Susanna Raitano
Katholieke Universiteit Leuven
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Susanna Raitano.
Stem cell reports | 2015
Susanna Raitano; Laura Ordovas; Louis De Muynck; Wenting Guo; Ira Espuny-Camacho; Martine Geraerts; Satish Khurana; Kim Vanuytsel; Balázs István Tóth; Thomas Voets; Rik Vandenberghe; Toni Cathomen; Ludo Van Den Bosch; Pierre Vanderhaeghen; Philip Van Damme; Catherine M. Verfaillie
Summary To understand how haploinsufficiency of progranulin (PGRN) causes frontotemporal dementia (FTD), we created induced pluripotent stem cells (iPSCs) from patients carrying the GRNIVS1+5G > C mutation (FTD-iPSCs). FTD-iPSCs were fated to cortical neurons, the cells most affected in FTD. Although generation of neuroprogenitors was unaffected, their further differentiation into CTIP2-, FOXP2-, or TBR1-TUJ1 double-positive cortical neurons, but not motorneurons, was significantly decreased in FTD-neural progeny. Zinc finger nuclease-mediated introduction of GRN cDNA into the AAVS1 locus corrected defects in cortical neurogenesis, demonstrating that PGRN haploinsufficiency causes inefficient cortical neuron generation. RNA sequencing analysis confirmed reversal of the altered gene expression profile following genetic correction. We identified the Wnt signaling pathway as one of the top defective pathways in FTD-iPSC-derived neurons, which was reversed following genetic correction. Differentiation of FTD-iPSCs in the presence of a WNT inhibitor mitigated defective corticogenesis. Therefore, we demonstrate that PGRN haploinsufficiency hampers corticogenesis in vitro.
Molecular Psychiatry | 2016
S Nageshappa; Cassiano Carromeu; Cleber A. Trujillo; Pinar Mesci; Ira Espuny-Camacho; E Pasciuto; Pierre Vanderhaeghen; Catherine M. Verfaillie; Susanna Raitano; Anujith Kumar; C M B Carvalho; Claudia Bagni; Mb Ramocki; B H S Araujo; L B Torres; James R. Lupski; H. Van Esch; Alysson R. Muotri
Increased dosage of methyl-CpG-binding protein-2 (MeCP2) results in a dramatic neurodevelopmental phenotype with onset at birth. We generated induced pluripotent stem cells (iPSCs) from patients with the MECP2 duplication syndrome (MECP2dup), carrying different duplication sizes, to study the impact of increased MeCP2 dosage in human neurons. We show that cortical neurons derived from these different MECP2dup iPSC lines have increased synaptogenesis and dendritic complexity. In addition, using multi-electrodes arrays, we show that neuronal network synchronization was altered in MECP2dup-derived neurons. Given MeCP2 functions at the epigenetic level, we tested whether these alterations were reversible using a library of compounds with defined activity on epigenetic pathways. One histone deacetylase inhibitor, NCH-51, was validated as a potential clinical candidate. Interestingly, this compound has never been considered before as a therapeutic alternative for neurological disorders. Our model recapitulates early stages of the human MECP2 duplication syndrome and represents a promising cellular tool to facilitate therapeutic drug screening for severe neurodevelopmental disorders.
Stem cell reports | 2015
Laura Ordovas; Ruben Boon; Mariaelena Pistoni; Yemiao Chen; Esther Wolfs; Wenting Guo; Rangarajan Sambathkumar; Sylwia Bobis-Wozowicz; Nicky Helsen; Jolien Vanhove; Pieter Berckmans; Qing Cai; Kim Vanuytsel; Kristel Eggermont; Veerle Vanslembrouck; Béla Z. Schmidt; Susanna Raitano; Ludo Van Den Bosch; Yaakov Nahmias; Toni Cathomen; Tom Struys; Catherine M. Verfaillie
Summary Tools for rapid and efficient transgenesis in “safe harbor” loci in an isogenic context remain important to exploit the possibilities of human pluripotent stem cells (hPSCs). We created hPSC master cell lines suitable for FLPe recombinase-mediated cassette exchange (RMCE) in the AAVS1 locus that allow generation of transgenic lines within 15 days with 100% efficiency and without random integrations. Using RMCE, we successfully incorporated several transgenes useful for lineage identification, cell toxicity studies, and gene overexpression to study the hepatocyte lineage. However, we observed unexpected and variable transgene expression inhibition in vitro, due to DNA methylation and other unknown mechanisms, both in undifferentiated hESC and differentiating hepatocytes. Therefore, the AAVS1 locus cannot be considered a universally safe harbor locus for reliable transgene expression in vitro, and using it for transgenesis in hPSC will require careful assessment of the function of individual transgenes.
Stem Cells Translational Medicine | 2014
Qing Cai; Paola Bonfanti; Rangarajan Sambathkumar; Kim Vanuytsel; Jolien Vanhove; Conny Gysemans; Maria Debiec-Rychter; Susanna Raitano; Harry Heimberg; Laura Ordovas; Catherine M. Verfaillie
Pancreatic endocrine progenitors obtained from human embryonic stem cells (hESCs) represent a promising source to develop cell‐based therapies for diabetes. Although endocrine pancreas progenitor cells have been isolated from mouse pancreata on the basis of Ngn3 expression, human endocrine progenitors have not been isolated yet. As substantial differences exist between human and murine pancreas biology, we investigated whether it is possible to isolate pancreatic endocrine progenitors from differentiating hESC cultures by lineage tracing of NGN3. We targeted the 3′ end of NGN3 using zinc finger nuclease‐mediated homologous recombination to allow selection of NGN3eGFP+ cells without disrupting the coding sequence of the gene. Isolated NGN3eGFP+ cells express PDX1, NKX6.1, and chromogranin A and differentiate in vivo toward insulin, glucagon, and somatostatin single hormone‐expressing cells but not to ductal or exocrine pancreatic cells or other endodermal, mesodermal, or ectodermal lineages. This confirms that NGN3+ cells represent pancreatic endocrine progenitors in humans. In addition, this hESC reporter line constitutes a unique tool that may aid in gaining insight into the developmental mechanisms underlying fate choices in human pancreas and in developing cell‐based therapies.
Frontiers in Physiology | 2013
Susanna Raitano; Catherine M. Verfaillie; Anna Petryk
In 2011, Remboutsika et al. (2011) published an elegant paper in which they demonstrated the unique importance of Sox2 in self-renewal of neural stem cells (NSC) in vitro and in vivo. They demonstrated that Sox2 helps maintain the cortical identity of NSC ex vivo as evidenced by the expression of Pax6 and the nestin-linked epitope RC2. Only the Sox2+ cells isolated from Soxβ−geo/+ neurospheres were capable of generating secondary neurospheres while the Sox2− fraction could not, confirming that only the Sox2+ cells can self-renew in vitro. Sox2+ neurospheres differentiated towards Tuj1+ cells with long axons like cortical neurons, while wild type and Soxβ−geo/+ neurospheres derived-neurons developed short axons, showing that these cells had distinct developmental and differentiation potential. When transplanted into mouse and chick embryos, wild type and Soxβ−geo/+ cells generated neural crest cells while Sox2+ cells did not. Moreover, Sox2 overexpression in the neuroepithelium of chick embryo prevented neuroepithelial delamination and migration and restricted the contribution of neuroepithelium to the neural tube only, suggesting that Sox2 inhibits neural crest cell generation by blocking NSC differentiation.
Journal of Visualized Experiments | 2016
Laura Ordovas; Ruben Boon; Mariaelena Pistoni; Yemiao Chen; Rangarajan Sambathkumar; Nicky Helsen; Jolien Vanhove; Pieter Berckmans; Qing Cai; Kim Vanuytsel; Susanna Raitano; Catherine M. Verfaillie
Even with the revolution of gene-targeting technologies led by CRISPR-Cas9, genetic modification of human pluripotent stem cells (hPSCs) is still time consuming. Comparative studies that use recombinant lines with transgenes integrated into safe harbor loci could benefit from approaches that use site-specific targeted recombinases, like Cre or FLPe, which are more rapid and less prone to off-target effects. Such methods have been described, although they do not significantly outperform gene targeting in most aspects. Using Zinc-finger nucleases, we previously created a master cell line in the AAVS1 locus of hPSCs that contains a GFP-Hygromycin-tk expressing cassette, flanked by heterotypic FRT sequences. Here, we describe the procedures to perform FLPe recombinase-mediated cassette exchange (RMCE) using this line. The master cell line is transfected with a RMCE donor vector, which contains a promoterless Puromycin resistance, and with FLPe recombinase. Application of both a positive (Puromycin) and negative (FIAU) selection program leads to the selection of RMCE without random integrations. RMCE generates fully characterized pluripotent polyclonal transgenic lines in 15 d with 100% efficiency. Despite the recently described limitations of the AAVS1 locus, the ease of the system paves the way for hPSC transgenesis in isogenic settings, is necessary for comparative studies, and enables semi-high-throughput genetic screens for gain/loss of function analysis that would otherwise be highly time consuming.
The Journal of Nuclear Medicine | 2017
Esther Wolfs; Bryan Holvoet; Laura Ordovas; Natacha Breuls; Nicky Helsen; Matthias Schönberger; Susanna Raitano; Tom Struys; B Vanbilloen; Cindy Casteels; Maurilio Sampaolesi; Koen Van Laere; Ivo Lambrichts; Catherine M. Verfaillie; Christophe Deroose
Molecular imaging is indispensable for determining the fate and persistence of engrafted stem cells. Standard strategies for transgene induction involve the use of viral vectors prone to silencing and insertional mutagenesis or the use of nonhuman genes. Methods: We used zinc finger nucleases to induce stable expression of human imaging reporter genes into the safe-harbor locus adeno-associated virus integration site 1 in human embryonic stem cells. Plasmids were generated carrying reporter genes for fluorescence, bioluminescence imaging, and human PET reporter genes. Results: In vitro assays confirmed their functionality, and embryonic stem cells retained differentiation capacity. Teratoma formation assays were performed, and tumors were imaged over time with PET and bioluminescence imaging. Conclusion: This study demonstrates the application of genome editing for targeted integration of human imaging reporter genes in human embryonic stem cells for long-term molecular imaging.
Neuron | 2018
Jorge S. Valadas; Giovanni Esposito; Dirk Vandekerkhove; Katarzyna Miskiewicz; Liesbeth Deaulmerie; Susanna Raitano; Philip Seibler; Christine Klein; Patrik Verstreken
Online abstracts | 2017
Cedric Thues; Mala Isrie; Susanna Raitano; Julia Manetsberger; Nils Schoovaerts; Patrik Verstreken; Hilde Van Esch
Archive | 2013
Catherine M. Verfaillie; Susanna Raitano; Elke Stappers; Annelies Crabbe