41 results on '"de Vrij, Femke M S"'
Search Results
2. Differential susceptibility of human motor neurons to infection with Usutu and West Nile virus
- Author
-
Marshall, Eleanor M., Bauer, Lisa, Nelemans, Tessa, Sooksawasdi Na Ayudhya, Syriam, Benavides, Feline, Lanko, Kristina, de Vrij, Femke M. S., Kushner, Steven A., Koopmans, Marion, van Riel, Debby, and Rockx, Barry
- Published
- 2024
- Full Text
- View/download PDF
3. In vitro and in vivo differences in neurovirulence between D614G, Delta And Omicron BA.1 SARS-CoV-2 variants
- Author
-
Bauer, Lisa, Rissmann, Melanie, Benavides, Feline F. W., Leijten, Lonneke, van Run, Peter, Begeman, Lineke, Veldhuis Kroeze, Edwin J. B., Lendemeijer, Bas, Smeenk, Hilde, de Vrij, Femke M. S., Kushner, Steven A., Koopmans, Marion P. G., Rockx, Barry, and van Riel, Debby
- Published
- 2022
- Full Text
- View/download PDF
4. Human Pluripotent Stem Cell-Derived Astrocyte Functionality Compares Favorably with Primary Rat Astrocytes.
- Author
-
Lendemeijer, Bas, Unkel, Maurits, Smeenk, Hilde, Mossink, Britt, Hijazi, Sara, Gordillo-Sampedro, Sara, Shpak, Guy, Slump, Denise E., van den Hout, Mirjam C. G. N., van IJcken, Wilfred F. J., Bindels, Eric M. J., Hoogendijk, Witte J. G., Kasri, Nael Nadif, de Vrij, Femke M. S., and Kushner, Steven A.
- Published
- 2024
- Full Text
- View/download PDF
5. Second-tier trio exome sequencing after negative solo clinical exome sequencing: an efficient strategy to increase diagnostic yield and decipher molecular bases in undiagnosed developmental disorders
- Author
-
Tran Mau-Them, Frederic, Moutton, Sebastien, Racine, Caroline, Vitobello, Antonio, Bruel, Ange-Line, Nambot, Sophie, Kushner, Steven A., de Vrij, Femke M. S., Lehalle, Daphné, Jean-Marçais, Nolwenn, Lecoquierre, François, Delanne, Julian, Thevenon, Julien, Poe, Charlotte, Jouan, Thibaut, Chevarin, Martin, Geneviève, David, Willems, Marjolaine, Coubes, Christine, Houcinat, Nada, Masurel-Paulet, Alice, Mosca-Boidron, Anne-Laure, Tisserant, Emilie, Callier, Patrick, Sorlin, Arthur, Duffourd, Yannis, Faivre, Laurence, Philippe, Christophe, and Thauvin-Robinet, Christel
- Published
- 2020
- Full Text
- View/download PDF
6. Loss of nuclear UBE3A causes electrophysiological and behavioral deficits in mice and is associated with Angelman syndrome
- Author
-
Avagliano Trezza, Rossella, Sonzogni, Monica, Bossuyt, Stijn N. V., Zampeta, F. Isabella, Punt, A. Mattijs, van den Berg, Marlene, Rotaru, Diana C., Koene, Linda M. C., Munshi, Shashini T., Stedehouder, Jeffrey, Kros, Johan M., Williams, Mark, Heussler, Helen, de Vrij, Femke M. S., Mientjes, Edwin J., van Woerden, Geeske M., Kushner, Steven A., Distel, Ben, and Elgersma, Ype
- Published
- 2019
- Full Text
- View/download PDF
7. Activity-based protein profiling reveals off-target proteins of the FAAH inhibitor BIA 10-2474
- Author
-
van Esbroeck, Annelot C. M., Janssen, Antonius P. A., Cognetta, Armand B., Ogasawara, Daisuke, Shpak, Guy, van der Kroeg, Mark, Kantae, Vasudev, Baggelaar, Marc P., de Vrij, Femke M. S., Deng, Hui, Allarà, Marco, Fezza, Filomena, Lin, Zhanmin, van der Wel, Tom, Soethoudt, Marjolein, Mock, Elliot D., den Dulk, Hans, Baak, Ilse L., Florea, Bogdan I., Hendriks, Giel, De Petrocellis, Luciano, Overkleeft, Herman S., Hankemeier, Thomas, De Zeeuw, Chris I., Di Marzo, Vincenzo, Maccarrone, Mauro, Cravatt, Benjamin F., Kushner, Steven A., and van der Stelt, Mario
- Published
- 2017
8. Hepatitis E Virus Infects Neurons and Brains
- Author
-
Zhou, Xinying, Huang, Fen, Xu, Lei, Lin, Zhanmin, de Vrij, Femke M. S., Ayo-Martin, Ane C., van der Kroeg, Mark, Zhao, Manzhi, Yin, Yuebang, Wang, Wenshi, Cao, Wanlu, Wang, Yijin, Kushner, Steven A., Peron, Jean Marie, Alric, Laurent, de Man, Robert A., Jacobs, Bart C., van Eijk, Jeroen J., Aronica, Eleonora M. A., Sprengers, Dave, Metselaar, Herold J., de Zeeuw, Chris I., Dalton, Harry R., Kamar, Nassim, Peppelenbosch, Maikel P., and Pan, Qiuwei
- Published
- 2017
9. A simplified protocol for the generation of cortical brain organoids
- Author
-
Eigenhuis, Kristel N., primary, Somsen, Hedda B., additional, van der Kroeg, Mark, additional, Smeenk, Hilde, additional, Korporaal, Anne L., additional, Kushner, Steven A., additional, de Vrij, Femke M. S., additional, and van den Berg, Debbie L. C., additional
- Published
- 2023
- Full Text
- View/download PDF
10. A simplified protocol for the generation of cortical brain organoids
- Author
-
Eigenhuis, Kristel N, Somsen, Hedda B, van der Kroeg, Mark, Smeenk, Hilde, Korporaal, Anne L, Kushner, Steven A, de Vrij, Femke M S, van den Berg, Debbie L C, Eigenhuis, Kristel N, Somsen, Hedda B, van der Kroeg, Mark, Smeenk, Hilde, Korporaal, Anne L, Kushner, Steven A, de Vrij, Femke M S, and van den Berg, Debbie L C
- Abstract
Human brain organoid technology has the potential to generate unprecedented insight into normal and aberrant brain development. It opens up a developmental time window in which the effects of gene or environmental perturbations can be experimentally tested. However, detection sensitivity and correct interpretation of phenotypes are hampered by notable batch-to-batch variability and low reproducibility of cell and regional identities. Here, we describe a detailed, simplified protocol for the robust and reproducible generation of brain organoids with cortical identity from feeder-independent induced pluripotent stem cells (iPSCs). This self-patterning approach minimizes media supplements and handling steps, resulting in cortical brain organoids that can be maintained over prolonged periods and that contain radial glial and intermediate progenitors, deep and upper layer neurons, and astrocytes.
- Published
- 2023
11. Recommendations, guidelines, and best practice for the use of human induced pluripotent stem cells for neuropharmacological studies of neuropsychiatric disorders
- Author
-
Dutan Polit, Lucia, Eidhof, Ilse; https://orcid.org/0000-0002-1567-3078, McNeill, Rhiannon V; https://orcid.org/0000-0002-3297-9212, Warre-Cornish, Katherine M; https://orcid.org/0000-0001-7892-4295, Yde Ohki, Cristine Marie, Walter, Natalie Monet, Sala, Carlo; https://orcid.org/0000-0003-0662-9523, Verpelli, Chiara, Radtke, Franziska, Galderisi, Silvana, Mucci, Armida; https://orcid.org/0000-0002-6195-9456, Collo, Ginetta, Edenhofer, Frank, Castrén, Maija L, Réthelyi, János M, Ejlersen, Morten, Hohmann, Sonja Simone, Ilieva, Mirolyuba S; https://orcid.org/0000-0002-7992-8885, Lukjanska, Renate; https://orcid.org/0000-0002-4981-7789, Matuleviciute, Rugile; https://orcid.org/0000-0001-5618-5700, Michel, Tanja Maria, de Vrij, Femke M S, Kushner, Steven A; https://orcid.org/0000-0002-9777-3338, Lendemeijer, Bas, Kittel-Schneider, Sarah, Ziegler, Georg C, Gruber-Schoffnegger, Doris, Pasterkamp, R Jeroen; https://orcid.org/0000-0003-1631-6440, Kasri, Amal, Potier, Marie-Claude, et al, Grünblatt, Edna; https://orcid.org/0000-0001-8505-7265, Dutan Polit, Lucia, Eidhof, Ilse; https://orcid.org/0000-0002-1567-3078, McNeill, Rhiannon V; https://orcid.org/0000-0002-3297-9212, Warre-Cornish, Katherine M; https://orcid.org/0000-0001-7892-4295, Yde Ohki, Cristine Marie, Walter, Natalie Monet, Sala, Carlo; https://orcid.org/0000-0003-0662-9523, Verpelli, Chiara, Radtke, Franziska, Galderisi, Silvana, Mucci, Armida; https://orcid.org/0000-0002-6195-9456, Collo, Ginetta, Edenhofer, Frank, Castrén, Maija L, Réthelyi, János M, Ejlersen, Morten, Hohmann, Sonja Simone, Ilieva, Mirolyuba S; https://orcid.org/0000-0002-7992-8885, Lukjanska, Renate; https://orcid.org/0000-0002-4981-7789, Matuleviciute, Rugile; https://orcid.org/0000-0001-5618-5700, Michel, Tanja Maria, de Vrij, Femke M S, Kushner, Steven A; https://orcid.org/0000-0002-9777-3338, Lendemeijer, Bas, Kittel-Schneider, Sarah, Ziegler, Georg C, Gruber-Schoffnegger, Doris, Pasterkamp, R Jeroen; https://orcid.org/0000-0003-1631-6440, Kasri, Amal, Potier, Marie-Claude, et al, and Grünblatt, Edna; https://orcid.org/0000-0001-8505-7265
- Abstract
The number of individuals suffering from neuropsychiatric disorders (NPDs) has increased worldwide, with 3 million disability-adjusted life-years calculated in 2019. Though research using various approaches including genetics, imaging, clinical and animal models has advanced our knowledge regarding NPDs, we still lack basic knowledge regarding the underlying pathophysiological mechanisms. Moreover, there is an urgent need for highly effective therapeutics for NPDs. Human induced pluripotent stem cells (hiPSCs) generated from somatic cells enabled scientists to create brain cells in a patient-specific manner. However, there are challenges to the use of hiPSCs that need to be addressed. In the current paper, consideration of best practices for neuropharmacological and neuropsychiatric research using hiPSCs will be discussed. Specifically, we provide recommendations for best practice in patient recruitment, including collecting demographic, clinical, medical (before and after treatment and response), diagnostic (including scales) and genetic data from the donors. We highlight considerations regarding donor genetics and sex, in addition to discussing biological and technical replicates. Furthermore, we present our views on selecting control groups/lines, experimental designs, and considerations for conducting neuropharmacological studies using hiPSC-based models in the context of NPDs. In doing so, we explore key issues in the field concerning reproducibility, statistical analysis, and how to translate in vitro studies into clinically relevant observations. The aim of this article is to provide a key resource for hiPSC researchers to perform robust and reproducible neuropharmacological studies, with the ultimate aim of improving identification and clinical translation of novel therapeutic drugs for NPDs.
- Published
- 2023
12. Additional file 1 of In vitro and in vivo differences in neurovirulence between D614G, Delta And Omicron BA.1 SARS-CoV-2 variants
- Author
-
Bauer, Lisa, Rissmann, Melanie, Benavides, Feline F. W., Leijten, Lonneke, van Run, Peter, Begeman, Lineke, Veldhuis Kroeze, Edwin J. B., Lendemeijer, Bas, Smeenk, Hilde, de Vrij, Femke M. S., Kushner, Steven A., Koopmans, Marion P. G., Rockx, Barry, and van Riel, Debby
- Abstract
Additional file 1. Figure S1-S6 and Table S1-S2.
- Published
- 2022
- Full Text
- View/download PDF
13. Replication Kinetics, Cell Tropism, and Associated Immune Responses in SARS-CoV-2- and H5N1 Virus-Infected Human Induced Pluripotent Stem Cell-Derived Neural Models
- Author
-
Bauer, Lisa, primary, Lendemeijer, Bas, additional, Leijten, Lonneke, additional, Embregts, Carmen W. E., additional, Rockx, Barry, additional, Kushner, Steven A., additional, de Vrij, Femke M. S., additional, and van Riel, Debby, additional
- Published
- 2021
- Full Text
- View/download PDF
14. Conserved UBE3A subcellular distribution between human and mice is facilitated by non-homologous isoforms
- Author
-
Zampeta, F Isabella, primary, Sonzogni, Monica, additional, Niggl, Eva, additional, Lendemeijer, Bas, additional, Smeenk, Hilde, additional, de Vrij, Femke M S, additional, Kushner, Steven A, additional, Distel, Ben, additional, and Elgersma, Ype, additional
- Published
- 2020
- Full Text
- View/download PDF
15. Activity-based protein profiling reveals off-target proteins of the FAAH inhibitor BIA 10-2474
- Author
-
van Esbroeck, Annelot C M, Janssen, Antonius P A, Cognetta, Armand B, Ogasawara, Daisuke, Shpak, Guy, van der Kroeg, Mark, Kantae, Vasudev, Baggelaar, Marc P, de Vrij, Femke M S, Deng, Hui, Allarà, Marco, Fezza, Filomena, Lin, Zhanmin, van der Wel, Tom, Soethoudt, Marjolein, Mock, Elliot D, den Dulk, Hans, Baak, Ilse L, Florea, Bogdan I, Hendriks, Giel, De Petrocellis, Luciano, Overkleeft, Herman S, Hankemeier, Thomas, De Zeeuw, Chris I, Di Marzo, Vincenzo, Maccarrone, Mauro, Cravatt, Benjamin F, Kushner, Steven A, van der Stelt, Mario, van Esbroeck, Annelot C M, Janssen, Antonius P A, Cognetta, Armand B, Ogasawara, Daisuke, Shpak, Guy, van der Kroeg, Mark, Kantae, Vasudev, Baggelaar, Marc P, de Vrij, Femke M S, Deng, Hui, Allarà, Marco, Fezza, Filomena, Lin, Zhanmin, van der Wel, Tom, Soethoudt, Marjolein, Mock, Elliot D, den Dulk, Hans, Baak, Ilse L, Florea, Bogdan I, Hendriks, Giel, De Petrocellis, Luciano, Overkleeft, Herman S, Hankemeier, Thomas, De Zeeuw, Chris I, Di Marzo, Vincenzo, Maccarrone, Mauro, Cravatt, Benjamin F, Kushner, Steven A, and van der Stelt, Mario
- Abstract
A recent phase 1 trial of the fatty acid amide hydrolase (FAAH) inhibitor BIA 10-2474 led to the death of one volunteer and produced mild-to-severe neurological symptoms in four others. Although the cause of the clinical neurotoxicity is unknown, it has been postulated, given the clinical safety profile of other tested FAAH inhibitors, that off-target activities of BIA 10-2474 may have played a role. Here we use activity-based proteomic methods to determine the protein interaction landscape of BIA 10-2474 in human cells and tissues. This analysis revealed that the drug inhibits several lipases that are not targeted by PF04457845, a highly selective and clinically tested FAAH inhibitor. BIA 10-2474, but not PF04457845, produced substantial alterations in lipid networks in human cortical neurons, suggesting that promiscuous lipase inhibitors have the potential to cause metabolic dysregulation in the nervous system.
- Published
- 2017
16. An expandable embryonic stem cell-derived Purkinje neuron progenitor population that exhibits in vivo maturation in the adult mouse cerebellum
- Author
-
Higuera, Gustavo A, Iaffaldano, Grazia, Bedar, Meiwand, Shpak, Guy, Broersen, Robin, Munshi, Shashini T, Dupont, Catherine, Gribnau, Joost, de Vrij, Femke M S, Kushner, Steven A, De Zeeuw, Chris I, Higuera, Gustavo A, Iaffaldano, Grazia, Bedar, Meiwand, Shpak, Guy, Broersen, Robin, Munshi, Shashini T, Dupont, Catherine, Gribnau, Joost, de Vrij, Femke M S, Kushner, Steven A, and De Zeeuw, Chris I
- Abstract
The directed differentiation of patient-derived induced pluripotent stem cells into cell-type specific neurons has inspired the development of therapeutic discovery for neurodegenerative diseases. Many forms of ataxia result from degeneration of cerebellar Purkinje cells, but thus far it has not been possible to efficiently generate Purkinje neuron (PN) progenitors from human or mouse pluripotent stem cells, let alone to develop a methodology for in vivo transplantation in the adult cerebellum. Here, we present a protocol to obtain an expandable population of cerebellar neuron progenitors from mouse embryonic stem cells. Our protocol is characterized by applying factors that promote proliferation of cerebellar progenitors. Cerebellar progenitors isolated in culture from cell aggregates contained a stable subpopulation of PN progenitors that could be expanded for up to 6 passages. When transplanted into the adult cerebellum of either wild-type mice or a strain lacking Purkinje cells (L7cre-ERCC1 knockout), GFP-labeled progenitors differentiated in vivo to establish a population of calbindin-positive cells in the molecular layer with dendritic trees typical of mature PNs. We conclude that this protocol may be useful for the generation and maturation of PNs, highlighting the potential for development of a regenerative medicine approach to the treatment of cerebellar neurodegenerative diseases.
- Published
- 2017
17. Phenotypic Differences between Asian and African Lineage Zika Viruses in Human Neural Progenitor Cells
- Author
-
Anfasa, Fatih, primary, Siegers, Jurre Y., additional, van der Kroeg, Mark, additional, Mumtaz, Noreen, additional, Stalin Raj, V., additional, de Vrij, Femke M. S., additional, Widagdo, W., additional, Gabriel, Gülsah, additional, Salinas, Sara, additional, Simonin, Yannick, additional, Reusken, Chantal, additional, Kushner, Steven A., additional, Koopmans, Marion P. G., additional, Haagmans, Bart, additional, Martina, Byron E. E., additional, and van Riel, Debby, additional
- Published
- 2017
- Full Text
- View/download PDF
18. An expandable embryonic stem cell-derived Purkinje neuron progenitor population that exhibits in vivo maturation in the adult mouse cerebellum
- Author
-
Higuera, Gustavo A., primary, Iaffaldano, Grazia, additional, Bedar, Meiwand, additional, Shpak, Guy, additional, Broersen, Robin, additional, Munshi, Shashini T., additional, Dupont, Catherine, additional, Gribnau, Joost, additional, de Vrij, Femke M. S., additional, Kushner, Steven A., additional, and De Zeeuw, Chris I., additional
- Published
- 2017
- Full Text
- View/download PDF
19. Novel genetic loci affecting facial shape variation in humans.
- Author
-
Ziyi Xiong, Dankova, Gabriela, Howe, Laurence J., Myoung Keun Lee, Hysi, Pirro G., de Jong, Markus A., Gu Zhu, Adhikari, Kaustubh, Dan Li, Yi Li, Bo Pan, Feingold, Eleanor, Marazita, Mary L., Shaffer, John R., McAloney, Kerrie, Shu-Hua Xu, Li Jin, Sijia Wang, de Vrij, Femke M. S., and Lendemeijer, Bas
- Published
- 2019
- Full Text
- View/download PDF
20. Epigenetic characterization of the FMR1 promoter in induced pluripotent stem cells from human fibroblasts carrying an unmethylated full mutation
- Author
-
de Esch, Celine E F, Ghazvini, Mehrnaz, Loos, Friedemann, Schelling-Kazaryan, Nune, Widagdo, W, Munshi, Shashini T, van der Wal, Erik, Douben, Hannie, Gunhanlar, Nilhan, Kushner, Steven A, Pijnappel, W W M Pim, de Vrij, Femke M S, Geijsen, Niels, Gribnau, Joost, Willemsen, Rob, de Esch, Celine E F, Ghazvini, Mehrnaz, Loos, Friedemann, Schelling-Kazaryan, Nune, Widagdo, W, Munshi, Shashini T, van der Wal, Erik, Douben, Hannie, Gunhanlar, Nilhan, Kushner, Steven A, Pijnappel, W W M Pim, de Vrij, Femke M S, Geijsen, Niels, Gribnau, Joost, and Willemsen, Rob
- Abstract
Silencing of the FMR1 gene leads to fragile X syndrome, the most common cause of inherited intellectual disability. To study the epigenetic modifications of the FMR1 gene during silencing in time, we used fibroblasts and induced pluripotent stem cells (iPSCs) of an unmethylated full mutation (uFM) individual with normal intelligence. The uFM fibroblast line carried an unmethylated FMR1 promoter region and expressed normal to slightly increased FMR1 mRNA levels. The FMR1 expression in the uFM line corresponds with the increased H3 acetylation and H3K4 methylation in combination with a reduced H3K9 methylation. After reprogramming, the FMR1 promoter region was methylated in all uFM iPSC clones. Two clones were analyzed further and showed a lack of FMR1 expression, whereas the presence of specific histone modifications also indicated a repressed FMR1 promoter. In conclusion, these findings demonstrate that the standard reprogramming procedure leads to epigenetic silencing of the fully mutated FMR1 gene.
- Published
- 2014
21. Epigenetic characterization of the FMR1 promoter in induced pluripotent stem cells from human fibroblasts carrying an unmethylated full mutation
- Author
-
Onderzoek, Tissue Repair, CSCA TR1, de Esch, Celine E F, Ghazvini, Mehrnaz, Loos, Friedemann, Schelling-Kazaryan, Nune, Widagdo, W, Munshi, Shashini T, van der Wal, Erik, Douben, Hannie, Gunhanlar, Nilhan, Kushner, Steven A, Pijnappel, W W M Pim, de Vrij, Femke M S, Geijsen, Niels, Gribnau, Joost, Willemsen, Rob, Onderzoek, Tissue Repair, CSCA TR1, de Esch, Celine E F, Ghazvini, Mehrnaz, Loos, Friedemann, Schelling-Kazaryan, Nune, Widagdo, W, Munshi, Shashini T, van der Wal, Erik, Douben, Hannie, Gunhanlar, Nilhan, Kushner, Steven A, Pijnappel, W W M Pim, de Vrij, Femke M S, Geijsen, Niels, Gribnau, Joost, and Willemsen, Rob
- Published
- 2014
22. Hepatitis E Virus Infects Neurons and Brains.
- Author
-
Xinying Zhou, Fen Huang, Lei Xu, Zhanmin Lin, de Vrij, Femke M. S., Ayo-Martin, Ane C., van der Kroeg, Mark, Manzhi Zhao, Yuebang Yin, Wenshi Wang, Wanlu Cao, Yijin Wang, Kushner, Steven A., Peron, Jean Marie, Alric, Laurent, de Man, Robert A., Jacobs, Bart C., van Eijk, Jeroen J., Aronica, Eleonora M. A., and Sprengers, Dave
- Subjects
HEPATITIS E virus ,NEURONS ,CELL lines ,LABORATORY mice ,ANTIVIRAL agents ,DIFFERENTIAL diagnosis ,RNA analysis ,ANIMALS ,BRAIN ,CEREBROSPINAL fluid ,HEPATITIS E ,HEPATITIS viruses ,LIVER ,MICE ,GUILLAIN-Barre syndrome ,PRIMATES ,PROTEINS ,RIBAVIRIN ,VIRAL physiology ,PHARMACODYNAMICS - Abstract
Hepatitis E virus (HEV), as a hepatotropic virus, is supposed to exclusively infect the liver and only cause hepatitis. However, a broad range of extrahepatic manifestations (in particular, idiopathic neurological disorders) have been recently reported in association with its infection. In this study, we have demonstrated that various human neural cell lines (embryonic stem cell-derived neural lineage cells) induced pluripotent stem cell-derived human neurons and primary mouse neurons are highly susceptible to HEV infection. Treatment with interferon-α or ribavirin, the off-label antiviral drugs for chronic hepatitis E, exerted potent antiviral activities against HEV infection in neural cells. More importantly, in mice and monkey peripherally inoculated with HEV particles, viral RNA and protein were detected in brain tissues. Finally, patients with HEV-associated neurological disorders shed the virus into cerebrospinal fluid, indicating a direct infection of their nervous system. Thus, HEV is neurotropic in vitro, and in mice, monkeys, and possibly humans. These results challenge the dogma of HEV as a pure hepatotropic virus and suggest that HEV infection should be considered in the differential diagnosis of idiopathic neurological disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
23. Mutant ubiquitin expressed in Alzheimer's disease causes neuronal death
- Author
-
de Vrij, Femke M S, Sluijs, J A, Gregori, L, Fischer, D.F., Hermens, W.T.J.M.C., Goldgaber, D, Verhaagen, J, Van Leeuwen, F W, Hol, E M, and Netherlands Institute for Neuroscience (NIN)
- Subjects
Cell Nucleus ,Neurons ,Proteasome Endopeptidase Complex ,Ubiquitin ,Recombinant Fusion Proteins ,Research Support, Non-U.S. Gov't ,Blotting, Western ,Genetic Vectors ,Proteins ,Apoptosis ,Adenoviridae ,Cysteine Endopeptidases ,Adenosine Triphosphate ,nervous system ,Lac Operon ,Alzheimer Disease ,Multienzyme Complexes ,Mutation ,Tumor Cells, Cultured ,Journal Article ,Humans ,Signal Transduction - Abstract
Ubiquitin-B+1 (UBB+1) is a mutant ubiquitin that accumulates in the neurones of patients with Alzheimer's disease (AD). Here we report on the biochemical and functional differences between ubiquitin and UBB+1 and the effect of the mutant protein on neuronal cells. UBB+1 lacks the capacity to ubiquitinate, and although it is ubiquitinated itself, UBB+1 is not degraded by the ubiquitin-proteasomal system and is quite stable in neuronal cells. Overexpression of UBB+1 in neuroblastoma cells significantly induces nuclear fragmentation and cell death. Our results demonstrate that accumulation of UBB+1 in neurones is detrimental and may contribute to neuronal dysfunction in AD patients.
- Published
- 2001
24. Dose-dependent inhibition of proteasome activity by a mutant ubiquitin associated with neurodegenerative disease
- Author
-
van Tijn, Paula, primary, de Vrij, Femke M. S., additional, Schuurman, Karianne G., additional, Dantuma, Nico P., additional, Fischer, David F., additional, van Leeuwen, Fred W., additional, and Hol, Elly M., additional
- Published
- 2007
- Full Text
- View/download PDF
25. Disease-specific accumulation of mutant ubiquitin as a marker for proteasomal dysfunction in the brain.
- Author
-
Fischer, David F., De Vos, Rob A. I., Van Dijk, Renske, De Vrij, Femke M. S., Proper, Evelien A., Sonnemans, Marc A. F., Verhage, Marian C., Sluijs, Jacqueline A., Hobo, Barbara, Zouambia, Mohamed, Steur, Ernst N. H. Jansen, Kamphorst, Wouter, Hol, Elly M., and Van Leeuwen, Fred W.
- Subjects
UBIQUITIN ,GENE expression ,RNA ,NUCLEIC acids ,NEUTRAL proteinases ,ALZHEIMER'S disease ,BRAIN diseases ,PATHOLOGICAL physiology - Abstract
Molecular misreading of the ubiquitin-B (UBB) gene results in a dinucleotide deletion in UBB mRNA. The resulting mutant protein, UBB&sup+1;, accumulates in the neuropathological hallmarks of Alzheimer disease. In vitro, UBB&sup+1; inhibits proteasomal proteolysis, although it is also an ubiquitin fusion degradation substrate for the proteasome. Using the ligase chain reaction to detect dinucleotide deletions, we report here that UBB&sup+1; transcripts are present in each neurodegenerative disease studied (tauo- and synucleinopathies) and even in control brain samples. In contrast to UBB&sup+1; transcripts, UBB&sup+1; protein accumulation in the ubiquitin-containing neuropathological hallmarks is restricted to the tauopathies such as Pick disease, frontotemporal dementia, progressive supranuclear palsy, and argyrophilic grain disease. Remarkably, UBB&sup+1; protein is not detected in the major forms of synucleinopathies (Lewy body disease and multiple system atrophy). The neurologically intact brain can cope with UBB&sup+1; as lentivirally delivered UBB&sup+1; protein is rapidly degraded in rat hippocampus, whereas the K29,48R mutant of UBB&sup+1;, which is not ubiquitinated, is abundantly expressed. The finding that UBB&sup+1; protein only accumulates in tauopathies thus implies that the ubiquitin-proteasome system is impaired specifically in this group of neurodegenerative diseases and not in synucleinopathies and that the presence of UBB&sup+1; protein reports proteasomal dysfunction in the brain. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
26. Novel genetic loci affecting facial shape variation in humans
- Author
-
Xiong, Ziyi, Dankova, Gabriela, Howe, Laurence J., Lee, Myoung Keun, Hysi, Pirro G., de Jong, Markus A., Zhu, Gu, Adhikari, Kaustubh, Li, Dan, Li, Yi, Pan, Bo, Feingold, Eleanor, Marazita, Mary L., Shaffer, John R., McAloney, Kerrie, Xu, Shu-Hua, Jin, Li, Wang, Sijia, de Vrij, Femke M. S., Lendemeijer, Bas, Richmond, Stephen, Zhurov, Alexei, Lewis, Sarah, Sharp, Gemma C., Paternoster, Lavinia, Thompson, Holly, Gonzalez-Jose, Rolando, Bortolini, Maria Catira, Canizales-Quinteros, Samuel, Gallo, Carla, Poletti, Giovanni, Bedoya, Gabriel, Rothhammer, Francisco, Uitterlinden, André G., Ikram, M. Arfan, Wolvius, Eppo, Kushner, Steven A., Nijsten, Tamar E. C., Palstra, Robert-Jan T. S., Boehringer, Stefan, Medland, Sarah E., Tang, Kun, Ruiz-Linares, Andrés, Martin, Nicholas G., Spector, Timothy D., Stergiakouli, Evie, Weinberg, Seth M., Liu, Fan, Kayser, Manfred, Xiong, Ziyi, Dankova, Gabriela, Howe, Laurence J., Lee, Myoung Keun, Hysi, Pirro G., de Jong, Markus A., Zhu, Gu, Adhikari, Kaustubh, Li, Dan, Li, Yi, Pan, Bo, Feingold, Eleanor, Marazita, Mary L., Shaffer, John R., McAloney, Kerrie, Xu, Shu-Hua, Jin, Li, Wang, Sijia, de Vrij, Femke M. S., Lendemeijer, Bas, Richmond, Stephen, Zhurov, Alexei, Lewis, Sarah, Sharp, Gemma C., Paternoster, Lavinia, Thompson, Holly, Gonzalez-Jose, Rolando, Bortolini, Maria Catira, Canizales-Quinteros, Samuel, Gallo, Carla, Poletti, Giovanni, Bedoya, Gabriel, Rothhammer, Francisco, Uitterlinden, André G., Ikram, M. Arfan, Wolvius, Eppo, Kushner, Steven A., Nijsten, Tamar E. C., Palstra, Robert-Jan T. S., Boehringer, Stefan, Medland, Sarah E., Tang, Kun, Ruiz-Linares, Andrés, Martin, Nicholas G., Spector, Timothy D., Stergiakouli, Evie, Weinberg, Seth M., Liu, Fan, and Kayser, Manfred
- Abstract
The human face represents a combined set of highly heritable phenotypes, but knowledge on its genetic architecture remains limited, despite the relevance for various fields. A series of genome-wide association studies on 78 facial shape phenotypes quantified from 3-dimensional facial images of 10,115 Europeans identified 24 genetic loci reaching study-wide suggestive association (p<5x10-8), among which 17 were previously unreported. A follow-up multi-ethnic study in additional 7,917 individuals confirmed 10 loci including 6 unreported ones (padjusted<2.1x10-3). A global map of derived polygenic face scores assembled facial features in major continental groups consistent with anthropological knowledge. Analyses of epigenomic datasets from cranial neural crest cells revealed abundant cis-regulatory activities at the face-associated genetic loci. Luciferase reporter assays in neural crest progenitor cells highlighted enhancer activities of several face-associated DNA variants. These results substantially advance our understanding of the genetic basis underlying human facial variation and provide candidates for future in-vivo functional studies.
27. Novel genetic loci affecting facial shape variation in humans
- Author
-
Xiong, Ziyi, Dankova, Gabriela, Howe, Laurence J., Lee, Myoung Keun, Hysi, Pirro G., de Jong, Markus A., Zhu, Gu, Adhikari, Kaustubh, Li, Dan, Li, Yi, Pan, Bo, Feingold, Eleanor, Marazita, Mary L., Shaffer, John R., McAloney, Kerrie, Xu, Shu-Hua, Jin, Li, Wang, Sijia, de Vrij, Femke M. S., Lendemeijer, Bas, Richmond, Stephen, Zhurov, Alexei, Lewis, Sarah, Sharp, Gemma C., Paternoster, Lavinia, Thompson, Holly, Gonzalez-Jose, Rolando, Bortolini, Maria Catira, Canizales-Quinteros, Samuel, Gallo, Carla, Poletti, Giovanni, Bedoya, Gabriel, Rothhammer, Francisco, Uitterlinden, André G., Ikram, M. Arfan, Wolvius, Eppo, Kushner, Steven A., Nijsten, Tamar E. C., Palstra, Robert-Jan T. S., Boehringer, Stefan, Medland, Sarah E., Tang, Kun, Ruiz-Linares, Andrés, Martin, Nicholas G., Spector, Timothy D., Stergiakouli, Evie, Weinberg, Seth M., Liu, Fan, Kayser, Manfred, Xiong, Ziyi, Dankova, Gabriela, Howe, Laurence J., Lee, Myoung Keun, Hysi, Pirro G., de Jong, Markus A., Zhu, Gu, Adhikari, Kaustubh, Li, Dan, Li, Yi, Pan, Bo, Feingold, Eleanor, Marazita, Mary L., Shaffer, John R., McAloney, Kerrie, Xu, Shu-Hua, Jin, Li, Wang, Sijia, de Vrij, Femke M. S., Lendemeijer, Bas, Richmond, Stephen, Zhurov, Alexei, Lewis, Sarah, Sharp, Gemma C., Paternoster, Lavinia, Thompson, Holly, Gonzalez-Jose, Rolando, Bortolini, Maria Catira, Canizales-Quinteros, Samuel, Gallo, Carla, Poletti, Giovanni, Bedoya, Gabriel, Rothhammer, Francisco, Uitterlinden, André G., Ikram, M. Arfan, Wolvius, Eppo, Kushner, Steven A., Nijsten, Tamar E. C., Palstra, Robert-Jan T. S., Boehringer, Stefan, Medland, Sarah E., Tang, Kun, Ruiz-Linares, Andrés, Martin, Nicholas G., Spector, Timothy D., Stergiakouli, Evie, Weinberg, Seth M., Liu, Fan, and Kayser, Manfred
- Abstract
The human face represents a combined set of highly heritable phenotypes, but knowledge on its genetic architecture remains limited, despite the relevance for various fields. A series of genome-wide association studies on 78 facial shape phenotypes quantified from 3-dimensional facial images of 10,115 Europeans identified 24 genetic loci reaching study-wide suggestive association (p<5x10-8), among which 17 were previously unreported. A follow-up multi-ethnic study in additional 7,917 individuals confirmed 10 loci including 6 unreported ones (padjusted<2.1x10-3). A global map of derived polygenic face scores assembled facial features in major continental groups consistent with anthropological knowledge. Analyses of epigenomic datasets from cranial neural crest cells revealed abundant cis-regulatory activities at the face-associated genetic loci. Luciferase reporter assays in neural crest progenitor cells highlighted enhancer activities of several face-associated DNA variants. These results substantially advance our understanding of the genetic basis underlying human facial variation and provide candidates for future in-vivo functional studies.
28. iPSC-derived healthy human astrocytes selectively load miRNAs targeting neuronal genes into extracellular vesicles.
- Author
-
Gordillo-Sampedro S, Antounians L, Wei W, Mufteev M, Lendemeijer B, Kushner SA, de Vrij FMS, Zani A, and Ellis J
- Subjects
- Humans, Cell Differentiation, Cells, Cultured, Neural Stem Cells metabolism, Neural Stem Cells cytology, Extracellular Vesicles metabolism, Induced Pluripotent Stem Cells metabolism, MicroRNAs metabolism, MicroRNAs genetics, Astrocytes metabolism, Neurons metabolism
- Abstract
Astrocytes are in constant communication with neurons during the establishment and maturation of functional networks in the developing brain. Astrocytes release extracellular vesicles (EVs) containing microRNA (miRNA) cargo that regulates transcript stability in recipient cells. Astrocyte released factors are thought to be involved in neurodevelopmental disorders. Healthy astrocytes partially rescue Rett Syndrome (RTT) neuron function. EVs isolated from stem cell progeny also correct aspects of RTT. EVs cross the blood-brain barrier (BBB) and their cargo is found in peripheral blood which may allow non-invasive detection of EV cargo as biomarkers produced by healthy astrocytes. Here we characterize miRNA cargo and sequence motifs in healthy human astrocyte derived EVs (ADEVs). First, human induced Pluripotent Stem Cells (iPSC) were differentiated into Neural Progenitor Cells (NPCs) and subsequently into astrocytes using a rapid differentiation protocol. iPSC derived astrocytes expressed specific markers, displayed intracellular calcium transients and secreted ADEVs. miRNAs were identified by RNA-Seq on astrocytes and ADEVs and target gene pathway analysis detected brain and immune related terms. The miRNA profile was consistent with astrocyte identity, and included approximately 80 miRNAs found in astrocytes that were relatively depleted in ADEVs suggestive of passive loading. About 120 miRNAs were relatively enriched in ADEVs and motif analysis discovered binding sites for RNA binding proteins FUS, SRSF7 and CELF5. miR-483-5p was the most significantly enriched in ADEVs. This miRNA regulates MECP2 expression in neurons and has been found differentially expressed in blood samples from RTT patients. Our results identify potential miRNA biomarkers selectively sorted into ADEVs and implicate RNA binding protein sequence dependent mechanisms for miRNA cargo loading., Competing Interests: Declaration of competing interest The authors declare they have no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
29. Modulating mutational outcomes and improving precise gene editing at CRISPR-Cas9-induced breaks by chemical inhibition of end-joining pathways.
- Author
-
Schimmel J, Muñoz-Subirana N, Kool H, van Schendel R, van der Vlies S, Kamp JA, de Vrij FMS, Kushner SA, Smith GCM, Boulton SJ, and Tijsterman M
- Subjects
- DNA Breaks, Double-Stranded, Mutation genetics, DNA End-Joining Repair, Gene Editing, CRISPR-Cas Systems genetics
- Abstract
Gene editing through repair of CRISPR-Cas9-induced chromosomal breaks offers a means to correct a wide range of genetic defects. Directing repair to produce desirable outcomes by modulating DNA repair pathways holds considerable promise to increase the efficiency of genome engineering. Here, we show that inhibition of non-homologous end joining (NHEJ) or polymerase theta-mediated end joining (TMEJ) can be exploited to alter the mutational outcomes of CRISPR-Cas9. We show robust inhibition of TMEJ activity at CRISPR-Cas9-induced double-strand breaks (DSBs) using ART558, a potent polymerase theta (Polϴ) inhibitor. Using targeted sequencing, we show that ART558 suppresses the formation of microhomology-driven deletions in favor of NHEJ-specific outcomes. Conversely, NHEJ deficiency triggers the formation of large kb-sized deletions, which we show are the products of mutagenic TMEJ. Finally, we show that combined chemical inhibition of TMEJ and NHEJ increases the efficiency of homology-driven repair (HDR)-mediated precise gene editing. Our work reports a robust strategy to improve the fidelity and safety of genome engineering., Competing Interests: Declaration of interests S.J.B. is a co-founder, shareholder, and VP of Science Strategy at Artios Pharma, Ltd., Babraham Research Campus, UK. G.C.M.S. is chief scientific officer and shareholder of Artios Pharma, Ltd., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
30. The neuroinvasiveness, neurotropism, and neurovirulence of SARS-CoV-2.
- Author
-
Bauer L, Laksono BM, de Vrij FMS, Kushner SA, Harschnitz O, and van Riel D
- Subjects
- Humans, SARS-CoV-2, COVID-19, Nervous System Diseases
- Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection is associated with a diverse spectrum of neurological complications during the acute and postacute stages. The pathogenesis of these complications is complex and dependent on many factors. For accurate and consistent interpretation of experimental data in this fast-growing field of research, it is essential to use terminology consistently. In this article, we outline the distinctions between neuroinvasiveness, neurotropism, and neurovirulence. Additionally, we discuss current knowledge of these distinct features underlying the pathogenesis of SARS-CoV-2-associated neurological complications. Lastly, we briefly discuss the advantages and limitations of different experimental models, and how these approaches can further be leveraged to advance the field., Competing Interests: Declaration of interests The authors declare no conflicts of interest, (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
31. A functional variant in the miR-142 promoter modulating its expression and conferring risk of Alzheimer disease.
- Author
-
Ghanbari M, Munshi ST, Ma B, Lendemeijer B, Bansal S, Adams HH, Wang W, Goth K, Slump DE, van den Hout MCGN, van IJcken WFJ, Bellusci S, Pan Q, Erkeland SJ, de Vrij FMS, Kushner SA, and Ikram MA
- Subjects
- Alleles, Alzheimer Disease metabolism, Animals, Cell Line, Chromosome Mapping, Computational Biology methods, Disease Models, Animal, Gene Expression Regulation, Genetic Association Studies, Genome-Wide Association Study, Hippocampus metabolism, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Mice, Mice, Knockout, Models, Biological, Neural Stem Cells cytology, Neural Stem Cells metabolism, Polymorphism, Single Nucleotide, RNA Interference, RNA, Untranslated, Alzheimer Disease genetics, Genetic Predisposition to Disease, Genetic Variation, MicroRNAs genetics, Promoter Regions, Genetic
- Abstract
Noncoding RNAs have been widely recognized as essential mediators of gene regulation. However, in contrast to protein-coding genes, much less is known about the influence of noncoding RNAs on human diseases. Here we examined the association of genetic variants located in primary microRNA sequences and long noncoding RNAs (lncRNAs) with Alzheimer disease (AD) by leveraging data from the largest genome-wide association meta-analysis of late-onset AD. Variants annotated to 5 miRNAs and 10 lncRNAs (in seven distinct loci) exceeded the Bonferroni-corrected significance threshold (p < 1.02 × 10
-6 ). Among these, a leading variant (rs2526377:A>G) at the 17q22 locus annotated to two noncoding RNAs (MIR142 and BZRAP1-AS) was significantly associated with a reduced risk of AD and fulfilled predefined criteria for being a functional variant. Our functional genomic analyses revealed that rs2526377 affects the promoter activity and decreases the expression of miR-142. Moreover, differential expression analysis by RNA-Seq in human iPSC-derived neural progenitor cells and the hippocampus of miR-142 knockout mice demonstrated multiple target genes of miR-142 in the brain that are likely to be involved in the inflammatory and neurodegenerative manifestations of AD. These include TGFBR1 and PICALM, of which their derepression in the brain due to reduced expression levels of miR-142-3p may reduce the risk of AD., (© 2019 Wiley Periodicals, Inc.)- Published
- 2019
- Full Text
- View/download PDF
32. ACO2 homozygous missense mutation associated with complicated hereditary spastic paraplegia.
- Author
-
Bouwkamp CG, Afawi Z, Fattal-Valevski A, Krabbendam IE, Rivetti S, Masalha R, Quadri M, Breedveld GJ, Mandel H, Tailakh MA, Beverloo HB, Stevanin G, Brice A, van IJcken WFJ, Vernooij MW, Dolga AM, de Vrij FMS, Bonifati V, and Kushner SA
- Abstract
Objective: To identify the clinical characteristics and genetic etiology of a family affected with hereditary spastic paraplegia (HSP)., Methods: Clinical, genetic, and functional analyses involving genome-wide linkage coupled to whole-exome sequencing in a consanguineous family with complicated HSP., Results: A homozygous missense mutation was identified in the ACO2 gene (c.1240T>G p.Phe414Val) that segregated with HSP complicated by intellectual disability and microcephaly. Lymphoblastoid cell lines of homozygous carrier patients revealed significantly decreased activity of the mitochondrial aconitase enzyme and defective mitochondrial respiration. ACO2 encodes mitochondrial aconitase, an essential enzyme in the Krebs cycle. Recessive mutations in this gene have been previously associated with cerebellar ataxia., Conclusions: Our findings nominate ACO2 as a disease-causing gene for autosomal recessive complicated HSP and provide further support for the central role of mitochondrial defects in the pathogenesis of HSP.
- Published
- 2018
- Full Text
- View/download PDF
33. A novel fragile X syndrome mutation reveals a conserved role for the carboxy-terminus in FMRP localization and function.
- Author
-
Okray Z, de Esch CE, Van Esch H, Devriendt K, Claeys A, Yan J, Verbeeck J, Froyen G, Willemsen R, de Vrij FM, and Hassan BA
- Subjects
- Animals, Cell Line, Transformed, Cell Line, Tumor, Drosophila melanogaster, Humans, Male, Mice, Protein Structure, Tertiary, Protein Transport genetics, Cell Nucleus genetics, Cell Nucleus metabolism, Cell Nucleus pathology, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome genetics, Fragile X Syndrome metabolism, Fragile X Syndrome pathology, Mutation, Nuclear Localization Signals genetics, Nuclear Localization Signals metabolism, Trinucleotide Repeat Expansion
- Abstract
Loss of function of the FMR1 gene leads to fragile X syndrome (FXS), the most common form of intellectual disability. The loss of FMR1 function is usually caused by epigenetic silencing of the FMR1 promoter leading to expansion and subsequent methylation of a CGG repeat in the 5' untranslated region. Very few coding sequence variations have been experimentally characterized and shown to be causal to the disease. Here, we describe a novel FMR1 mutation and reveal an unexpected nuclear export function for the C-terminus of FMRP. We screened a cohort of patients with typical FXS symptoms who tested negative for CGG repeat expansion in the FMR1 locus. In one patient, we identified a guanine insertion in FMR1 exon 15. This mutation alters the open reading frame creating a short novel C-terminal sequence, followed by a stop codon. We find that this novel peptide encodes a functional nuclear localization signal (NLS) targeting the patient FMRP to the nucleolus in human cells. We also reveal an evolutionarily conserved nuclear export function associated with the endogenous C-terminus of FMRP. In vivo analyses in Drosophila demonstrate that a patient-mimetic mutation alters the localization and function of Dfmrp in neurons, leading to neomorphic neuronal phenotypes., (© 2015 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2015
- Full Text
- View/download PDF
34. Epigenetic characterization of the FMR1 promoter in induced pluripotent stem cells from human fibroblasts carrying an unmethylated full mutation.
- Author
-
de Esch CE, Ghazvini M, Loos F, Schelling-Kazaryan N, Widagdo W, Munshi ST, van der Wal E, Douben H, Gunhanlar N, Kushner SA, Pijnappel WW, de Vrij FM, Geijsen N, Gribnau J, and Willemsen R
- Subjects
- Adolescent, Animals, Case-Control Studies, Cell Line, Cellular Reprogramming, Child, Child, Preschool, Female, Fibroblasts cytology, Fragile X Mental Retardation Protein metabolism, Histones metabolism, Humans, Induced Pluripotent Stem Cells cytology, Male, Mice, Promoter Regions, Genetic, RNA, Messenger genetics, RNA, Messenger metabolism, DNA Methylation, Fibroblasts metabolism, Fragile X Mental Retardation Protein genetics, Gene Silencing, Induced Pluripotent Stem Cells metabolism, Mutation
- Abstract
Silencing of the FMR1 gene leads to fragile X syndrome, the most common cause of inherited intellectual disability. To study the epigenetic modifications of the FMR1 gene during silencing in time, we used fibroblasts and induced pluripotent stem cells (iPSCs) of an unmethylated full mutation (uFM) individual with normal intelligence. The uFM fibroblast line carried an unmethylated FMR1 promoter region and expressed normal to slightly increased FMR1 mRNA levels. The FMR1 expression in the uFM line corresponds with the increased H3 acetylation and H3K4 methylation in combination with a reduced H3K9 methylation. After reprogramming, the FMR1 promoter region was methylated in all uFM iPSC clones. Two clones were analyzed further and showed a lack of FMR1 expression, whereas the presence of specific histone modifications also indicated a repressed FMR1 promoter. In conclusion, these findings demonstrate that the standard reprogramming procedure leads to epigenetic silencing of the fully mutated FMR1 gene., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
35. AFQ056, a new mGluR5 antagonist for treatment of fragile X syndrome.
- Author
-
Levenga J, Hayashi S, de Vrij FM, Koekkoek SK, van der Linde HC, Nieuwenhuizen I, Song C, Buijsen RA, Pop AS, Gomezmancilla B, Nelson DL, Willemsen R, Gasparini F, and Oostra BA
- Subjects
- Animals, Cells, Cultured, Dendritic Spines drug effects, Dendritic Spines metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Hippocampus metabolism, Mice, Mice, Knockout, Neurons metabolism, Receptor, Metabotropic Glutamate 5, Hippocampus drug effects, Neurons drug effects, Receptors, Metabotropic Glutamate antagonists & inhibitors, Reflex, Startle drug effects, Sensory Gating drug effects
- Abstract
Fragile X syndrome, the most common form of inherited intellectual disability, is caused by a lack of FMRP, which is the product of the Fmr1 gene. FMRP is an RNA-binding protein and a component of RNA-granules found in the dendrites of neurons. At the synapse, FMRP is involved in regulation of translation of specific target mRNAs upon stimulation of mGluR5 receptors. In this study, we test the effects of a new mGluR5 antagonist (AFQ056) on the prepulse inhibition of startle response in mice. We show that Fmr1 KO mice have a deficit in inhibition of the startle response after a prepulse and that AFQ056 can rescue this phenotype. We also studied the effect of AFQ056 on cultured Fmr1 KO hippocampal neurons; untreated neurons showed elongated spines and treatment resulted in shortened spines. These results suggest that AFQ056 might be a potent mGluR5 antagonist to rescue various aspects of the fragile X phenotype., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
36. Subregion-specific dendritic spine abnormalities in the hippocampus of Fmr1 KO mice.
- Author
-
Levenga J, de Vrij FM, Buijsen RA, Li T, Nieuwenhuizen IM, Pop A, Oostra BA, and Willemsen R
- Subjects
- Animals, CA1 Region, Hippocampal metabolism, CA3 Region, Hippocampal cytology, CA3 Region, Hippocampal metabolism, Dendritic Spines genetics, Fragile X Mental Retardation Protein genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Pyramidal Cells cytology, Pyramidal Cells metabolism, CA1 Region, Hippocampal cytology, Dendritic Spines classification, Fragile X Mental Retardation Protein metabolism, Pyramidal Cells growth & development
- Abstract
Fragile X syndrome (FXS) is the most common inherited form of mental retardation and is caused by the lack of fragile X mental retardation protein (FMRP). In the brain, spine abnormalities have been reported in both patients with FXS and Fmr1 knockout mice. This altered spine morphology has been linked to disturbed synaptic transmission related to altered signaling in the excitatory metabotropic glutamate receptor 5 (mGluR5) pathway. We investigated hippocampal protrusion morphology in adult Fmr1 knockout mice. Our results show a hippocampal CA1-specific altered protrusion phenotype, which was absent in the CA3 region of the hippocampus. This suggests a subregion-specific function of FMRP in synaptic plasticity in the brain., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
37. Potential therapeutic interventions for fragile X syndrome.
- Author
-
Levenga J, de Vrij FM, Oostra BA, and Willemsen R
- Subjects
- Animals, Benzodiazepines therapeutic use, Excitatory Amino Acid Agonists therapeutic use, Fragile X Syndrome drug therapy, Fragile X Syndrome genetics, Humans, Models, Biological, Receptor, Metabotropic Glutamate 5, Receptors, GABA metabolism, Receptors, Metabotropic Glutamate agonists, Signal Transduction drug effects, Fragile X Syndrome metabolism, Receptors, Metabotropic Glutamate metabolism
- Abstract
Fragile X syndrome (FXS) is caused by a lack of the fragile X mental retardation protein (FMRP); FMRP deficiency in neurons of patients with FXS causes intellectual disability (IQ<70) and several behavioural problems, including hyperactivity and autistic-like features. In the brain, no gross morphological malformations have been found, although subtle spine abnormalities have been reported. FXS has been linked to altered group I metabotropic glutamate receptor (mGluR)-dependent and independent forms of synaptic plasticity. Here, we discuss potential targeted therapeutic strategies developed to specifically correct disturbances in the excitatory mGluR and the inhibitory gamma-aminobutyric (GABA) receptor pathways that have been tested in animal models and/or in clinical trials with patients with FXS., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
38. Ultrastructural analysis of the functional domains in FMRP using primary hippocampal mouse neurons.
- Author
-
Levenga J, Buijsen RA, Rifé M, Moine H, Nelson DL, Oostra BA, Willemsen R, and de Vrij FM
- Subjects
- Animals, Cells, Cultured, Dendrites ultrastructure, Fragile X Mental Retardation Protein chemistry, Fragile X Mental Retardation Protein genetics, Hippocampus cytology, Immunohistochemistry, In Situ Hybridization, Mice, Mice, Knockout, Microscopy, Confocal, Mutation, Neurons cytology, Nucleic Acid Conformation, Protein Isoforms chemistry, Protein Isoforms metabolism, Protein Structure, Tertiary, RNA, Messenger metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Transfection, Dendrites metabolism, Fragile X Mental Retardation Protein metabolism, Hippocampus metabolism, Neurons metabolism, RNA Transport
- Abstract
Fragile X syndrome is caused by lack of the protein FMRP. FMRP mediates mRNA binding, dendritic mRNA transport and translational control at spines. We examined the role of functional domains of FMRP in neuronal RNA-granule formation and dendritic transport using different FMRP variants, including the mutant FMRP_I304N and the splice-variant FMRP_Iso12. Both variants are absent from dendritic RNA-granules in Fmr1 knockout neurons. Co-transfection experiments showed that wild-type FMRP recruits both FMRP variants into dendritic RNA-granules. Co-transfection of FXR2, an FMRP homologue, also resulted in redistribution of both variants into dendritic RNA-granules. Furthermore, the capacity of the variants to transport their mRNAs and the mRNA localization of an FMR1 construct containing silent point-mutations affecting only the G-quartet-structure were investigated. In conclusion, we show that wild-type FMRP and FXR2P are able to recruit FMRP variants into RNA-granules and that the G-quartet-structure in FMR1 mRNA is not essential for its incorporation in RNA-granules.
- Published
- 2009
- Full Text
- View/download PDF
39. Genes and pathways differentially expressed in the brains of Fxr2 knockout mice.
- Author
-
Cavallaro S, Paratore S, Fradale F, de Vrij FM, Willemsen R, and Oostra BA
- Subjects
- Animals, Blotting, Western, Brain pathology, Cerebellum metabolism, Cerebellum pathology, Down-Regulation, Gene Expression Profiling, Hippocampus metabolism, Hippocampus pathology, Immunohistochemistry, In Situ Hybridization, Mice, Mice, Knockout, Oligonucleotide Array Sequence Analysis, RNA-Binding Proteins genetics, Receptors, AMPA genetics, Reverse Transcriptase Polymerase Chain Reaction, Brain metabolism, Gene Expression Regulation, RNA-Binding Proteins physiology
- Abstract
Fragile X syndrome is a common inherited form of mental retardation and originates from the absence of expression of the FMR1 gene. This gene and its two homologues, FXR1 and FXR2, encode for a family of fragile X related (FXR) proteins with similar tissue distribution, together with sequence and functional homology. Based on these characteristics, it has been suggested that these proteins might partly complement one another. To unravel the function of Fxr2 protein, the expression pattern of 12,588 genes was studied in the brains of wild-type and Fxr2 knockout mice, an animal model which shows behavioral abnormalities partly similar to those observed in Fmr1-knockout mice. By genome expression profiling and stringent significance tests we identify genes and gene groups de-regulated in the brains of Fxr2 knockout mice. Differential expression of candidate genes was validated by real-time PCR, in situ hybridization, immunohistochemistry and western blot analysis. A number of differentially expressed genes associated with the Fxr2 phenotype have been previously involved in other memory or cognitive disorders.
- Published
- 2008
- Full Text
- View/download PDF
40. Rescue of behavioral phenotype and neuronal protrusion morphology in Fmr1 KO mice.
- Author
-
de Vrij FM, Levenga J, van der Linde HC, Koekkoek SK, De Zeeuw CI, Nelson DL, Oostra BA, and Willemsen R
- Subjects
- Animals, Cells, Cultured, Fragile X Syndrome pathology, Hippocampus drug effects, Hippocampus pathology, Imidazoles pharmacology, Mice, Mice, Knockout, Microscopy, Confocal, Neurons drug effects, Phenotype, Receptor, Metabotropic Glutamate 5, Receptors, Metabotropic Glutamate antagonists & inhibitors, Reflex, Startle drug effects, Behavior, Animal drug effects, Excitatory Amino Acid Antagonists pharmacology, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome drug therapy, Neurons pathology, Pyridines pharmacology
- Abstract
Lack of fragile X mental retardation protein (FMRP) causes Fragile X Syndrome, the most common form of inherited mental retardation. FMRP is an RNA-binding protein and is a component of messenger ribonucleoprotein complexes, associated with brain polyribosomes, including dendritic polysomes. FMRP is therefore thought to be involved in translational control of specific mRNAs at synaptic sites. In mice lacking FMRP, protein synthesis-dependent synaptic plasticity is altered and structural malformations of dendritic protrusions occur. One hypothesized cause of the disease mechanism is based on exaggerated group I mGluR receptor activation. In this study, we examined the effect of the mGluR5 antagonist MPEP on Fragile X related behavior in Fmr1 KO mice. Our results demonstrate a clear defect in prepulse inhibition of startle in Fmr1 KO mice, that could be rescued by MPEP. Moreover, we show for the first time a structural rescue of Fragile X related protrusion morphology with two independent mGluR5 antagonists.
- Published
- 2008
- Full Text
- View/download PDF
41. Mutant ubiquitin found in neurodegenerative disorders is a ubiquitin fusion degradation substrate that blocks proteasomal degradation.
- Author
-
Lindsten K, de Vrij FM, Verhoef LG, Fischer DF, van Leeuwen FW, Hol EM, Masucci MG, and Dantuma NP
- Subjects
- Cell Cycle, Green Fluorescent Proteins, HeLa Cells, Humans, Luminescent Proteins genetics, Lysine metabolism, Multienzyme Complexes antagonists & inhibitors, Neurodegenerative Diseases enzymology, Neurodegenerative Diseases genetics, Proteasome Endopeptidase Complex, Proteins metabolism, Substrate Specificity, Tumor Cells, Cultured, Ubiquitin antagonists & inhibitors, Ubiquitin metabolism, Cysteine Endopeptidases metabolism, Multienzyme Complexes metabolism, Mutation, Neurodegenerative Diseases metabolism, Neurons metabolism, Ubiquitin genetics
- Abstract
Loss of neurons in neurodegenerative diseases is usually preceded by the accumulation of protein deposits that contain components of the ubiquitin/proteasome system. Affected neurons in Alzheimer's disease often accumulate UBB(+1), a mutant ubiquitin carrying a 19-amino acid C-terminal extension generated by a transcriptional dinucleotide deletion. Here we show that UBB(+1) is a potent inhibitor of ubiquitin-dependent proteolysis in neuronal cells, and that this inhibitory activity correlates with induction of cell cycle arrest. Surprisingly, UBB(+1) is recognized as a ubiquitin fusion degradation (UFD) proteasome substrate and ubiquitinated at Lys29 and Lys48. Full blockade of proteolysis requires both ubiquitination sites. Moreover, the inhibitory effect was enhanced by the introduction of multiple UFD signals. Our findings suggest that the inhibitory activity of UBB(+1) may be an important determinant of neurotoxicity and contribute to an environment that favors the accumulation of misfolded proteins.
- 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.