sharkey, LM, Sharma, D, Sharma, G, Sharma, K, Sharma, P, Sharma, S, Shen, H-M, Shen, H, Shen, J, Shen, M, Shen, W, Shen, Z, Sheng, R, Sheng, Z, Sheng, Z-H, Shi, J, Shi, X, Shi, Y-H, Shiba-Fukushima, K, Shieh, J-J, Shimada, Y, Shimizu, S, Shimozawa, M, Shintani, T, Shoemaker, CJ, Shojaei, S, Shoji, I, Shravage, BV, Shridhar, V, Shu, C-W, Shu, H-B, Shui, K, Shukla, AK, Shutt, TE, Sica, V, Siddiqui, A, Sierra, A, Sierra-Torre, V, Signorelli, S, Sil, P, Silva, BJDA, Silva, JD, Silva-Pavez, E, Silvente-Poirot, S, Simmonds, RE, Simon, AK, Simon, H-U, Simons, M, Singh, A, Singh, LP, Singh, R, Singh, SV, Singh, SK, Singh, SB, Singh, S, Singh, SP, Sinha, D, Sinha, RA, Sinha, S, Sirko, A, Sirohi, K, Sivridis, EL, Skendros, P, Skirycz, A, Slaninová, I, Smaili, SS, Smertenko, A, Smith, MD, Soenen, SJ, Sohn, EJ, Sok, SPM, Solaini, G, Soldati, T, Soleimanpour, SA, Soler, RM, Solovchenko, A, Somarelli, JA, Sonawane, A, Song, F, Song, HK, Song, J-X, Song, K, Song, Z, Soria, LR, Sorice, M, Soukas, AA, Soukup, S-F, Sousa, D, Sousa, N, Spagnuolo, PA, Spector, SA, Srinivas Bharath, MM, St Clair, D, Stagni, V, Staiano, L, Stalnecker, CA, Stankov, MV, Stathopulos, PB, Stefan, K, Stefan, SM, Stefanis, L, Steffan, JS, Steinkasserer, A, Stenmark, H, Sterneckert, J, Stevens, C, Stoka, V, Storch, S, Stork, B, Strappazzon, F, Strohecker, AM, Stupack, DG, Su, H, Su, L-Y, Su, L, Suarez-Fontes, AM, Subauste, CS, Subbian, S, Subirada, PV, Sudhandiran, G, Sue, CM, Sui, X, Summers, C, Sun, G, Sun, J, Sun, K, Sun, M-X, Sun, Q, Sun, Y, Sun, Z, Sunahara, KKS, Sundberg, E, Susztak, K, Sutovsky, P, Suzuki, H, Sweeney, G, Symons, JD, Sze, SCW, Szewczyk, NJ, Tabęcka-Łonczynska, A, Tabolacci, C, Tacke, F, Taegtmeyer, H, Tafani, M, Tagaya, M, Tai, H, Tait, SWG, Takahashi, Y, Takats, S, Talwar, P, Tam, C, Tam, SY, Tampellini, D, Tamura, A, Tan, CT, Tan, E-K, Tan, Y-Q, Tanaka, M, Tang, D, Tang, J, Tang, T-S, Tanida, I, Tao, Z, Taouis, M, Tatenhorst, L, Tavernarakis, N, Taylor, A, Taylor, GA, Taylor, JM, Tchetina, E, Tee, AR, Tegeder, I, Teis, D, Teixeira, N, Teixeira-Clerc, F, Tekirdag, KA, Tencomnao, T, Tenreiro, S, Tepikin, AV, Testillano, PS, Tettamanti, G, Tharaux, P-L, Thedieck, K, Thekkinghat, AA, Thellung, S, Thinwa, JW, Thirumalaikumar, VP, Thomas, SM, Thomes, PG, Thorburn, A, Thukral, L, Thum, T, Thumm, M, Tian, L, Tichy, A, Till, A, Timmerman, V, Titorenko, VI, Todi, SV, Todorova, K, Toivonen, JM, Tomaipitinca, L, Tomar, D, Tomas-Zapico, C, Tomić, S, Tong, BC-K, Tong, C, Tong, X, Tooze, SA, Torgersen, ML, Torii, S, Torres-López, L, Torriglia, A, Towers, CG, Towns, R, Toyokuni, S, Trajkovic, V, Tramontano, D, Tran, Q-G, Travassos, LH, Trelford, CB, Tremel, S, Trougakos, IP, Tsao, BP, Tschan, MP, Tse, H-F, Tse, TF, Tsugawa, H, Tsvetkov, AS, Tumbarello, DA, Tumtas, Y, Tuñón, MJ, Turcotte, S, Turk, B, Turk, V, Turner, BJ, Tuxworth, RI, Tyler, JK, Tyutereva, EV, Uchiyama, Y, Ugun-Klusek, A, Uhlig, HH, Ułamek-Kozioł, M, Ulasov, IV, Umekawa, M, Ungermann, C, Unno, R, Urbe, S, Uribe-Carretero, E, Üstün, S, Uversky, VN, Vaccari, T, Vaccaro, MI, Vahsen, BF, Vakifahmetoglu-Norberg, H, Valdor, R, Valente, MJ, Valko, A, Vallee, RB, Valverde, AM, Van den Berghe, G, van der Veen, S, Van Kaer, L, van Loosdregt, J, van Wijk, SJL, Vandenberghe, W, Vanhorebeek, I, Vannier-Santos, MA, Vannini, N, Vanrell, MC, Vantaggiato, C, Varano, G, Varela-Nieto, I, Varga, M, Vasconcelos, MH, Vats, S, Vavvas, DG, Vega-Naredo, I, Vega-Rubin-de-Celis, S, Velasco, G, Velázquez, AP, Vellai, T, Vellenga, E, Velotti, F, Verdier, M, Verginis, P, Vergne, I, Verkade, P, Verma, M, Verstreken, P, Vervliet, T, Vervoorts, J, Vessoni, AT, Victor, VM, Vidal, M, Vidoni, C, Vieira, OV, Vierstra, RD, Viganó, S, Vihinen, H, Vijayan, V, Vila, M, Vilar, M, Villalba, JM, Villalobo, A, Villarejo-Zori, B, Villarroya, F, Villarroya, J, Vincent, O, Vindis, C, Viret, C, Viscomi, MT, Visnjic, D, Vitale, I, Vocadlo, DJ, Voitsekhovskaja, OV, Volonté, C, Volta, M, Vomero, M, Von Haefen, C, Vooijs, MA, Voos, W, Vucicevic, L, Wade-Martins, R, Waguri, S, Waite, KA, Wakatsuki, S, Walker, DW, Walker, MJ, Walker, SA, Walter, J, Wandosell, FG, Wang, B, Wang, C-Y, Wang, C, Wang, D, Wang, F, Wang, G, Wang, H, Wang, H-G, Wang, J, Wang, K, Wang, L, Wang, MH, Wang, M, Wang, N, Wang, P, Wang, QJ, Wang, Q, Wang, QK, Wang, QA, Wang, W-T, Wang, W, Wang, X, Wang, Y, Wang, Y-Y, Wang, Z, Warnes, G, Warnsmann, V, Watada, H, Watanabe, E, Watchon, M, Wawrzyńska, A, Weaver, TE, Wegrzyn, G, Wehman, AM, Wei, H, Wei, L, Wei, T, Wei, Y, Weiergräber, OH, Weihl, CC, Weindl, G, Weiskirchen, R, Wells, A, Wen, RH, Wen, X, Werner, A, Weykopf, B, Wheatley, SP, Whitton, JL, Whitworth, AJ, Wiktorska, K, Wildenberg, ME, Wileman, T, Wilkinson, S, Willbold, D, Williams, B, Williams, RSB, Williams, RL, Williamson, PR, Wilson, RA, Winner, B, Winsor, NJ, Witkin, SS, Wodrich, H, Woehlbier, U, Wollert, T, Wong, E, Wong, JH, Wong, RW, Wong, VKW, Wong, WW-L, Wu, A-G, Wu, C, Wu, J, Wu, KK, Wu, M, Wu, S-Y, Wu, S, Wu, WKK, Wu, X, Wu, Y-W, Wu, Y, Xavier, RJ, Xia, H, Xia, L, Xia, Z, Xiang, G, Xiang, J, Xiang, M, Xiang, W, Xiao, B, Xiao, G, Xiao, H, Xiao, H-T, Xiao, J, Xiao, L, Xiao, S, Xiao, Y, Xie, B, Xie, C-M, Xie, M, Xie, Y, Xie, Z, Xilouri, M, Xu, C, Xu, E, Xu, H, Xu, J, Xu, L, Xu, WW, Xu, X, Xue, Y, Yakhine-Diop, SMS, Yamaguchi, M, Yamaguchi, O, Yamamoto, A, Yamashina, S, Yan, S, Yan, S-J, Yan, Z, Yanagi, Y, Yang, C, Yang, D-S, Yang, H, Yang, H-T, Yang, J-M, Yang, J, Yang, L, Yang, M, Yang, P-M, Yang, Q, Yang, S, Yang, S-F, Yang, W, Yang, WY, Yang, X, Yang, Y, Yao, H, Yao, S, Yao, X, Yao, Y-G, Yao, Y-M, Yasui, T, Yazdankhah, M, Yen, PM, Yi, C, Yin, X-M, Yin, Y, Yin, Z, Ying, M, Ying, Z, Yip, CK, Yiu, SPT, Yoo, YH, Yoshida, K, Yoshii, SR, Yoshimori, T, Yousefi, B, Yu, B, Yu, H, Yu, J, Yu, L, Yu, M-L, Yu, S-W, Yu, VC, Yu, WH, Yu, Z, Yuan, J, Yuan, L-Q, Yuan, S, Yuan, S-SF, Yuan, Y, Yuan, Z, Yue, J, Yue, Z, Yun, J, Yung, RL, Zacks, DN, Zaffagnini, G, Zambelli, VO, Zanella, I, Zang, QS, Zanivan, S, Zappavigna, S, Zaragoza, P, Zarbalis, KS, Zarebkohan, A, Zarrouk, A, Zeitlin, SO, Zeng, J, Zeng, J-D, Žerovnik, E, Zhan, L, Zhang, B, Zhang, DD, Zhang, H, Zhang, H-L, Zhang, J, Zhang, J-P, Zhang, KYB, Zhang, LW, Zhang, L, Zhang, M, Zhang, P, Zhang, S, Zhang, W, Zhang, X, Zhang, X-W, Zhang, XD, Zhang, Y, Zhang, Y-D, Zhang, Y-Y, Zhang, Z, Zhao, H, Zhao, L, Zhao, S, Zhao, T, Zhao, X-F, Zhao, Y, Zheng, G, Zheng, K, Zheng, L, Zheng, S, Zheng, X-L, Zheng, Y, Zheng, Z-G, Zhivotovsky, B, Zhong, Q, Zhou, A, Zhou, B, Zhou, C, Zhou, G, Zhou, H, Zhou, J, Zhou, K, Zhou, R, Zhou, X-J, Zhou, Y, Zhou, Z-Y, Zhou, Z, Zhu, B, Zhu, C, Zhu, G-Q, Zhu, H, Zhu, W-G, Zhu, Y, Zhuang, H, Zhuang, X, Zientara-Rytter, K, Zimmermann, CM, Ziviani, E, Zoladek, T, Zong, W-X, Zorov, DB, Zorzano, A, Zou, W, Zou, Z, Zuryn, S, Zwerschke, W, Brand-Saberi, B, Dong, XC, Kenchappa, CS, Lin, Y, Oshima, S, Rong, Y, Sluimer, JC, Stallings, CL, and Tong, C-K
- Abstract
In 2008, we published the first set of guidelines for standardizing research in autophagy. Since then, this topic has received increasing attention, and many scientists have entered the field. Our knowledge base and relevant new technologies have also been expanding. Thus, it is important to formulate on a regular basis updated guidelines for monitoring autophagy in different organisms. Despite numerous reviews, there continues to be confusion regarding acceptable methods to evaluate autophagy, especially in multicellular eukaryotes. Here, we present a set of guidelines for investigators to select and interpret methods to examine autophagy and related processes, and for reviewers to provide realistic and reasonable critiques of reports that are focused on these processes. These guidelines are not meant to be a dogmatic set of rules, because the appropriateness of any assay largely depends on the question being asked and the system being used. Moreover, no individual assay is perfect for every situation, calling for the use of multiple techniques to properly monitor autophagy in each experimental setting. Finally, several core components of the autophagy machinery have been implicated in distinct autophagic processes (canonical and noncanonical autophagy), implying that genetic approaches to block autophagy should rely on targeting two or more autophagy-related genes that ideally participate in distinct steps of the pathway. Along similar lines, because multiple proteins involved in autophagy also regulate other cellular pathways including apoptosis, not all of them can be used as a specific marker for bona fide autophagic responses. Here, we critically discuss current methods of assessing autophagy and the information they can, or cannot, provide. Our ultimate goal is to encourage intellectual and technical innovation in the field.
- Published
- 2021
3. Against the Norm: Do Not Rely on Serum C-Reactive Protein and White Blood Cell Count Only When Assessing Eradication of Periprosthetic Joint Infection
- Author
-
Farouk Khury, Moritz Oltmanns, Michael Fuchs, Janina Leiprecht, Heiko Reichel, and Martin Faschingbauer
- Subjects
periprosthetic joint infection ,diagnostic criteria ,CRP ,WBC ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Introduction: Periprosthetic joint infections (PJI) following primary arthroplasty continue to be a serious complication, despite advances in diagnostics and treatment. Two-stage revision arthroplasty has been commonly used as the gold standard for the treatment of PJI. However, much discussion persists regarding the interim of the two-stage procedure and the optimal timing of reimplantation. Serology markers have been proposed as defining parameters for a successful reimplantation. The objective of this matched-pair analysis was to assess the role of serum C-reactive protein (CRP) and white blood cell count (WBC) in determining infection eradication and proper timing of reimplantation. We investigated the delta (∆) change in CRP and WBC values prior to both stages of two-stage revision arthroplasty as a useful marker of infection eradication. Methods: We analyzed 39 patients and 39 controls, matched by propensity score matching (BMI, age, ASA-classification), with a minimum follow-up of 24 months and treated with a two-stage revision THA or TKA in our institution. Data of serum CRP and WBC values were gathered at two selected time points: prior to the explantation of the implant (preexplantation) and following the completion of antibiotic treatment regimen, both systemic and with a drug-eluting cement spacer (prereimplantation). Patient records were reviewed electronically for preexisting comorbidities, overall health status, synovial fluid cultures, inflammatory serologies, revision surgeries, and recurrent or persistent infection based on the modified Musculoskeletal Infection Society criteria. Patient demographics, ∆CRP, ∆WBC, and time interval to reimplantation were statistically analyzed using receiver operator curves (ROC), Pearson’s correlation coefficient, Levene’s test, and Student’s t-test. Results: Infection-free patients exhibited higher mean CRP and WBC than did patients who were reinfected at both time points. When comparing preexplantation with prereimplantation values, the median ∆CRP was 9.48 mg/L (interquartile range (IQR) 2.3–36.6 mg/L) for patients who did not develop a reinfection versus 2.74 mg/L (IQR 1.4–14.2 mg/L) for patients who developed reinfection (p = 0.069). The median ∆WBC was 1.5 × 109/L (IQR 0.6–4.0 × 109/L) for patients who remained infection-free versus 1.2 × 109/L (IQR 0.8–2.2 109/L) for patients who developed reinfection (p = 0.072). Analysis of areas under the curve (AUC) using ROC demonstrated poor prediction of persistent infection by ∆CRP (AUC = 0.654) and ∆WBC (AUC = 0.573). Although a highly significant correlation was found between the interim interval and infection persistence (r = 0.655, p < 0.01), analysis using ROC failed to result in a specific threshold time to reimplantation above which patients are at significantly higher risk for reinfection (AUC = 0.507). Conclusion: No association could be determined between the delta change in serum CRP and WBC before and after two-stage revision arthroplasty for PJI and reinfection risk. Even though inflammatory serologies demonstrate a downtrending pattern prior to reimplantation, the role of CRP and WBC in determining the optimal timing of reimplantation seems to be dispensable. Planning a second-stage reimplantation requires assessing multiple variables rather than relying on specific numeric changes in these inflammatory marker values.
- Published
- 2022
- Full Text
- View/download PDF
4. Numerical Analysis of Hot Cracking in Laser-Hybrid Welded Tubes
- Author
-
Moritz Oliver Gebhardt, Andrey Gumenyuk, and Michael Rethmeier
- Subjects
Materials of engineering and construction. Mechanics of materials ,TA401-492 - Abstract
In welding experiments conducted on heavy wall pipes, the penetration mode (full or partial penetration) occurred to be a significant factor influencing appearance of solidification cracks. To explain the observed phenomena and support further optimization of manufacturing processes, a computational model was developed, which used a sophisticated strategy to model the material. High stresses emerged in the models in regions which showed cracking during experiments. In partial penetration welding, they were caused by the prevention of weld shrinkage due to the cold and strong material below the joint. Another identified factor having an influence on high stress localization is bulging of the weld.
- Published
- 2013
- Full Text
- View/download PDF
5. Prominin-1 null Xenopus laevis develop subretinal drusenoid-like deposits, cone-rod dystrophy, and RPE atrophy.
- Author
-
Carr BJ, Skitsko D, Kriese LM, Song J, Li Z, Ju MJ, and Moritz OL
- Abstract
PROMININ-1 (PROM1) mutations are associated with inherited, non-syndromic vision loss. We used CRISPR/Cas9 to induce prom1-null mutations in Xenopus laevis and then tracked retinal disease progression from the ages of 6 weeks to 3 years old. Prom1-null associated retinal degeneration in frogs is age-dependent and involves RPE dysfunction preceding photoreceptor degeneration. Before photoreceptor degeneration occurs, aging prom1-null frogs develop increasing size and numbers of cellular debris deposits in the subretinal space and outer segment layer, which resemble subretinal drusenoid deposits (SDD) in their location, histology, and representation in color fundus photography and optical coherence tomography (OCT). Evidence for an RPE origin of these deposits includes infiltration of pigment granules into the deposits, thinning of RPE as measured by OCT, and RPE disorganization as measured by histology and OCT. The appearance and accumulation of SDD-like deposits and RPE thinning and disorganization in our animal model suggests an underlying disease mechanism for prom1-null mediated blindness of death and dysfunction of the RPE preceding photoreceptor degeneration, instead of direct effects upon photoreceptor outer segment morphogenesis, as was previously hypothesized., (© 2024. Published by The Company of Biologists Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
6. Synchronized Photoactivation of T4K Rhodopsin Causes a Chromophore-Dependent Retinal Degeneration That Is Moderated by Interaction with Phototransduction Cascade Components.
- Author
-
Tam BM, Burns P, Chiu CN, and Moritz OL
- Subjects
- Animals, Female, Male, Light Signal Transduction, Light adverse effects, Retinal Rod Photoreceptor Cells metabolism, Retinal Rod Photoreceptor Cells pathology, cis-trans-Isomerases metabolism, cis-trans-Isomerases genetics, Cell Death, Rhodopsin metabolism, Rhodopsin genetics, Retinal Degeneration metabolism, Retinal Degeneration pathology, Retinal Degeneration genetics, Xenopus laevis
- Abstract
Multiple mutations in the Rhodopsin gene cause sector retinitis pigmentosa in humans and a corresponding light-exacerbated retinal degeneration (RD) in animal models. Previously we have shown that T4K rhodopsin requires photoactivation to exert its toxic effect. Here we further investigated the mechanisms involved in rod cell death caused by T4K rhodopsin in mixed male and female Xenopus laevis In this model, RD was prevented by rearing animals in constant darkness but surprisingly also in constant light. RD was maximized by light cycles containing at least 1 h of darkness and 20 min of light exposure, light intensities >750 lux, and by a sudden light onset. Under conditions of frequent light cycling, RD occurred rapidly and synchronously, with massive shedding of ROS fragments into the RPE initiated within hours and subsequent death and phagocytosis of rod cell bodies. RD was minimized by reduced light levels, pretreatment with constant light, and gradual light onset. RD was prevented by genetic ablation of the retinal isomerohydrolase RPE65 and exacerbated by ablation of phototransduction components GNAT1, SAG, and GRK1. Our results indicate that photoactivated T4K rhodopsin is toxic, that cell death requires synchronized photoactivation of T4K rhodopsin, and that toxicity is mitigated by interaction with other rod outer segment proteins regardless of whether they participate in activation or shutoff of phototransduction. In contrast, RD caused by P23H rhodopsin does not require photoactivation of the mutant protein, as it was exacerbated by RPE65 ablation, suggesting that these phenotypically similar disorders may require different treatment strategies., Competing Interests: The authors declare no competing financial interests., (Copyright © 2024 the authors.)
- Published
- 2024
- Full Text
- View/download PDF
7. Identification and cellular localization in Xenopus laevis photoreceptors of three Peripherin-2 family members, Prph2, Rom1 and Gp2l, which arose from gene duplication events in the common ancestors of jawed vertebrates.
- Author
-
Tam BM, Taylor JS, and Moritz OL
- Subjects
- Animals, Gene Duplication, Mammals, Peripherins genetics, Peripherins metabolism, Phylogeny, Retinal Cone Photoreceptor Cells metabolism, Tetraspanins genetics, Xenopus laevis genetics, Xenopus laevis metabolism, Retinal Degeneration metabolism
- Abstract
Rod and cone photoreceptors are named for the distinct morphologies of their outer segment organelles, which are either cylindrical or conical, respectively. The morphologies of the stacked disks that comprise the rod and cone outer segments also differ: rod disks are completely sealed and are discontinuous from the plasma membrane, while cone disks remain partially open to the extracellular space. These morphological differences between photoreceptor types are more prominent in non-mammalian vertebrates, whose cones typically possess a greater proportion of open disks and are more tapered in shape. In mammals, the tetraspanin prph2 generates and maintains the highly curved disk rim regions by forming extended oligomeric structures with itself and a structurally similar paralog, rom1. Here we determined that in addition to these two proteins, there is a third Prph2 family paralog in most non-mammalian vertebrate species, including X. laevis: Glycoprotein 2-like protein or "Gp2l". A survey of multiple genome databases revealed a single invertebrate Prph2 'pro-ortholog' in Amphioxus, several echinoderms and in a diversity of protostomes indicating an ancient divergence from other tetraspanins. Based on phylogenetic analysis, duplication of the vertebrate predecessor likely gave rise to the Gp2l and Prph2/Rom1 clades, with a further duplication distinguishing the Prph2 and Rom1 clades. Mammals have lost Gp2l and their Rom1 has undergone a period of accelerated evolution such that it has lost several features that are retained in non-mammalian vertebrate Rom1. Specifically, Prph2, Gp2l and non-mammalian Rom1 encode proteins with consensus N-linked glycosylation and outer segment localization signals; mammalian rom1 lacks these motifs. We determined that X. laevis gp2l is expressed exclusively in cones and green rods, while X. laevis rom1 is expressed exclusively in rods, and prph2 is present in both rods and cones. The presence of three Prph2-related genes with distinct expression patterns as well as the rapid evolution of mammalian Rom1, may contribute to the more pronounced differences in morphology between rod and cone outer segments and rod and cone disks observed in non-mammalian versus mammalian vertebrates., (Copyright © 2023. Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
8. Distinct roles for prominin-1 and photoreceptor cadherin in outer segment disc morphogenesis in CRISPR-altered X. laevis .
- Author
-
Carr BJ, Stanar P, and Moritz OL
- Subjects
- AC133 Antigen genetics, Animals, Morphogenesis genetics, Nerve Tissue Proteins, Xenopus laevis, Cadherins genetics, Clustered Regularly Interspaced Short Palindromic Repeats
- Abstract
Mutations in prominin-1 ( prom1 ) and photoreceptor cadherin ( cdhr1 ) are associated with inherited retinal degenerative disorders but their functions remain unknown. Here, we used CRISPR-Cas9 to generate prom1 -null, cdhr1 -null, and prom1 plus cdhr1 double-null Xenopus laevis and then documented the effects of these mutations on photoreceptor structure and function. Prom1 -null mutations resulted in severely dysmorphic photoreceptors comprising overgrown and disorganized disc membranes. Cone outer segments were more severely affected than rods and had an impaired electroretinogram response. Cdhr1 -null photoreceptors did not appear grossly dysmorphic, but ultrastructural analysis revealed that some disc membranes were overgrown or oriented vertically within the plasma membrane. Double-null mutants did not differ significantly from prom1 -null mutants. Our results indicate that neither prom1 nor cdhr1 are necessary for outer segment disc membrane evagination or the fusion event that controls disc sealing. Rather, they are necessary for the higher-order organization of the outer segment. Prom1 may align and reinforce interactions between nascent disc leading edges, a function more critical in cones for structural support. Cdhr1 may secure discs in a horizontal orientation prior to fusion and regulate cone lamellae size.This article has an associated First Person interview with the first author of the paper., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2021. Published by The Company of Biologists Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
9. Germline CRISPR/Cas9-Mediated Gene Editing Prevents Vision Loss in a Novel Mouse Model of Aniridia.
- Author
-
Mirjalili Mohanna SZ, Hickmott JW, Lam SL, Chiu NY, Lengyell TC, Tam BM, Moritz OL, and Simpson EM
- Abstract
Aniridia is a rare eye disorder, which is caused by mutations in the paired box 6 ( PAX6 ) gene and results in vision loss due to the lack of a long-term vision-saving therapy. One potential approach to treating aniridia is targeted CRISPR-based genome editing. To enable the Pax6 small eye ( Sey ) mouse model of aniridia, which carries the same mutation found in patients, for preclinical testing of CRISPR-based therapeutic approaches, we endogenously tagged the Sey allele, allowing for the differential detection of protein from each allele. We optimized a correction strategy in vitro then tested it in vivo in the germline of our new mouse to validate the causality of the Sey mutation. The genomic manipulations were analyzed by PCR, as well as by Sanger and next-generation sequencing. The mice were studied by slit lamp imaging, immunohistochemistry, and western blot analyses. We successfully achieved both in vitro and in vivo germline correction of the Sey mutation, with the former resulting in an average 34.8% ± 4.6% SD correction, and the latter in restoration of 3xFLAG-tagged PAX6 expression and normal eyes. Hence, in this study we have created a novel mouse model for aniridia, demonstrated that germline correction of the Sey mutation alone rescues the mutant phenotype, and developed an allele-distinguishing CRISPR-based strategy for aniridia., (© 2020 The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
10. Autophagy in Xenopus laevis rod photoreceptors is independently regulated by phototransduction and misfolded RHO P23H .
- Author
-
Wen RH, Stanar P, Tam B, and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, Autophagosomes metabolism, Autophagosomes radiation effects, Circadian Rhythm genetics, Circadian Rhythm radiation effects, Fluorescence, Green Fluorescent Proteins chemistry, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Heterotrimeric GTP-Binding Proteins genetics, Heterotrimeric GTP-Binding Proteins metabolism, Larva genetics, Larva metabolism, Larva ultrastructure, Light Signal Transduction radiation effects, Mutation, Retinal Rod Photoreceptor Cells cytology, Retinal Rod Photoreceptor Cells radiation effects, Retinal Rod Photoreceptor Cells ultrastructure, Retinitis Pigmentosa genetics, Rhodopsin chemistry, Rhodopsin genetics, Rhodopsin radiation effects, Time Factors, Xenopus laevis, cis-trans-Isomerases genetics, cis-trans-Isomerases metabolism, Autophagy genetics, Autophagy radiation effects, Light Signal Transduction genetics, Retinal Rod Photoreceptor Cells metabolism, Retinitis Pigmentosa metabolism, Rhodopsin metabolism
- Abstract
We previously reported autophagic structures in rod photoreceptors expressing a misfolding RHO (rhodopsin) mutant (RHO
P23H ), suggesting that autophagy may play a role in degrading the mutant RHO and/or be involved in photoreceptor cell death. To further examine autophagy in normal and diseased rods, we generated transgenic Xenopus laevis tadpoles expressing the dually fluorescent autophagy marker mRFP-eGFP-LC3 in rods, which changes from green to yellow and finally red as autophagic structures develop and mature. Using transgenic lines with constitutive and inducible expression, we determined the time-course of autophagy in rod photoreceptors: autophagosomes last for 6 to 8 hours before fusing with lysosomes, and acidified autolysosomes last for about 28 hours before being degraded. Autophagy was diurnally regulated in normal rods, with more autophagic structures generated during periods of light, and this regulation was non-circadian. We also found that more autophagosomes were produced in rods expressing the misfolding RHOP23H mutant. The RHO chromophore absorbs photons to initiate phototransduction, and is consumed in this process; it also promotes RHO folding. To determine whether increased autophagy in light-exposed normal rods is caused by increased RHO misfolding or phototransduction, we used CRISPR/Cas9 to knock out the RPE65 and GNAT1 genes, which are essential for chromophore biosynthesis and phototransduction respectively. Both knockouts suppressed light-induced autophagy, indicating that although light and misfolded rhodopsin can both induce autophagy in rods, light-induced autophagy is not due to misfolding of RHO, but rather due to phototransduction. Abbreviations : CYCS: cytochrome c; bRHOP23H : bovine RHOP23H ; Cas9: CRISPR associated protein 9; dpf: days post-fertilization; eGFP: enhanced green fluorescent protein; GNAT1: guanine nucleotide-binding protein G(t) subunit alpha-1 aka rod alpha-transducin; HSPA1A/hsp70: heat shock protein of 70 kilodaltons; LAMP1: lysosomal-associated membrane protein 1; LC3: microtubule-associated protein 1A/1B light chain 3; mRFP: monomeric red fluorescent protein; RHO: rhodopsin; RP: retinitis pigmentosa; RPE65: retinal pigment epithelium-specific 65 kDa protein: sfGFP: superfolding GFP; sgRNA: single guide RNA; WGA: wheat germ agglutinin; RHOp : the Xenopus laevis RHO.2.L promoter.- Published
- 2019
- Full Text
- View/download PDF
11. Cell Death Pathways in Mutant Rhodopsin Rat Models Identifies Genotype-Specific Targets Controlling Retinal Degeneration.
- Author
-
Viringipurampeer IA, Gregory-Evans CY, Metcalfe AL, Bashar E, Moritz OL, and Gregory-Evans K
- Subjects
- Animals, Cell Death, Disease Models, Animal, Genotype, Rats, Rats, Transgenic, Retinal Cone Photoreceptor Cells pathology, Retinal Degeneration genetics, Retinal Degeneration pathology, Retinal Rod Photoreceptor Cells pathology, Retinitis Pigmentosa genetics, Retinitis Pigmentosa pathology, Rhodopsin genetics, Retinal Cone Photoreceptor Cells metabolism, Retinal Degeneration metabolism, Retinal Rod Photoreceptor Cells metabolism, Retinitis Pigmentosa metabolism, Rhodopsin metabolism
- Abstract
Retinitis pigmentosa (RP) is a group of inherited neurological disorders characterized by rod photoreceptor cell death, followed by secondary cone cell death leading to progressive blindness. Currently, there are no viable treatment options for RP. Due to incomplete knowledge of the molecular signaling pathways associated with RP pathogenesis, designing therapeutic strategies remains a challenge. In particular, preventing secondary cone photoreceptor cell loss is a key goal in designing potential therapies. In this study, we identified the main drivers of rod cell death and secondary cone loss in the transgenic S334ter rhodopsin rat model, tested the efficacy of specific cell death inhibitors on retinal function, and compared the effect of combining drugs to target multiple pathways in the S334ter and P23H rhodopsin rat models. The primary driver of early rod cell death in the S334ter model was a caspase-dependent process, whereas cone cell death occurred though RIP3-dependent necroptosis. In comparison, rod cell death in the P23H model was via necroptotic signaling, whereas cone cell loss occurred through inflammasome activation. Combination therapy of four drugs worked better than the individual drugs in the P23H model but not in the S334ter model. These differences imply that treatment modalities need to be tailored for each genotype. Taken together, our data demonstrate that rationally designed genotype-specific drug combinations will be an important requisite to effectively target primary rod cell loss and more importantly secondary cone survival.
- Published
- 2019
- Full Text
- View/download PDF
12. Electrophysiological Changes During Early Steps of Retinitis Pigmentosa.
- Author
-
Bocchero U, Tam BM, Chiu CN, Torre V, and Moritz OL
- Subjects
- Animals, Dark Adaptation physiology, Disease Models, Animal, Electrophysiological Phenomena, Electroretinography, Female, Male, Microscopy, Confocal, Photic Stimulation, Retinitis Pigmentosa genetics, Xenopus laevis, Animals, Genetically Modified, Retinitis Pigmentosa physiopathology, Rhodopsin genetics, Rod Cell Outer Segment physiology, Vision, Ocular physiology
- Abstract
Purpose: The rhodopsin mutation P23H is responsible for a significant portion of autosomal-dominant retinitis pigmentosa, a disorder characterized by rod photoreceptor death. The mechanisms of toxicity remain unclear; previous studies implicate destabilization of P23H rhodopsin during light exposure, causing decreased endoplasmic reticulum (ER) exit and ER stress responses. Here, we probed phototransduction in Xenopus laevis rods expressing bovine P23H rhodopsin, in which retinal degeneration is inducible by light exposure, in order to examine early physiological changes that occur during retinal degeneration., Methods: We recorded single-cell and whole-retina responses to light stimuli using electrophysiology. Moreover, we monitored morphologic changes in rods after different periods of light exposure., Results: Initially, P23H rods had almost normal photoresponses, but following a brief light exposure varying from 4 to 32 photoisomerizations per disc, photoresponses became irreversibly prolonged. In intact retinas, rods began to shed OS fragments after a rod-saturating exposure of 12 minutes, corresponding to approximately 10 to 100 times more photoisomerizations., Conclusions: Our results indicate that in P23H rods light-induced degeneration occurs in at least two stages, the first involving impairment of phototransduction and the second involving initiation of morphologic changes.
- Published
- 2019
- Full Text
- View/download PDF
13. Autophagy Induction by HDAC Inhibitors Is Unlikely to be the Mechanism of Efficacy in Prevention of Retinal Degeneration Caused by P23H Rhodopsin.
- Author
-
Wen RH, Loewen AD, Vent-Schmidt RYJ, and Moritz OL
- Subjects
- Animals, Disease Models, Animal, Larva, Retinal Degeneration chemically induced, Xenopus laevis, Autophagy, Histone Deacetylase Inhibitors pharmacology, Retinal Degeneration prevention & control, Rhodopsin adverse effects
- Abstract
We previously found that valproic acid (VPA) and other histone deacetylase inhibitors (HDACis) ameliorate retinal degeneration (RD) caused by P23H rhodopsin in Xenopus laevis larvae and hypothesized that this may be due to enhancement of autophagy. Here we use X. laevis expressing an autophagy marker to assess effects of HDACis on autophagy. We also assess the effects of non-HDACi activators and inducers of autophagy on RD caused by P23H rhodopsin.
- Published
- 2019
- Full Text
- View/download PDF
14. Generation and Analysis of Xenopus laevis Models of Retinal Degeneration Using CRISPR/Cas9.
- Author
-
Feehan JM, Stanar P, Tam BM, Chiu C, and Moritz OL
- Subjects
- Animals, Disease Models, Animal, Fluorescent Antibody Technique, Gene Editing, Gene Expression, Gene Knockout Techniques, Genes, Reporter, Humans, Mice, Phenotype, RNA, Guide, CRISPR-Cas Systems, Retinal Degeneration pathology, Xenopus laevis, CRISPR-Cas Systems, Retinal Degeneration genetics, Retinal Degeneration metabolism
- Abstract
Xenopus laevis have proven to be a useful system for rapid generation and analysis of transgenic models of human retinal disease. However, experimental approaches in this system were limited by lack of a robust knockdown or knockout technology. Here we describe a protocol for generation of Cas9-edited X. laevis embryos. The technique introduces point mutations into the genome of X. laevis resulting in in-frame and out-of-frame insertions and deletions that allow modeling of both dominant and recessive human diseases and efficiently generates gene knockdown and knockout. Our techniques can produce high-frequency gene editing in X. laevis, permitting analysis in the F0 generation.
- Published
- 2019
- Full Text
- View/download PDF
15. Prominin-1 and Photoreceptor Cadherin Localization in Xenopus laevis: Protein-Protein Relationships and Function.
- Author
-
Carr BJ, Yang LL, and Moritz OL
- Subjects
- Animals, Retinal Degeneration, Xenopus laevis, AC133 Antigen genetics, Cadherins genetics, Retinal Cone Photoreceptor Cells physiology, Retinal Rod Photoreceptor Cells physiology
- Abstract
Retinal degenerative diseases are genetically diverse and rare inherited disorders that cause the death of rod and cone photoreceptors, resulting in progressive vision loss and blindness. This review will focus on two retinal degeneration-causing genes: prominin-1 (prom1) and photoreceptor cadherin (prCAD). We will discuss protein localization, potential roles in photoreceptor outer segment disc morphogenesis, and areas for future investigation.
- Published
- 2019
- Full Text
- View/download PDF
16. An interaction network between the SNARE VAMP7 and Rab GTPases within a ciliary membrane-targeting complex.
- Author
-
Kandachar V, Tam BM, Moritz OL, and Deretic D
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Organelles metabolism, Protein Transport physiology, R-SNARE Proteins metabolism, Rhodopsin metabolism, ADP-Ribosylation Factors metabolism, Cilia metabolism, Membrane Fusion physiology, SNARE Proteins metabolism
- Abstract
The Arf4-rhodopsin complex (mediated by the VxPx motif in rhodopsin) initiates expansion of vertebrate rod photoreceptor cilia-derived light-sensing organelles through stepwise assembly of a conserved trafficking network. Here, we examine its role in the sorting of VAMP7 (also known as TI-VAMP) - an R-SNARE possessing a regulatory longin domain (LD) - into rhodopsin transport carriers (RTCs). During RTC formation and trafficking, VAMP7 colocalizes with the ciliary cargo rhodopsin and interacts with the Rab11-Rabin8-Rab8 trafficking module. Rab11 and Rab8 bind the VAMP7 LD, whereas Rabin8 (also known as RAB3IP) interacts with the SNARE domain. The Arf/Rab11 effector FIP3 (also known as RAB11FIP3) regulates VAMP7 access to Rab11. At the ciliary base, VAMP7 forms a complex with the cognate SNAREs syntaxin 3 and SNAP-25. When expressed in transgenic animals, a GFP-VAMP7ΔLD fusion protein and a Y45E phosphomimetic mutant colocalize with endogenous VAMP7. The GFP-VAMP7-R150E mutant displays considerable localization defects that imply an important role of the R-SNARE motif in intracellular trafficking, rather than cognate SNARE pairing. Our study defines the link between VAMP7 and the ciliary targeting nexus that is conserved across diverse cell types, and contributes to general understanding of how functional Arf and Rab networks assemble SNAREs in membrane trafficking., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2018. Published by The Company of Biologists Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
17. Modeling Dominant and Recessive Forms of Retinitis Pigmentosa by Editing Three Rhodopsin-Encoding Genes in Xenopus Laevis Using Crispr/Cas9.
- Author
-
Feehan JM, Chiu CN, Stanar P, Tam BM, Ahmed SN, and Moritz OL
- Subjects
- Animals, CRISPR-Associated Protein 9 genetics, CRISPR-Cas Systems, Female, Genes, Dominant, Genes, Recessive, Green Fluorescent Proteins genetics, Humans, Male, Phenotype, Point Mutation, RNA, Guide, CRISPR-Cas Systems genetics, Retinitis Pigmentosa pathology, Sequence Deletion, Xenopus Proteins genetics, Xenopus laevis embryology, Disease Models, Animal, Gene Editing methods, Retinitis Pigmentosa genetics, Rhodopsin genetics
- Abstract
The utility of Xenopus laevis, a common research subject for developmental biology, retinal physiology, cell biology, and other investigations, has been limited by lack of a robust gene knockout or knock-down technology. Here we describe manipulation of the X. laevis genome using CRISPR/Cas9 to model the human disorder retinitis pigmentosa, and to introduce point mutations or exogenous DNA sequences. We introduced and characterized in-frame and out-of-frame insertions and deletions in three genes encoding rhodopsin by co-injection of Cas9 mRNA, eGFP mRNA, and single guide RNAs into fertilized eggs. Deletions were characterized by direct sequencing and cloning; phenotypes were assessed by assays of rod opsin in retinal extracts, and confocal microscopy of cryosectioned and immunolabeled contralateral eyes. We obtained germline transmission of editing to F1 offspring. In-frame deletions frequently caused dominant retinal degeneration associated with rhodopsin biosynthesis defects, while frameshift phenotypes were consistent with knockout. We inserted eGFP or point mutations into rhodopsin genes by co-injection of repair fragments with homology to the Cas9 target sites. Our techniques can produce high frequency gene editing in X. laevis, permitting analysis in the F0 generation, and advancing the utility of X. laevis as a subject for biological research and disease modeling.
- Published
- 2017
- Full Text
- View/download PDF
18. Opposing Effects of Valproic Acid Treatment Mediated by Histone Deacetylase Inhibitor Activity in Four Transgenic X. laevis Models of Retinitis Pigmentosa.
- Author
-
Vent-Schmidt RYJ, Wen RH, Zong Z, Chiu CN, Tam BM, May CG, and Moritz OL
- Subjects
- Animals, Autophagosomes metabolism, Histone Deacetylase Inhibitors pharmacology, Histones metabolism, Humans, Photoreceptor Cells drug effects, Photoreceptor Cells metabolism, Photoreceptor Cells ultrastructure, Retinitis Pigmentosa genetics, Retinitis Pigmentosa metabolism, Rhodopsin genetics, Valproic Acid pharmacology, Xenopus laevis, Histone Deacetylase Inhibitors therapeutic use, Retinitis Pigmentosa drug therapy, Valproic Acid therapeutic use
- Abstract
Retinitis pigmentosa (RP) is an inherited retinal degeneration (RD) that leads to blindness for which no treatment is available. RP is frequently caused by mutations in Rhodopsin ; in some animal models, RD is exacerbated by light. Valproic acid (VPA) is a proposed treatment for RP and other neurodegenerative disorders, with a phase II trial for RP under way. However, the therapeutic mechanism is unclear, with minimal research supporting its use in RP. We investigated the effects of VPA on Xenopus laevis models of RP expressing human P23H, T17M, T4K, and Q344ter rhodopsins, which are associated with RP in humans. VPA ameliorated RD associated with P23H rhodopsin and promoted clearing of mutant rhodopsin from photoreceptors. The effect was equal to that of dark rearing, with no additive effect observed. Rescue of visual function was confirmed by electroretinography. In contrast, VPA exacerbated RD caused by T17M rhodopsin in light, but had no effect in darkness. Effects in T4K and Q344ter rhodopsin models were also negative. These effects of VPA were paralleled by treatment with three additional histone deacetylase (HDAC) inhibitors, but not other antipsychotics, chemical chaperones, or VPA structural analogues. In WT retinas, VPA treatment increased histone H3 acetylation. In addition, electron microscopy showed increased autophagosomes in rod inner segments with HDAC inhibitor (HDACi) treatment, potentially linking the therapeutic effects in P23H rhodopsin animals and negative effects in other models with autophagy. Our results suggest that the success or failure of VPA treatment is dependent on genotype and that HDACi treatment is contraindicated for some RP cases. SIGNIFICANCE STATEMENT Retinitis pigmentosa (RP) is an inherited, degenerative retinal disease that leads to blindness for which no therapy is available. We determined that valproic acid (VPA), currently undergoing a phase II trial for RP, has both beneficial and detrimental effects in animal models of RP depending on the underlying disease mechanism and that both effects are due to histone deacetylase (HDAC) inhibition possibly linked to autophagy regulation. Off-label use of VPA and other HDAC inhibitors for the treatment of RP should be limited to the research setting until this effect is understood and can be predicted. Our study suggests that, unless genotype is accounted for, clinical trials for RP treatments may give negative results due to multiple disease mechanisms with differential responses to therapeutic interventions., (Copyright © 2017 the authors 0270-6474/17/371039-16$15.00/0.)
- Published
- 2017
- Full Text
- View/download PDF
19. Molecular basis for photoreceptor outer segment architecture.
- Author
-
Goldberg AF, Moritz OL, and Williams DS
- Subjects
- Animals, Humans, Nerve Tissue Proteins metabolism, Rhodopsin metabolism, Rod Cell Outer Segment metabolism
- Abstract
To serve vision, vertebrate rod and cone photoreceptors must detect photons, convert the light stimuli into cellular signals, and then convey the encoded information to downstream neurons. Rods and cones are sensory neurons that each rely on specialized ciliary organelles to detect light. These organelles, called outer segments, possess elaborate architectures that include many hundreds of light-sensitive membranous disks arrayed one atop another in precise register. These stacked disks capture light and initiate the chain of molecular and cellular events that underlie normal vision. Outer segment organization is challenged by an inherently dynamic nature; these organelles are subject to a renewal process that replaces a significant fraction of their disks (up to ∼10%) on a daily basis. In addition, a broad range of environmental and genetic insults can disrupt outer segment morphology to impair photoreceptor function and viability. In this chapter, we survey the major progress that has been made for understanding the molecular basis of outer segment architecture. We also discuss key aspects of organelle lipid and protein composition, and highlight distributions, interactions, and potential structural functions of key OS-resident molecules, including: kinesin-2, actin, RP1, prominin-1, protocadherin 21, peripherin-2/rds, rom-1, glutamic acid-rich proteins, and rhodopsin. Finally, we identify key knowledge gaps and challenges that remain for understanding how normal outer segment architecture is established and maintained., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
20. NLRP3 inflammasome activation drives bystander cone photoreceptor cell death in a P23H rhodopsin model of retinal degeneration.
- Author
-
Viringipurampeer IA, Metcalfe AL, Bashar AE, Sivak O, Yanai A, Mohammadi Z, Moritz OL, Gregory-Evans CY, and Gregory-Evans K
- Subjects
- Animals, Bystander Effect drug effects, Cell Death drug effects, Cell Survival, Disease Models, Animal, Gene Expression Regulation, Humans, Imidazoles pharmacology, Indoles pharmacology, Rats, Rats, Transgenic, Retinal Cone Photoreceptor Cells drug effects, Retinal Degeneration genetics, Retinal Degeneration metabolism, Retinal Rod Photoreceptor Cells drug effects, Signal Transduction drug effects, Inflammasomes metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Retinal Cone Photoreceptor Cells cytology, Retinal Degeneration pathology, Retinal Rod Photoreceptor Cells cytology, Rhodopsin genetics
- Abstract
The molecular signaling leading to cell death in hereditary neurological diseases such as retinal degeneration is incompletely understood. Previous neuroprotective studies have focused on apoptotic pathways; however, incomplete suppression of cell death with apoptosis inhibitors suggests that other mechanisms are at play. Here, we report that different signaling pathways are activated in rod and cone photoreceptors in the P23H rhodopsin mutant rat, a model representing one of the commonest forms of retinal degeneration. Up-regulation of the RIP1/RIP3/DRP1 axis and markedly improved survival with necrostatin-1 treatment highlighted necroptosis as a major cell-death pathway in degenerating rod photoreceptors. Conversely, up-regulation of NLRP3 and caspase-1, expression of mature IL-1β and IL-18 and improved cell survival with N-acetylcysteine treatment suggested that inflammasome activation and pyroptosis was the major cause of cone cell death. This was confirmed by generation of the P23H mutation on an Nlrp3-deficient background, which preserved cone viability. Furthermore, Brilliant Blue G treatment inhibited inflammasome activation, indicating that the 'bystander cell death' phenomenon was mediated through the P2RX7 cell-surface receptor. Here, we identify a new pathway in cones for bystander cell death, a phenomenon important in development and disease in many biological systems. In other retinal degeneration models different cell-death pathways are activated, which suggests that the particular pathways that are triggered are to some extent genotype-specific. This also implies that neuroprotective strategies to limit retinal degeneration need to be customized; thus, different combinations of inhibitors will be needed to target the specific pathways in any given disease., (© The Author 2016. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
21. Light Induces Ultrastructural Changes in Rod Outer and Inner Segments, Including Autophagy, in a Transgenic Xenopus laevis P23H Rhodopsin Model of Retinitis Pigmentosa.
- Author
-
Bogéa TH, Wen RH, and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, Caspase 9 metabolism, Disease Models, Animal, Photoperiod, Retinal Photoreceptor Cell Inner Segment ultrastructure, Retinal Rod Photoreceptor Cells, Retinitis Pigmentosa chemically induced, Retinitis Pigmentosa pathology, Rod Cell Outer Segment metabolism, Rod Cell Outer Segment ultrastructure, Tacrolimus analogs & derivatives, Xenopus laevis, Autophagy radiation effects, Light adverse effects, Radiation Injuries, Experimental physiopathology, Retinal Photoreceptor Cell Inner Segment radiation effects, Retinitis Pigmentosa physiopathology, Rhodopsin metabolism, Rod Cell Outer Segment radiation effects
- Abstract
Purpose: We previously reported a transgenic Xenopus laevis model of retinitis pigmentosa in which tadpoles express the bovine form of P23H rhodopsin (bP23H) in rod photoreceptors. In this model, retinal degeneration was dependent on light exposure. Here, we investigated ultrastructural changes that occurred in the rod photoreceptors of these retinas when exposed to light., Methods: Tadpoles expressing bP23H in rods were transferred from constant darkness to a 12-hour light:12-hour dark (12L:12D) regimen. For comparison, transgenic tadpoles expressing an inducible form of caspase 9 (iCasp9) were reared in a 12L:12D regimen, and retinal degeneration was induced by administration of the drug AP20187. Tadpoles were euthanized at various time points, and eyes were processed for confocal light and transmission electron microscopy., Results: We observed defects in outer and inner segments of rods expressing bP23H that were aggravated by light exposure. Rod outer segments exhibited vesiculations throughout and were rapidly phagocytosed by the retinal pigment epithelium. In rod inner segments, we observed autophagic compartments adjacent to the endoplasmic reticulum and extensive vesiculation at later time points. These defects were not found in rods expressing iCasp9, which completely degenerated within 36 hours after drug administration., Conclusions: Our results indicate that ultrastructural defects in outer and inner segment membranes of bP23H expressing rods differ from those observed in drug-induced apoptosis. We suggest that light-induced retinal degeneration caused by P23H rhodopsin occurs via cell death with autophagy, which may represent an attempt to eliminate the mutant rhodopsin and/or damaged cellular compartments from the secretory pathway.
- Published
- 2015
- Full Text
- View/download PDF
22. Kinesin family 17 (osmotic avoidance abnormal-3) is dispensable for photoreceptor morphology and function.
- Author
-
Jiang L, Tam BM, Ying G, Wu S, Hauswirth WW, Frederick JM, Moritz OL, and Baehr W
- Subjects
- Amino Acid Sequence, Animals, HEK293 Cells, Humans, Kinesins chemistry, Kinesins genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Molecular Sequence Data, NIH 3T3 Cells, Protein Transport, Rhodopsin metabolism, Xenopus laevis, Kinesins physiology, Photoreceptor Cells, Vertebrate physiology
- Abstract
In Caenorhabditis elegans, homodimeric [kinesin family (KIF) 17, osmotic avoidance abnormal-3 (OSM-3)] and heterotrimeric (KIF3) kinesin-2 motors are required to establish sensory cilia by intraflagellar transport (IFT) where KIF3 and KIF17 cooperate to build the axoneme core and KIF17 builds the distal segments. However, the function of KIF17 in vertebrates is unresolved. We expressed full-length and motorless KIF17 constructs in mouse rod photoreceptors using adeno-associated virus in Xenopus laevis rod photoreceptors using a transgene and in ciliated IMCD3 cells. We found that tagged KIF17 localized along the rod outer segment axoneme when expressed in mouse and X. laevis photoreceptors, whereas KIF3A was restricted to the proximal axoneme. Motorless KIF3A and KIF17 mutants caused photoreceptor degeneration, likely through dominant negative effects on IFT. KIF17 mutant lacking the motor domain translocated to nuclei after exposure of a C-terminal nuclear localization signal. Germ-line deletion of Kif17 in mouse did not affect photoreceptor function. A rod-specific Kif3/Kif17 double knockout mouse demonstrated that KIF17 and KIF3 do not act synergistically and did not prevent rhodopsin trafficking to rod outer segments. In summary, the nematode model of KIF3/KIF17 cooperation apparently does not apply to mouse photoreceptors in which the photosensory cilium is built exclusively by KIF3., (© FASEB.)
- Published
- 2015
- Full Text
- View/download PDF
23. Photoreceptors at a glance.
- Author
-
Molday RS and Moritz OL
- Subjects
- Animals, Humans, Light Signal Transduction, Protein Transport, Retinal Degeneration metabolism, Retinal Rod Photoreceptor Cells metabolism
- Abstract
Retinal photoreceptor cells contain a specialized outer segment (OS) compartment that functions in the capture of light and its conversion into electrical signals in a process known as phototransduction. In rods, photoisomerization of 11-cis to all-trans retinal within rhodopsin triggers a biochemical cascade culminating in the closure of cGMP-gated channels and hyperpolarization of the cell. Biochemical reactions return the cell to its 'dark state' and the visual cycle converts all-trans retinal back to 11-cis retinal for rhodopsin regeneration. OS are continuously renewed, with aged membrane removed at the distal end by phagocytosis and new membrane added at the proximal end through OS disk morphogenesis linked to protein trafficking. The molecular basis for disk morphogenesis remains to be defined in detail although several models have been proposed, and molecular mechanisms underlying protein trafficking are under active investigation. The aim of this Cell Science at a Glance article and the accompanying poster is to highlight our current understanding of photoreceptor structure, phototransduction, the visual cycle, OS renewal, protein trafficking and retinal degenerative diseases., (© 2015. Published by The Company of Biologists Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
24. Preparation of Xenopus laevis retinal cryosections for electron microscopy.
- Author
-
Tam BM, Yang LL, Bogėa TH, Ross B, Martens G, and Moritz OL
- Subjects
- Animals, Histocytological Preparation Techniques, Microscopy, Electron, Transmission, Tissue Embedding, Tissue Fixation methods, Cryoultramicrotomy methods, Retina ultrastructure, Xenopus laevis
- Abstract
Transmission electron microscopy is the gold standard for examination of photoreceptor outer segment morphology and photoreceptor outer segment abnormalities in transgenic animal models of retinal disease. Small vertebrates such as zebrafish and Xenopus laevis tadpoles have been used to generate retinal disease models and to study outer segment processes such as protein trafficking, and their breeding capabilities facilitate experiments involving large numbers of animals and conditions. However, electron microscopy processing and analysis of these very small eyes can be challenging. Here we present a methodology that facilitates processing of X. laevis tadpole eyes for electron microscopy by introducing an intermediate cryosectioning step. This method reproducibly provides a well-oriented tissue block that can be sectioned with minimal effort by a non-expert, and also allows retroactive analysis of samples collected on slides for light microscopy., (Copyright © 2015. Published by Elsevier Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
25. Photoactivation-induced instability of rhodopsin mutants T4K and T17M in rod outer segments underlies retinal degeneration in X. laevis transgenic models of retinitis pigmentosa.
- Author
-
Tam BM, Noorwez SM, Kaushal S, Kono M, and Moritz OL
- Subjects
- Analysis of Variance, Animals, Animals, Genetically Modified, COS Cells, Chlorocebus aethiops, Disease Models, Animal, Humans, Microscopy, Confocal, Retinal Degeneration diet therapy, Statistics, Nonparametric, Transfection, Vitamin A administration & dosage, Vitamin A metabolism, Wheat Germ Agglutinins metabolism, Xenopus laevis, Light, Mutation genetics, Retinal Degeneration etiology, Retinitis Pigmentosa complications, Retinitis Pigmentosa genetics, Retinitis Pigmentosa pathology, Rhodopsin genetics, Rod Cell Outer Segment pathology
- Abstract
Retinitis pigmentosa (RP) is an inherited neurodegenerative disease involving progressive vision loss, and is often linked to mutations in the rhodopsin gene. Mutations that abolish N-terminal glycosylation of rhodopsin (T4K and T17M) cause sector RP in which the inferior retina preferentially degenerates, possibly due to greater light exposure of this region. Transgenic animal models expressing rhodopsin glycosylation mutants also exhibit light exacerbated retinal degeneration (RD). In this study, we used transgenic Xenopus laevis to investigate the pathogenic mechanism connecting light exposure and RD in photoreceptors expressing T4K or T17M rhodopsin. We demonstrate that increasing the thermal stability of these rhodopsins via a novel disulfide bond resulted in significantly less RD. Furthermore, T4K or T17M rhodopsins that were constitutively inactive (due to lack of the chromophore-binding site or dietary deprivation of the chromophore precursor vitamin A) induced less toxicity. In contrast, variants in the active conformation accumulated in the ER and caused RD even in the absence of light. In vitro, T4K and T17M rhodopsins showed reduced ability to regenerate pigment after light exposure. Finally, although multiple amino acid substitutions of T4 abolished glycosylation at N2 but were not toxic, similar substitutions of T17 were not tolerated, suggesting that the carbohydrate moiety at N15 is critical for cell viability. Our results identify a novel pathogenic mechanism in which the glycosylation-deficient rhodopsins are destabilized by light activation. These results have important implications for proposed RP therapies, such as vitamin A supplementation, which may be ineffective or even detrimental for certain RP genotypes., (Copyright © 2014 the authors 0270-6474/14/3413336-13$15.00/0.)
- Published
- 2014
- Full Text
- View/download PDF
26. Mutant ELOVL4 that causes autosomal dominant stargardt-3 macular dystrophy is misrouted to rod outer segment disks.
- Author
-
Agbaga MP, Tam BM, Wong JS, Yang LL, Anderson RE, and Moritz OL
- Subjects
- Animals, DNA Mutational Analysis, Disease Models, Animal, Eye Proteins metabolism, Immunohistochemistry, Macular Degeneration genetics, Macular Degeneration metabolism, Macular Degeneration pathology, Membrane Proteins metabolism, Mice, Mice, Transgenic, Microscopy, Confocal, Rod Cell Outer Segment pathology, Xenopus laevis genetics, DNA genetics, Eye Proteins genetics, Macular Degeneration congenital, Membrane Proteins genetics, Mutation, Rod Cell Outer Segment metabolism
- Abstract
Purpose: Autosomal dominant Stargardt macular dystrophy caused by mutations in the Elongation of Very Long Chain fatty acids (ELOVL4) gene results in macular degeneration, leading to early childhood blindness. Transgenic mice and pigs expressing mutant ELOVL4 develop progressive photoreceptor degeneration. The mechanism by which these mutations cause macular degeneration remains unclear, but have been hypothesized to involve the loss of an ER-retention dilysine motif located in the extreme C-terminus. Dominant negative mechanisms and reduction in retinal polyunsaturated fatty acids also have been suggested. To understand the molecular mechanisms involved in disease progression in vivo, we addressed the hypothesis that the disease-linked C-terminal truncation mutant of ELOVL4 exerts a dominant negative effect on wild-type (WT) ELOVL4, altering its subcellular localization and function, which subsequently induces retinal degeneration and loss of vision., Methods: We generated transgenic Xenopus laevis that overexpress HA-tagged murine ELOVL4 variants in rod photoreceptors., Results: Tagged or untagged WT ELOVL4 localized primarily to inner segments. However, the mutant protein lacking the dilysine motif was mislocalized to post-Golgi compartments and outer segment disks. Coexpression of mutant and WT ELOVL4 in rods did not result in mislocalization of the WT protein to outer segments or in the formation of aggregates. Full-length HA-tagged ELOVL4 lacking the dilysine motif (K308R/K310R) necessary for targeting the WT ELOVL4 protein to the endoplasmic reticulum was similarly mislocalized to outer segments., Conclusions: We propose that expression and outer segment mislocalization of the disease-linked 5-base-pair deletion mutant ELOVL4 protein alters photoreceptor structure and function, which subsequently results in retinal degeneration, and suggest three possible mechanisms by which mutant ELOVL4 may induce retinal degeneration in STGD3., (Copyright 2014 The Association for Research in Vision and Ophthalmology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
27. Xenopus laevis tadpoles can regenerate neural retina lost after physical excision but cannot regenerate photoreceptors lost through targeted ablation.
- Author
-
Lee DC, Hamm LM, and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, Apoptosis, Blotting, Western, Disease Models, Animal, Immunohistochemistry, Larva, Microscopy, Confocal, Retina surgery, Retinitis Pigmentosa genetics, Xenopus laevis genetics, Microsurgery, Regeneration, Retina physiology, Retinal Cone Photoreceptor Cells physiology, Retinal Rod Photoreceptor Cells physiology, Retinitis Pigmentosa pathology
- Abstract
Purpose: To determine whether the Xenopus laevis retina is capable of regenerating photoreceptor cells lost through apoptotic cell death in an inducible transgenic X. laevis model of retinitis pigmentosa (RP)., Methods: Acute rod photoreceptor apoptosis was induced in transgenic X. laevis expressing drug-inducible caspase 9. We subsequently monitored the ability of the retina to regenerate lost photoreceptors in the absence of drug, and in combination with physical injury or ectopic supplementation of basic fibroblast growth factor (FGF2)., Results: Direct activation of caspase 9 in rod photoreceptors resulted in the initiation of apoptosis and complete removal of rod photoreceptors within 4 days. Photoreceptors lost by apoptosis were not replaced over a 4-week recovery time frame. In contrast, physical disruption of rod-ablated retina was repaired by the end of a 3-week time frame, but did not result in rod photoreceptor regeneration other than at the site of injury. Furthermore, ectopic supplementation of FGF2 did not stimulate regeneration of photoreceptors lost by apoptosis. However, FGF2 supplementation increased the rate of regeneration of retina (including rod photoreceptors) in eyes from which retinal tissue was surgically removed., Conclusions: In the X. laevis retina, rod photoreceptors that undergo drug-induced caspase-9-mediated apoptosis are permanently lost and do not regenerate. In contrast, the neural retina (including rod photoreceptors) can regenerate in injured or retinectomized eyes, and this regeneration is promoted by supplementation with FGF2. However, FGF2 does not promote regeneration of rod photoreceptors that are selectively lost by apoptosis.
- Published
- 2013
- Full Text
- View/download PDF
28. Targeting inflammation in emerging therapies for genetic retinal disease.
- Author
-
Viringipurampeer IA, Bashar AE, Gregory-Evans CY, Moritz OL, and Gregory-Evans K
- Abstract
Genetic retinal diseases such as age-related macular degeneration and monogenic diseases such as retinitis pigmentosa account for some of the commonest causes of blindness in the developed world. Diverse genetic abnormalities and environmental causes have been implicated in triggering multiple pathological mechanisms such as oxidative stress, lipofuscin deposits, neovascularisation, and programmed cell death. In recent years, inflammation has also been highlighted although whether inflammatory mediators play a central role in pathogenesis or a more minor secondary role has yet to be established. Despite this, numerous interventional studies, particularly targeting the complement system, are underway with the promise of novel therapeutic strategies for these important blinding conditions.
- Published
- 2013
- Full Text
- View/download PDF
29. Generation of transgenic X. laevis models of retinal degeneration.
- Author
-
Tam BM, Lai CC, Zong Z, and Moritz OL
- Subjects
- Animals, Humans, Transgenes, Animals, Genetically Modified genetics, Disease Models, Animal, Retinal Degeneration genetics, Xenopus laevis genetics
- Abstract
Transgenic models are invaluable tools for researching retinal degenerative disease mechanisms. However, they are time-consuming and expensive to generate and maintain. We have developed an alternative to transgenic rodent models of retinal degeneration using transgenic Xenopus laevis. We have optimized this system to allow rapid analysis of transgene effects in primary transgenic animals, thereby providing an alternative to establishing transgenic lines, and simultaneously allowing rigorous comparisons between the effects of different transgenes.
- Published
- 2013
- Full Text
- View/download PDF
30. Influence of Iron Oxide Nanoparticles on Innate and Genetically Modified Secretion Profiles of Mesenchymal Stem Cells.
- Author
-
Bashar AE, Metcalfe A, Yanai A, Laver C, Häfeli UO, Gregory-Evans CY, Moritz OL, Matsubara JA, and Gregory-Evans K
- Abstract
Mesenchymal stem cells (MSCs) have well-established paracrine effects that are proving to be therapeutically useful. This potential is based on the ability of MSCs to secrete a range of neuroprotective and anti-inflammatory molecules. Previous work in our laboratory has demonstrated that intravenous injection of MSCs, treated with superparamagnetic iron oxide nanoparticle fluidMAG-D resulted in enhanced levels of glial-derived neurotrophic factor, ciliary neurotrophic factor, hepatocyte growth factor and interleukin-10 in the dystrophic rat retina. In this present study we investigated whether the concentration of fluidMAG-D in cell culture media affects the secretion of these four molecules in vitro . In addition, we assessed the effect of fluidMAG-D concentration on retinoschisin secretion from genetically modified MSCs. ELISA-assayed secretion of these molecules was measured using escalating concentrations of fluidMAG-D which resulted in MSC iron loads of 0, 7, 120, or 274 pg iron oxide per cell respectively. Our results demonstrated glial-derived neurotrophic factor and hepatocyte growth factor secretion was significantly decreased but only at the 96 hour's time-point whereas no statistically significant effect was seen with ciliary neurotrophic factor secretion. Whereas no effect was observed on culture media concentrations of retinoschisin with increasing iron oxide load, a statistically significant increase in cell lysate retinoschisin concentration (p = 0.01) was observed suggesting that increasing fluidMAG-D concentration did increase retinoschisin production but this did not lead to greater secretion. We hypothesize that higher concentrations of iron-oxide nanoparticle fluidMAG-D have an effect on the innate ability of MSCs to secrete therapeutically useful molecules and also on secretion from genetically modified cells. Further work is required to verify these in vitro finding using in vivo model systems.
- Published
- 2013
- Full Text
- View/download PDF
31. Dysmorphic photoreceptors in a P23H mutant rhodopsin model of retinitis pigmentosa are metabolically active and capable of regenerating to reverse retinal degeneration.
- Author
-
Lee DC, Vazquez-Chona FR, Ferrell WD, Tam BM, Jones BW, Marc RE, and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, Disease Models, Animal, Histidine genetics, Nerve Regeneration genetics, Proline genetics, Retinal Degeneration metabolism, Retinal Rod Photoreceptor Cells physiology, Retinitis Pigmentosa metabolism, Rhodopsin genetics, Rhodopsin physiology, Xenopus laevis, Amino Acid Substitution genetics, Mutation genetics, Retinal Degeneration genetics, Retinal Rod Photoreceptor Cells metabolism, Retinitis Pigmentosa genetics, Rhodopsin metabolism
- Abstract
This study evaluated the capacity of Xenopus laevis retina to regenerate photoreceptor cells after cyclic light-mediated acute rod photoreceptor degeneration in a transgenic P23H mutant rhodopsin model of retinits pigmentosa. After discontinuation of cyclic light exposure, we monitored histologic progression of retinal regeneration over a 3 week recovery period. To assess their metabolomic states, contralateral eyes were processed for computational molecular phenotyping. We found that retinal degeneration in the P23H rhodopsin mutation could be partially reversed, with regeneration of rod photoreceptors recovering normal morphology (including full-length rod outer segments) by the end of the 3 week recovery period. In contrast, retinal degeneration mediated by directly induced apoptosis did not recover in the 3 week recovery period. Dystrophic rod photoreceptors with truncated rod outer segments were identified as the likely source of rod photoreceptor regeneration in the P23H retinas. These dystrophic photoreceptors remain metabolically active despite having lost most of their outer segments.
- Published
- 2012
- Full Text
- View/download PDF
32. Focused magnetic stem cell targeting to the retina using superparamagnetic iron oxide nanoparticles.
- Author
-
Yanai A, Häfeli UO, Metcalfe AL, Soema P, Addo L, Gregory-Evans CY, Po K, Shan X, Moritz OL, and Gregory-Evans K
- Subjects
- Animals, Cell Survival drug effects, Hepatocyte Growth Factor metabolism, Interleukin-10 metabolism, Magnetite Nanoparticles toxicity, Mesenchymal Stem Cell Transplantation, Mesenchymal Stem Cells chemistry, Microscopy, Confocal, Rats, Rats, Transgenic, Retinal Degeneration metabolism, Retinal Degeneration pathology, Ferric Compounds chemistry, Magnetite Nanoparticles chemistry, Mesenchymal Stem Cells cytology, Retinal Degeneration therapy
- Abstract
Developing new ways of delivering cells to diseased tissue will be a key factor in translating cell therapeutics research into clinical use. Magnetically targeting cells enables delivery of significant numbers of cells to key areas of specific organs. To demonstrate feasibility in neurological tissue, we targeted cells magnetically to the upper hemisphere of the rodent retina. Rat mesenchymal stem cells (MSCs) were magnetized using superparamagnetic iron oxide nanoparticles (SPIONs). In vitro studies suggested that magnetization with fluidMAG-D was well tolerated, that cells remained viable, and they retained their differentiation capabilities. FluidMAG-D-labeled MSCs were injected intravitreally or via the tail vein of the S334ter-4 transgenic rat model of retinal degeneration with or without placing a gold-plated neodymium disc magnet within the orbit, but outside the eye. Retinal flatmount and cryosection imaging demonstrated that after intravitreal injection cells localized to the inner retina in a tightly confined area corresponding to the position of the orbital magnet. After intravenous injection, similar retinal localization was achieved and remarkably was associated with a tenfold increase in magnetic MSC delivery to the retina. Cryosections demonstrated that cells had migrated into both the inner and outer retina. Magnetic MSC treatment with orbital magnet also resulted in significantly higher retinal concentrations of anti-inflammatory molecules interleukin-10 and hepatocyte growth factor. This suggested that intravenous MSC therapy also resulted in significant therapeutic benefit in the dystrophic retina. With minimal risk of collateral damage, these results suggest that magnetic cell delivery is the best approach for controlled delivery of cells to the outer retina-the focus for disease in age-related macular degeneration and retinitis pigmentosa.
- Published
- 2012
- Full Text
- View/download PDF
33. Targeting of mouse guanylate cyclase 1 (Gucy2e) to Xenopus laevis rod outer segments.
- Author
-
Karan S, Tam BM, Moritz OL, and Baehr W
- Subjects
- Animals, Animals, Genetically Modified, Cell Membrane metabolism, Membrane Fusion Proteins metabolism, Molecular Sequence Data, Protein Sorting Signals genetics, Protein Sorting Signals physiology, Xenopus laevis genetics, Guanylate Cyclase metabolism, Receptors, Cell Surface metabolism, Rod Cell Outer Segment metabolism, Xenopus laevis metabolism
- Abstract
Photoreceptor guanylate cyclase (GC1) is a transmembrane protein and responsible for synthesis of cGMP, the secondary messenger of phototransduction. It consists of an extracellular domain, a single transmembrane domain, and an intracellular domain. It is unknown how GC1 targets to the outer segments where it resides. To identify a putative GC1 targeting signal, we generated a series of peripheral membrane and transmembrane constructs encoding extracellular and intracellular mouse GC1 fragments fused to EGFP. The constructs were expressed in Xenopus laevis rod photoreceptors under the control of the rhodopsin promoter. We examined the localization of GFP-GC1 fusion proteins containing the complete GC1 sequence, or partial GC1 sequences, which were membrane-associated via either the GC1 transmembrane domain or the rhodopsin C-terminal palmitoyl chains. Full-length GFP-GC1 targeted to the rod outer segment disk rims. As a group, fusion proteins containing the entire cytoplasmic domain of GC1 targeted to the OS, whereas other fusion proteins containing portions of the cytoplasmic or the extracellular domains did not. We conclude that GC1 likely has no single linear peptide-based OS targeting signal. Our results suggest targeting is due to either multiple weak signals in the cytoplasmic domain of GC1, or co-transport to the OS with an accessory protein., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
34. In situ visualization of protein interactions in sensory neurons: glutamic acid-rich proteins (GARPs) play differential roles for photoreceptor outer segment scaffolding.
- Author
-
Ritter LM, Khattree N, Tam B, Moritz OL, Schmitz F, and Goldberg AF
- Subjects
- Alcohol Oxidoreductases, Animals, Animals, Genetically Modified, Cell Line, Transformed, Co-Repressor Proteins, DNA-Binding Proteins metabolism, Humans, Immunoprecipitation methods, Larva, Luminescent Proteins genetics, Luminescent Proteins metabolism, Microscopy, Immunoelectron methods, Phosphoproteins metabolism, Photoreceptor Cells, Vertebrate ultrastructure, Protein Transport genetics, Protein Transport physiology, Retinal Photoreceptor Cell Outer Segment ultrastructure, Rhodopsin metabolism, Transfection, Wheat Germ Agglutinin-Horseradish Peroxidase Conjugate metabolism, Xenopus, Cell Membrane metabolism, Photoreceptor Cells, Vertebrate cytology, Retinal Photoreceptor Cell Outer Segment metabolism, Sensory Receptor Cells metabolism
- Abstract
Vertebrate photoreceptors initiate vision via a G-protein-mediated signaling cascade organized within a specialized cilium, the outer segment (OS). The membranous "stacked pancake" architecture of this organelle must be partially renewed daily to maintain cell function and viability; however, neither its static structure nor renewal process is well described in molecular terms. Glutamic acid-rich proteins (GARPs), including the cyclic nucleotide-gated cation channel (CNGB1) and GARP2 (a CNGB1 splice-variant), are proposed to contribute to OS organization in concert with peripherin/rds (P/rds), a retinal tetraspanin. We developed and applied an in situ fluorescence complementation approach that offers an unprecedented glimpse at the formation, trafficking, and localization of GARP-P/rds interactions in transgenic Xenopus laevis rod photoreceptors. Interactions for these (and other) proteins could be readily visualized using confocal microscopy. Nearly all associations, including CNGB1-P/rds interaction, were initiated within inner segments (ISs) before trafficking to OSs. In contrast, GARP2-P/rds interactions were only observed downstream, at or near sites of disk morphogenesis. These results suggest that GARP2-P/rds interaction participates directly in structuring disk stacks but CNGB1-P/rds interaction does not and instead serves mainly to localize plasma membrane ion channels. Altogether, the results lead us to propose that differential interaction of GARPs with P/rds may contribute to the broad phenotypic heterogeneity produced by inherited defects in P/rds. Analogous experiments applied to the synaptic protein RIBEYE suggest that monomers can oligomerize at the level of the IS before ribbon assembly and demonstrate the general applicability of this strategy for in situ analysis of protein interactions in sensory neurons.
- Published
- 2011
- Full Text
- View/download PDF
35. The dependence of retinal degeneration caused by the rhodopsin P23H mutation on light exposure and vitamin a deprivation.
- Author
-
Tam BM, Qazalbash A, Lee HC, and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, Male, Microscopy, Confocal, Photometry, Radiation Injuries, Experimental genetics, Retina radiation effects, Retinal Degeneration genetics, Transgenes, Vitamin A physiology, Xenopus laevis, Light, Mutation, Missense, Radiation Injuries, Experimental etiology, Retinal Degeneration etiology, Rhodopsin genetics, Rhodopsin radiation effects, Vitamin A Deficiency complications
- Abstract
Purpose: To characterize the influence of light and vitamin A on retinal degeneration in an animal model of retinitis pigmentosa caused by the rhodopsin P23H mutation., Methods: Retinal degeneration was examined in transgenic Xenopus laevis expressing P23H rhodopsin, in which retinal degeneration is completely rescued by preventing light exposure. The sensitivity of this retinal degeneration to varying intensities, wavelengths, and durations of light exposure, and to vitamin A deprivation was characterized., Results: Green light was the most effective inducer of retinal degeneration in this model. Retinal degeneration was induced by prolonged exposure to green light and was prevented by filters that block short wavelengths. Reducing the duration of light exposure prevented retinal degeneration, even when the light intensity was proportionally increased. Vitamin A deprivation also induced retinal degeneration associated with defects in P23H rhodopsin biosynthesis. Vitamin A deprivation did not cause retinal degeneration in nontransgenic animals., Conclusions: The mechanism of retinal degeneration in this animal model of RP involves the interaction of light with rhodopsin rather than with free chromophore or bleached rhodopsin. These results may explain the clinical benefits of vitamin A for patients with retinitis pigmentosa and may indicate that pharmacological chaperones are a viable approach to RP therapy. Results also suggest strategies for minimizing RD in patients through controlling light exposure duration or wavelengths.
- Published
- 2010
- Full Text
- View/download PDF
36. Fourier domain optical coherence tomography as a noninvasive means for in vivo detection of retinal degeneration in Xenopus laevis tadpoles.
- Author
-
Lee DC, Xu J, Sarunic MV, and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, Caspase 9 genetics, Disease Models, Animal, Female, Fluorescent Antibody Technique, Indirect, Fourier Analysis, Larva, Male, Microscopy, Confocal, Retinal Degeneration genetics, Rhodopsin genetics, Xenopus laevis genetics, Phagosomes pathology, Retinal Degeneration diagnosis, Retinal Pigment Epithelium pathology, Rod Cell Outer Segment pathology, Tomography, Optical Coherence
- Abstract
Purpose: To determine the efficacy of Fourier domain optical coherence tomography (FD-OCT) as a noninvasive, nonlethal method for detecting in vivo, pathologic signs of retinal degeneration in Xenopus laevis larvae., Methods: A prototype OCT system using FD detection customized for tadpole imaging was used to noninvasively obtain retinal scans in two different transgenic X. laevis models of retinal degeneration. FD-OCT retinal scans were compared with laser scanning confocal micrographs of histologic sections of the same eye. Retinal thickness was measured in the histologic micrographs and compared with in vivo measurements acquired with FD-OCT., Results: In vivo retinal images of X. laevis tadpoles were obtained that visualized the major retinal layers. FD-OCT successfully detected the ablation of rod outer segments (OS) in degenerating tadpole eyes. Measurements from FD-OCT and histology showed a decrease in retinal thickness in transgenic mutant tadpoles relative to the wild-type control. The accumulation of phagosomes from dying rod OS was also visualized in the retinal pigment epithelium (RPE) in a degenerating tadpole retina., Conclusions: This report demonstrates that FD-OCT is a viable technique for screening, diagnosing, and monitoring retinal degeneration in X. laevis tadpoles in vivo.
- Published
- 2010
- Full Text
- View/download PDF
37. Recent insights into the mechanisms underlying light-dependent retinal degeneration from X. laevis models of retinitis pigmentosa.
- Author
-
Moritz OL and Tam BM
- Subjects
- Animals, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum pathology, Endoplasmic Reticulum radiation effects, Models, Biological, Mutant Proteins metabolism, Retinal Degeneration pathology, Retinitis Pigmentosa pathology, Rhodopsin genetics, Rhodopsin metabolism, Disease Models, Animal, Light, Retinal Degeneration complications, Retinitis Pigmentosa complications, Xenopus laevis
- Abstract
We have recently developed transgenic X. laevis models of retinitis pigmentosa based on the rhodopsin P23H mutation in the context of rhodopsin cDNAs derived from several different species. The mutant rhodopsin in these animals is expressed at low levels, with levels of export from the endoplasmic reticulum to the outer segment that depend on the cDNA context. Retinal degeneration in these models demonstrates varying degrees of light dependence, with the highest light dependence coinciding with the highest ER export efficiency. Rescue of light dependent retinal degeneration by dark rearing is in turn dependent on the capacity of the mutant rhodopsin to bind chromophore. Our results indicate that rhodopsin chromophore can act in vivo as a pharmacological chaperone for P23H rhodopsin, and that light-dependent retinal degeneration caused by P23H rhodopsin is due to reduced chromophore binding.
- Published
- 2010
- Full Text
- View/download PDF
38. The role of rhodopsin glycosylation in protein folding, trafficking, and light-sensitive retinal degeneration.
- Author
-
Tam BM and Moritz OL
- Subjects
- Amino Acids genetics, Animals, Animals, Genetically Modified, Disease Models, Animal, Glycosylation, Green Fluorescent Proteins genetics, Humans, Larva, Mutation genetics, Photoreceptor Cells, Vertebrate metabolism, Protein Transport genetics, Protein Transport physiology, Retinal Degeneration genetics, Retinal Degeneration pathology, Rhodopsin genetics, Xenopus, Light adverse effects, Protein Folding, Retinal Degeneration metabolism, Rhodopsin metabolism
- Abstract
Several mutations in the N terminus of the G-protein-coupled receptor rhodopsin disrupt NXS/T consensus sequences for N-linked glycosylation (located at N2 and N15) and cause sector retinitis pigmentosa in which the inferior retina preferentially degenerates. Here we examined the role of rhodopsin glycosylation in biosynthesis, trafficking, and retinal degeneration (RD) using transgenic Xenopus laevis expressing glycosylation-defective human rhodopsin mutants. Although mutations T4K and T4N caused RD, N2S and T4V did not, demonstrating that glycosylation at N2 was not required for photoreceptor viability. In contrast, similar mutations eliminating glycosylation at N15 (N15S and T17M) caused rod death. Expression of T17M was more toxic than T4K to transgenic photoreceptors, further suggesting that glycosylation at N15 plays a more important physiological role than glycosylation at N2. Together, these results indicate that the structure of the rhodopsin N terminus must be maintained by an appropriate amino acid sequence surrounding N2 and may require a carbohydrate moiety at N15. The mutant rhodopsins were rendered less toxic in their dark inactive states, because RD was abolished or significantly reduced when transgenic tadpoles expressing T4K, T17M, and N2S/N15S were protected from light exposure. Regardless of their effect on rod viability, all of the mutants primarily localized to the outer segment and Golgi and showed little or no endoplasmic reticulum accumulation. Thus, glycosylation was not crucial for rhodopsin biosynthesis or trafficking. Interestingly, expression of similar bovine rhodopsin mutants did not cause rod cell death, possibly attributable to greater stability of bovine rhodopsin.
- Published
- 2009
- Full Text
- View/download PDF
39. Ciliary targeting motif VxPx directs assembly of a trafficking module through Arf4.
- Author
-
Mazelova J, Astuto-Gribble L, Inoue H, Tam BM, Schonteich E, Prekeris R, Moritz OL, Randazzo PA, and Deretic D
- Subjects
- Actin Cytoskeleton metabolism, Adaptor Proteins, Signal Transducing metabolism, Amino Acid Motifs, Amino Acid Sequence, Animals, Animals, Genetically Modified, Cilia ultrastructure, GTPase-Activating Proteins metabolism, Guanosine Triphosphate metabolism, Hydrolysis, I-kappa B Kinase chemistry, I-kappa B Kinase metabolism, Intracellular Membranes metabolism, Intracellular Membranes ultrastructure, Molecular Sequence Data, Mutant Proteins metabolism, Organ Specificity, Protein Binding, Protein Sorting Signals, Protein Structure, Tertiary, Protein Transport, Retinal Degeneration metabolism, Xenopus genetics, rab GTP-Binding Proteins metabolism, trans-Golgi Network ultrastructure, ADP-Ribosylation Factors metabolism, Cilia metabolism, Rhodopsin chemistry, Rhodopsin metabolism, Xenopus Proteins metabolism
- Abstract
Dysfunctions of primary cilia and cilia-derived sensory organelles underlie a multitude of human disorders, including retinal degeneration, yet membrane targeting to the cilium remains poorly understood. Here, we show that the newly identified ciliary targeting VxPx motif present in rhodopsin binds the small GTPase Arf4 and regulates its association with the trans-Golgi network (TGN), which is the site of assembly and function of a ciliary targeting complex. This complex is comprised of two small GTPases, Arf4 and Rab11, the Rab11/Arf effector FIP3, and the Arf GTPase-activating protein ASAP1. ASAP1 mediates GTP hydrolysis on Arf4 and functions as an Arf4 effector that regulates budding of post-TGN carriers, along with FIP3 and Rab11. The Arf4 mutant I46D, impaired in ASAP1-mediated GTP hydrolysis, causes aberrant rhodopsin trafficking and cytoskeletal and morphological defects resulting in retinal degeneration in transgenic animals. As the VxPx motif is present in other ciliary membrane proteins, the Arf4-based targeting complex is most likely a part of conserved machinery involved in the selection and packaging of the cargo destined for delivery to the cilium.
- Published
- 2009
- Full Text
- View/download PDF
40. Controlled rod cell ablation in transgenic Xenopus laevis.
- Author
-
Hamm LM, Tam BM, and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, Blotting, Western, Caspase 9 genetics, Caspase 9 metabolism, Disease Models, Animal, Electroretinography, Enzyme Activation, Gene Expression Regulation, Enzymologic physiology, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Microscopy, Confocal, Opsins genetics, Promoter Regions, Genetic, Retina enzymology, Retinal Cone Photoreceptor Cells enzymology, Retinal Rod Photoreceptor Cells enzymology, Retinitis Pigmentosa enzymology, Retinitis Pigmentosa genetics, Tacrolimus pharmacology, Xenopus laevis genetics, Apoptosis drug effects, Retina physiopathology, Retinal Cone Photoreceptor Cells pathology, Retinal Rod Photoreceptor Cells pathology, Retinitis Pigmentosa pathology, Tacrolimus analogs & derivatives
- Abstract
Purpose: Because of their high cone/rod ratio, Xenopus laevis may be a useful system for examining rod-cone interactions during retinal degeneration and mechanisms that underlie secondary cone degeneration. The authors developed an inducible model of retinitis pigmentosa (RP) in X. laevis to investigate these issues., Methods: The authors generated transgenic X. laevis that express a modified caspase-9 (iCasp9) under the control of the X. laevis rod opsin promoter. iCasp9 is activated by the compound AP20187, resulting in an apoptotic cascade. Confocal microscopy, Western blot analysis, and electroretinography (ERG) were used to determine the effects of AP20187 on transgenic retinas., Results: AP20187 induced rod cell apoptosis in transgenic tadpoles and postmetamorphic frogs. Longitudinal results indicate rod cell death led to cone cell dysfunction within 3 months; however, cone function was reinstated after 6 months. Returning cone function may be associated with increased numbers of morphologically normal cone cells and thickening of the inner nuclear layer., Conclusions: These studies indicate that X. laevis may be a useful system for examining cone dysfunction associated with rod death in RP and longer term regeneration of cone responses. This inducible model of RP is unique in that rod death proceeds through a well-understood mechanism, rod death can be carefully controlled to occur at any stage of development, and the stimulus for rod death can be removed at any time.
- Published
- 2009
- Full Text
- View/download PDF
41. CRX controls retinal expression of the X-linked juvenile retinoschisis (RS1) gene.
- Author
-
Langmann T, Lai CC, Weigelt K, Tam BM, Warneke-Wittstock R, Moritz OL, and Weber BH
- Subjects
- Acetylation, Animals, Base Sequence, Binding Sites, Cell Line, Dogs, Eye Proteins biosynthesis, Histones metabolism, Humans, Mice, Molecular Sequence Data, Photoreceptor Cells, Vertebrate metabolism, Response Elements, Retinal Bipolar Cells metabolism, Transcriptional Activation, Xenopus laevis, p300-CBP Transcription Factors metabolism, Eye Proteins genetics, Homeodomain Proteins metabolism, Promoter Regions, Genetic, Retina metabolism, Trans-Activators metabolism
- Abstract
X-linked juvenile retinoschisis is a heritable condition of the retina in males caused by mutations in the RS1 gene. Still, the cellular function and retina-specific expression of RS1 are poorly understood. To address the latter issue, we characterized the minimal promoter driving expression of RS1 in the retina. Binding site prediction, site-directed mutagenesis, and reporter assays suggest an essential role of two nearby cone-rod homeobox (CRX)-responsive elements (CRE) in the proximal -177/+32 RS1 promoter. Chromatin immunoprecipitation associates the RS1 promoter in vivo with CRX, the coactivators CBP, P300, GCN5 and acetylated histone H3. Transgenic Xenopus laevis expressing a green fluorescent protein (GFP) reporter under the control of RS1 promoter sequences show that the -177/+32 fragment drives GFP expression in photoreceptors and bipolar cells. Mutating either of the two conserved CRX binding sites results in strongly decreased RS1 expression. Despite the presence of sequence motifs in the promoter, NRL and NR2E3 appear not to be essential for RS1 expression. Together, our in vitro and in vivo results indicate that two CRE sites in the minimal RS1 promoter region control retinal RS1 expression and establish CRX as a key factor driving this expression.
- Published
- 2008
- Full Text
- View/download PDF
42. Dark rearing rescues P23H rhodopsin-induced retinal degeneration in a transgenic Xenopus laevis model of retinitis pigmentosa: a chromophore-dependent mechanism characterized by production of N-terminally truncated mutant rhodopsin.
- Author
-
Tam BM and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, Cattle, Cell Line, Transformed, Disease Models, Animal, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Gene Expression Regulation radiation effects, Humans, Mice, Microscopy, Electron, Scanning methods, Peptide Fragments genetics, Peptide Fragments metabolism, Retinal Rod Photoreceptor Cells metabolism, Retinal Rod Photoreceptor Cells physiopathology, Retinal Rod Photoreceptor Cells ultrastructure, Retinaldehyde pharmacology, Transfection methods, Xenopus laevis, Darkness, Histidine genetics, Mutation, Proline genetics, Retinitis Pigmentosa genetics, Retinitis Pigmentosa pathology, Retinitis Pigmentosa therapy, Rhodopsin genetics
- Abstract
To elucidate the molecular mechanisms underlying the light-sensitive retinal degeneration caused by the rhodopsin mutation P23H, which causes retinitis pigmentosa (RP) in humans, we expressed Xenopus laevis, bovine, human, and murine forms of P23H rhodopsin in transgenic X. laevis rod photoreceptors. All P23H rhodopsins caused aggressive retinal degeneration associated with low expression levels and retention of P23H rhodopsin in the endoplasmic reticulum (ER), suggesting involvement of protein misfolding and ER stress. However, light sensitivity varied dramatically between these RP models, with complete or partial rescue by dark rearing in the case of bovine and human P23H rhodopsin, and no rescue for X. laevis P23H rhodopsin. Rescue by dark rearing required an intact 11-cis-retinal chromophore binding site within the mutant protein and was associated with truncation of the P23H rhodopsin N terminus. This yielded an abundant nontoxic approximately 27 kDa form that escaped the ER and was transported to the rod outer segment. The truncated protein was produced in the greatest quantities in dark-reared retinas expressing bovine P23H rhodopsin and was not observed with X. laevis P23H rhodopsin. These results are consistent with a mechanism involving enhanced protein folding in the presence of 11-cis-retinal chromophore, with ER exit assisted by proteolytic truncation of the N terminus. This study provides a molecular mechanism for light sensitivity observed in other transgenic models of RP and for phenotypic variation among RP patients.
- Published
- 2007
- Full Text
- View/download PDF
43. Characterization of rhodopsin P23H-induced retinal degeneration in a Xenopus laevis model of retinitis pigmentosa.
- Author
-
Tam BM and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, Blotting, Western, Cell Death, Cell Line, Dark Adaptation, Dose-Response Relationship, Drug, Fluorescent Antibody Technique, Indirect, Kidney embryology, Microscopy, Confocal, Photoreceptor Cells, Vertebrate metabolism, Photoreceptor Cells, Vertebrate pathology, Retinal Degeneration metabolism, Retinal Degeneration pathology, Retinitis Pigmentosa metabolism, Retinitis Pigmentosa pathology, Rhodopsin metabolism, Transducin metabolism, Transfection, Transgenes, Xenopus laevis, Disease Models, Animal, Mutation, Retinal Degeneration genetics, Retinitis Pigmentosa genetics, Rhodopsin genetics
- Abstract
Purpose: To investigate the pathogenic mechanisms that underlie retinal degeneration induced by the rhodopsin mutation P23H in a Xenopus laevis model of RP., Methods: Transgenic X. laevis were generated that expressed the rhodopsin mutants rhoP23H and rhoP23H/K29R (a variant incapable of transducin activation). Using quantitative dot blot assay, transgenic rhodopsin levels and the extent of retinal degeneration were determined. The contribution of rhodopsin signal transduction to cell death was assessed by comparison of rhoP23H and rhoP23H/K296R effects and by dark rearing of rhoP23H tadpoles. Intracellular localization and the oligomeric state of rhoP23H were determined by confocal immunofluorescence microscopy and Western blot analysis., Results: RhoP23H induced retinal degeneration in a dose-dependent manner whereas expression of a control rhodopsin did not, indicating that rod photoreceptor death was specific to the P23H mutation and was not caused by the overexpression of rhodopsin. Neither abolishment of rhoP23H photosensitivity and ability to activate transducin nor dark rearing rescued rod viability. RhoP23H was localized primarily to the endoplasmic reticulum (ER) of inner segments. Western blot analysis of transgenic retinas showed that rhoP23H was prone to form dimers and higher molecular weight oligomers. However, aggresomes were not observed in rhoP23H transgenic retinal sections, despite their being reported in cultured cells expressing rhoP23H., Conclusions: These results support a role for rhoP23H misfolding and inner segment accumulation in rod death, possibly by ER overload or other cellular stress pathways rather than by altered rhodopsin signal transduction or aggresome formation.
- Published
- 2006
- Full Text
- View/download PDF
44. Mislocalized rhodopsin does not require activation to cause retinal degeneration and neurite outgrowth in Xenopus laevis.
- Author
-
Tam BM, Xie G, Oprian DD, and Moritz OL
- Subjects
- Animals, Animals, Genetically Modified, COS Cells, Chlorocebus aethiops, Female, Male, Neurites chemistry, Point Mutation, Retinal Degeneration genetics, Retinitis Pigmentosa genetics, Retinitis Pigmentosa metabolism, Rhodopsin genetics, Rhodopsin metabolism, Xenopus Proteins genetics, Xenopus Proteins metabolism, Xenopus laevis, Neurites metabolism, Retinal Degeneration metabolism, Rhodopsin physiology, Xenopus Proteins physiology
- Abstract
Mutations in the C terminus of rhodopsin disrupt a rod outer segment localization signal, causing rhodopsin mislocalization and aggressive forms of retinitis pigmentosa (RP). Studies of cultured photoreceptors suggest that activated mislocalized rhodopsin can cause cell death via inappropriate G-protein-coupled signaling. To determine whether this pathway occurs in vivo, we developed a transgenic Xenopus laevis model of RP based on the class I rhodopsin mutation Q344Ter (Q350Ter in X. laevis). We used a second mutation, K296R, to block the ability of rhodopsin to bind chromophore and activate transducin. We compared the effects of expression of both mutants on X. laevis retinas alone and in combination. K296R did not significantly alter the cellular distribution of rhodopsin and did not induce retinal degeneration. Q350Ter caused rhodopsin mislocalization and induced an RP-like degeneration, including loss of rods and development of sprouts or neurites in some remaining rods, but did not affect the distribution of endogenous rhodopsin. The double mutant K296R/Q350Ter caused a similar degeneration and neurite outgrowth. In addition, we found no protective effects of dark rearing in these animals. Our results demonstrate that the degenerative effects of mislocalized rhodopsin are not mediated by the activated form of rhodopsin and therefore do not proceed via conventional G-protein-coupled signaling.
- Published
- 2006
- Full Text
- View/download PDF
45. Uncoupling of photoreceptor peripherin/rds fusogenic activity from biosynthesis, subunit assembly, and targeting: a potential mechanism for pathogenic effects.
- Author
-
Ritter LM, Boesze-Battaglia K, Tam BM, Moritz OL, Khattree N, Chen SC, and Goldberg AF
- Subjects
- Amino Acid Sequence, Animals, Animals, Genetically Modified, COS Cells, Cell Membrane metabolism, Disulfides metabolism, Escherichia coli, Fluorescent Antibody Technique, Glutathione Transferase genetics, Molecular Sequence Data, Mutagenesis, Peripherins, Recombinant Fusion Proteins genetics, Xenopus Proteins, Intermediate Filament Proteins genetics, Intermediate Filament Proteins metabolism, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Retinal Rod Photoreceptor Cells physiology, Xenopus laevis physiology
- Abstract
Inherited defects in the RDS gene cause a multiplicity of progressive retinal diseases in humans. The gene product, peripherin/rds (P/rds), is a member of the tetraspanin protein family required for normal vertebrate photoreceptor outer segment (OS) architecture. Although its molecular function remains uncertain, P/rds has been suggested to catalyze membrane fusion events required for the OS renewal process. This study investigates the importance of two charged residues within a predicted C-terminal helical region for protein biosynthesis, localization, and interaction with model membranes. Targeted mutagenesis was utilized to neutralize charges at Glu(321) and Lys(324) individually and in combination to generate three mutant variants. Studies were conducted on variants expressed as 1) full-length P/rds in COS-1 cells, 2) glutathione S-transferase fusion proteins in Escherichia coli, and 3) membrane-associated green fluorescent protein fusion proteins in transgenic Xenopus laevis. None of the mutations affected biosynthesis of full-length P/rds in COS-1 cells as assessed by Western blotting, sedimentation velocity, and immunofluorescence microscopy. Although all mutations reside within a recently identified localization signal, none altered the ability of this region to direct OS targeting in transgenic X. laevis retinas. In contrast, individual or simultaneous neutralization of the charged amino acids Glu(321) and Lys(324) abolished the ability of the C-terminal domain to promote model membrane fusion as assayed by lipid mixing. These results demonstrate that, although overlapping, C-terminal determinants responsible for OS targeting and fusogenicity are separable and that fusogenic activity has been uncoupled from other protein properties. The observation that subunit assembly and OS targeting can both proceed normally in the absence of fusogenic activity suggests that properly assembled and targeted yet functionally altered proteins could potentially generate pathogenic effects within the vertebrate photoreceptor.
- Published
- 2004
- Full Text
- View/download PDF
46. The C terminus of peripherin/rds participates in rod outer segment targeting and alignment of disk incisures.
- Author
-
Tam BM, Moritz OL, and Papermaster DS
- Subjects
- Amino Acid Sequence, Animals, COS Cells, Cattle, Cell Division, Green Fluorescent Proteins, Humans, Immunohistochemistry, Luminescent Proteins chemistry, Luminescent Proteins metabolism, Microscopy, Confocal, Microscopy, Electron, Microscopy, Immunoelectron, Molecular Sequence Data, Peripherins, Protein Structure, Tertiary, Reactive Oxygen Species, Time Factors, Transgenes, Xenopus Proteins, Xenopus laevis, Intermediate Filament Proteins chemistry, Membrane Glycoproteins chemistry, Nerve Tissue Proteins chemistry, Rod Cell Outer Segment metabolism
- Abstract
Protein targeting is essential for domain specialization in polarized cells. In photoreceptors, three distinct membrane domains exist in the outer segment: plasma membrane, disk lamella, and disk rim. Peripherin/retinal degeneration slow (rds) and rom-1 are photoreceptor-specific members of the transmembrane 4 superfamily of transmembrane proteins, which participate in disk morphogenesis and localize to rod outer segment (ROS) disk rims. We examined the role of their C termini in targeting by generating transgenic Xenopus laevis expressing green fluorescent protein (GFP) fusion proteins. A GFP fusion containing residues 317-336 of peripherin/rds localized uniformly to disk membranes. A longer fusion (residues 307-346) also localized to the ROS but exhibited higher affinity for disk rims than disk lamella. In contrast, the rom-1 C terminus did not promote ROS localization. The GFP-peripherin/rds fusion proteins did not immunoprecipitate with peripherin/rds or rom-1, suggesting this region does not form intermolecular interactions and is not involved in subunit assembly. Presence of GFP-peripherin/rds fusions correlated with disrupted incisures, disordered ROS tips, and membrane whorls. These abnormalities may reflect competition of the fusion proteins for other proteins that interact with peripherin/rds. This work describes novel roles for the C terminus of peripherin/rds in targeting and maintaining ROS structure and its potential involvement in inherited retinal degenerations.
- Published
- 2004
- Full Text
- View/download PDF
47. The role of subunit assembly in peripherin-2 targeting to rod photoreceptor disk membranes and retinitis pigmentosa.
- Author
-
Loewen CJ, Moritz OL, Tam BM, Papermaster DS, and Molday RS
- Subjects
- Animals, Animals, Genetically Modified, COS Cells, Chlorocebus aethiops, Cloning, Molecular, Green Fluorescent Proteins, Luminescent Proteins, Mutation, Peripherins, Protein Subunits metabolism, Retinitis Pigmentosa genetics, Xenopus Proteins, Xenopus laevis, Intermediate Filament Proteins metabolism, Membrane Glycoproteins metabolism, Nerve Tissue Proteins metabolism, Retinitis Pigmentosa metabolism, Rod Cell Outer Segment metabolism
- Abstract
Peripherin-2 is a member of the tetraspanin family of membrane proteins that plays a critical role in photoreceptor outer segment disk morphogenesis. Mutations in peripherin-2 are responsible for various retinal degenerative diseases including autosomal dominant retinitis pigmentosa (ADRP). To identify determinants required for peripherin-2 targeting to disk membranes and elucidate mechanisms underlying ADRP, we have generated transgenic Xenopus tadpoles expressing wild-type and ADRP-linked peripherin-2 mutants as green fluorescent fusion proteins in rod photoreceptors. Wild-type peripherin-2 and P216L and C150S mutants, which assemble as tetramers, targeted to disk membranes as visualized by confocal and electron microscopy. In contrast the C214S and L185P mutants, which form homodimers, but not tetramers, were retained in the rod inner segment. Only the P216L disease mutant induced photoreceptor degeneration. These results indicate that tetramerization is required for peripherin-2 targeting and incorporation into disk membranes. Tetramerization-defective mutants cause ADRP through a deficiency in wild-type peripherin-2, whereas tetramerization-competent P216L peripherin-2 causes ADRP through a dominant negative effect, possibly arising from the introduction of a new oligosaccharide chain that destabilizes disks. Our results further indicate that a checkpoint between the photoreceptor inner and outer segments allows only correctly assembled peripherin-2 tetramers to be incorporated into nascent disk membranes.
- Published
- 2003
- Full Text
- View/download PDF
48. Arrestin migrates in photoreceptors in response to light: a study of arrestin localization using an arrestin-GFP fusion protein in transgenic frogs.
- Author
-
Peterson JJ, Tam BM, Moritz OL, Shelamer CL, Dugger DR, McDowell JH, Hargrave PA, Papermaster DS, and Smith WC
- Subjects
- Animals, Animals, Genetically Modified, Dark Adaptation physiology, Green Fluorescent Proteins, Light, Luminescent Proteins, Microscopy, Confocal, Photic Stimulation, Recombinant Fusion Proteins metabolism, Rod Cell Outer Segment metabolism, Xenopus laevis, Adaptation, Ocular physiology, Arrestin metabolism, Retinal Rod Photoreceptor Cells metabolism
- Abstract
Subcellular translocation of phototransduction proteins in response to light has previously been detected by immunocytochemistry. This movement is consistent with the hypothesis that migration is part of a basic cellular mechanism regulating photoreceptor sensitivity. In order to monitor the putative migration of arrestin in response to light, we expressed a functional fusion between the signal transduction protein arrestin and green fluorescent protein (GFP) in rod photoreceptors of transgenic Xenopus laevis. In addition to confirming reports that arrestin is translocated, this alternative approach generated unique observations, raising new questions regarding the nature and time scale of migration. Confocal fluorescence microscopy was performed on fixed frozen retinal sections from tadpoles exposed to three different lighting conditions. A consistent pattern of localization emerged in each case. During early light exposure, arrestin-GFP levels diminished in the inner segments (ISs) and simultaneously increased in the outer segments (OSs), initially at the base and eventually at the distal tips as time progressed. Arrestin-GFP reached the distal tips of the photoreceptors by 45-75 min at which time the ratio of arrestin-GFP fluorescence in the OSs compared to the ISs was maximal. When dark-adaptation was initiated after 45 min of light exposure, arrestin-GFP rapidly re-localized to the ISs and axoneme within 30 min. Curiously, prolonged periods of light exposure also resulted in re-localization of arrestin-GFP. Between 150 and 240 min of light adaptation the arrestin-GFP in the ROS gradually declined until the pattern of arrestin-GFP localization was indistinguishable from that of dark-adapted photoreceptors. This distribution pattern was observed over a wide range of lighting intensity (25-2700 lux). Immunocytochemical analysis of arrestin in wild-type Xenopus retinas gave similar results.
- Published
- 2003
- Full Text
- View/download PDF
49. Xenopus laevis red cone opsin and Prph2 promoters allow transgene expression in amphibian cones, or both rods and cones.
- Author
-
Moritz OL, Peck A, and Tam BM
- Subjects
- Animals, Animals, Genetically Modified, Base Sequence, Cloning, Molecular, Gene Expression, Green Fluorescent Proteins, Luminescent Proteins genetics, Microscopy, Fluorescence, Molecular Sequence Data, Peripherins, RNA, Messenger genetics, RNA, Messenger metabolism, Recombinant Fusion Proteins genetics, Reverse Transcriptase Polymerase Chain Reaction, Xenopus Proteins, Intermediate Filament Proteins genetics, Membrane Glycoproteins, Nerve Tissue Proteins genetics, Photoreceptor Cells, Vertebrate metabolism, Promoter Regions, Genetic genetics, Rod Opsins genetics, Transgenes genetics, Xenopus laevis genetics
- Abstract
We have cloned the promoter regions of two Xenopus laevis genes, Prph2 (also called RDS) and red cone opsin (RCO) using a polymerase chain reaction-based gene-walking method. The proteins coded by these genes are expressed exclusively in retinal photoreceptors. Although these promoter sequences are evolutionarily distant from previously described homologues, potentially informative similarities were noted that suggest conserved binding sites of the transcription factors Crx and Rx. The promoters were tested for function in transgenic X. laevis. RCO-driven expression was restricted to cones and pinealocytes, while the Prph2 promoter drove expression of a reporter green fluorescent protein transgene in both rod and cone photoreceptors, as well as low levels of expression in muscle tissue. This is the first description of transgene expression driven by a Prph2 promoter homologue from any species. In combination with the previously reported X. laevis opsin and arrestin promoters, these sequences will facilitate the development and analysis of X. laevis models of inherited retinal degeneration.
- Published
- 2002
- Full Text
- View/download PDF
50. Selection of transgenic Xenopus laevis using antibiotic resistance.
- Author
-
Moritz OL, Biddle KE, and Tam BM
- Subjects
- Animals, Biomarkers, Gentamicins pharmacology, Xenopus laevis genetics, Animals, Genetically Modified, Drug Resistance genetics
- Abstract
We previously established lines of transgenic Xenopus laevis expressing green fluorescent protein (GFP) or GFP fusion proteins in the rod photoreceptors of their retinas under control of the X. laevis opsin promoter, which permits easy identification of transgenic animals by fluorescence microscopy. However, GFP tags can alter the properties of fusion partners, and in many circumstances a second selectable marker would be useful. The transgene constructs we used also encode a gene that confers resistance to the antibiotic G418 in cultured mammalian cells. In this study, we show that F2 transgenic offspring of these animals are more resistant to G418 toxicity than their non-transgenic siblings, as are primary transgenic X. laevis. G418 resistance can be used as a selectable marker in transgenic X. laevis, and possibly other aquatic transgenic animals.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.