129 results on '"Benedikt Berninger"'
Search Results
2. Corrigendum: Enhanced proliferation of oligodendrocyte progenitor cells following retrovirus mediated Achaete-scute complex-like 1 overexpression in the postnatal cerebral cortex in vivo
- Author
-
Chiara Galante, Nicolás Marichal, Franciele Franco Scarante, Litsa Maria Ghayad, Youran Shi, Carol Schuurmans, Benedikt Berninger, and Sophie Péron
- Subjects
astrocyte ,gliogenesis ,lineage reprogramming ,neurogenesis ,proliferation ,proneural ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2023
- Full Text
- View/download PDF
3. Enhanced proliferation of oligodendrocyte progenitor cells following retrovirus mediated Achaete-scute complex-like 1 overexpression in the postnatal cerebral cortex in vivo
- Author
-
Chiara Galante, Nicolás Marichal, Franciele Franco Scarante, Litsa Maria Ghayad, Youran Shi, Carol Schuurmans, Benedikt Berninger, and Sophie Péron
- Subjects
astrocyte ,gliogenesis ,lineage reprogramming ,neurogenesis ,proliferation ,proneural ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The proneural transcription factor Achaete-scute complex-like 1 (Ascl1) is a major regulator of neural fate decisions, implicated both in neurogenesis and oligodendrogliogenesis. Focusing on its neurogenic activity, Ascl1 has been widely used to reprogram non-neuronal cells into induced neurons. In vitro, Ascl1 induces efficient reprogramming of proliferative astroglia from the early postnatal cerebral cortex into interneuron-like cells. Here, we examined whether Ascl1 can similarly induce neuronal reprogramming of glia undergoing proliferation in the postnatal mouse cerebral cortex in vivo. Toward this goal, we targeted cortical glia during the peak of proliferative expansion (i.e., postnatal day 5) by injecting a retrovirus encoding for Ascl1 into the mouse cerebral cortex. In contrast to the efficient reprogramming observed in vitro, in vivo Ascl1-transduced glial cells were converted into doublecortin-immunoreactive neurons only with very low efficiency. However, we noted a drastic shift in the relative number of retrovirus-transduced Sox10-positive oligodendrocyte progenitor cells (OPCs) as compared to glial fibrillary acidic protein (GFAP)-positive astrocytes. Genetic fate mapping demonstrated that this increase in OPCs was not due to Ascl1-mediated astrocyte-to-OPC fate conversion. Rather, EdU incorporation experiments revealed that Ascl1 caused a selective increase in proliferative activity of OPCs, but not astrocytes. Our data indicate that rather than inducing neuronal reprogramming of glia in the early postnatal cortex, Ascl1 is a selective enhancer of OPC proliferation.
- Published
- 2022
- Full Text
- View/download PDF
4. Extensive transcriptional and chromatin changes underlie astrocyte maturation in vivo and in culture
- Author
-
Michael Lattke, Robert Goldstone, James K. Ellis, Stefan Boeing, Jerónimo Jurado-Arjona, Nicolás Marichal, James I. MacRae, Benedikt Berninger, and Francois Guillemot
- Subjects
Science - Abstract
Astrocytes have functions crucial for brain homeostasis, which are disrupted in many neurological disorders, but how these functions are established during astrocyte maturation is largely unknown. Here the authors show transcriptional and chromatin changes underlying astrocyte maturation in mice and identify transcription factors regulating maturation of cultured astrocytes.
- Published
- 2021
- Full Text
- View/download PDF
5. Gatekeeping astrocyte identity
- Author
-
Alexis Cooper and Benedikt Berninger
- Subjects
PTBP1 ,astrocyte-to-neuron conversion ,lineage reprogramming ,parkinson's disease ,brain repair ,astrocyte ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
New findings cast doubt on whether suppressing the RNA-binding protein PTBP1 can force astrocytes to become dopaminergic neurons.
- Published
- 2022
- Full Text
- View/download PDF
6. Bifunctional Hydrogels Containing the Laminin Motif IKVAV Promote Neurogenesis
- Author
-
Aleeza Farrukh, Felipe Ortega, Wenqiang Fan, Nicolás Marichal, Julieta I. Paez, Benedikt Berninger, Aránzazu del Campo, and Marcelo J. Salierno
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Engineering of biomaterials with specific biological properties has gained momentum as a means to control stem cell behavior. Here, we address the effect of bifunctionalized hydrogels comprising polylysine (PL) and a 19-mer peptide containing the laminin motif IKVAV (IKVAV) on embryonic and adult neuronal progenitor cells under different stiffness regimes. Neuronal differentiation of embryonic and adult neural progenitors was accelerated by adjusting the gel stiffness to 2 kPa and 20 kPa, respectively. While gels containing IKVAV or PL alone failed to support long-term cell adhesion, in bifunctional gels, IKVAV synergized with PL to promote differentiation and formation of focal adhesions containing β1-integrin in embryonic cortical neurons. Furthermore, in adult neural stem cell culture, bifunctionalized gels promoted neurogenesis via the expansion of neurogenic clones. These data highlight the potential of synthetic matrices to steer stem and progenitor cell behavior via defined mechano-adhesive properties. : In this article, Farrukh and colleagues show that bifunctionalization of hydrogel substrates with polylysine and a 19-mer peptide containing the laminin motif IKVAV promotes neurogenesis from embryonic neuroblasts and adult neural stem cells. Neurogenesis and neurite outgrowth can be further optimized by adjusting gel stiffness in a cell-type-specific manner. Keywords: biomaterials, bioengineering, hydrogels, neural stem cells, cell differentiation, laminin, IKVAV, polylysine, β1-integrin, neurogenesis
- Published
- 2017
- Full Text
- View/download PDF
7. Neurovascular EGFL7 regulates adult neurogenesis in the subventricular zone and thereby affects olfactory perception
- Author
-
Frank Bicker, Verica Vasic, Guilherme Horta, Felipe Ortega, Hendrik Nolte, Atria Kavyanifar, Stefanie Keller, Nevenka Dudvarski Stankovic, Patrick N. Harter, Rui Benedito, Beat Lutz, Tobias Bäuerle, Jens Hartwig, Jan Baumgart, Marcus Krüger, Konstantin Radyushkin, Lavinia Alberi, Benedikt Berninger, and Mirko H. H. Schmidt
- Subjects
Science - Abstract
The vascular stem cell niche regulates the proliferation and differentiation of neural stem cells (NSCs) in the adult subventricular zone. Here the authors identify EGFL7 as a neurovascular regulator of NSCsin vivo; EGFL7-knockout mice show reduced neurogenesis, and exhibit impaired olfactory perception and behaviour.
- Published
- 2017
- Full Text
- View/download PDF
8. CEND1 and NEUROGENIN2 Reprogram Mouse Astrocytes and Embryonic Fibroblasts to Induced Neural Precursors and Differentiated Neurons
- Author
-
Katerina Aravantinou-Fatorou, Felipe Ortega, Dafni Chroni-Tzartou, Nasia Antoniou, Cornelia Poulopoulou, Panagiotis K. Politis, Benedikt Berninger, Rebecca Matsas, and Dimitra Thomaidou
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Recent studies demonstrate that astroglia from non-neurogenic brain regions can be reprogrammed into functional neurons through forced expression of neurogenic factors. Here we explored the effect of CEND1 and NEUROG2 on reprogramming of mouse cortical astrocytes and embryonic fibroblasts. Forced expression of CEND1, NEUROG2, or both resulted in acquisition of induced neuronal cells expressing subtype-specific markers, while long-term live-cell imaging highlighted the existence of two different modes of neuronal trans-differentiation. Of note, a subpopulation of CEND1 and NEUROG2 double-transduced astrocytes formed spheres exhibiting neural stem cell properties. mRNA and protein expression studies revealed a reciprocal feedback loop existing between the two molecules, while knockdown of endogenous CEND1 demonstrated that it is a key mediator of NEUROG2-driven neuronal reprogramming. Our data suggest that common reprogramming mechanisms exist driving the conversion of lineage-distant somatic cell types to neurons and reveal a critical role for CEND1 in NEUROG2-driven astrocytic reprogramming.
- Published
- 2015
- Full Text
- View/download PDF
9. Ascl1 Coordinately Regulates Gene Expression and the Chromatin Landscape during Neurogenesis
- Author
-
Alexandre A.S.F. Raposo, Francisca F. Vasconcelos, Daniela Drechsel, Corentine Marie, Caroline Johnston, Dirk Dolle, Angela Bithell, Sébastien Gillotin, Debbie L.C. van den Berg, Laurence Ettwiller, Paul Flicek, Gregory E. Crawford, Carlos M. Parras, Benedikt Berninger, Noel J. Buckley, François Guillemot, and Diogo S. Castro
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The proneural transcription factor Ascl1 coordinates gene expression in both proliferating and differentiating progenitors along the neuronal lineage. Here, we used a cellular model of neurogenesis to investigate how Ascl1 interacts with the chromatin landscape to regulate gene expression when promoting neuronal differentiation. We find that Ascl1 binding occurs mostly at distal enhancers and is associated with activation of gene transcription. Surprisingly, the accessibility of Ascl1 to its binding sites in neural stem/progenitor cells remains largely unchanged throughout their differentiation, as Ascl1 targets regions of both readily accessible and closed chromatin in proliferating cells. Moreover, binding of Ascl1 often precedes an increase in chromatin accessibility and the appearance of new regions of open chromatin, associated with de novo gene expression during differentiation. Our results reveal a function of Ascl1 in promoting chromatin accessibility during neurogenesis, linking the chromatin landscape at Ascl1 target regions with the temporal progression of its transcriptional program.
- Published
- 2015
- Full Text
- View/download PDF
10. Pharmacogenomic identification of small molecules for lineage specific manipulation of subventricular zone germinal activity.
- Author
-
Kasum Azim, Diane Angonin, Guillaume Marcy, Francesca Pieropan, Andrea Rivera, Vanessa Donega, Claudio Cantù, Gareth Williams, Benedikt Berninger, Arthur M Butt, and Olivier Raineteau
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Strategies for promoting neural regeneration are hindered by the difficulty of manipulating desired neural fates in the brain without complex genetic methods. The subventricular zone (SVZ) is the largest germinal zone of the forebrain and is responsible for the lifelong generation of interneuron subtypes and oligodendrocytes. Here, we have performed a bioinformatics analysis of the transcriptome of dorsal and lateral SVZ in early postnatal mice, including neural stem cells (NSCs) and their immediate progenies, which generate distinct neural lineages. We identified multiple signaling pathways that trigger distinct downstream transcriptional networks to regulate the diversity of neural cells originating from the SVZ. Next, we used a novel in silico genomic analysis, searchable platform-independent expression database/connectivity map (SPIED/CMAP), to generate a catalogue of small molecules that can be used to manipulate SVZ microdomain-specific lineages. Finally, we demonstrate that compounds identified in this analysis promote the generation of specific cell lineages from NSCs in vivo, during postnatal life and adulthood, as well as in regenerative contexts. This study unravels new strategies for using small bioactive molecules to direct germinal activity in the SVZ, which has therapeutic potential in neurodegenerative diseases.
- Published
- 2017
- Full Text
- View/download PDF
11. Sox2-Mediated Conversion of NG2 Glia into Induced Neurons in the Injured Adult Cerebral Cortex
- Author
-
Christophe Heinrich, Matteo Bergami, Sergio Gascón, Alexandra Lepier, Francesca Viganò, Leda Dimou, Bernd Sutor, Benedikt Berninger, and Magdalena Götz
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
The adult cerebral cortex lacks the capacity to replace degenerated neurons following traumatic injury. Conversion of nonneuronal cells into induced neurons has been proposed as an innovative strategy toward brain repair. Here, we show that retrovirus-mediated expression of the transcription factors Sox2 and Ascl1, but strikingly also Sox2 alone, can induce the conversion of genetically fate-mapped NG2 glia into induced doublecortin (DCX)+ neurons in the adult mouse cerebral cortex following stab wound injury in vivo. In contrast, lentiviral expression of Sox2 in the unlesioned cortex failed to convert oligodendroglial and astroglial cells into DCX+ cells. Neurons induced following injury mature morphologically and some acquire NeuN while losing DCX. Patch-clamp recording of slices containing Sox2- and/or Ascl1-transduced cells revealed that a substantial fraction of these cells receive synaptic inputs from neurons neighboring the injury site. Thus, NG2 glia represent a potential target for reprogramming strategies toward cortical repair.
- Published
- 2014
- Full Text
- View/download PDF
12. Cultured subventricular zone progenitor cells transduced with neurogenin-2 become mature glutamatergic neurons and integrate into the dentate gyrus.
- Author
-
Xia Chen, Alexandra Lepier, Benedikt Berninger, Aviva M Tolkovsky, and Joe Herbert
- Subjects
Medicine ,Science - Abstract
We have previously shown that transplantation of immature DCX+/NeuN+/Prox1+ neurons (found in the neonatal DG), but not undifferentiated neuronal progenitor cells (NPCs) from ventral subventricular zone (SVZ), results in neuronal maturation in vivo within the dentate niche. Here we investigated whether we could enhance the integration of SVZ NPCs by forced expression of the proneural gene Neurogenin 2 (NEUROG2). NPCs cultured from neonatal GFP-transgenic rat SVZ for 7 days in a non-differentiating medium were transduced with a retrovirus encoding NEUROG2 and DsRed or the DsRed reporter gene alone (control). By 3 days post-transduction, the NEUROG2-transduced cells maintained in culture contained mostly immature neurons (91% DCX+; 76% NeuN+), whereas the control virus-transduced cells remained largely undifferentiated (30% DCX+;
- Published
- 2012
- Full Text
- View/download PDF
13. Directing astroglia from the cerebral cortex into subtype specific functional neurons.
- Author
-
Christophe Heinrich, Robert Blum, Sergio Gascón, Giacomo Masserdotti, Pratibha Tripathi, Rodrigo Sánchez, Steffen Tiedt, Timm Schroeder, Magdalena Götz, and Benedikt Berninger
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Astroglia from the postnatal cerebral cortex can be reprogrammed in vitro to generate neurons following forced expression of neurogenic transcription factors, thus opening new avenues towards a potential use of endogenous astroglia for brain repair. However, in previous attempts astroglia-derived neurons failed to establish functional synapses, a severe limitation towards functional neurogenesis. It remained therefore also unknown whether neurons derived from reprogrammed astroglia could be directed towards distinct neuronal subtype identities by selective expression of distinct neurogenic fate determinants. Here we show that strong and persistent expression of neurogenic fate determinants driven by silencing-resistant retroviral vectors instructs astroglia from the postnatal cortex in vitro to mature into fully functional, synapse-forming neurons. Importantly, the neurotransmitter fate choice of astroglia-derived neurons can be controlled by selective expression of distinct neurogenic transcription factors: forced expression of the dorsal telencephalic fate determinant neurogenin-2 (Neurog2) directs cortical astroglia to generate synapse-forming glutamatergic neurons; in contrast, the ventral telencephalic fate determinant Dlx2 induces a GABAergic identity, although the overall efficiency of Dlx2-mediated neuronal reprogramming is much lower compared to Neurog2, suggesting that cortical astroglia possess a higher competence to respond to the dorsal telencephalic fate determinant. Interestingly, however, reprogramming of astroglia towards the generation of GABAergic neurons was greatly facilitated when the astroglial cells were first expanded as neurosphere cells prior to transduction with Dlx2. Importantly, this approach of expansion under neurosphere conditions and subsequent reprogramming with distinct neurogenic transcription factors can also be extended to reactive astroglia isolated from the adult injured cerebral cortex, allowing for the selective generation of glutamatergic or GABAergic neurons. These data provide evidence that cortical astroglia can undergo a conversion across cell lineages by forced expression of a single neurogenic transcription factor, stably generating fully differentiated neurons. Moreover, neuronal reprogramming of astroglia is not restricted to postnatal stages but can also be achieved from terminally differentiated astroglia of the adult cerebral cortex following injury-induced reactivation.
- Published
- 2010
- Full Text
- View/download PDF
14. The transcriptional co‐activator Yap1 promotes adult hippocampal neural stem cell activation
- Author
-
Wenqiang Fan, Jerónimo Jurado‐Arjona, Gregorio Alanis‐Lobato, Sophie Péron, Christian Berger, Miguel A Andrade‐Navarro, Sven Falk, and Benedikt Berninger
- Subjects
General Immunology and Microbiology ,General Neuroscience ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
15. Tuning the neurogenesis channel
- Author
-
Nicolás Marichal and Benedikt Berninger
- Subjects
Diazepam Binding Inhibitor ,Neurons ,Diazepam ,General Neuroscience ,Neurogenesis ,Receptors, GABA-A ,gamma-Aminobutyric Acid - Abstract
Earlier work has implicated the neurotransmitter GABA in controlling forebrain progenitor proliferation. In this issue of Neuron, Everlien et al. (2022) demonstrate that diazepam binding inhibitor acts to keep the neurogenesis-promoting effect of GABA at bay.
- Published
- 2022
16. cAAVe phaenomena: Beware of appearances!
- Author
-
Filippo Calzolari and Benedikt Berninger
- Subjects
medicine.anatomical_structure ,nervous system ,viruses ,Cell ,medicine ,Endogeny ,Biology ,Neuroscience ,General Biochemistry, Genetics and Molecular Biology ,Brain repair - Abstract
In this issue of Cell, Wang et al. come to the unsettling conclusion that adeno-associated viruses, despite being engineered for glia-specific expression, can become widely active in endogenous neurons, misleading researchers in their quest for efficient conversion of glia into neurons for brain repair.
- Published
- 2021
- Full Text
- View/download PDF
17. Editorial overview: Fluidity of cell fates – from reprogramming to repair
- Author
-
Benedikt Berninger, Stefan H. Stricker, and Magdalena Götz
- Subjects
2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Cell ,Biology ,Cellular Reprogramming ,Models, Biological ,Virology ,medicine.anatomical_structure ,Genetics ,medicine ,Animals ,Humans ,Cell Lineage ,Reprogramming ,Developmental Biology - Published
- 2021
- Full Text
- View/download PDF
18. Low-efficiency conversion of proliferative glia into induced neurons by Ascl1 in the postnatal mouse cerebral cortex in vivo
- Author
-
Chiara Galante, Nicolás Marichal, Carol Schuurmans, Benedikt Berninger, and Sophie Péron
- Subjects
nervous system - Abstract
The proneural transcription factor Achaete-scute complex-like 1 (Ascl1) is a major regulator of neural progenitor fate, implicated both in neurogenesis and oligodendrogliogenesis. Ascl1 has been widely used to reprogram non-neuronal cells into induced neurons. In vitro, Ascl1 induces efficient reprogramming of proliferative astroglia from the early postnatal cerebral cortex into interneuron-like cells. Here, we examined whether Ascl1 can similarly induce neuronal reprogramming of glia undergoing proliferation in the postnatal mouse cerebral cortex in vivo. Toward this, we targeted cortical glia at the peak of proliferative expansion (i.e., postnatal day 5) by injecting a retrovirus encoding for Ascl1 into the mouse cerebral cortex. In sharp contrast to the very efficient reprogramming in vitro, Ascl1-transduced glial cells were converted into doublecortin-immunoreactive neurons only with low efficiency in vivo. Interfering with the phosphorylation of Ascl1 by mutation of six conserved proline-directed serine/threonine phosphorylation sites (Ascl1SA6) has been previously shown to increase its neurogenic activity in the early embryonic cerebral cortex. We therefore tested whether transduction of proliferative glia with a retrovirus encoding Ascl1SA6 improved their conversion into neurons. While in vitro glia-to-neuron conversion was markedly enhanced, in vivo reprogramming efficiency remained low. However, both wild-type and mutant Ascl1 reduced the relative number of cells expressing the astrocytic marker glial fibrillary acidic protein (GFAP) and increased the relative number of cells expressing the oligodendroglial marker Sox10 in vivo. Together, our results indicate that the enhanced neurogenic response of proliferative postnatal glia to Ascl1SA6 versus Ascl1 observed in vitro is not recapitulated in vivo.
- Published
- 2022
- Full Text
- View/download PDF
19. Reprogramming cellular identity in vivo
- Author
-
Sydney Leaman, Nicolás Marichal, and Benedikt Berninger
- Subjects
Neurons ,Genetic Vectors ,Animals ,Cell Lineage ,Dependovirus ,Cellular Reprogramming ,Regenerative Medicine ,Neuroglia ,Molecular Biology ,Cell Proliferation ,Developmental Biology - Abstract
Cellular identity is established through complex layers of genetic regulation, forged over a developmental lifetime. An expanding molecular toolbox is allowing us to manipulate these gene regulatory networks in specific cell types in vivo. In principle, if we found the right molecular tricks, we could rewrite cell identity and harness the rich repertoire of possible cellular functions and attributes. Recent work suggests that this rewriting of cell identity is not only possible, but that newly induced cells can mitigate disease phenotypes in animal models of major human diseases. So, is the sky the limit, or do we need to keep our feet on the ground? This Spotlight synthesises key concepts emerging from recent efforts to reprogramme cellular identity in vivo. We provide our perspectives on recent controversies in the field of glia-to-neuron reprogramming and identify important gaps in our understanding that present barriers to progress.
- Published
- 2022
- Full Text
- View/download PDF
20. Role of Yap1 in adult neural stem cell activation
- Author
-
Wenqiang Fan, Jerónimo Jurado-Arjona, Gregorio Alanis-Lobato, Sophie Péron, Christian Berger, Miguel A. Andrade-Navarro, Sven Falk, and Benedikt Berninger
- Subjects
nervous system ,reproductive and urinary physiology ,nervous system diseases - Abstract
Most adult hippocampal neural stem cells (NSCs) remain quiescent with only a minor portion undergoing active proliferation and neurogenesis. The molecular mechanisms that trigger eventually the transition from quiescence to activation are still poorly understood. Here, we found the activity of the transcriptional activator Yap1 to be enriched in active NSCs. Genetic deletion of Yap1 led to a significant reduction in the relative proportion of active NSCs supporting a physiological role of Yap1 in regulating the transition from quiescence to activation. Overexpression of wild type Yap1 in adult NSCs did not induce NSC activation suggesting tight upstream control mechanisms, but overexpression of a gain-of-function mutant (Yap1-5SA) elicited cell cycle entry in NSCs and hilar astrocytes. Consistent with a role of Yap1 in NSC activation, single cell RNA sequencing revealed the partial induction of an activated NSC gene expression program. Yet, Yap1-5SA expression also induced Taz and other key components of the Yap/Taz regulon previously identified in glioblastoma stem cell-like cells. Consequently, dysregulated Yap1 activity led to repression of hippocampal neurogenesis, promoting aberrant differentiation instead.
- Published
- 2022
- Full Text
- View/download PDF
21. Decision letter: Repressing PTBP1 fails to convert reactive astrocytes to dopaminergic neurons in a 6-hydroxydopamine mouse model of Parkinson’s disease
- Author
-
Benedikt Berninger
- Published
- 2021
- Full Text
- View/download PDF
22. Reprogramming reactive glia into interneurons reduces chronic seizure activity in a mouse model of mesial temporal lobe epilepsy
- Author
-
Karl-Klaus Conzelmann, Nicolás Marichal, Charlotte Verrier, Olivier Raineteau, Rory Vignoles, Sylvie Rival-Gervier, Antoine Depaulis, Louis Foucault, Christophe Heinrich, Marie-Madeleine Trottmann, Marie d’Orange, Célia Lentini, Benedikt Berninger, Céline Massera, Institut cellule souche et cerveau / Stem Cell and Brain Research Institute (U1208 Inserm - UCBL1 / SBRI - USC 1361 INRAE), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), King‘s College London, Dynamique et Structure du Cytosquelette Neuronal, [GIN] Grenoble Institut des Neurosciences (GIN), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Grenoble Alpes (UGA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Grenoble Alpes (UGA), Ludwig-Maximilians University [Munich] (LMU), Institut cellule souche et cerveau / Stem Cell and Brain Research Institute (SBRI), Institute of Psychiatry, Psychology & Neuroscience, King's College London, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Grenoble Alpes (UGA), Max Von Pettenkofer Institute (MVP), Ludwig-Maximilians-Universität München (LMU), University Medical Center of the Johannes Gutenberg-University Mainz, This work was supported by ANR ReprogramEpi (ANR-14-CE13-0001), LabEx CORTEX (ANR-11-LABX-0042) of Lyon University (ANR-11-IDEX-0007), FRC, FFRE, and CURE (Award ID: 262178) to C.H., Wellcome Trust (206410/Z/17/Z) and DFG (BE 4182/8-1 and CRC1080) to B.B., DFG (ID 118803580 and SFB 870 Z1) to K.-K.C., and European Community’s Framework Program Neurinox (FP7, 278611) to A.D. R.V., C.L., and N.M. were supported by fellowships from Région Rhône-Alpes (R.V., ARC2 16-005489-01), LFCE (C.L.), and Human Frontier Science Program (N.M., LT000646/2015)., ANR-14-CE13-0001,ReprogramEpi,La reprogrammation des astrocytes réactionnels en neurones GABAergiques: une nouvelle approche thérapeutique de l'épilepsie(2014), ANR-11-LABX-0042,CORTEX,Construction, Fonction Cognitive et Réhabilitation du Cerveau(2011), ANR-11-IDEX-0007,Avenir L.S.E.,PROJET AVENIR LYON SAINT-ETIENNE(2011), European Project: 278611,EC:FP7:HEALTH,FP7-HEALTH-2011-two-stage,NEURINOX(2012), heinrich, christophe, Appel à projets générique - La reprogrammation des astrocytes réactionnels en neurones GABAergiques: une nouvelle approche thérapeutique de l'épilepsie - - ReprogramEpi2014 - ANR-14-CE13-0001 - Appel à projets générique - VALID, Construction, Fonction Cognitive et Réhabilitation du Cerveau - - CORTEX2011 - ANR-11-LABX-0042 - LABX - VALID, PROJET AVENIR LYON SAINT-ETIENNE - - Avenir L.S.E.2011 - ANR-11-IDEX-0007 - IDEX - VALID, NOX enzymes as mediators of inflammation-triggered neurodegeneration: modulating NOX enzymes as novel therapies - NEURINOX - - EC:FP7:HEALTH2012-01-01 - 2016-12-31 - 278611 - VALID, Institut cellule souche et cerveau (U846 Inserm - UCBL1), Institut National de la Recherche Agronomique (INRA)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
regeneration and repair in the nervous system ,[SDV]Life Sciences [q-bio] ,Hippocampus ,regenerative medicine ,Context (language use) ,Hippocampal formation ,Biology ,glia-to-neuron conversion ,Article ,Mice ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Interneurons ,Seizures ,Genetics ,medicine ,Animals ,Humans ,GABAergic Neurons ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,0303 health sciences ,therapy-resistant epilepsy ,DLX2 ,Cell Biology ,medicine.disease ,Hand ,gene therapy ,[SDV] Life Sciences [q-bio] ,ASCL1 ,Epilepsy, Temporal Lobe ,nervous system ,direct lineage reprogramming ,Molecular Medicine ,GABAergic ,Neuroglia ,Neuroscience ,Reprogramming ,030217 neurology & neurosurgery - Abstract
Summary Reprogramming brain-resident glial cells into clinically relevant induced neurons (iNs) is an emerging strategy toward replacing lost neurons and restoring lost brain functions. A fundamental question is now whether iNs can promote functional recovery in pathological contexts. We addressed this question in the context of therapy-resistant mesial temporal lobe epilepsy (MTLE), which is associated with hippocampal seizures and degeneration of hippocampal GABAergic interneurons. Using a MTLE mouse model, we show that retrovirus-driven expression of Ascl1 and Dlx2 in reactive hippocampal glia in situ, or in cortical astroglia grafted in the epileptic hippocampus, causes efficient reprogramming into iNs exhibiting hallmarks of interneurons. These induced interneurons functionally integrate into epileptic networks and establish GABAergic synapses onto dentate granule cells. MTLE mice with GABAergic iNs show a significant reduction in both the number and cumulative duration of spontaneous recurrent hippocampal seizures. Thus glia-to-neuron reprogramming is a potential disease-modifying strategy to reduce seizures in therapy-resistant epilepsy., Graphical abstract, Highlights • Retroviruses target reactive hippocampal glia proliferating in a mouse model of mesial temporal lobe epilepsy • Ascl1 and Dlx2 reprogram reactive glia into GABAergic interneurons in the epileptic hippocampus • Induced interneurons establish GABAergic synapses onto dentate granule cells • Induced interneurons reduce chronic epileptic activity in the hippocampus, Mesial temporal lobe epilepsy belongs to treatment-refractory forms of human epilepsy. Lentini et al. show that reactive glia proliferating in the epileptic hippocampus can be reprogrammed into GABAergic induced neurons that reduce chronic seizure activity. This study uncovers glia-to-neuron reprogramming as a potential disease-modifying strategy to reduce intractable seizures.
- Published
- 2021
- Full Text
- View/download PDF
23. Brain-derived neurotrophic factor expression in serotonergic neurons improves stress resilience and promotes adult hippocampal neurogenesis
- Author
-
Julia Leschik, Antonietta Gentile, Cigdem Cicek, Sophie Péron, Margaryta Tevosian, Annika Beer, Konstantin Radyushkin, Anna Bludau, Karl Ebner, Inga Neumann, Nicolas Singewald, Benedikt Berninger, Volkmar Lessmann, and Beat Lutz
- Subjects
Mice ,Brain-Derived Neurotrophic Factor ,Fluoxetine ,Neurogenesis ,General Neuroscience ,Animals ,Mice, Transgenic ,Hippocampus ,Antidepressive Agents ,Serotonergic Neurons - Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) stimulates adult neurogenesis, but also influences structural plasticity and function of serotonergic neurons. Both, BDNF/TrkB signaling and the serotonergic system modulate behavioral responses to stress and can lead to pathological states when dysregulated. The two systems have been shown to mediate the therapeutic effect of antidepressant drugs and to regulate hippocampal neurogenesis. To elucidate the interplay of both systems at cellular and behavioral levels, we generated a transgenic mouse line that overexpresses BDNF in serotonergic neurons in an inducible manner. Besides displaying enhanced hippocampus-dependent contextual learning, transgenic mice were less affected by chronic social defeat stress (CSDS) compared to wild-type animals. In parallel, we observed enhanced serotonergic axonal sprouting in the dentate gyrus and increased neural stem/progenitor cell proliferation, which was uniformly distributed along the dorsoventral axis of the hippocampus. In the forced swim test, BDNF-overexpressing mice behaved similarly as wild-type mice treated with the antidepressant fluoxetine. Our data suggest that BDNF released from serotonergic projections exerts this effect partly by enhancing adult neurogenesis. Furthermore, independently of the genotype, enhanced neurogenesis positively correlated with the social interaction time after the CSDS, a measure for stress resilience.
- Published
- 2022
- Full Text
- View/download PDF
24. Decision letter: Transcriptional regulation of neural stem cell expansion in the adult hippocampus
- Author
-
Benedikt Berninger
- Published
- 2021
- Full Text
- View/download PDF
25. Direct In Vitro Reprogramming of Astrocytes into Induced Neurons
- Author
-
Nesrin, Sharif, Filippo, Calzolari, and Benedikt, Berninger
- Subjects
Neurons ,Mice ,Astrocytes ,Primary Cell Culture ,Animals ,Cell Differentiation ,Neocortex ,Cell Separation ,Cellular Reprogramming ,Neuroglia ,Cells, Cultured ,Transcription Factors - Abstract
Spontaneous neuronal replacement is almost absent in the postnatal mammalian nervous system. However, several studies have shown that both early postnatal and adult astroglia can be reprogrammed in vitro or in vivo by forced expression of proneural transcription factors, such as Neurogenin-2 or Achaete-scute homolog 1 (Ascl1), to acquire a neuronal fate. The reprogramming process stably induces properties such as distinctly neuronal morphology, expression of neuron-specific proteins, and the gain of mature neuronal functional features. Direct conversion of astroglia into neurons thus possesses potential as a basis for cell-based strategies against neurological diseases. In this chapter, we describe a well-established protocol used for direct reprogramming of postnatal cortical astrocytes into functional neurons in vitro and discuss available tools and approaches to dissect molecular and cell biological mechanisms underlying the reprogramming process.
- Published
- 2021
26. Astrocytes and neurons share region-specific transcriptional signatures that confer regional identity to neuronal reprogramming
- Author
-
María Figueres-Oñate, José P. López-Atalaya, Benedikt Berninger, Laura López-Mascaraque, Alejandro Sempere-Ferràndez, Ana Espinosa, Guillermina López-Bendito, Lorenzo Puche-Aroca, Laia Torres-Masjoan, Eduardo Leyva-Díaz, Alvaro Herrero-Navarro, Rafael Susín, Verónica Moreno-Juan, Marisa Karow, Ministerio de Ciencia, Innovación y Universidades (España), European Commission, German Research Foundation, and European Research Council
- Subjects
Epigenomics ,Cell ,Thalamus ,Neocortex ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,medicine ,Epigenetics ,Progenitor cell ,Neural cell ,Research Articles ,030304 developmental biology ,Neurons ,0303 health sciences ,Multidisciplinary ,SciAdv r-articles ,medicine.anatomical_structure ,nervous system ,Astrocytes ,Neuroscience ,Reprogramming ,030217 neurology & neurosurgery ,Research Article - Abstract
Region-specific gene expression shared with neurons imparts to astrocytes competence for region-specific neuronal reprogramming., Neural cell diversity is essential to endow distinct brain regions with specific functions. During development, progenitors within these regions are characterized by specific gene expression programs, contributing to the generation of diversity in postmitotic neurons and astrocytes. While the region-specific molecular diversity of neurons and astrocytes is increasingly understood, whether these cells share region-specific programs remains unknown. Here, we show that in the neocortex and thalamus, neurons and astrocytes express shared region-specific transcriptional and epigenetic signatures. These signatures not only distinguish cells across these two brain regions but are also detected across substructures within regions, such as distinct thalamic nuclei, where clonal analysis reveals the existence of common nucleus-specific progenitors for neurons and astrocytes. Consistent with their shared molecular signature, regional specificity is maintained following astrocyte-to-neuron reprogramming. A detailed understanding of these regional-specific signatures may thus inform strategies for future cell-based brain repair.
- Published
- 2021
27. Direct In Vitro Reprogramming of Astrocytes into Induced Neurons
- Author
-
Benedikt Berninger, Filippo Calzolari, and Nesrin Sharif
- Subjects
Mammalian nervous system ,ASCL1 ,medicine.anatomical_structure ,nervous system ,In vivo ,Functional features ,Cell ,medicine ,Biology ,Transcription factor ,Reprogramming ,In vitro ,Cell biology - Abstract
Spontaneous neuronal replacement is almost absent in the postnatal mammalian nervous system. However, several studies have shown that both early postnatal and adult astroglia can be reprogrammed in vitro or in vivo by forced expression of proneural transcription factors, such as Neurogenin-2 or Achaete-scute homolog 1 (Ascl1), to acquire a neuronal fate. The reprogramming process stably induces properties such as distinctly neuronal morphology, expression of neuron-specific proteins, and the gain of mature neuronal functional features. Direct conversion of astroglia into neurons thus possesses potential as a basis for cell-based strategies against neurological diseases. In this chapter, we describe a well-established protocol used for direct reprogramming of postnatal cortical astrocytes into functional neurons in vitro and discuss available tools and approaches to dissect molecular and cell biological mechanisms underlying the reprogramming process.
- Published
- 2021
- Full Text
- View/download PDF
28. Extensive transcriptional and chromatin changes underlie astrocyte maturation in vivo and in culture
- Author
-
François Guillemot, Nicolás Marichal, James I. MacRae, Benedikt Berninger, Stefan Boeing, Michael Lattke, James K. Ellis, Jerónimo Jurado-Arjona, and Robert J. Goldstone
- Subjects
0301 basic medicine ,Male ,Molecular biology ,Cell Culture Techniques ,General Physics and Astronomy ,Gene Expression ,Epigenesis, Genetic ,0302 clinical medicine ,Chemical Biology & High Throughput ,Cerebral Cortex ,Multidisciplinary ,Stem Cells ,Genome Integrity & Repair ,Cell Differentiation ,Chromatin ,Cell biology ,medicine.anatomical_structure ,Chromatin Immunoprecipitation Sequencing ,Single-Cell Analysis ,Genetics & Genomics ,Astrocyte ,Model organisms ,Science ,Biology ,Biochemistry & Proteomics ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cellular neuroscience ,Developmental biology ,medicine ,Animals ,Gene ,Transcription factor ,Computational & Systems Biology ,FOS: Clinical medicine ,Neurosciences ,Development of the nervous system ,General Chemistry ,Cell Biology ,Tumour Biology ,In vitro ,Mice, Inbred C57BL ,030104 developmental biology ,Astrocytes ,030217 neurology & neurosurgery ,Homeostasis ,Transcription Factors ,Neuroscience - Abstract
Astrocytes have essential functions in brain homeostasis that are established late in differentiation, but the mechanisms underlying the functional maturation of astrocytes are not well understood. Here we identify extensive transcriptional changes that occur during murine astrocyte maturation in vivo that are accompanied by chromatin remodelling at enhancer elements. Investigating astrocyte maturation in a cell culture model revealed that in vitro-differentiated astrocytes lack expression of many mature astrocyte-specific genes, including genes for the transcription factors Rorb, Dbx2, Lhx2 and Fezf2. Forced expression of these factors in vitro induces distinct sets of mature astrocyte-specific transcripts. Culturing astrocytes in a three-dimensional matrix containing FGF2 induces expression of Rorb, Dbx2 and Lhx2 and improves astrocyte maturity based on transcriptional and chromatin profiles. Therefore, extrinsic signals orchestrate the expression of multiple intrinsic regulators, which in turn induce in a modular manner the transcriptional and chromatin changes underlying astrocyte maturation., Astrocytes have functions crucial for brain homeostasis, which are disrupted in many neurological disorders, but how these functions are established during astrocyte maturation is largely unknown. Here the authors show transcriptional and chromatin changes underlying astrocyte maturation in mice and identify transcription factors regulating maturation of cultured astrocytes.
- Published
- 2020
29. Astrocytes and neurons share brain region-specific transcriptional signatures
- Author
-
Verónica Moreno-Juan, Laia Torres-Masjoan, Marisa Karow, Eduardo Leyva-Díaz, Alvaro Herrero-Navarro, José P. López-Atalaya, Laura López-Mascaraque, Alejandro Sempere-Ferràndez, Rafael Susín, Guillermina López-Bendito, Ana Espinosa, Benedikt Berninger, Lorenzo Puche-Aroca, and María Figueres-Oñate
- Subjects
0303 health sciences ,Neocortex ,Cell ,Thalamus ,Biology ,03 medical and health sciences ,Brain region ,0302 clinical medicine ,medicine.anatomical_structure ,nervous system ,Gene expression ,medicine ,Epigenetics ,Progenitor cell ,Reprogramming ,Neuroscience ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
SUMMARYNeuronal cell diversity is essential to endow distinct brain regions with specific functions. During development, progenitors within these regions are characterised by specific gene expression programs, contributing to the generation of diversity in postmitotic neurons and glia. While the region-specific molecular diversity of neurons and astrocytes is increasingly understood, whether these cells share region-specific programs remains unknown. Here, we show that in the neocortex and thalamus, neurons and astrocytes express shared region-specific transcriptional and epigenetic signatures. These signatures not only distinguish cells across brain regions but are also detected across substructures within regions, such as distinct thalamic nuclei, where clonal analysis revealed the existence of common nucleus-specific progenitors for neurons and glia. Consistent with their shared molecular signature, regional specificity was maintained following astrocyte-to-neuron reprogramming. A detailed understanding of these regional-specific signatures may thus inform strategies for future cell-based brain repair.
- Published
- 2020
- Full Text
- View/download PDF
30. Human SPG11 cerebral organoids reveal cortical neurogenesis impairment
- Author
-
Wenqiang Fan, Francesc Pérez-Brangulí, Isabel Y. Buchsbaum, Silvia Cappello, Daniela Gräf, Annika Schray, Zacharias Kohl, Jürgen Winkler, Tom Börstler, Tatyana Pozner, Benedikt Berninger, Martin Regensburger, Beate Winner, and Himanshu K. Mishra
- Subjects
Genotype ,Hereditary spastic paraplegia ,Neurogenesis ,Fluorescent Antibody Technique ,Biology ,03 medical and health sciences ,Glycogen Synthase Kinase 3 ,Genetics ,Organoid ,medicine ,Spastic ,Humans ,Molecular Biology ,Genetics (clinical) ,Alleles ,beta Catenin ,Cerebral Cortex ,0303 health sciences ,030305 genetics & heredity ,Proteins ,General Medicine ,Human brain ,medicine.disease ,Neural stem cell ,nervous system diseases ,Organoids ,medicine.anatomical_structure ,Phenotype ,Mutation ,General Article ,Disease Susceptibility ,Paraplegia ,Cognition Disorders ,Neuroscience ,Neural development ,Biomarkers - Abstract
Spastic paraplegia gene 11(SPG11)-linked hereditary spastic paraplegia is a complex monogenic neurodegenerative disease that in addition to spastic paraplegia is characterized by childhood onset cognitive impairment, thin corpus callosum and enlarged ventricles. We have previously shown impaired proliferation of SPG11 neural progenitor cells (NPCs). For the delineation of potential defect in SPG11 brain development we employ 2D culture systems and 3D human brain organoids derived from SPG11 patients’ iPSC and controls. We reveal that an increased rate of asymmetric divisions of NPCs leads to proliferation defect, causing premature neurogenesis. Correspondingly, SPG11 organoids appeared smaller than controls and had larger ventricles as well as thinner germinal wall. Premature neurogenesis and organoid size were rescued by GSK3 inhibititors including the Food and Drug Administration-approved tideglusib. These findings shed light on the neurodevelopmental mechanisms underlying disease pathology.
- Published
- 2018
- Full Text
- View/download PDF
31. Changing the Face of Modern Medicine: Stem Cells and Gene Therapy Abstract Author Index
- Author
-
Vijay K. Tiwari, Barbara Treutlein, J. Gray Camp, Benedikt Berninger, Christian Schichor, Abhijeet Pataskar, and Marisa Karow
- Subjects
ASCL1 ,medicine.anatomical_structure ,SOX2 ,Genetics ,medicine ,Molecular Medicine ,GABAergic ,Pericyte ,Human brain ,Biology ,Molecular Biology ,Reprogramming ,Cell biology - Published
- 2016
- Full Text
- View/download PDF
32. TOX3 regulates neural progenitor identity
- Author
-
Vijay K. Tiwari, Pablo Bora, Benedikt Berninger, Alireza Pouya, Jan Baumgart, Teresa Schacht, Sanjeeb Kumar Sahu, Zsuzsa Kovacs, Verena Wüllner, Alina Fritz, Neha Tiwari, Axel Methner, and Sophie Péron
- Subjects
0301 basic medicine ,Neurogenesis ,Biophysics ,Notch signaling pathway ,Subventricular zone ,Mice, Transgenic ,Biology ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,SOX2 ,Pregnancy ,Structural Biology ,Genetics ,medicine ,Animals ,RNA, Small Interfering ,Progenitor cell ,Molecular Biology ,Cells, Cultured ,reproductive and urinary physiology ,Neurons ,Gene Expression Regulation, Developmental ,Nestin ,Embryo, Mammalian ,Molecular biology ,Neural stem cell ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,embryonic structures ,Trans-Activators ,Female ,Stem cell ,Apoptosis Regulatory Proteins ,Receptors, Progesterone ,030217 neurology & neurosurgery - Abstract
The human genomic locus for the transcription factor TOX3 has been implicated in susceptibility to restless legs syndrome and breast cancer in genome-wide association studies, but the physiological role of TOX3 remains largely unknown. We found Tox3 to be predominantly expressed in the developing mouse brain with a peak at embryonic day E14 where it co-localizes with the neural stem and progenitor markers Nestin and Sox2 in radial glia of the ventricular zone and intermediate progenitors of the subventricular zone. Tox3 is also expressed in neural progenitor cells obtained from the ganglionic eminence of E15 mice that express Nestin, and it specifically binds the Nestin promoter in chromatin immunoprecipitation assays. In line with this, over-expression of Tox3 increased Nestin promoter activity, which was cooperatively enhanced by treatment with the stem cell self-renewal promoting Notch ligand Jagged and repressed by pharmacological inhibition of Notch signaling. Knockdown of Tox3 in the subventricular zone of E12.5 mouse embryos by in utero electroporation of Tox3 shRNA revealed a reduced Nestin expression and decreased proliferation at E14 and a reduced migration to the cortical plate in E16 embryos in electroporated cells. Together, these results argue for a role of Tox3 in the development of the nervous system.
- Published
- 2016
- Full Text
- View/download PDF
33. Prox1 Is Required for Oligodendrocyte Cell Identity in Adult Neural Stem Cells of the Subventricular Zone
- Author
-
Shima Safaiyan, Athanasios Stergiopoulos, Laura Gonzalez Cano, Jens Christian Schwamborn, Gökhan Ertaylan, Sandra Völs, Felipe Ortega, Panagiotis K. Politis, Mikael Simons, Marianne van Cann, Benedikt Berninger, Maria Angeliki S. Pavlou, Antonio del Sol, Eva C. Bunk, Sciences, RS: FSE MaCSBio, RS: FPN MaCSBio, and Maastricht Centre for Systems Biology
- Subjects
0301 basic medicine ,Adult neurogenesis ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Cell Movement ,Lateral Ventricles ,Promoter Regions, Genetic ,Cells, Cultured ,MOUSE-BRAIN ,Receptors, Notch ,Oligodendrocytes ,Neurogenesis ,Cell Differentiation ,LINEAGE ,Anatomy ,Olfactory Bulb ,Neural stem cell ,Cell biology ,Neuroepithelial cell ,Adult Stem Cells ,Oligodendroglia ,DIFFERENTIATION ,Enhancer Elements, Genetic ,medicine.anatomical_structure ,Gene Knockdown Techniques ,Molecular Medicine ,SPINAL-CORD ,Stem cell ,SUBCELLULAR-LOCALIZATION ,Protein Binding ,Adult stem cell ,OLIG2 ,Subventricular zone ,Biology ,03 medical and health sciences ,Neurosphere ,Prox1 ,medicine ,Animals ,Cell Lineage ,OLFACTORY-BULB ,Body Patterning ,Homeodomain Proteins ,Tumor Suppressor Proteins ,Cell Biology ,MAMMALIAN BRAIN ,Oligodendrocyte Transcription Factor 2 ,030104 developmental biology ,Neuropoiesis ,PROGENITOR CELLS ,Gene Expression Regulation ,nervous system ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Adult neural stem cells with the ability to generate neurons and glia cells are active throughout life in both the dentate gyrus (DG) and the subventricular zone (SVZ). Differentiation of adult neural stem cells is induced by cell fate determinants like the transcription factor Prox1. Evidence has been provided for a function of Prox1 as an inducer of neuronal differentiation within the DG. We now show that within the SVZ Prox1 induces differentiation into oligodendrocytes. Moreover, we find that loss of Prox1 expression in vivo reduces cell migration into the corpus callosum, where the few Prox1 deficient SVZ-derived remaining cells fail to differentiate into oligodendrocytes. Thus, our work uncovers a novel function of Prox1 as a fate determinant for oligodendrocytes in the adult mammalian brain. These data indicate that the neurogenic and oligodendrogliogenic lineages in the two adult neurogenic niches exhibit a distinct requirement for Prox1, being important for neurogenesis in the DG but being indispensable for oligodendrogliogenesis in the SVZ.
- Published
- 2016
- Full Text
- View/download PDF
34. Identification and Successful Negotiation of a Metabolic Checkpoint in Direct Neuronal Reprogramming
- Author
-
Susan Gascon, Magdalena Götz, Martin Irmler, Felipe Ortega, David Petrik, Stephen P. Robertson, Johannes Beckers, Gianluca Luigi Russo, Aditi Deshpande, Marisa Karow, Carsten Berndt, Marcus Conrad, Giacomo Masserdotti, Timm Schroeder, Benedikt Berninger, José Pedro Friedmann Angeli, Christophe Heinrich, and Elisa Murenu
- Subjects
0301 basic medicine ,Genetics ,Programmed cell death ,Cell type ,Cellular Reprogramming Techniques ,Mutant ,Cell Biology ,Biology ,In vitro ,Cell biology ,03 medical and health sciences ,Transduction (genetics) ,030104 developmental biology ,In vivo ,Molecular Medicine ,Reprogramming - Abstract
Despite the widespread interest in direct neuronal reprogramming, the mechanisms underpinning fate conversion remain largely unknown. Our study revealed a critical time point after which cells either successfully convert into neurons or succumb to cell death. Co-transduction with Bcl-2 greatly improved negotiation of this critical point by faster neuronal differentiation. Surprisingly, mutants with reduced or no affinity for Bax demonstrated that Bcl-2 exerts this effect by an apoptosis-independent mechanism. Consistent with a caspase-independentrole, ferroptosis inhibitors potently increased neuronal reprogramming by inhibiting lipid peroxidation occurring during fate conversion. Genome-wide expression analysis confirmed that treatments promoting neuronal reprogramming elicit an anti-oxidative stress response. Importantly, co-expression of Bcl-2 and anti-oxidative treatments leads to an unprecedented improvement in glial-to-neuron conversion after traumatic brain injury invivo, underscoring the relevance of these pathways in cellular reprograming irrespective of cell type invitro and invivo.
- Published
- 2016
- Full Text
- View/download PDF
35. An increase in neural stem cells and olfactory bulb adult neurogenesis improves discrimination of highly similar odorants
- Author
-
Federico Calegari, Benedikt Berninger, Simone Massalini, Sara Bragado Alonso, Nicolás Marichal, Janine K. Reinert, and Thomas Kuner
- Subjects
Male ,Neurogenesis ,Subventricular zone ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Discrimination Learning ,Mice ,Neural Stem Cells ,odor discrimination ,medicine ,Animals ,Cyclin D1 ,News & Views ,Molecular Biology ,neural stem cells ,General Immunology and Microbiology ,General Neuroscience ,Cyclin-Dependent Kinase 4 ,Cell cycle ,Olfactory Bulb ,Neural stem cell ,Olfactory bulb ,adult neurogenesis ,Disease Models, Animal ,Electrophysiology ,medicine.anatomical_structure ,nervous system ,Odor ,Odorants ,Neuroscience ,Adult stem cell - Abstract
Adult neurogenesis is involved in cognitive performance but studies that manipulated this process to improve brain function are scarce. Here, we characterized a genetic mouse model in which neural stem cells (NSC) of the subventricular zone (SVZ) were temporarily expanded by conditional expression of the cell cycle regulators Cdk4/cyclinD1, thus increasing neurogenesis. We found that supernumerary neurons matured and integrated in the olfactory bulb similarly to physiologically generated newborn neurons displaying a correct expression of molecular markers, morphology and electrophysiological activity. Olfactory performance upon increased neurogenesis was unchanged when mice were tested on relatively easy tasks using distinct odor stimuli. In contrast, intriguingly, increasing neurogenesis improved the discrimination ability of mice when challenged with a difficult task using mixtures of highly similar odorants. Together, our study provides a mammalian model to control the expansion of somatic stem cells that can in principle be applied to any tissue for basic research and models of therapy. By applying this to NSC of the SVZ, we highlighted the importance of adult neurogenesis to specifically improve performance in a challenging olfactory task.
- Published
- 2019
- Full Text
- View/download PDF
36. Quiescence Modulates Stem Cell Maintenance and Regenerative Capacity in the Aging Brain
- Author
-
Srikanth Ravichandran, Peter Schmezer, Wenqiang Fan, Jan Bolz, Benedikt Berninger, Enric Llorens-Bobadilla, Georgios Kalamakis, Antonio del Sol, Birgit Berger, Sheng Zhao, Janina Kupke, Jan-Philipp Mallm, Frederik Ziebell, Katharina Bauer, Urs Christen, Ana Martin-Villalba, Simon Anders, Daniel Brüne, Thomas Stiehl, Stefanie Limpert, Anna Marciniak-Czochra, and Francisco Catalá-Martinez
- Subjects
Male ,Neurogenesis ,Subventricular zone ,Inflammation ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neural Stem Cells ,medicine ,Aging brain ,sFRP5 ,stem cell aging ,Animals ,Homeostasis ,quiescence ,Stem Cell Niche ,reproductive and urinary physiology ,Cellular Senescence ,030304 developmental biology ,neural stem cells ,Cell Proliferation ,0303 health sciences ,Wnt signaling pathway ,Age Factors ,subventricular zone ,Brain ,modeling ,Cell Differentiation ,interferon ,Wnt signaling ,Neural stem cell ,Cell biology ,nervous system diseases ,Nerve Regeneration ,Mice, Inbred C57BL ,medicine.anatomical_structure ,nervous system ,inflammation ,simulations ,medicine.symptom ,Stem cell ,biological phenomena, cell phenomena, and immunity ,single-cell transcriptomics ,030217 neurology & neurosurgery ,Cell Division ,Adult stem cell - Abstract
The function of somatic stem cells declines with age. Understanding the molecular underpinnings of this decline is key to counteract age-related disease. Here, we report a dramatic drop in the neural stem cells (NSCs) number in the aging murine brain. We find that this smaller stem cell reservoir is protected from full depletion by an increase in quiescence that makes old NSCs more resistant to regenerate the injured brain. Once activated, however, young and old NSCs show similar proliferation and differentiation capacity. Single-cell transcriptomics of NSCs indicate that aging changes NSCs minimally. In the aging brain, niche-derived inflammatory signals and the Wnt antagonist sFRP5 induce quiescence. Indeed, intervention to neutralize them increases activation of old NSCs during homeostasis and following injury. Our study identifies quiescence as a key feature of old NSCs imposed by the niche and uncovers ways to activate NSCs to repair the aging brain.
- Published
- 2018
- Full Text
- View/download PDF
37. Direct pericyte-to-neuron reprogramming via unfolding of a neural stem cell-like program
- Author
-
Therese Riedemann, Sven Falk, Barbara Treutlein, J. Gray Camp, Tobias Gerber, Andrej Smiyakin, Magdalena Götz, Christian Schichor, Angela Garding, Agnieska Brazovskaja, Vijay K. Tiwari, Abhijeet Pataskar, Wenqiang Fan, Benedikt Berninger, Malgorzata Gac-Santel, Marisa Karow, Jorge Kageyama, Antonella Casamassa, Karow, Marisa, Camp, J Gray, Falk, Sven, Gerber, Tobia, Pataskar, Abhijeet, Gac-Santel, Malgorzata, Kageyama, Jorge, Brazovskaja, Agnieska, Garding, Angela, Fan, Wenqiang, Riedemann, Therese, Casamassa, Antonella, Smiyakin, Andrej, Schichor, Christian, Götz, Magdalena, Tiwari, Vijay K, Treutlein, Barbara, and Berninger, Benedikt
- Subjects
Adult ,Male ,0301 basic medicine ,Somatic cell ,Cellular differentiation ,Basic Helix-Loop-Helix Transcription Factor ,SOXB1 Transcription Factor ,Biology ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,SOX2 ,Basic Helix-Loop-Helix Transcription Factors ,Humans ,Cell Lineage ,Neural Stem Cell ,Aged ,Pericyte ,Neurons ,SOXB1 Transcription Factors ,General Neuroscience ,Cell Differentiation ,Middle Aged ,Neuron ,Cellular Reprogramming ,Neural stem cell ,ASCL1 ,030104 developmental biology ,Gene Expression Regulation ,Female ,Ectopic expression ,Pericytes ,Neural development ,Reprogramming ,Neuroscience ,030217 neurology & neurosurgery ,Human - Abstract
Ectopic expression of defined transcription factors can force direct cell-fate conversion from one lineage to another in the absence of cell division. Several transcription factor cocktails have enabled successful reprogramming of various somatic cell types into induced neurons (iNs) of distinct neurotransmitter phenotype. However, the nature of the intermediate states that drive the reprogramming trajectory toward distinct iN types is largely unknown. Here we show that successful direct reprogramming of adult human brain pericytes into functional iNs by Ascl1 and Sox2 encompasses transient activation of a neural stem cell-like gene expression program that precedes bifurcation into distinct neuronal lineages. During this transient state, key signaling components relevant for neural induction and neural stem cell maintenance are regulated by and functionally contribute to iN reprogramming and maturation. Thus, Ascl1- and Sox2-mediated reprogramming into a broad spectrum of iN types involves the unfolding of a developmental program via neural stem cell-like intermediates. ISSN:1097-6256 ISSN:1546-1726
- Published
- 2018
- Full Text
- View/download PDF
38. Reawakening the sleeping beauty in the adult brain: neurogenesis from parenchymal glia
- Author
-
Benedikt Berninger and Sophie Péron
- Subjects
Neurogenesis ,Striatum ,Biology ,Parenchyma ,Genetics ,medicine ,Animals ,Humans ,Regeneration ,Neurons ,Regulation of gene expression ,Regeneration (biology) ,Gene Expression Regulation, Developmental ,Anatomy ,Cellular Reprogramming ,medicine.anatomical_structure ,nervous system ,Cerebral cortex ,Astrocytes ,Brain Injuries ,Neuroglia ,Neuroscience ,Reprogramming ,Developmental Biology - Abstract
Life-long neurogenesis is highly restricted to specialized niches in the adult mammalian brain and therefore the brain's capacity for spontaneous regeneration is extremely limited. However, recent work has demonstrated that under certain circumstances parenchymal astrocytes and NG2 glia can generate neuronal progeny. In the striatum, stroke or excitotoxic lesions can reawaken in astrocytes a latent neurogenic program resulting in the genesis of new neurons. By contrast, in brain areas that fail to mount a neurogenic response following injury, such as the cerebral cortex, forced expression of neurogenic reprogramming factors can lineage convert local glia into induced neurons. Yet, injury-induced and reprogramming-induced neurogenesis exhibit intriguing commonalities, suggesting that they may converge on similar mechanisms.
- Published
- 2015
- Full Text
- View/download PDF
39. <scp>JNK</scp> ‐dependent gene regulatory circuitry governs mesenchymal fate
- Author
-
Vijay K. Tiwari, Marcus Schmidt, Joern Toedling, Neha Tiwari, Nikolai Schmarowski, Felipe Ortega, Angela Garding, Susanne Gebhard, Robert Nitsch, Benedikt Berninger, Sudhir Thakurela, and Sanjeeb Kumar Sahu
- Subjects
MAP Kinase Kinase 4 ,MAP Kinase Signaling System ,Cellular differentiation ,Gene regulatory network ,Biology ,Time-Lapse Imaging ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mesoderm ,Transcriptome ,transcription factors ,metastasis ,Humans ,Gene Regulatory Networks ,Epithelial–mesenchymal transition ,Molecular Biology ,Transcription factor ,JNK signaling ,Genetics ,Regulation of gene expression ,General Immunology and Microbiology ,Gene Expression Profiling ,General Neuroscience ,Cell Cycle ,EMT ,Cell Differentiation ,Articles ,3. Good health ,Chromatin ,Cell biology ,embryonic structures ,gene regulation ,Reprogramming - Abstract
The epithelial to mesenchymal transition (EMT) is a biological process in which cells lose cell-cell contacts and become motile. EMT is used during development, for example, in triggering neural crest migration, and in cancer metastasis. Despite progress, the dynamics of JNK signaling, its role in genomewide transcriptional reprogramming, and involved downstream effectors during EMT remain largely unknown. Here, we show that JNK is not required for initiation, but progression of phenotypic changes associated with EMT. Such dependency resulted from JNK-driven transcriptional reprogramming of critical EMT genes and involved changes in their chromatin state. Furthermore, we identified eight novel JNK-induced transcription factors that were required for proper EMT. Three of these factors were also highly expressed in invasive cancer cells where they function in gene regulation to maintain mesenchymal identity. These factors were also induced during neuronal development and function in neuronal migration in vivo. These comprehensive findings uncovered a kinetically distinct role for the JNK pathway in defining the transcriptome that underlies mesenchymal identity and revealed novel transcription factors that mediate these responses during development and disease.
- Published
- 2015
- Full Text
- View/download PDF
40. Transcriptional Mechanisms of Proneural Factors and REST in Regulating Neuronal Reprogramming of Astrocytes
- Author
-
Benedikt Berninger, Sébastien Gillotin, Magdalena Götz, Helle F. Jørgensen, Johannes Beckers, Bernd Sutor, Giacomo Masserdotti, Daniela Drechsel, Martin Irmler, Steffen Sass, François Guillemot, Fabian J. Theis, and Apollo - University of Cambridge Repository
- Subjects
Transcription, Genetic ,Repressor ,Nerve Tissue Proteins ,Cell fate determination ,Biology ,DNA-binding protein ,Article ,Mice ,Glutamatergic ,Basic Helix-Loop-Helix Transcription Factors ,Genetics ,Animals ,Humans ,Promoter Regions, Genetic ,Transcription factor ,Cells, Cultured ,Neurons ,Cell Biology ,Cellular Reprogramming ,Molecular biology ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,ASCL1 ,Astrocytes ,embryonic structures ,Molecular Medicine ,GABAergic ,Reprogramming ,Transcription Factors - Abstract
Summary Direct lineage reprogramming induces dramatic shifts in cellular identity, employing poorly understood mechanisms. Recently, we demonstrated that expression of Neurog2 or Ascl1 in postnatal mouse astrocytes generates glutamatergic or GABAergic neurons. Here, we take advantage of this model to study dynamics of neuronal cell fate acquisition at the transcriptional level. We found that Neurog2 and Ascl1 rapidly elicited distinct neurogenic programs with only a small subset of shared target genes. Within this subset, only NeuroD4 could by itself induce neuronal reprogramming in both mouse and human astrocytes, while co-expression with Insm1 was required for glutamatergic maturation. Cultured astrocytes gradually became refractory to reprogramming, in part by the repressor REST preventing Neurog2 from binding to the NeuroD4 promoter. Notably, in astrocytes refractory to Neurog2 activation, the underlying neurogenic program remained amenable to reprogramming by exogenous NeuroD4. Our findings support a model of temporal hierarchy for cell fate change during neuronal reprogramming., Graphical Abstract, Highlights • Neurog2 and Ascl1 regulate largely non-overlapping neurogenic targets • A subset of genes is required and sufficient to convert astrocytes and fibroblasts • Neurog2 and REST compete for binding to the NeuroD4 promoter • REST deletion enhances Neurog2-mediated reprogramming in vitro, Masserdotti et al. analyzed early transcriptional changes mediated by Neurog2 and Ascl1 during direct reprogramming of murine postnatal astrocytes into distinct neuronal subtypes in vitro. This led to the identification of shared downstream targets, including NeuroD4, capable of neuronal reprogramming of fibroblasts and human astrocytes, as well as mechanistic insight into how the repressor REST functions as a barrier in direct neuronal reprogramming.
- Published
- 2015
- Full Text
- View/download PDF
41. Programming of neural progenitors of the adult subependymal zone towards a glutamatergic identity by Neurogenin2
- Author
-
Miyakoshi Lm, Susan Gascon, Monika S. Brill, Marisa Karow, Felipe Ortega, Benedikt Berninger, and Sophie Péron
- Subjects
medicine.medical_specialty ,education.field_of_study ,biology ,Rostral migratory stream ,Neurogenesis ,Population ,Neural stem cell ,Glutamatergic ,medicine.anatomical_structure ,Endocrinology ,Internal medicine ,embryonic structures ,medicine ,biology.protein ,Subependymal zone ,TBR1 ,Neuron ,education ,Neuroscience - Abstract
While the adult subependymal zone (SEZ) harbors pools of distinct neural stem cells that generate different types of GABAergic interneurons, a small progenitor population in the dorsal SEZ expresses Neurog2 and gives rise to glutamatergic neurons. Here we investigated whether SEZ progenitors can be programmed towards glutamatergic neurogenesis through forced expression of Neurog2. Retrovirus-mediated expression of Neurog2 induced the glutamatergic neuron lineage markers Tbr2 and Tbr1 in cultured SEZ progenitors which subsequently differentiated into functional glutamatergic neurons. Likewise, retrovirus-mediated expression of Neurog2 in dividing SEZ progenitors within the adult SEZ induced Tbr2 and Tbr1 expression, hallmarking entry into the glutamatergic lineage also in vivo. Intriguingly, Neurog2-expressing progenitors failed to enter the rostral migratory stream (RMS) and instead differentiated directly within the SEZ or the adjacent striatum. In sharp contrast, lentivirus-mediated postmitotic expression of Neurog2 failed to reprogram early SEZ neurons, which instead maintained their GABAergic identity and migrated along the RMS towards the olfactory bulb. Thus, our data show that Neurog2 can program SEZ progenitors towards a glutamatergic identity, but fails to reprogram their postmitotic progeny.Summary statementOur study identifies a critical developmental time window during which progenitors of the adult subependymal zone, specified for generating GABAergic neurons, can be reprogrammed towards glutamatergic neurogenesis.
- Published
- 2017
- Full Text
- View/download PDF
42. Stage-Specific Transcription Factors Drive Astrogliogenesis by Remodeling Gene Regulatory Landscapes
- Author
-
Neha, Tiwari, Abhijeet, Pataskar, Sophie, Péron, Sudhir, Thakurela, Sanjeeb Kumar, Sahu, María, Figueres-Oñate, Nicolás, Marichal, Laura, López-Mascaraque, Vijay K, Tiwari, and Benedikt, Berninger
- Subjects
Male ,cell fate ,Activating Transcription Factor 3 ,Neurogenesis ,epigenetic mechanisms ,Core Binding Factor Alpha 1 Subunit ,Article ,Mice, Inbred C57BL ,astrogliogenesis ,Mice ,NFI Transcription Factors ,astrocyte ,Gene Expression Regulation ,transcription factors ,Animals ,gene regulation ,Cells, Cultured ,neural stem cells - Abstract
Summary A broad molecular framework of how neural stem cells are specified toward astrocyte fate during brain development has proven elusive. Here we perform comprehensive and integrated transcriptomic and epigenomic analyses to delineate gene regulatory programs that drive the developmental trajectory from mouse embryonic stem cells to astrocytes. We report molecularly distinct phases of astrogliogenesis that exhibit stage- and lineage-specific transcriptomic and epigenetic signatures with unique primed and active chromatin regions, thereby revealing regulatory elements and transcriptional programs underlying astrocyte generation and maturation. By searching for transcription factors that function at these elements, we identified NFIA and ATF3 as drivers of astrocyte differentiation from neural precursor cells while RUNX2 promotes astrocyte maturation. These transcription factors facilitate stage-specific gene expression programs by switching the chromatin state of their target regulatory elements from primed to active. Altogether, these findings provide integrated insights into the genetic and epigenetic mechanisms steering the trajectory of astrogliogenesis., Graphical Abstract, Highlights • Distinct transcriptional programs define consecutive stages of astrogliogenesis • Stage- and lineage-specific enhancers promote the distinct gene expression programs • ATF3 and NFIA drive astrocyte differentiation, while RUNX2 promotes astrocyte maturation • These TFs switch the enhancers from a primed to an active chromatin state, Neural stem cells give rise to both neurons and glia. Modeling gliogenesis in vitro, Tiwari et al. show that the generation of astrocytes involves several transcriptionally and epigenetically distinct stages. The authors found that the transcription factors NFIA, ATF3, and RUNX2 play pivotal roles in establishing these stages.
- Published
- 2017
43. Interplay between a Mental Disorder Risk Gene and Developmental Polarity Switch of GABA Action Leads to Excitation-Inhibition Imbalance
- Author
-
Benedikt Berninger, Guo Li Ming, Jennifer H. Lee, Jaesuk Park, Qassim Hussani, Shaoyu Ge, Yu-Ting Lin, Hongjun Song, Kuei Sen Hsu, Weidong Li, Juan Song, Yan Gu, Kimberly M. Christian, and Eunchai Kang
- Subjects
Male ,0301 basic medicine ,Neurogenesis ,Mutant ,Nerve Tissue Proteins ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,DISC1 ,Glutamatergic ,0302 clinical medicine ,Risk Factors ,mental disorders ,Animals ,Genetic Predisposition to Disease ,GABAergic Neurons ,gamma-Aminobutyric Acid ,Neurons ,Gene knockdown ,biology ,Mental Disorders ,Cell Polarity ,Neural Inhibition ,Depolarization ,Synaptic Potentials ,Mice, Inbred C57BL ,030104 developmental biology ,nervous system ,Gene Knockdown Techniques ,Synapses ,biology.protein ,Excitatory postsynaptic potential ,GABAergic ,Female ,Neuroscience ,030217 neurology & neurosurgery ,Parvalbumin - Abstract
Excitation-inhibition (E-I) imbalance is considered a hallmark of various neurodevelopmental disorders, including schizophrenia and autism. How genetic risk factors disrupt coordinated glutamatergic and GABAergic synapse formation to cause an E-I imbalance is not well understood. Here, we show that knockdown of Disrupted-in-schizophrenia 1 (DISC1), a risk gene for major mental disorders, leads to E-I imbalance in mature dentate granule neurons. We found that excessive GABAergic inputs from parvalbumin-, but not somatostatin-, expressing interneurons enhance the formation of both glutamatergic and GABAergic synapses in immature mutant neurons. Following the switch in GABAergic signaling polarity from depolarizing to hyperpolarizing during neuronal maturation, heightened inhibition from excessive parvalbumin(+) GABAergic inputs causes loss of excitatory glutamatergic synapses in mature mutant neurons, resulting in an E-I imbalance. Our findings provide insights into the developmental role of depolarizing GABA in establishing E-I balance and how it can be influenced by genetic risk factors for mental disorders.
- Published
- 2019
- Full Text
- View/download PDF
44. The art of forging neurons: direct reprogramming of somatic cells into induced neuronal cells
- Author
-
Marisa Karow and Benedikt Berninger
- Subjects
Induced stem cells ,medicine.anatomical_structure ,Somatic cell ,Cellular differentiation ,Transdifferentiation ,Cell ,medicine ,Biology ,Neuroscience ,Reprogramming ,Neural cell ,Neural stem cell - Abstract
Cellular reprogramming has shed new light on the plasticity of terminally differentiated cells and unearthed novel strategies for cell-based therapies to treat neurological disorders. With accumulating knowledge of the programs underlying the genesis of the distinct neural cell types, particularly the identification of crucial transcription factors and microRNAs, reprogramming of somatic cells of different origins into induced neuronal cells or neural stem cells has been successfully achieved. Starting with the general concept of reprogramming, we discuss three different paradigms: (1) direct conversion of central nervous system (CNS) foreign cells such as skin fibroblasts into induced neuronal cells or neural stem cells; (2) transdifferentiation of CNS resident cells such as astrocytes and brain pericytes into induced neuronal cells; (3) reprogramming of one neuronal subtype into another. The latter has already been successfully achieved in vivo during early brain development, providing a strong impulse to attempt direct reprogramming in situ for future brain repair.
- Published
- 2013
- Full Text
- View/download PDF
45. Die Kunst des Neuronenschmiedens: Direkte Reprogrammierung somatischer Zellen in induzierte neuronale Zellen
- Author
-
Benedikt Berninger and Marisa Karow
- Subjects
Chemistry ,Transdifferentiation ,Molecular biology - Abstract
The art of forging neurons: direct reprogramming of somatic cells into induced neuronal cells. Cellular reprogramming has shed new light on the plasticity of terminally differentiated cells and discloses novel strategies for cell-based therapies for neurological disorders. With accumulating knowledge of the programs underlying the genesis of the distinct neural cell types, especially with the identification of relevant transcription factors and microRNAs, reprogramming of somatic cells of different origins into induced neuronal cells or neural stem cells has been successfully achieved. Starting with the general concept of reprogramming we discuss here three different paradigms: 1) direct conversion of CNS-foreign cells such as skin fibroblasts into induced neuronal cells or neural stem cells; 2) transdifferentiation of CNS resident cells such as astrocytes and brain pericytes into induced neuronal cells; 3) reprogramming of one neuronal subtype into another. The latter has already been successfully achieved in vivo during early brain development, providing strong impulse for the attempt to succeed in direct reprogramming in situ for future brain repair.
- Published
- 2013
- Full Text
- View/download PDF
46. Tuning neural circuits by turning the interneuron knob
- Author
-
Nicolás Marichal, Nathalie Dehorter, Benedikt Berninger, and Oscar Marín
- Subjects
0301 basic medicine ,Neuronal Plasticity ,Interneuron ,genetic structures ,General Neuroscience ,fungi ,Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,nervous system ,Interneurons ,medicine ,Biological neural network ,Premovement neuronal activity ,Animals ,Homeostasis ,Humans ,Nervous System Diseases ,Reprogramming ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Interneurons play a critical role in sculpting neuronal circuit activity and their dysfunction can result in neurological and neuropsychiatric disorders. To temporally structure and balance neuronal activity in the adult brain interneurons display a remarkable degree of subclass-specific plasticity, of which the underlying molecular mechanisms have recently begun to be elucidated. Grafting new interneurons to pre-existing neuronal networks allows for amelioration of circuit dysfunction in rodent models of neurological disease and can reopen critical windows for circuit plasticity. The crucial contribution of specific classes of interneurons to circuit homeostasis and plasticity in health and disease underscores their generation as an important objective for emerging strategies of lineage reprogramming in vivo.
- Published
- 2017
- Full Text
- View/download PDF
47. List of Contributors
- Author
-
Daniel G. Abernathy, Benedikt Berninger, Pascal Bielefeld, Irene Bozzoni, Murray J. Cairns, Nathalie Coré, Harold Cremer, Antoine de Chevigny, Davide De Pietri Tonelli, Andrea Erni, Carlos P. Fitzsimons, Michael Geaghan, Georgios Georgakilas, Stefanie Grosswendt, Artemis G. Hatzigeorgiou, Wieland B. Huttner, Dimitra Karagkouni, Ivano Legnini, Yangjian Liu, Qing Richard Lu, Federica Marinaro, Debora Napoli, Stefania Nicoli, Tomasz Jan Nowakowski, Maria D. Paraskevopoulou, Amy E. Pasquinelli, Tommaso Pizzorusso, Meritxell Pons-Espinal, Thomas Pratt, David Jonathan Price, Ben Pustjens, Nikolaus Rajewsky, Emma Ristori, Chiara Rolando, Marijn Schouten, William P. Schreiner, Spyros Tastsoglou, Stefania Tavano, Verdon Taylor, Neha Tiwari, Ioannis S. Vlachos, Haibo Wang, Andrew S. Yoo, and Xianghui Zhao
- Published
- 2017
- Full Text
- View/download PDF
48. Bifunctional poly(acrylamide) hydrogels through orthogonal coupling chemistries
- Author
-
Wenqiang Fan, Aránzazu del Campo, Marcelo Salierno, Aleeza Farrukh, Benedikt Berninger, and Julieta I. Paez
- Subjects
0301 basic medicine ,Polymers and Plastics ,Polymers ,Otras Ciencias Biológicas ,Poly(acrylamide) ,Acrylic Resins ,Biocompatible Materials ,Bioengineering ,INGENIERÍAS Y TECNOLOGÍAS ,02 engineering and technology ,Biotecnología Industrial ,Ciencias Biológicas ,Biomaterials ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Ultraviolet visible spectroscopy ,Polymer chemistry ,Materials Chemistry ,Copolymer ,Animals ,Polylysine ,Bifunctional ,Cells, Cultured ,Acrylic acid ,Neurons ,chemistry.chemical_classification ,Otras Ciencias Químicas ,Biomolecule ,Ciencias Químicas ,Hydrogels ,021001 nanoscience & nanotechnology ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Chemical engineering ,Acrylamide ,Self-healing hydrogels ,Amine gas treating ,Laminin ,0210 nano-technology ,CIENCIAS NATURALES Y EXACTAS - Abstract
Biomaterials for cell culture allowing simple and quantitative presentation of instructive cues enable rationalization of the interplay between cells and their surrounding microenvironment. Poly(acrylamide) (PAAm) hydrogels are popular 2D-model substrates for this purpose. However, quantitative and reproducible biofunctionalization of PAAm hydrogels with multiple ligands in a trustable, controlled, and independent fashion is not trivial. Here, we describe a method for bifunctional modification of PAAm hydrogels with thiol- and amine- containing biomolecules with controlled densities in an independent, orthogonal manner. We developed copolymer networks of AAm with 9% acrylic acid and 2% N-(4-(5-(methylsulfonyl)-1,3,4-oxadiazol-2-yl)phenyl)acrylamide. The covalent binding of thiol- and amine-containing chromophores at tunable concentrations was demonstrated and quantified by UV spectroscopy. The morphology, mechanical properties, and homogeneity of the copolymerized hydrogels were characterized by scanning electron microscopy, dynamic mechanical analysis, and confocal microscopy studies. Our copolymer hydrogels were bifunctionalized with polylysine and a laminin-mimetic peptide using the specific chemistries. We analyzed the effect of binding protocol of the two components in the maturation of cultured postmitotic cortical neurons. Our substrates supported neuronal attachment, proliferation, and neuronal differentiation. We found that neurons cultured on our hydrogels bifunctionalized with ligand-specific chemistries in a sequential fashion exhibited higher maturation at comparable culture times than using a simultaneous bifunctionalization strategy, displaying a higher number of neurites, branches, and dendritic filopodia. These results demonstrate the relevance of quantitative and optimized coupling chemistries for the performance of simple biomaterials and with sensitive cell types. Fil: Farrukh, Aleeza. Leibniz Institute for New Materials; Alemania. Max-Planck-Institut für Polymerforschung; Alemania Fil: Paez, Julieta Irene. Leibniz Institute for New Materials; Alemania. Max-Planck-Institut für Polymerforschung; Alemania Fil: Salierno, Marcelo Javier. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina. Max-Planck-Institut für Polymerforschung; Alemania. Johannes Gutenberg University; Alemania. Johannes Gutenberg University Mainz; Alemania Fil: Fan, Wenqiang. Johannes Gutenberg University; Alemania. Johannes Gutenberg University Mainz; Alemania Fil: Berninger, Benedikt. Johannes Gutenberg University; Alemania. Johannes Gutenberg University Mainz; Alemania Fil: del Campo, Aránzazu. Leibniz Institute for New Materials; Alemania. Max-Planck-Institut für Polymerforschung; Alemania. Saarland University; Alemania
- Published
- 2017
- Full Text
- View/download PDF
49. Transcriptional and Epigenetic Control of Astrogliogenesis
- Author
-
Benedikt Berninger and Neha Tiwari
- Subjects
0301 basic medicine ,Genetics ,Neurogenesis ,Biology ,Neural stem cell ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Histone ,medicine.anatomical_structure ,DNA methylation ,microRNA ,biology.protein ,medicine ,Epigenetics ,Neuroscience ,Transcription factor ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Astrocytes exert pivotal functions in the brain ranging from homeostasis to plasticity and their malfunctioning may contribute to neurodegenerative diseases. With increased recognition of their importance, more efforts are being dedicated to decoding the molecular mechanisms that control the generation of astrocytes from neural stem cells, a process referred to as astrogliogenesis. In this chapter, we highlight the discoveries that have shed light on the role of transcription factors, DNA methylation, histone modifications, and microRNAs in driving the transcriptional programs that underlie astrocyte generation. We further discuss the current understanding of gene regulatory pathways that control the switch from neurogenesis to astrogliogenesis during development.
- Published
- 2017
- Full Text
- View/download PDF
50. Neurovascular EGFL7 regulates adult neurogenesis in the subventricular zone and thereby affects olfactory perception
- Author
-
Beat Lutz, Marcus Krüger, Benedikt Berninger, Guilherme Horta, Felipe Ortega, Jan Baumgart, Stefanie Keller, Patrick N. Harter, Tobias Bäuerle, Nevenka Dudvarski Stankovic, Frank Bicker, Konstantin Radyushkin, Mirko H. H. Schmidt, Lavinia Alberi, Rui Benedito, Hendrik Nolte, Atria Kavyanifar, Jens Hartwig, Verica Vasic, and Deutsche Forschungsgemeinschaft (Alemania)
- Subjects
Male ,0301 basic medicine ,General Physics and Astronomy ,NEURAL STEM-CELLS ,MOUSE ,Mice ,SUBEPENDYMAL ZONE ,Neural Stem Cells ,Lateral Ventricles ,LINEAGE PROGRESSION ,BRAIN ,IN-VIVO ,Mice, Knockout ,Neuronal Plasticity ,Multidisciplinary ,Cell Cycle ,Neurogenesis ,NICHE ,Anatomy ,Neural stem cell ,Cell biology ,Adult Stem Cells ,medicine.anatomical_structure ,Signal Transduction ,STIMULATES NEUROGENESIS ,EGF Family of Proteins ,Science ,Notch signaling pathway ,Subventricular zone ,Biology ,Inhibitory postsynaptic potential ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Neuroplasticity ,medicine ,Biological neural network ,Animals ,Calcium-Binding Proteins ,Proteins ,General Chemistry ,Olfactory Perception ,ENDOTHELIAL-CELLS ,nervous system diseases ,Olfactory bulb ,Mice, Inbred C57BL ,SELF-RENEWAL ,030104 developmental biology ,nervous system - Abstract
Adult neural stem cells reside in a specialized niche in the subventricular zone (SVZ). Throughout life they give rise to adult-born neurons in the olfactory bulb (OB), thus contributing to neural plasticity and pattern discrimination. Here, we show that the neurovascular protein EGFL7 is secreted by endothelial cells and neural stem cells (NSCs) of the SVZ to shape the vascular stem-cell niche. Loss of EGFL7 causes an accumulation of activated NSCs, which display enhanced activity and re-entry into the cell cycle. EGFL7 pushes activated NSCs towards quiescence and neuronal progeny towards differentiation. This is achieved by promoting Dll4-induced Notch signalling at the blood vessel-stem cell interface. Fewer inhibitory neurons form in the OB of EGFL7-knockout mice, which increases the absolute signal conducted from the mitral cell layer of the OB but decreases neuronal network synchronicity. Consequently, EGFL7-knockout mice display severe physiological defects in olfactory behaviour and perception., The vascular stem cell niche regulates the proliferation and differentiation of neural stem cells (NSCs) in the adult subventricular zone. Here the authors identify EGFL7 as a neurovascular regulator of NSCs in vivo; EGFL7-knockout mice show reduced neurogenesis, and exhibit impaired olfactory perception and behaviour.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.