116 results on '"Vleminckx, K."'
Search Results
2. Hearing loss in Waardenburg syndrome: a systematic review
- Author
-
Song, J., Feng, Y., Acke, F. R., Coucke, P., Vleminckx, K., and Dhooge, I. J.
- Published
- 2016
- Full Text
- View/download PDF
3. PO-206 Modelling T cell acute lymphoblastic leukaemia using CRISPR/Cas9 mediated genome editing in Xenopus tropicalis
- Author
-
Dimitrakopoulou, D., primary, Tulkens, D., additional, Naert, T., additional, Vlierberghe, P. Van, additional, and Vleminckx, K., additional
- Published
- 2018
- Full Text
- View/download PDF
4. Late abstracts 186–187
- Author
-
Jaehne, J., Meyer, H. -J., Wittekind, Ch., Maschek, H., Pichlmayr, R., Jacobi, G., Weiermann, G., Vitzthum, H. Gräfin, Schwabe, D., Manegold, Ch., Krempien, B., Kaufmann, M., Bailly, M., Doré, J. -F., Fodstad, Ø., Kjønniksen, I., Brøgger, A., Flørenes, V. A., Pihl, A., Aamdal, S., Nesland, J. M., Geldof, A. A., Rao, B. R., De Giovanni, C., Lollini, P. -L., Del Re, B., Scotlandi, K., Nicoletti, G., Nanni, P., Van Muijen, G. N. P., Van Der Wiel-Miezenbeek, J. M., Cornelissen, L. M. H. A., Jansen, C. F. J., Ruiter, D. J., Kieler, J., Oda, Y., Tokuriki, Y., Tenang, E. M., Lamb, J. F., Galante, E., Zanoni, F., Galluzzi, D., Cerrotta, A., Martelli, G., Guzzon, A., Reduzzi, D., Barberá-Guillem, E., Barceló, J. R., Urcelay, B., Alonso-Varona, A. I., Vidal-Vanaclocha, F., Bassukas, I. D., Maurer-Schultze, B., Storeng, R., Manzotti, C., Pratesi, G., Schachert, G., Fidler, I. J., Grimstad, I. A., Rutt, G. Th., Riesinger, P., Frank, J., Neumann, G., Wissler, J. H., Bastert, G., Liebrich, W., Lehner, B., Gonzer, S., Schlag, P., Vehmeyer, K., Hajto, T., Gabius, H. -J., Funke, I., Schlimok, G., Bock, B., Dreps, A., Schweiberer, B., Riethmüller, G., Nicolai, U., Vykoupil, K. -F., Wolf, M., Havemann, K., Georgii, A., Bertrand, S., N'Guyen, M. -J., Siracky, J., Kysela, B., Siracka, E., Pflüger, E., Schirrmacher, V., Boyano, M. D., Hanania, N., Poupon, M. F., Sherbet, G. V., Lakshmi, M. S., Van Roy, F., Vleminckx, K., Fiers, W., Dragonetti, C., De Bruyne, G., Messiaen, L., Mareel, M., Kuhn, S., Choritz, H., Schmid, U., Bihl, H., Griesbach, A., Matzku, S., Eccles, S. A., Purvies, H. P., Miller, F. R., McEachern, D., Ponton, A., Waghorne, C., Coulombe, B., Kerbel, R. S., Breitman, M., Skup, D., Gingras, M. C., Jarolim, L., Wright, J. A., Greenberg, A. H., N'Guyen, M. J., Allavena, G., Melchiori, A., Aresu, O., Percario, M., Parodi, S., Schmidt, J., Kars, P., Chader, G., Albini, A., Zöller, M., Lissitzky, J. C., Bouzon, M., Martin, P. M., Grossi, I. M., Taylor, J. D., Honn, K. V., Koch, B., Baum, W., Giedl, J., Gabius, H. J., Kalden, J. R., Hakim, A. A., LadÁnyi, A., Timár, J., Moczar, E., Lapis, K., Müller, K., Wolf, M. F., Benz, B., Schumacher, K., Kemmner, W., Morgenthaler, J., Brossmer, R., Hagmar, B., Burns, G., Erkell§, L. J., Ryd, W., Paku, S., Rot, A., Hilario, E., Unda, F., Simón, J., Aliño, S. F., Sargent, N. S. E., Burger, M. M., Altevogt, P., Kowitz, A., Chopra, H., Bandlow, G., Nagel, G. A., Lotan, R., Carralero, D., Lotan, D., Raz, A., Skubitz, A. P. N., Koliakos, G. G., Furcht, L. T., Charonis, A. S., Hamann, A., Jablonski-Westrich, D., Jonas, P., Harder, R., Butcher, E. C., Thiele, H. G., Breillout, F., Antoine, E., Lascaux, V., Boxberger, H. -J., Paweletz, N., Bracke, M., Vyncke, B., Opdenakker, G., Castronovo, V., Foidart, J. -M., Camacho, M., Fras, A. Fabra, Llorens, A., Rutllant, M. L., Erkell, L. J., Brunner, G., Heredia, A., Imhoff, J. M., Burtin, P., Nakajima, M., Lunec, J., Parker, C., Fennelly, J. A., Smith, K., Roossien, F. F., La Rivière, G., Roos, E., Erdel, M., Trefz, G., Spiess, E., Ebert, W., Verhaegen, S., Remels, L., Verschueren, H., Dekegel, D., De Baetselier, P., Van Hecke, D., Hannecart-Pokorni, E., Falkvoll, K. H., Alonso, A., Baroja, A., Sebbag, U., Barbera-Guillem, E., Behrens, J., Mareel, M. M., Birchmeier, W., Waterhouse, P., Khokha, R., Chambers, A., Yagel, S., Lala, P. K., Denhardt, D. T., Hennes, R., Frantzen, F., Keller, R., Schwartz-Albiez, R., Fondaneche, M. C., Mignatti, P., Tsuboi, R., Robbins, E., Rifkin, D. B., Overall, C. M., Sacchi, A., Falcioni, R., Piaggio, G., Rizzo, M. G., Perrotti, N., Kennel, S. J., Girschick, H., Müller-Hermelink, H. K., Vollmers, H. P., Wenzel, A., Liu, S., Günthert, U., Wesch, V., Giles, M., Ponta, H., Herrlich, P., Stade, B., Hupke, U., Holzmann, B., Johnson, J. P., Sauer, A., Roller, E., Klumpp, B., Güttler, N., Lison, A., Walk, A., Redini, F., Moczar, M., Leoni, F., Da Dalt, M. G., Ménard, S., Canevari, S., Miotti, S., Tagliabue, E., Colnaghi, M. I., Ostmeier, H., Suter, L., Possati, L., Rosciani, C., Recanatini, E., Beatrici, V., Diambrini, M., Polito, M., Rothbächer, U., Eisenbach, L., Plaksin, D., Gelber, C., Kushtai, G., Gubbay, J., Feldman, M., Benke, R., Benedetto, A., Elia, G., Sala, A., Belardelli, F., Lehmann, J. M., Ladanyi, A., Hanisch, F. -G., Sölter, J., Jansen, V., Böhmer, G., Peter-Katalinic, J., Uhlenbruck, G., O'Connor, R., Müller, J., Kirchner, T., Bover, B., Tucker, G., Valles, A. M., Gavrilovic, J., Thiery, J. P., Kaufmann, A. M., Volm, M., Edel, G., Zühlsdorf, M., Voss, H., Wörmann, B., Hiddemann, W., De Neve, W., Van Den Berge, D., Van Loon, R., Storme, G., Zacharski, L. R., Wojtukiewicz, M. Z., Memoli, V., Kisiel, W., Kudryk, B. J., Stump, D., Piñol, G., Gonzalez-Garrigues, M., Fabra, A., Marti, F., Rueda, F., Lichtner, R. B., Khazaie, K., Timar, J., Greenzhevskaya, S. N., Shmalko, Yu. P., Hill, S. E., Rees, R. C., MacNeil, S., Millon, R., Muller, D., Eber, M., Abecassis, J., Betzler, M., Bahtsky, K. P., Umansky, V. Yu., Krivorotov, A. A., Balitskaya, E. K., Pridatko, O. E., Smelkova, M. I., Smirnov, I. M., Korczak, B., Fisher, C., Thody, A. J., Young, S. D., Hill, R. P., Frixen, U., Gopas, J., Segal, S., Hammerling, G., Bar-Eli, M., Rager-Zisman, B., Har-Vardi, I., Alon, Y., Hämmerling, G. J., Perez, M., Algarra, I., Collado, Ma. D., Peran, E., Caballero, A., Garrido, F., Turner, G. A., Blackmore, M., Stern, P. L., Thompson, S., Levin, I., Kuperman, O., Eyal, A., Kaneti, J., Notter, M., Knuth, A., Martin, M., Chauffert, B., Caignard, A., Hammann, A., Martin, F., Dearden, M. T., Pelletier, H., Dransfield, I., Jacob, G., Rogers, K., Pérez-Yarza, G., Cañavate, M. L., Lucas, R., Bouwens, L., Mantovani, G., Serri, F. G., Macciò, A., Zucca, M. V., Del Giacco, G. S., Pérez, M., Kärre, K., Apt, D., Traversari, C., Sensi, M., Carbone, G., Parmiani, G., Hainaut, P., Weynants, P., Degiovanni, G., Boon, T., Marquardt, P., Stulle, K., Wölfel, T., Herin, M., Van den Eynde, B., Klehmann, E., Büschenfelde, K. -H. Meyer zum, Samija, M., Gerenčer, M., Eljuga, D., Bašić, I., Heacock, C. S., Blake, A. M., D'Aleo, C. J., Alvarez, V. L., Gresser, I., Maury, C., Moss, J., Woodrow, D., von Ardenne, M., Krüger, W., Möller, P., Schachert, H. K., Itaya, T., Frost, P., Rodolfo, M., Salvi, C., Bassi, C., Huland, E., Huland, H., Sersa, G., Willingham, V., Hunter, N., Milas, L., Schild, H., von Hoegen, P., Mentges, B., Bätz, W., Suzuki, N., Mizukoshi, T., Sava, G., Ceschia, V., Zabucchi, G., Farkas-Himsley, H., Schaal, O., Klenner, T., Keppler, B., Alvarez-Diaz, A., Bizzari, J. P., Barbera-Guillem, F., Osterloh, B., Bartkowski, R., LÖhrke, H., Schwahn, E., Schafmayer, A., Goerttler, K., Cillo, C., Ling, V., Giavazzi, R., Vecchi, A., Luini, W., Garofalo, A., Iwakawa, M., Arundel, C., Tofilon, P., Giraldi, T., Perissin, L., Zorzet, S., Piccini, P., Pacor, S., Rapozzi, V., Fink, U., Zeuner, H., Dancygier, H., Classen, M., Lersch, C., Reuter, M., Hammer, C., Brendel, W., Mathé, G., Bourut, C., Chenu, E., Kidani, Y., Mauvernay, Y., Schally, A. V., Reizenstein, P., Gastiaburu, J., Comaru-Schally, A. M., Cupissol, D., Jasmin, C., Missot, J. L., Wingen, F., Schmähl, D., Pauwels-Vergely, C., Poupon, M. -F., Gasic, T. B., Ewaskiewicz, J. I., Gasic, G. J., Pápay, J., Mauvernay, R., Schally, A., Keiling, R., Hagipantelli, R., Busuttil, M., VoVan, M. L., Misset, J. L., Lévi, F., Musset, M., Ribaud, P., Hilgard, P., Reissmann, T., Stekar, J., Voegeli, R., Den Otter, W., Maas, H. A., Dullens, H. F. J., Merriman, R. L., Tanzer, L. R., Shackelford, K. A., Bemis, K. G., Campbell, J. B., and Matsumoto, K.
- Published
- 1988
- Full Text
- View/download PDF
5. Hearing loss in Waardenburg syndrome: a systematic review
- Author
-
Song, J., primary, Feng, Y., additional, Acke, F.R., additional, Coucke, P., additional, Vleminckx, K., additional, and Dhooge, I.J., additional
- Published
- 2015
- Full Text
- View/download PDF
6. A novel RIPK4–IRF6 connection is required to prevent epithelial fusions characteristic for popliteal pterygium syndromes
- Author
-
De Groote, P, primary, Tran, H T, additional, Fransen, M, additional, Tanghe, G, additional, Urwyler, C, additional, De Craene, B, additional, Leurs, K, additional, Gilbert, B, additional, Van Imschoot, G, additional, De Rycke, R, additional, Guérin, C J, additional, Holland, P, additional, Berx, G, additional, Vandenabeele, P, additional, Lippens, S, additional, Vleminckx, K, additional, and Declercq, W, additional
- Published
- 2014
- Full Text
- View/download PDF
7. The Wnt signaling mediator tcf1 is required for expression of foxd3 during Xenopus gastrulation
- Author
-
Janssens, S., Van den Broek, O., Davenport, I.R., Akkers, R.C., Liu, F., Veenstra, G.J.C., Hoppler, S., Vleminckx, K., Destree, O., Janssens, S., Van den Broek, O., Davenport, I.R., Akkers, R.C., Liu, F., Veenstra, G.J.C., Hoppler, S., Vleminckx, K., and Destree, O.
- Abstract
Item does not contain fulltext
- Published
- 2013
8. Harmonisatie van de statistische concepten in de sociale zekerheid
- Author
-
Boos, C.L.J., Booghmans, A.M., Nachtergaele, W., Verhalle, I., Vleminckx, K., and Sociology
- Published
- 2002
9. Reducing child poverty in the European Union
- Author
-
Immervoll, H., Sutherland, H., de Vos, K., Vleminckx, K., Smeeding, T., and Research Group: Econometrics
- Published
- 2001
10. The chronic toxicity of molybdate to freshwater organisms. I. Generating reliable effects data
- Author
-
De Schamphelaere, K.A.C., primary, Stubblefield, W., additional, Rodriguez, P., additional, Vleminckx, K., additional, and Janssen, C.R., additional
- Published
- 2010
- Full Text
- View/download PDF
11. A novel RIPK4-IRF6 connection is required to prevent epithelial fusions characteristic for popliteal pterygium syndromes.
- Author
-
De Groote, P, Tran, H T, Fransen, M, Tanghe, G, Urwyler, C, De Craene, B, Leurs, K, Gilbert, B, Van Imschoot, G, De Rycke, R, Guérin, C J, Holland, P, Berx, G, Vandenabeele, P, Lippens, S, Vleminckx, K, and Declercq, W
- Subjects
RECEPTOR-interacting proteins ,INTERFERON regulatory factors ,PTERYGIUM ,EPITHELIAL cells ,PROTEIN kinases ,XENOPUS ,LABORATORY mice - Abstract
Receptor-interacting protein kinase 4 (RIPK4)-deficient mice have epidermal defects and fusion of all external orifices. These are similar to Bartsocas-Papas syndrome and popliteal pterygium syndrome (PPS) in humans, for which causative mutations have been documented in the RIPK4 and IRF6 (interferon regulatory factor 6) gene, respectively. Although genetically distinct, these syndromes share the anomalies of marked pterygia, syndactyly, clefting and hypoplastic genitalia. Despite the strong resemblance of these two syndromes, no molecular connection between the transcription factor IRF6 and the kinase RIPK4 was known and the mechanism underlying the phenotype was unclear. Here we describe that RIPK4 deficiency in mice causes epithelial fusions associated with abnormal periderm development and aberrant ectopic localization of E-cadherin on the apical membrane of the outer peridermal cell layers. In Xenopus, RIPK4 depletion causes the absence of ectodermal epiboly and concomitant gastrulation defects that phenocopy ectopic expression of dominant-negative IRF6. We found that IRF6 controls RIPK4 expression and that wild-type, but not kinase-dead, RIPK4 can complement the gastrulation defect in Xenopus caused by IRF6 malfunctioning. In contrast to the mouse, we observed only minor effects on cadherin membrane expression in Xenopus RIPK4 morphants. However, gastrulation defects were associated with a virtual absence of cortical actin in the ectodermal cells that face the blastocoel cavity and this was phenocopied in embryos expressing dominant-negative IRF6. A role for RIPK4 in actin cytoskeleton organization was also revealed in mouse epidermis and in human epithelial HaCaT cells. In conclusion, we showed that in mice RIPK4 is implicated in cortical actin organization and in E-cadherin localization or function, which can explain the characteristic epithelial fusions observed in PPSs. In addition, we provide a novel molecular link between IRF6 and RIPK4 that unifies the different PPSs to a common molecular pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
12. Syndecan-1 expression in mammary epithelial tumor cells is E-cadherin-dependent
- Author
-
Leppa, S., primary, Vleminckx, K., additional, Van Roy, F., additional, and Jalkanen, M., additional
- Published
- 1996
- Full Text
- View/download PDF
13. Insulin-like growth factor I activates the invasion suppressor function of E-cadherin in MCF-7 human mammary carcinoma cells in vitro
- Author
-
Bracke, ME, primary, Vyncke, BM, additional, Bruyneel, EA, additional, Vermeulen, SJ, additional, De Bruyne, GK, additional, Van Larebeke, NA, additional, Vleminckx, K, additional, Van Roy, FM, additional, and Mareel, MM, additional
- Published
- 1993
- Full Text
- View/download PDF
14. E-cadherin versus tumor invasion
- Author
-
Van Roy, F., primary, Vleminckx, K., additional, Berx, G., additional, Keirsebilck, A., additional, Bracke, M., additional, Deman, J., additional, Vandenbossche, G., additional, and Mareel, M., additional
- Published
- 1993
- Full Text
- View/download PDF
15. 3.3 Homotypic cell-cell adhesion molecules and tumor invasion
- Author
-
Mareel, M., primary, Vleminckx, K., additional, Vermeulen, S., additional, Gao, Y., additional, Vakaet, L., additional, Bracke, M., additional, and van Roy, F., additional
- Published
- 1992
- Full Text
- View/download PDF
16. Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role
- Author
-
Vleminckx, K., primary, Vakaet, L., additional, Mareel, M., additional, Fiers, W., additional, and Van Roy, F., additional
- Published
- 1991
- Full Text
- View/download PDF
17. Hormone sensitivity in vitro and in vivo of v‐ras‐transfected mcf‐7 cell derivatives
- Author
-
Van Roy, F., primary, Mareel, M., additional, Vleminckx, K., additional, Beyaert, R., additional, Fiers, W., additional, Devleeschouwer, N., additional, Muquardt, C., additional, Legros, N., additional, Bracke, M., additional, and Leclercq, G., additional
- Published
- 1990
- Full Text
- View/download PDF
18. Expression of E-cadherin in eptthelial tumor cells abrogates malignant invasion
- Author
-
VANROY, F, primary, VLEMINCKX, K, additional, FIERS, W, additional, and MAREEL, M, additional
- Published
- 1990
- Full Text
- View/download PDF
19. The C-terminal transactivation domain of -catenin is necessary and sufficient for signaling by the LEF-1/ -catenin complex in Xenopus laevis
- Author
-
Vleminckx, K., Kemler, R., and Hecht, A.
- Published
- 1999
- Full Text
- View/download PDF
20. Effect of oncogene transfection or passage in vivo on malignant phenotypes of rat2 cells
- Author
-
Fm, Roy, Peter Coopman, Suffys P, Liebaut G, Vleminckx K, Gao J, Ch, Dragonetti, Fiers W, and Mm, Mareel
- Subjects
Cell Transformation, Neoplastic ,Genes, ras ,Phenotype ,Animals ,DNA ,Oncogenes ,Cosmids ,Embryo, Mammalian ,Transfection ,Thymidine Kinase ,Cell Division ,Cell Line ,Rats - Abstract
Rat2 cells are thymidine kinase-deficient derivatives from the immortalized rat embryo cell line Rat1. They show no phenotypic correlates of malignancy in vitro and produce tumors in syngeneic Fischer rats after long latency periods. We have investigated how transfection with oncogenes would alter the in vitro and in vivo behavior of Rat2 cells. Thus we have manipulated Rat2 cultures in various ways. The cell lines obtained were categorized as parental, in vitro subclones, untransfected in vivo derivatives, non-oncogene (neor and tk) transfectants, oncogene (mutated c-Ha-ras, polyoma middle-T, FBR v-gag-fos-fox) transfectants, and in vivo derivatives of transfectants. They were tested in vitro for morphotype, colony formation in soft agar, growth in organ culture, invasion in organ culture, and in vivo for latency period of tumor formation, tumor growth rate, invasiveness, and metastasis. Differences between the consequences of various manipulations were found in the number of malignancy-related phenotypic alterations. The following trend could be deduced from our data: induction of invasiveness in organ culture by all manipulations; morphotypic transformation and shortening of tumor-latency period by all oncogene transfections and by passage with tumor formation in vivo; growth in organ culture and increased tumor growth rate in vivo by transfection with ras-, or fos-oncogenes and by passage in vivo. Metastatic capability (present in parental Rat2 cell tumors) and colony formation in soft agar (absent in Rat2 cells) were not affected by the present manipulations. We concluded that differences between the oncogene-transfectants and the untransfected in vivo derivatives do not lie in the expression of malignancy-related phenotypes but in the time needed to acquire them.
- Published
- 1989
21. Overlevingspensioen en/of arbeid: noden en behoeften van weduwen
- Author
-
Koen Ponnet, Dimitri Mortelmans, Paul Peter Vermeiren, and Vleminckx, K.
- Subjects
Sociology
22. Proffered papers and posters presented at the Sixth International Symposium on Hereditary Breast and Ovarian Cancer—BRCA: Challenges and Opportunities : Presented by the Hereditary Breast and Ovarian Cancer Foundation in collaboration with the Program in Cancer Genetics, McGill University; Centre Mont-Royal, Montreal, QC; 10–13 May 2016
- Author
-
Starita L, Islam M, Fields S, Parvin J, Shendure J, Vierstraete J, Willaert A, Vleminckx K, Vermassen P, Pieters L, Coucke P, Vral A, Claes K, and Chalas E
23. Mechanism underlying synergic activation of tyrosinase promoter by MITF and IRF4
- Author
-
Song, J., Liu, X., Li, J., Liu, H., Peng, Z., Liu, Y., Mei, L., He, C., Cai, X., Hongsheng Chen, Vleminckx, K., and Feng, Y.
- Subjects
EXPRESSION ,TYR ,MITF ,integumentary system ,MUTATIONS ,EUROPEANS ,EPITHELIUM ,Biology and Life Sciences ,MELANOMA ,synergy effects ,IRF4 ,MICROPHTHALMIA TRANSCRIPTION FACTOR ,Medicine and Health Sciences ,GENETIC-DETERMINANTS ,PIGMENTATION ,transcriptional activation ,WAARDENBURG SYNDROME TYPE-2 ,IN-VIVO - Abstract
Background: The transcription factor interferon regulatory factor 4 (IRF4) was identified to be involved in human pigmentation by genome-wide association studies (GWASs). The rs12203592-[T/C], which is located in intron 4 of IRF4, shows the strongest link to these pigmentation phenotypes including freckling, sun sensitivity, eye and hair color. Previous studies indicated a functional cooperation of IRF4 with Microphthalmia-associated transcription factor (MITF), a causing gene of Waardenburg syndrome (WS), to synergistically trans-activate Tyrosinase (TYR). However, the underlying mechanism is still unknown. Methods: To investigate the importance of DNA binding in the synergic effect of IRF4. Reporter plasmids with mutant TYR promoters was generated to locate the IRF4 DNA binding sites in the Tyrosinase minimal promoter. By building MITF and IRF4 truncated mutations plasmids, the necessary regions of the synergy functions of these two proteins were also located. Results: The cooperative effect between MITF and IRF4 was specific for TYR promoter. The DNA-binding of IRF4 was critical for the synergic function. IRF4 DNA binding sites in TYR promoter were identified. The Trans-activation domains in IRF4 (aa134-207, aa300-420) were both important for the synergic function, whereas the auto-mask domain (aa207-300) appeared to mask the synergic effect. Mutational analysis in MITF indicated that both DNA-binding and transcriptional activation domains were both required for this synergic effect. Conclusions: Here we showed that IRF4 potently synergized with MITF to activate the TYR promoter, which was dependent on DNA binding of IRF4. The synergic domains in both IRF4 and MITF were identified by mutational analysis. This identification of IRF4 as a partner for MITF in regulation of TYR may provide an important molecular function for IRF4 in the genesis of melanocytes and the pathogenic mechanism in WS.
24. Deletion upstream of MAB21L2 highlights the importance of evolutionarily conserved non-coding sequences for eye development.
- Author
-
Ceroni F, Cicekdal MB, Holt R, Sorokina E, Chassaing N, Clokie S, Naert T, Talbot LV, Muheisen S, Bax DA, Kesim Y, Kivuva EC, Vincent-Delorme C, Lienkamp SS, Plaisancié J, De Baere E, Calvas P, Vleminckx K, Semina EV, and Ragge NK
- Subjects
- Animals, Female, Humans, Male, Mice, Anophthalmos genetics, Conserved Sequence, Evolution, Molecular, Phenotype, Sequence Deletion, Xenopus genetics, Coloboma genetics, Eye, Microphthalmos genetics, Pedigree, Zebrafish genetics
- Abstract
Anophthalmia, microphthalmia and coloboma (AMC) comprise a spectrum of developmental eye disorders, accounting for approximately 20% of childhood visual impairment. While non-coding regulatory sequences are increasingly recognised as contributing to disease burden, characterising their impact on gene function and phenotype remains challenging. Furthermore, little is known of the nature and extent of their contribution to AMC phenotypes. We report two families with variants in or near MAB21L2, a gene where genetic variants are known to cause AMC in humans and animal models. The first proband, presenting with microphthalmia and coloboma, has a likely pathogenic missense variant (c.338 G > C; p.[Trp113Ser]), segregating within the family. The second individual, presenting with microphthalmia, carries an ~ 113.5 kb homozygous deletion 19.38 kb upstream of MAB21L2. Modelling of the deletion results in transient small lens and coloboma as well as midbrain anomalies in zebrafish, and microphthalmia and coloboma in Xenopus tropicalis. Using conservation analysis, we identify 15 non-coding conserved elements (CEs) within the deleted region, while ChIP-seq data from mouse embryonic stem cells demonstrates that two of these (CE13 and 14) bind Otx2, a protein with an established role in eye development. Targeted disruption of CE14 in Xenopus tropicalis recapitulates an ocular coloboma phenotype, supporting its role in eye development. Together, our data provides insights into regulatory mechanisms underlying eye development and highlights the importance of non-coding sequences as a source of genetic diagnoses in AMC., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
25. Evolutionary origin of Hoxc13-dependent skin appendages in amphibians.
- Author
-
Carron M, Sachslehner AP, Cicekdal MB, Bruggeman I, Demuynck S, Golabi B, De Baere E, Declercq W, Tschachler E, Vleminckx K, and Eckhart L
- Subjects
- Animals, Humans, Skin metabolism, Hair metabolism, Keratins genetics, Keratins metabolism, Amphibians, Mammals metabolism, Keratins, Hair-Specific, Transcription Factors metabolism
- Abstract
Cornified skin appendages, such as hair and nails, are major evolutionary innovations of terrestrial vertebrates. Human hair and nails consist largely of special intermediate filament proteins, known as hair keratins, which are expressed under the control of the transcription factor Hoxc13. Here, we show that the cornified claws of Xenopus frogs contain homologs of hair keratins and the genes encoding these keratins are flanked by promoters in which binding sites of Hoxc13 are conserved. Furthermore, these keratins and Hoxc13 are co-expressed in the claw-forming epithelium of frog toe tips. Upon deletion of hoxc13, the expression of hair keratin homologs is abolished and the development of cornified claws is abrogated in X. tropicalis. These results indicate that Hoxc13-dependent expression of hair keratin homologs evolved already in stem tetrapods, presumably as a mechanism for protecting toe tips, and that this ancestral genetic program was coopted to the growth of hair in mammals., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
26. Mutations in the histone methyltransferase Ezh2 drive context-dependent leukemia in Xenopus tropicalis.
- Author
-
Tulkens D, Boelens M, Naert T, Carron M, Demuynck S, Dewaele S, Van Isterdael G, Creytens D, Pieters T, Goossens S, Van Vlierberghe P, and Vleminckx K
- Subjects
- Animals, Humans, Histone Methyltransferases genetics, Xenopus genetics, Enhancer of Zeste Homolog 2 Protein genetics, Mutation, RNA, Guide, CRISPR-Cas Systems, Leukemia, Myeloid, Acute
- Abstract
CRISPR-mediated simultaneous targeting of candidate tumor suppressor genes in Xenopus tropicalis allows fast functional assessment of co-driver genes for various solid tumors. Genotyping of tumors that emerge in the mosaic mutant animals rapidly exposes the gene mutations under positive selection for tumor establishment. However, applying this simple approach to the blood lineage has not been attempted. Multiple hematologic malignancies have mutations in EZH2, encoding the catalytic subunit of the Polycomb Repressive Complex 2. Interestingly, EZH2 can act as an oncogene or a tumor suppressor, depending on cellular context and disease stage. We show here that mosaic CRISPR/Cas9 mediated ezh2 disruption in the blood lineage resulted in early and penetrant acute myeloid leukemia (AML) induction. While animals were co-targeted with an sgRNA that induces notch1 gain-of-function mutations, sequencing of leukemias revealed positive selection towards biallelic ezh2 mutations regardless of notch1 mutational status. Co-targeting dnm2, recurrently mutated in T/ETP-ALL, induced a switch from myeloid towards acute T-cell leukemia. Both myeloid and T-cell leukemias engrafted in immunocompromised hosts. These data underline the potential of Xenopus tropicalis for modeling human leukemia, where mosaic gene disruption, combined with deep amplicon sequencing of the targeted genomic regions, can rapidly and efficiently expose co-operating driver gene mutations., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
27. Multi-omics approach dissects cis-regulatory mechanisms underlying North Carolina macular dystrophy, a retinal enhanceropathy.
- Author
-
Van de Sompele S, Small KW, Cicekdal MB, Soriano VL, D'haene E, Shaya FS, Agemy S, Van der Snickt T, Rey AD, Rosseel T, Van Heetvelde M, Vergult S, Balikova I, Bergen AA, Boon CJF, De Zaeytijd J, Inglehearn CF, Kousal B, Leroy BP, Rivolta C, Vaclavik V, van den Ende J, van Schooneveld MJ, Gómez-Skarmeta JL, Tena JJ, Martinez-Morales JR, Liskova P, Vleminckx K, and De Baere E
- Subjects
- Adult, Animals, Humans, Pedigree, Retina metabolism, Xenopus laevis genetics, Tomography, Optical Coherence, Corneal Dystrophies, Hereditary
- Abstract
North Carolina macular dystrophy (NCMD) is a rare autosomal-dominant disease affecting macular development. The disease is caused by non-coding single-nucleotide variants (SNVs) in two hotspot regions near PRDM13 and by duplications in two distinct chromosomal loci, overlapping DNase I hypersensitive sites near either PRDM13 or IRX1. To unravel the mechanisms by which these variants cause disease, we first established a genome-wide multi-omics retinal database, RegRet. Integration of UMI-4C profiles we generated on adult human retina then allowed fine-mapping of the interactions of the PRDM13 and IRX1 promoters and the identification of eighteen candidate cis-regulatory elements (cCREs), the activity of which was investigated by luciferase and Xenopus enhancer assays. Next, luciferase assays showed that the non-coding SNVs located in the two hotspot regions of PRDM13 affect cCRE activity, including two NCMD-associated non-coding SNVs that we identified herein. Interestingly, the cCRE containing one of these SNVs was shown to interact with the PRDM13 promoter, demonstrated in vivo activity in Xenopus, and is active at the developmental stage when progenitor cells of the central retina exit mitosis, suggesting that this region is a PRDM13 enhancer. Finally, mining of single-cell transcriptional data of embryonic and adult retina revealed the highest expression of PRDM13 and IRX1 when amacrine cells start to synapse with retinal ganglion cells, supporting the hypothesis that altered PRDM13 or IRX1 expression impairs interactions between these cells during retinogenesis. Overall, this study provides insight into the cis-regulatory mechanisms of NCMD and supports that this condition is a retinal enhanceropathy., Competing Interests: Declaration of interests The authors declare no competing interests., (Crown Copyright © 2022. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
28. Engraftment of Allotransplanted Tumor Cells in Adult rag2 Mutant Xenopus tropicalis .
- Author
-
Tulkens D, Dimitrakopoulou D, Boelens M, Van Nieuwenhuysen T, Demuynck S, Toussaint W, Creytens D, Van Vlierberghe P, and Vleminckx K
- Abstract
Modeling human genetic diseases and cancer in lab animals has been greatly aided by the emergence of genetic engineering tools such as TALENs and CRISPR/Cas9. We have previously demonstrated the ease with which genetically engineered Xenopus models (GEXM) can be generated via injection of early embryos with Cas9 recombinant protein loaded with sgRNAs targeting single or multiple tumor suppressor genes. What has been lacking so far is the possibility to propagate and characterize the induced cancers via transplantation. Here, we describe the generation of a rag2 knockout line in Xenopus tropicalis that is deficient in functional T and B cells. This line was validated by means of allografting experiments with primary tp53
-/- and apc+/- /tp53-/- donor tumors. In addition, we optimized available protocols for the sub-lethal irradiation of wild-type X. tropicalis froglets. Irradiated animals also allowed the stable, albeit transient, engraftment of transplanted X. tropicalis tumor cells. The novel rag2-/- line and the irradiated wild-type froglets will further expand the experimental toolbox in the diploid amphibian X. tropicalis and help to establish it as a versatile and relevant model for exploring human cancer.- Published
- 2022
- Full Text
- View/download PDF
29. CRISPR-SID: Identifying EZH2 as a druggable target for desmoid tumors via in vivo dependency mapping.
- Author
-
Naert T, Tulkens D, Van Nieuwenhuysen T, Przybyl J, Demuynck S, van de Rijn M, Al-Jazrawe M, Alman BA, Coucke PJ, De Leeneer K, Vanhove C, Savvides SN, Creytens D, and Vleminckx K
- Subjects
- Abdominal Neoplasms genetics, Adenomatous Polyposis Coli genetics, Animals, Carcinogenesis genetics, Cell Line, Tumor, Cyclic AMP Response Element-Binding Protein, Fibromatosis, Aggressive genetics, Gene Expression Regulation, Neoplastic, Humans, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Nerve Tissue Proteins, Oncogenes, Polycomb Repressive Complex 2 metabolism, Transcription Factors genetics, Transcription Factors metabolism, Wnt Signaling Pathway, Xenopus, beta Catenin, CRISPR-Cas Systems, Clustered Regularly Interspaced Short Palindromic Repeats, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein isolation & purification, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Editing methods
- Abstract
Cancer precision medicine implies identification of tumor-specific vulnerabilities associated with defined oncogenic pathways. Desmoid tumors are soft-tissue neoplasms strictly driven by Wnt signaling network hyperactivation. Despite this clearly defined genetic etiology and the strict and unique implication of the Wnt/β-catenin pathway, no specific molecular targets for these tumors have been identified. To address this caveat, we developed fast, efficient, and penetrant genetic Xenopus tropicalis desmoid tumor models to identify and characterize drug targets. We used multiplexed CRISPR/Cas9 genome editing in these models to simultaneously target a tumor suppressor gene ( apc ) and candidate dependency genes. Our methodology CRISPR/Cas9 selection-mediated identification of dependencies (CRISPR-SID) uses calculated deviations between experimentally observed gene editing outcomes and deep-learning-predicted double-strand break repair patterns to identify genes under negative selection during tumorigenesis. This revealed EZH2 and SUZ12 , both encoding polycomb repressive complex 2 components, and the transcription factor CREB3L1 as genetic dependencies for desmoid tumors. In vivo EZH2 inhibition by Tazemetostat induced partial regression of established autochthonous tumors. In vitro models of patient desmoid tumor cells revealed a direct effect of Tazemetostat on Wnt pathway activity. CRISPR-SID represents a potent approach for in vivo mapping of tumor vulnerabilities and drug target identification., Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
30. Maximizing CRISPR/Cas9 phenotype penetrance applying predictive modeling of editing outcomes in Xenopus and zebrafish embryos.
- Author
-
Naert T, Tulkens D, Edwards NA, Carron M, Shaidani NI, Wlizla M, Boel A, Demuynck S, Horb ME, Coucke P, Willaert A, Zorn AM, and Vleminckx K
- Subjects
- Animals, CRISPR-Associated Protein 9 genetics, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Frameshift Mutation, Gene Frequency, HEK293 Cells, Humans, Mice, Mouse Embryonic Stem Cells metabolism, RNA, Guide, CRISPR-Cas Systems genetics, CRISPR-Cas Systems, Gene Editing methods, Penetrance, Xenopus laevis embryology, Xenopus laevis genetics, Zebrafish embryology, Zebrafish genetics
- Abstract
CRISPR/Cas9 genome editing has revolutionized functional genomics in vertebrates. However, CRISPR/Cas9 edited F
0 animals too often demonstrate variable phenotypic penetrance due to the mosaic nature of editing outcomes after double strand break (DSB) repair. Even with high efficiency levels of genome editing, phenotypes may be obscured by proportional presence of in-frame mutations that still produce functional protein. Recently, studies in cell culture systems have shown that the nature of CRISPR/Cas9-mediated mutations can be dependent on local sequence context and can be predicted by computational methods. Here, we demonstrate that similar approaches can be used to forecast CRISPR/Cas9 gene editing outcomes in Xenopus tropicalis, Xenopus laevis, and zebrafish. We show that a publicly available neural network previously trained in mouse embryonic stem cell cultures (InDelphi-mESC) is able to accurately predict CRISPR/Cas9 gene editing outcomes in early vertebrate embryos. Our observations can have direct implications for experiment design, allowing the selection of guide RNAs with predicted repair outcome signatures enriched towards frameshift mutations, allowing maximization of CRISPR/Cas9 phenotype penetrance in the F0 generation.- Published
- 2020
- Full Text
- View/download PDF
31. RBL1 (p107) functions as tumor suppressor in glioblastoma and small-cell pancreatic neuroendocrine carcinoma in Xenopus tropicalis.
- Author
-
Naert T, Dimitrakopoulou D, Tulkens D, Demuynck S, Carron M, Noelanders R, Eeckhout L, Van Isterdael G, Deforce D, Vanhove C, Van Dorpe J, Creytens D, and Vleminckx K
- Subjects
- Animals, Animals, Genetically Modified, CRISPR-Cas Systems genetics, Carcinoma, Neuroendocrine genetics, Carcinoma, Small Cell genetics, Disease Models, Animal, Gene Editing, Glioblastoma genetics, Humans, Pancreatic Neoplasms genetics, Retinoblastoma-Like Protein p107 genetics, Signal Transduction genetics, Xenopus, Xenopus Proteins genetics, Pancreatic Neoplasms, Carcinoma, Neuroendocrine pathology, Carcinoma, Small Cell pathology, Glioblastoma pathology, Pancreatic Neoplasms pathology, Retinoblastoma-Like Protein p107 metabolism, Xenopus Proteins metabolism
- Abstract
Alterations of the retinoblastoma and/or the p53 signaling network are associated with specific cancers such as high-grade astrocytoma/glioblastoma, small-cell lung cancer (SCLC), choroid plexus tumors, and small-cell pancreatic neuroendocrine carcinoma (SC-PaNEC). However, the intricate functional redundancy between RB1 and the related pocket proteins RBL1/p107 and RBL2/p130 in suppressing tumorigenesis remains poorly understood. Here we performed lineage-restricted parallel inactivation of rb1 and rbl1 by multiplex CRISPR/Cas9 genome editing in the true diploid Xenopus tropicalis to gain insight into this in vivo redundancy. We show that while rb1 inactivation is sufficient to induce choroid plexus papilloma, combined rb1 and rbl1 inactivation is required and sufficient to drive SC-PaNEC, retinoblastoma and astrocytoma. Further, using a novel Li-Fraumeni syndrome-mimicking tp53 mutant X. tropicalis line, we demonstrate increased malignancy of rb1/rbl1-mutant glioma towards glioblastoma upon concomitant inactivation of tp53. Interestingly, although clinical SC-PaNEC samples are characterized by abnormal p53 expression or localization, in the current experimental models, the tp53 status had little effect on the establishment and growth of SC-PaNEC, but may rather be essential for maintaining chromosomal stability. SCLC was only rarely observed in our experimental setup, indicating requirement of additional or alternative oncogenic insults. In conclusion, we used CRISPR/Cas9 to delineate the tumor suppressor properties of Rbl1, generating new insights in the functional redundancy within the retinoblastoma protein family in suppressing neuroendocrine pancreatic cancer and glioma/glioblastoma.
- Published
- 2020
- Full Text
- View/download PDF
32. Homozygous Null TBX4 Mutations Lead to Posterior Amelia with Pelvic and Pulmonary Hypoplasia.
- Author
-
Kariminejad A, Szenker-Ravi E, Lekszas C, Tajsharghi H, Moslemi AR, Naert T, Tran HT, Ahangari F, Rajaei M, Nasseri M, Haaf T, Azad A, Superti-Furga A, Maroofian R, Ghaderi-Sohi S, Najmabadi H, Abbaszadegan MR, Vleminckx K, Nikuei P, and Reversade B
- Subjects
- Abnormalities, Multiple pathology, Adolescent, Bone Diseases, Developmental pathology, Child, Ectromelia pathology, Female, Hip pathology, Humans, Ischium pathology, Lung pathology, Lung Diseases pathology, Male, Patella pathology, Pedigree, Pelvis pathology, Prognosis, Abnormalities, Multiple etiology, Bone Diseases, Developmental etiology, Ectromelia etiology, Hip abnormalities, Homozygote, Ischium abnormalities, Loss of Function Mutation, Lung abnormalities, Lung Diseases etiology, Patella abnormalities, Pelvis abnormalities, T-Box Domain Proteins genetics
- Abstract
The development of hindlimbs in tetrapod species relies specifically on the transcription factor TBX4. In humans, heterozygous loss-of-function TBX4 mutations cause dominant small patella syndrome (SPS) due to haploinsufficiency. Here, we characterize a striking clinical entity in four fetuses with complete posterior amelia with pelvis and pulmonary hypoplasia (PAPPA). Through exome sequencing, we find that PAPPA syndrome is caused by homozygous TBX4 inactivating mutations during embryogenesis in humans. In two consanguineous couples, we uncover distinct germline TBX4 coding mutations, p.Tyr113
∗ and p.Tyr127Asn, that segregated with SPS in heterozygous parents and with posterior amelia with pelvis and pulmonary hypoplasia syndrome (PAPPAS) in one available homozygous fetus. A complete absence of TBX4 transcripts in this proband with biallelic p.Tyr113∗ stop-gain mutations revealed nonsense-mediated decay of the endogenous mRNA. CRISPR/Cas9-mediated TBX4 deletion in Xenopus embryos confirmed its restricted role during leg development. We conclude that SPS and PAPPAS are allelic diseases of TBX4 deficiency and that TBX4 is an essential transcription factor for organogenesis of the lungs, pelvis, and hindlimbs in humans., (Copyright © 2019 American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
33. ΔN-Tp63 Mediates Wnt/β-Catenin-Induced Inhibition of Differentiation in Basal Stem Cells of Mucociliary Epithelia.
- Author
-
Haas M, Gómez Vázquez JL, Sun DI, Tran HT, Brislinger M, Tasca A, Shomroni O, Vleminckx K, and Walentek P
- Subjects
- Animals, Cell Differentiation, Humans, Mice, Epithelium metabolism, Transcription Factors genetics, Tumor Suppressor Proteins genetics, Wnt Signaling Pathway immunology, beta Catenin metabolism
- Abstract
Mucociliary epithelia provide a first line of defense against pathogens. Impaired regeneration and remodeling of mucociliary epithelia are associated with dysregulated Wnt/β-catenin signaling in chronic airway diseases, but underlying mechanisms remain elusive, and studies yield seemingly contradicting results. Employing the Xenopus mucociliary epidermis, the mouse airway, and human airway Basal cells, we characterize the evolutionarily conserved roles of Wnt/β-catenin signaling in vertebrates. In multiciliated cells, Wnt is required for cilia formation during differentiation. In Basal cells, Wnt prevents specification of epithelial cell types by activating ΔN-TP63, a master transcription factor, which is necessary and sufficient to mediate the Wnt-induced inhibition of specification and is required to retain Basal cells during development. Chronic Wnt activation leads to remodeling and Basal cell hyperplasia, which are reversible in vivo and in vitro, suggesting Wnt inhibition as a treatment option in chronic lung diseases. Our work provides important insights into mucociliary signaling, development, and disease., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
34. A new transgenic reporter line reveals Wnt-dependent Snai2 re-expression and cranial neural crest differentiation in Xenopus.
- Author
-
Li J, Perfetto M, Materna C, Li R, Thi Tran H, Vleminckx K, Duncan MK, and Wei S
- Subjects
- ADAM Proteins genetics, ADAM Proteins metabolism, Animals, Animals, Genetically Modified, Cell Differentiation drug effects, Cell Differentiation genetics, Embryo, Nonmammalian, Embryonic Development drug effects, Embryonic Development physiology, Gene Expression Regulation, Developmental drug effects, Gene Expression Regulation, Developmental genetics, Gene Knockdown Techniques, Genes, Reporter genetics, Green Fluorescent Proteins chemistry, Green Fluorescent Proteins genetics, Heterocyclic Compounds, 3-Ring pharmacology, Imides pharmacology, Membrane Proteins genetics, Membrane Proteins metabolism, Neural Crest cytology, Quinolines pharmacology, Wnt Proteins antagonists & inhibitors, Wnt Proteins genetics, Wnt Signaling Pathway drug effects, Wnt Signaling Pathway genetics, Wnt Signaling Pathway physiology, Xenopus Proteins genetics, Xenopus laevis genetics, Brain embryology, Neural Crest physiology, Transcription Factors metabolism, Wnt Proteins metabolism, Xenopus Proteins metabolism, Xenopus laevis embryology
- Abstract
During vertebrate embryogenesis, the cranial neural crest (CNC) forms at the neural plate border and subsequently migrates and differentiates into many types of cells. The transcription factor Snai2, which is induced by canonical Wnt signaling to be expressed in the early CNC, is pivotal for CNC induction and migration in Xenopus. However, snai2 expression is silenced during CNC migration, and its roles at later developmental stages remain unclear. We generated a transgenic X. tropicalis line that expresses enhanced green fluorescent protein (eGFP) driven by the snai2 promoter/enhancer, and observed eGFP expression not only in the pre-migratory and migrating CNC, but also the differentiating CNC. This transgenic line can be used directly to detect deficiencies in CNC development at various stages, including subtle perturbation of CNC differentiation. In situ hybridization and immunohistochemistry confirm that Snai2 is re-expressed in the differentiating CNC. Using a separate transgenic Wnt reporter line, we show that canonical Wnt signaling is also active in the differentiating CNC. Blocking Wnt signaling shortly after CNC migration causes reduced snai2 expression and impaired differentiation of CNC-derived head cartilage structures. These results suggest that Wnt signaling is required for snai2 re-expression and CNC differentiation.
- Published
- 2019
- Full Text
- View/download PDF
35. Corrigendum: Xenopus tropicalis : Joining the Armada in the Fight Against Blood Cancer.
- Author
-
Dimitrakopoulou D, Tulkens D, Van Vlierberghe P, and Vleminckx K
- Abstract
[This corrects the article DOI: 10.3389/fphys.2019.00048.].
- Published
- 2019
- Full Text
- View/download PDF
36. Xenopus tropicalis : Joining the Armada in the Fight Against Blood Cancer.
- Author
-
Dimitrakopoulou D, Tulkens D, Van Vlierberghe P, and Vleminckx K
- Abstract
Aquatic vertebrate organisms such as zebrafish have been used for over a decade to model different types of human cancer, including hematologic malignancies. However, the introduction of gene editing techniques such as CRISPR/Cas9 and TALEN, have now opened the road for other organisms featuring large externally developing embryos that are easily accessible. Thanks to its unique diploid genome that shows a high degree of synteny to the human, combined with its relatively short live cycle, Xenopus tropicalis has now emerged as an additional powerful aquatic model for studying human disease genes. Genome editing techniques are very simple and extremely efficient, permitting the fast and cheap generation of genetic models for human disease. Mosaic disruption of tumor suppressor genes allows the generation of highly penetrant and low latency cancer models. While models for solid human tumors have been recently generated, genetic models for hematologic malignancies are currently lacking for Xenopus. Here we describe our experimental pipeline, based on mosaic genome editing by CRISPR/Cas9, to generate innovative and high-performing leukemia models in X. tropicalis . These add to the existing models in zebrafish and will extend the experimental platform available in aquatic vertebrate organisms to contribute to the field of hematologic malignancies. This will extend our knowledge in the etiology of this cancer and assist the identification of molecular targets for therapeutic intervention.
- Published
- 2019
- Full Text
- View/download PDF
37. Author Correction: RSPO2 inhibition of RNF43 and ZNRF3 governs limb development independently of LGR4/5/6.
- Author
-
Szenker-Ravi E, Altunoglu U, Leushacke M, Bosso-Lefèvre C, Khatoo M, Thi Tran H, Naert T, Noelanders R, Hajamohideen A, Beneteau C, de Sousa SB, Karaman B, Latypova X, Başaran S, Yücel EB, Tan TT, Vlaminck L, Nayak SS, Shukla A, Girisha KM, Le Caignec C, Soshnikova N, Uyguner ZO, Vleminckx K, Barker N, Kayserili H, and Reversade B
- Abstract
In this Letter, the surname of author Lena Vlaminck was misspelled 'Vlaeminck'. In addition, author Kris Vleminckx should have been associated with affiliation 16 (Center for Medical Genetics, Ghent University, Ghent, Belgium). These have been corrected online.
- Published
- 2018
- Full Text
- View/download PDF
38. CRISPR/Cas9 disease models in zebrafish and Xenopus: The genetic renaissance of fish and frogs.
- Author
-
Naert T and Vleminckx K
- Subjects
- Animals, Gene Targeting, Genetic Therapy methods, Genomics, Mutation, Xenopus, Zebrafish, CRISPR-Cas Systems, Disease Models, Animal, Gene Editing
- Abstract
The speed by which clinical genomics is currently identifying novel potentially pathogenic variants is outperforming the speed by which these can be functionally (genotype-phenotype) annotated in animal disease models. However, over the past few years the emergence of CRISPR/Cas9 as a straight-forward genome editing technology has revolutionized disease modeling in vertebrate non-mammalian model organisms such as zebrafish, medaka and Xenopus. It is now finally possible, by CRISPR/Cas9, to rapidly establish clinically relevant disease models in these organisms. Interestingly, these can provide both cost-effective genotype-phenotype correlations for gene-(variants) and genomic rearrangements obtained from clinical practice, as well as be exploited to perform translational research to improve prospects of disease afflicted patients. In this review, we show an extensive overview of these new CRISPR/Cas9-mediated disease models and provide future prospects that will allow increasingly accurate modeling of human disease in zebrafish, medaka and Xenopus., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
39. RSPO2 inhibition of RNF43 and ZNRF3 governs limb development independently of LGR4/5/6.
- Author
-
Szenker-Ravi E, Altunoglu U, Leushacke M, Bosso-Lefèvre C, Khatoo M, Thi Tran H, Naert T, Noelanders R, Hajamohideen A, Beneteau C, de Sousa SB, Karaman B, Latypova X, Başaran S, Yücel EB, Tan TT, Vlaminck L, Nayak SS, Shukla A, Girisha KM, Le Caignec C, Soshnikova N, Uyguner ZO, Vleminckx K, Barker N, Kayserili H, and Reversade B
- Subjects
- Animals, DNA-Binding Proteins metabolism, Female, Fibroblasts, Gene Knockout Techniques, HEK293 Cells, Humans, Intercellular Signaling Peptides and Proteins genetics, Male, Mice, Oncogene Proteins antagonists & inhibitors, Oncogene Proteins metabolism, Phenotype, Receptors, G-Protein-Coupled deficiency, Ubiquitin-Protein Ligases metabolism, Xenopus genetics, DNA-Binding Proteins antagonists & inhibitors, Extremities embryology, Intercellular Signaling Peptides and Proteins metabolism, Limb Deformities, Congenital genetics, Receptors, G-Protein-Coupled metabolism, Ubiquitin-Protein Ligases antagonists & inhibitors
- Abstract
The four R-spondin secreted ligands (RSPO1-RSPO4) act via their cognate LGR4, LGR5 and LGR6 receptors to amplify WNT signalling
1-3 . Here we report an allelic series of recessive RSPO2 mutations in humans that cause tetra-amelia syndrome, which is characterized by lung aplasia and a total absence of the four limbs. Functional studies revealed impaired binding to the LGR4/5/6 receptors and the RNF43 and ZNRF3 transmembrane ligases, and reduced WNT potentiation, which correlated with allele severity. Unexpectedly, however, the triple and ubiquitous knockout of Lgr4, Lgr5 and Lgr6 in mice did not recapitulate the known Rspo2 or Rspo3 loss-of-function phenotypes. Moreover, endogenous depletion or addition of exogenous RSPO2 or RSPO3 in triple-knockout Lgr4/5/6 cells could still affect WNT responsiveness. Instead, we found that the concurrent deletion of rnf43 and znrf3 in Xenopus embryos was sufficient to trigger the outgrowth of supernumerary limbs. Our results establish that RSPO2, without the LGR4/5/6 receptors, serves as a direct antagonistic ligand to RNF43 and ZNRF3, which together constitute a master switch that governs limb specification. These findings have direct implications for regenerative medicine and WNT-associated cancers.- Published
- 2018
- Full Text
- View/download PDF
40. An atlas of Wnt activity during embryogenesis in Xenopus tropicalis.
- Author
-
Borday C, Parain K, Thi Tran H, Vleminckx K, Perron M, and Monsoro-Burq AH
- Subjects
- Animals, Gastrula metabolism, In Situ Hybridization, Neural Crest metabolism, Neural Tube metabolism, Wnt Proteins genetics, Xenopus genetics, Xenopus metabolism, Xenopus Proteins genetics, Embryonic Development physiology, Gene Expression Regulation, Developmental physiology, Wnt Proteins metabolism, Wnt Signaling Pathway physiology, Xenopus embryology, Xenopus Proteins metabolism
- Abstract
Wnt proteins form a family of highly conserved secreted molecules that are critical mediators of cell-cell signaling during embryogenesis. Partial data on Wnt activity in different tissues and at different stages have been reported in frog embryos. Our objective here is to provide a coherent and detailed description of Wnt activity throughout embryo development. Using a transgenic Xenopus tropicalis line carrying a Wnt-responsive reporter sequence, we depict the spatial and temporal dynamics of canonical Wnt activity during embryogenesis. We provide a comprehensive series of in situ hybridization in whole-mount embryos and in cross-sections, from gastrula to tadpole stages, with special focus on neural tube, retina and neural crest cell development. This collection of patterns will thus constitute a valuable resource for developmental biologists to picture the dynamics of Wnt activity during development.
- Published
- 2018
- Full Text
- View/download PDF
41. Targeted Genome Engineering in Xenopus Using the Transcription Activator-Like Effector Nuclease (TALEN) Technology.
- Author
-
Van Nieuwenhuysen T and Vleminckx K
- Subjects
- Animals, Base Sequence, Microinjections, RNA, Messenger biosynthesis, Genetic Engineering methods, Genome, Transcription Activator-Like Effector Nucleases metabolism, Xenopus genetics
- Abstract
Targeted genome engineering technologies are revolutionizing the field of functional genomics and have been extensively used in a variety of model organisms, including X. tropicalis and X. laevis. The original methods based on Zn-finger proteins coupled to endonuclease domains were initially replaced by the more efficient and straightforward transcription activator-like effector nucleases (TALENs), adapted from plant pathogenic Xanthomonas species. Although functional genomics are more recently dominated by the even faster and more convenient CRISPR/Cas9 technology, the use of TALENs may still be preferred in a number of cases. We have successfully implemented this technology in Xenopus and in this chapter we describe our working protocol for targeted genome editing in X. tropicalis using TALENs.
- Published
- 2018
- Full Text
- View/download PDF
42. Methods for CRISPR/Cas9 Xenopus tropicalis Tissue-Specific Multiplex Genome Engineering.
- Author
-
Naert T and Vleminckx K
- Subjects
- Animals, CRISPR-Associated Protein 9 metabolism, Gene Editing, Microinjections, Monophenol Monooxygenase metabolism, RNA, Guide, CRISPR-Cas Systems genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Ribonucleoproteins metabolism, Xenopus embryology, CRISPR-Cas Systems genetics, Genetic Engineering methods, Genome, Organ Specificity genetics, Xenopus genetics
- Abstract
In this chapter, we convey a state-of-the art update to the 2014 Nakayama protocol for CRISPR/Cas9 genome engineering in Xenopus tropicalis (X. tropicalis). We discuss in depth, gRNA design software and rules, gRNA synthesis, and procedures for tissue- and tissue-specific CRISPR/Cas9 genome editing by targeted microinjection in X. tropicalis embryos. We demonstrate the methodology by which any standard equipped Xenopus researcher with microinjection experience can generate F0 CRISPR/Cas9 mediated mosaic mutants (crispants) within one to two work-week(s). The described methodology allows CRISPR/Cas9 efficiencies to be high enough to read out phenotypic consequences, and thus perform gene function analysis, in the F0 crispant. Additionally, we provide the framework for performing multiplex tissue-specific CRISPR/Cas9 experiments generating crispants mosaic mutant in up to four genes simultaneously, which can be of importance for Laevis researchers aiming to target by CRISPR/Cas9 both the S and L homeolog of a gene simultaneously. Finally, we discuss off-target concerns, how to minimize these and ways to rapidly bypass reviewer off-target critique by exploiting the advantages of X. tropicalis.
- Published
- 2018
- Full Text
- View/download PDF
43. Cancer Models in Xenopus tropicalis by CRISPR/Cas9 Mediated Knockout of Tumor Suppressors.
- Author
-
Naert T and Vleminckx K
- Subjects
- Animals, Cell Proliferation, Disease Models, Animal, Gene Editing, Laser Capture Microdissection, Mutation genetics, Neoplasms pathology, Phenotype, Proliferating Cell Nuclear Antigen metabolism, RNA, Guide, CRISPR-Cas Systems metabolism, Xenopus genetics, CRISPR-Associated Protein 9 metabolism, CRISPR-Cas Systems genetics, Gene Knockout Techniques methods, Genes, Tumor Suppressor, Neoplasms genetics
- Abstract
The recent advent of CRISPR/Cas9 as a straightforward genome editing tool has allowed the establishment of the first bona fide genetic cancer models within the diploid aquatic model organism Xenopus tropicalis (X. tropicalis). Within this chapter, we demonstrate the methods for targeting tumor suppressors with the CRISPR/Cas9 system in the developing X. tropicalis embryo. We further illustrate genotyping and phenotyping of the resulting tumor-bearing F0 mosaic mutant animals (crispants). We focus in detail on the histopathological analysis of cancer neoplasms, the methodology to illustrate high proliferative index by proliferation marker immunofluorescence and how to isolate specific (tumor) cell populations by laser capture microdissection. As such, the described pipeline allows for rapid establishment of novel cancer models by CRISPR/Cas9 targeting of established tumor suppressor genes, or novel candidates obtained from clinical data. In conclusion, we thus provide the methodology for modeling human cancer with the highly efficient CRISPR/Cas9 system in F0 X. tropicalis.
- Published
- 2018
- Full Text
- View/download PDF
44. Cell Cycle Analysis of the Embryonic Brain of Fluorescent Reporter Xenopus tropicalis by Flow Cytometry.
- Author
-
Noelanders R and Vleminckx K
- Subjects
- Animals, DNA metabolism, Dissection, Fluorescence, Permeability, Staining and Labeling, Tissue Fixation, Brain embryology, Cell Cycle, Embryo, Nonmammalian cytology, Flow Cytometry methods, Genes, Reporter, Xenopus embryology
- Abstract
Many developmental signals are associated with changes in proliferative response. Also, growing organs and tissues can contain different cellular subpopulations with a defined status in the cell cycle, e.g., quiescent in stem cells, high proliferation in progenitors, cell cycle exit in differentiating cells. This chapter describes a method for isolation of individual cell populations from the Xenopus tadpole brain and determination of their cell cycle status using flow cytometry.
- Published
- 2018
- Full Text
- View/download PDF
45. Genotyping of CRISPR/Cas9 Genome Edited Xenopus tropicalis.
- Author
-
Naert T and Vleminckx K
- Subjects
- Animals, Breeding, Embryo, Nonmammalian metabolism, Homozygote, Mutation genetics, Nucleic Acid Denaturation, CRISPR-Associated Protein 9 metabolism, CRISPR-Cas Systems genetics, Gene Editing, Genotyping Techniques methods, Xenopus genetics
- Abstract
The targeted nuclease revolution (ZFN, TALEN, and CRISPR/Cas9) has led to a myriad of reports describing genotyping methodologies for genome edited founders (F0-crispants) and their offspring (F1). As such, choosing a specific genotyping methodology for your Xenopus CRISPR/Cas9 experiments can be challenging. In this chapter we will discuss, with emphasis on Xenopus tropicalis (X. tropicalis), different methods for assessing genome editing efficiencies within F0 CRISPR/Cas9 founders and for identification of their hetero-, compound hetero-, and homozygous mutant F1 offspring. For F0 crispants, we will provide the protocols and the respective (dis)advantages of genotyping with heteroduplex mobility assay (HMA), subclone Sanger sequencing, and sequence trace decomposition. Furthermore, we provide a previously unpublished pipe-line for rapid genotyping of F1 offspring-high resolution melting analysis (HRMA) and sequence trace decomposition-procured from breeding with F0 crispants. As such, we report here the current state-of-the-art cost- and time-effective approaches to perform genotyping of CRISPR/Cas9 experiments for the Xenopus tropicalis researcher.
- Published
- 2018
- Full Text
- View/download PDF
46. CRISPR/Cas9-Mediated Knockout of Rb1 in Xenopus tropicalis.
- Author
-
Naert T and Vleminckx K
- Subjects
- Animals, Retinoblastoma Protein genetics, Xenopus classification, CRISPR-Cas Systems, Gene Knockout Techniques methods, Genome, Retinoblastoma Protein antagonists & inhibitors, Xenopus genetics
- Abstract
At this time, no molecular targeted therapies exist for treatment of retinoblastoma. This can be, in part, attributed to the lack of animal models that allow for both rapid identification of novel therapeutic targets and hypothesis driven drug testing. Within this scope, we have recently reported the first genuine genetic nonmammalian retinoblastoma cancer model within the aquatic model organism Xenopus tropicalis (Naert et al., Sci Rep 6: 35263, 2016). Here we describe the methods to generate rb1 mosaic mutant Xenopus tropicalis by employing the CRISPR/Cas9 technology. In depth, we discuss short guide RNA (sgRNA) design parameters, generation, quality control, quantification, and delivery followed by several methods for assessing genome editing efficiencies. As such the reader should be capable, by minor changes to the methods described here, to (co-) target rb1 or any one or multiple gene(s) within the Xenopus tropicalis genome by multiplex CRISPR/Cas9 methodology.
- Published
- 2018
- Full Text
- View/download PDF
47. Caspase-9 has a nonapoptotic function in Xenopus embryonic primitive blood formation.
- Author
-
Tran HT, Fransen M, Dimitrakopoulou D, Van Imschoot G, Willemarck N, and Vleminckx K
- Subjects
- Animals, Apoptosis physiology, Cell Death physiology, Cell Differentiation physiology, Genes, Reporter, Green Fluorescent Proteins biosynthesis, Green Fluorescent Proteins genetics, HEK293 Cells, Humans, Signal Transduction, Transfection, Xenopus laevis embryology, Caspase 9 metabolism, Xenopus laevis blood
- Abstract
Caspases constitute a family of cysteine proteases centrally involved in programmed cell death, which is an integral part of normal embryonic and fetal development. However, it has become clear that specific caspases also have functions independent of cell death. In order to identify novel apoptotic and nonapoptotic developmental caspase functions, we designed and transgenically integrated novel fluorescent caspase reporter constructs in developing Xenopus embryos and tadpoles. This model organism has an external development, allowing direct and continuous monitoring. These studies uncovered a nonapoptotic role for the initiator caspase-9 in primitive blood formation. Functional experiments further corroborated that caspase-9, but possibly not the executioners caspase-3 and caspase-7, are required for primitive erythropoiesis in the early embryo. These data reveal a novel nonapoptotic function for the initiator caspase-9 and, for the first time, implicate nonapoptotic caspase activity in primitive blood formation., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2017. Published by The Company of Biologists Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
48. How Wnt Signaling Builds the Brain: Bridging Development and Disease.
- Author
-
Noelanders R and Vleminckx K
- Subjects
- Animals, Humans, Brain growth & development, Brain metabolism, Nervous System Diseases metabolism, Wnt Signaling Pathway
- Abstract
Wnt/β-catenin signaling plays a crucial role throughout all stages of brain development and remains important in the adult brain. Accordingly, many neurological disorders have been linked to Wnt signaling. Defects in Wnt signaling during neural development can give rise to birth defects or lead to neurological dysfunction later in life. Developmental signaling events can also be hijacked in the adult and result in disease. Moreover, knowledge about the physiological role of Wnt signaling in the brain might lead to new therapeutic strategies for neurological diseases. Especially, the important role for Wnt signaling in neural differentiation of pluripotent stem cells has received much attention as this might provide a cure for neurodegenerative disorders. In this review, we summarize the versatile role of Wnt/β-catenin signaling during neural development and discuss some recent studies linking Wnt signaling to neurological disorders.
- Published
- 2017
- Full Text
- View/download PDF
49. TALENs and CRISPR/Cas9 fuel genetically engineered clinically relevant Xenopus tropicalis tumor models.
- Author
-
Naert T, Van Nieuwenhuysen T, and Vleminckx K
- Subjects
- Animals, Disease Models, Animal, Gene Targeting, Humans, Neoplasms pathology, Xenopus genetics, CRISPR-Cas Systems genetics, Genetic Engineering, Neoplasms genetics, Transcription Activator-Like Effector Nucleases genetics
- Abstract
The targeted nuclease revolution (TALENs, CRISPR/Cas9) now allows Xenopus researchers to rapidly generate custom on-demand genetic knockout models. These novel methods to perform reverse genetics are unprecedented and are fueling a wide array of human disease models within the aquatic diploid model organism Xenopus tropicalis (X. tropicalis). This emerging technology review focuses on the tools to rapidly generate genetically engineered X. tropicalis models (GEXM), with a focus on establishment of genuine genetic and clinically relevant cancer models. We believe that due to particular advantageous characteristics, outlined within this review, GEXM will become a valuable alternative animal model for modeling human cancer. Furthermore, we provide perspectives of how GEXM will be used as a platform for elucidation of novel therapeutic targets and for preclinical drug validation. Finally, we also discuss some future prospects on how the recent expansions and adaptations of the CRISPR/Cas9 toolbox might influence and push forward X. tropicalis cancer research., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
50. CRISPR/Cas9 mediated knockout of rb1 and rbl1 leads to rapid and penetrant retinoblastoma development in Xenopus tropicalis.
- Author
-
Naert T, Colpaert R, Van Nieuwenhuysen T, Dimitrakopoulou D, Leoen J, Haustraete J, Boel A, Steyaert W, Lepez T, Deforce D, Willaert A, Creytens D, and Vleminckx K
- Subjects
- Animals, Disease Models, Animal, Eye Neoplasms genetics, Eye Neoplasms pathology, Gene Knockout Techniques methods, Retinoblastoma pathology, Retinoblastoma Protein genetics, CRISPR-Cas Systems genetics, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Retinoblastoma genetics, Retinoblastoma Binding Proteins genetics, Retinoblastoma-Like Protein p107 genetics, Xenopus genetics
- Abstract
Retinoblastoma is a pediatric eye tumor in which bi-allelic inactivation of the Retinoblastoma 1 (RB1) gene is the initiating genetic lesion. Although recently curative rates of retinoblastoma have increased, there are at this time no molecular targeted therapies available. This is, in part, due to the lack of highly penetrant and rapid retinoblastoma animal models that facilitate rapid identification of targets that allow therapeutic intervention. Different mouse models are available, all based on genetic deactivation of both Rb1 and Retinoblastoma-like 1 (Rbl1), and each showing different kinetics of retinoblastoma development. Here, we show by CRISPR/Cas9 techniques that similar to the mouse, neither rb1 nor rbl1 single mosaic mutant Xenopus tropicalis develop tumors, whereas rb1/rbl1 double mosaic mutant tadpoles rapidly develop retinoblastoma. Moreover, occasionally presence of pinealoblastoma (trilateral retinoblastoma) was detected. We thus present the first CRISPR/Cas9 mediated cancer model in Xenopus tropicalis and the first genuine genetic non-mammalian retinoblastoma model. The rapid kinetics of our model paves the way for use as a pre-clinical model. Additionally, this retinoblastoma model provides unique possibilities for fast elucidation of novel drug targets by triple multiplex CRISPR/Cas9 gRNA injections (rb1 + rbl1 + modifier gene) in order to address the clinically unmet need of targeted retinoblastoma therapy.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.