59 results on '"Bruno, Di Stefano"'
Search Results
2. Dynamics of alternative splicing during somatic cell reprogramming reveals functions for RNA-binding proteins CPSF3, hnRNP UL1, and TIA1
- Author
-
Claudia Vivori, Panagiotis Papasaikas, Ralph Stadhouders, Bruno Di Stefano, Anna Ribó Rubio, Clara Berenguer Balaguer, Serena Generoso, Anna Mallol, José Luis Sardina, Bernhard Payer, Thomas Graf, and Juan Valcárcel
- Subjects
Alternative splicing ,Somatic cell reprogramming ,CPSF3 ,hnRNP UL1 ,TIA1 ,Pluripotency ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background Somatic cell reprogramming is the process that allows differentiated cells to revert to a pluripotent state. In contrast to the extensively studied rewiring of epigenetic and transcriptional programs required for reprogramming, the dynamics of post-transcriptional changes and their associated regulatory mechanisms remain poorly understood. Here we study the dynamics of alternative splicing changes occurring during efficient reprogramming of mouse B cells into induced pluripotent stem (iPS) cells and compare them to those occurring during reprogramming of mouse embryonic fibroblasts. Results We observe a significant overlap between alternative splicing changes detected in the two reprogramming systems, which are generally uncoupled from changes in transcriptional levels. Correlation between gene expression of potential regulators and specific clusters of alternative splicing changes enables the identification and subsequent validation of CPSF3 and hnRNP UL1 as facilitators, and TIA1 as repressor of mouse embryonic fibroblasts reprogramming. We further find that these RNA-binding proteins control partially overlapping programs of splicing regulation, involving genes relevant for developmental and morphogenetic processes. Conclusions Our results reveal common programs of splicing regulation during reprogramming of different cell types and identify three novel regulators of this process and their targets.
- Published
- 2021
- Full Text
- View/download PDF
3. Editorial: Chromatin Regulation in Cell Fate Decisions
- Author
-
Justin Brumbaugh, Bruno Di Stefano, and José Luis Sardina
- Subjects
chromatin ,gene regulation ,epigenetics ,pluripotency ,differentiation ,cell fate ,Biology (General) ,QH301-705.5 - Published
- 2021
- Full Text
- View/download PDF
4. Direct Reprogramming of Mouse Fibroblasts into Functional Skeletal Muscle Progenitors
- Author
-
Ori Bar-Nur, Mattia F.M. Gerli, Bruno Di Stefano, Albert E. Almada, Amy Galvin, Amy Coffey, Aaron J. Huebner, Peter Feige, Cassandra Verheul, Priscilla Cheung, Duygu Payzin-Dogru, Sylvain Paisant, Anthony Anselmo, Ruslan I. Sadreyev, Harald C. Ott, Shahragim Tajbakhsh, Michael A. Rudnicki, Amy J. Wagers, and Konrad Hochedlinger
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Skeletal muscle harbors quiescent stem cells termed satellite cells and proliferative progenitors termed myoblasts, which play pivotal roles during muscle regeneration. However, current technology does not allow permanent capture of these cell populations in vitro. Here, we show that ectopic expression of the myogenic transcription factor MyoD, combined with exposure to small molecules, reprograms mouse fibroblasts into expandable induced myogenic progenitor cells (iMPCs). iMPCs express key skeletal muscle stem and progenitor cell markers including Pax7 and Myf5 and give rise to dystrophin-expressing myofibers upon transplantation in vivo. Notably, a subset of transplanted iMPCs maintain Pax7 expression and sustain serial regenerative responses. Similar to satellite cells, iMPCs originate from Pax7+ cells and require Pax7 itself for maintenance. Finally, we show that myogenic progenitor cell lines can be established from muscle tissue following small-molecule exposure alone. This study thus reports on a robust approach to derive expandable myogenic stem/progenitor-like cells from multiple cell types. : In this article, Hochedlinger and colleagues reprogrammed mouse fibroblasts into induced myogenic progenitors (iMPCs) by transient expression of MyoD and treatment with small molecules. iMPCs can be extensively propagated in vitro and exhibit skeletal muscle stem/progenitor cell characteristics, including the requirement for Pax7 function as well as the ability to sustain muscle regeneration upon repeated injury. Keywords: skeletal muscle, satellite cells, direct lineage reprogramming, induced muscle progenitor cells, MyoD, Pax7, small molecules, transplantation, muscular dystrophy
- Published
- 2018
- Full Text
- View/download PDF
5. Active DNA demethylation promotes cell fate specification and the DNA damage response
- Author
-
Dongpeng Wang, Wei Wu, Elsa Callen, Raphael Pavani, Nicholas Zolnerowich, Srikanth Kodali, Dali Zong, Nancy Wong, Santiago Noriega, William J. Nathan, Gabriel Matos-Rodrigues, Raj Chari, Michael J. Kruhlak, Ferenc Livak, Michael Ward, Keith Caldecott, Bruno Di Stefano, and André Nussenzweig
- Subjects
Neurons ,Multidisciplinary ,DNA Repair ,Macrophages ,Induced Pluripotent Stem Cells ,Cell Differentiation ,Article ,Thymine DNA Glycosylase ,DNA Demethylation ,Enhancer Elements, Genetic ,Cell Transdifferentiation ,5-Methylcytosine ,Humans ,DNA Breaks, Single-Stranded - Abstract
Neurons harbor high levels of single-strand DNA breaks (SSBs) that are targeted to neuronal enhancers, but the source of this endogenous damage remains unclear. Using two systems of postmitotic lineage specification—induced pluripotent stem cell–derived neurons and transdifferentiated macrophages—we show that thymidine DNA glycosylase (TDG)–driven excision of methylcytosines oxidized with ten-eleven translocation enzymes (TET) is a source of SSBs. Although macrophage differentiation favors short-patch base excision repair to fill in single-nucleotide gaps, neurons also frequently use the long-patch subpathway. Disrupting this gap-filling process using anti-neoplastic cytosine analogs triggers a DNA damage response and neuronal cell death, which is dependent on TDG. Thus, TET-mediated active DNA demethylation promotes endogenous DNA damage, a process that normally safeguards cell identity but can also provoke neurotoxicity after anticancer treatments.
- Published
- 2022
- Full Text
- View/download PDF
6. Chemical reprogramming takes the fast lane
- Author
-
Emily J. Park, Srikanth Kodali, and Bruno Di Stefano
- Subjects
Genetics ,Molecular Medicine ,Cell Biology - Published
- 2023
- Full Text
- View/download PDF
7. Single cell RNA-seq identifies the origins of heterogeneity in efficient cell transdifferentiation and reprogramming
- Author
-
Mirko Francesconi, Bruno Di Stefano, Clara Berenguer, Luisa de Andrés-Aguayo, Marcos Plana-Carmona, Maria Mendez-Lago, Amy Guillaumet-Adkins, Gustavo Rodriguez-Esteban, Marta Gut, Ivo G Gut, Holger Heyn, Ben Lehner, and Thomas Graf
- Subjects
single cell ,reprogramming ,transdifferentiation ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Forced transcription factor expression can transdifferentiate somatic cells into other specialised cell types or reprogram them into induced pluripotent stem cells (iPSCs) with variable efficiency. To better understand the heterogeneity of these processes, we used single-cell RNA sequencing to follow the transdifferentation of murine pre-B cells into macrophages as well as their reprogramming into iPSCs. Even in these highly efficient systems, there was substantial variation in the speed and path of fate conversion. We predicted and validated that these differences are inversely coupled and arise in the starting cell population, with Mychigh large pre-BII cells transdifferentiating slowly but reprogramming efficiently and Myclow small pre-BII cells transdifferentiating rapidly but failing to reprogram. Strikingly, differences in Myc activity predict the efficiency of reprogramming across a wide range of somatic cell types. These results illustrate how single cell expression and computational analyses can identify the origins of heterogeneity in cell fate conversion processes.
- Published
- 2019
- Full Text
- View/download PDF
8. Integrated loss- and gain-of-function screens define a core network governing human embryonic stem cell behavior
- Author
-
Mitzi I. Kuroda, Dominik Ricken, Bruno Di Stefano, Emma V. Watson, Marit A.C. de Kort, Eric C. Wooten, Mohammed Dezfulian, Timothy D. Martin, Konrad Hochedlinger, Kamila Naxerova, Jessica L. Makofske, and Stephen J. Elledge
- Subjects
Human Embryonic Stem Cells ,Cell Differentiation ,Germ layer ,Joint analysis ,Biology ,Regenerative medicine ,Lineage specification ,Embryonic stem cell ,Cell biology ,Gain of function ,Gain of Function Mutation ,Genetics ,Humans ,Developmental biology ,Embryonic Stem Cells ,Germ Layers ,Function (biology) ,Developmental Biology - Abstract
Understanding the genetic control of human embryonic stem cell function is foundational for developmental biology and regenerative medicine. Here we describe an integrated genome-scale loss- and gain-of-function screening approach to identify genetic networks governing embryonic stem cell proliferation and differentiation into the three germ layers. We identified a deep link between pluripotency maintenance and survival by showing that genetic alterations that cause pluripotency dissolution simultaneously increase apoptosis resistance. We discovered that the chromatin-modifying complex SAGA and in particular its subunit TADA2B are central regulators of pluripotency, survival, growth, and lineage specification. Joint analysis of all screens revealed that genetic alterations that broadly inhibit differentiation across multiple germ layers drive proliferation and survival under pluripotency-maintaining conditions and coincide with known cancer drivers. Our results show the power of integrated multilayer genetic screening for the robust mapping of complex genetic networks.
- Published
- 2021
- Full Text
- View/download PDF
9. Early Life Reprogramming-Based Treatment Promotes Longevity
- Author
-
Patrizia Pessina and Bruno Di Stefano
- Subjects
Cell Biology ,Developmental Biology ,Biotechnology - Published
- 2023
- Full Text
- View/download PDF
10. Cell identity and plasticity uncoupled
- Author
-
Bruno Di Stefano
- Subjects
Cell Biology - Published
- 2022
- Full Text
- View/download PDF
11. Dynamics of alternative splicing during somatic cell reprogramming reveals functions for RNA-binding proteins CPSF3, hnRNP UL1, and TIA1
- Author
-
Panagiotis Papasaikas, Anna Ribó Rubio, Clara Berenguer Balaguer, Ralph Stadhouders, Juan Valcárcel, Serena Francesca Generoso, Bernhard Payer, Bruno Di Stefano, Anna Mallol, Claudia Vivori, Thomas Graf, and Jose Luis Sardina
- Subjects
Pluripotency ,TIA1 ,QH301-705.5 ,Cellular differentiation ,RNA-binding protein ,CPSF3 ,Biology ,QH426-470 ,Heterogeneous-Nuclear Ribonucleoproteins ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,Animals ,Biology (General) ,Induced pluripotent stem cell ,030304 developmental biology ,B-Lymphocytes ,0303 health sciences ,Research ,Cleavage And Polyadenylation Specificity Factor ,Alternative splicing ,Gene Expression Regulation, Developmental ,Fibroblasts ,Cellular Reprogramming ,Embryo, Mammalian ,Embryonic stem cell ,hnRNP UL1 ,T-Cell Intracellular Antigen-1 ,Cell biology ,RNA splicing ,CCAAT-Enhancer-Binding Proteins ,Somatic cell reprogramming ,Reprogramming ,030217 neurology & neurosurgery - Abstract
C.V. was recipient of an FPI-Severo Ochoa Fellowship from the Spanish Ministry of Economy and Competitiveness. Work in J.V. laboratory is supported by the European Research Council (ERC AdvG 670146), AGAUR, Spanish Ministry of Economy and Competitiveness (BFU 2017 89308-P) and the Centre of Excellence Severo Ochoa. Work in T.G.'s laboratory was supported by the European Research Council FP7/2007-2013 (ERC Synergy Grant 4D-Genome) the Ministerio de Educación y Ciencia (SAF.2012-37167) and AGAUR. We acknowledge support of the Spanish Ministry of Science and Innovation to the EMBL partnership and the CERCA Programme / Generalitat de Catalunya. UDTRIAS Background: Somatic cell reprogramming is the process that allows differentiated cells to revert to a pluripotent state. In contrast to the extensively studied rewiring of epigenetic and transcriptional programs required for reprogramming, the dynamics of post-transcriptional changes and their associated regulatory mechanisms remain poorly understood. Here we study the dynamics of alternative splicing changes occurring during efficient reprogramming of mouse B cells into induced pluripotent stem (iPS) cells and compare them to those occurring during reprogramming of mouse embryonic fibroblasts. Results: We observe a significant overlap between alternative splicing changes detected in the two reprogramming systems, which are generally uncoupled from changes in transcriptional levels. Correlation between gene expression of potential regulators and specific clusters of alternative splicing changes enables the identification and subsequent validation of CPSF3 and hnRNP UL1 as facilitators, and TIA1 as repressor of mouse embryonic fibroblasts reprogramming. We further find that these RNA-binding proteins control partially overlapping programs of splicing regulation, involving genes relevant for developmental and morphogenetic processes. Conclusions: Our results reveal common programs of splicing regulation during reprogramming of different cell types and identify three novel regulators of this process and their targets.
- Published
- 2021
12. Dissecting dual roles of MyoD during lineage conversion to mature myocytes and myogenic stem cells
- Author
-
Simona Cristea, Fei Ji, Michael S. Hoetker, Bruno Di Stefano, Ruslan I. Sadreyev, Masakazu Yamamoto, Kathleen A. Messemer, Alexander Meissner, Aaron J. Huebner, Franziska Michor, Masaki Yagi, Naftali Horwitz, Anthony Patelunas, David J. Goldhamer, A. Almada, Nikolaos Tsopoulidis, Konrad Hochedlinger, Jocelyn Charlton, Ori Bar-Nur, and Amy J. Wagers
- Subjects
induced myogenic progenitor cells (iMPCs) ,transdifferentiation ,Muscle Fibers, Skeletal ,epigenetic reprogramming ,Biology ,MyoD ,Muscle Development ,dedifferentiation ,satellite cells ,DNA methylation ,Myoblasts ,Mice ,Genetics ,Animals ,Epigenetics ,Progenitor cell ,Muscle, Skeletal ,MyoD Protein ,Myogenesis ,Stem Cells ,Transdifferentiation ,Cell Differentiation ,Cell biology ,Stem cell ,Reprogramming ,Developmental Biology ,Research Paper - Abstract
The generation of myotubes from fibroblasts upon forced MyoD expression is a classic example of transcription factor-induced reprogramming. We recently discovered that additional modulation of signaling pathways with small molecules facilitates reprogramming to more primitive induced myogenic progenitor cells (iMPCs). Here, we dissected the transcriptional and epigenetic dynamics of mouse fibroblasts undergoing reprogramming to either myotubes or iMPCs using a MyoD-inducible transgenic model. Induction of MyoD in fibroblasts combined with small molecules generated Pax7(+) iMPCs with high similarity to primary muscle stem cells. Analysis of intermediate stages of iMPC induction revealed that extinction of the fibroblast program preceded induction of the stem cell program. Moreover, key stem cell genes gained chromatin accessibility prior to their transcriptional activation, and these regions exhibited a marked loss of DNA methylation dependent on the Tet enzymes. In contrast, myotube generation was associated with few methylation changes, incomplete and unstable reprogramming, and an insensitivity to Tet depletion. Finally, we showed that MyoD's ability to bind to unique bHLH targets was crucial for generating iMPCs but dispensable for generating myotubes. Collectively, our analyses elucidate the role of MyoD in myogenic reprogramming and derive general principles by which transcription factors and signaling pathways cooperate to rewire cell identity., Genes & Development, 35 (17-18), ISSN:0890-9369, ISSN:1549-5477
- Published
- 2021
- Full Text
- View/download PDF
13. Dynamics of alternative splicing during somatic cell reprogramming reveals functions for RNA-binding proteins CPSF3, hnRNP UL1 and TIA1
- Author
-
Bruno Di Stefano, Ralph Stadhouders, Thomas Graf, Panagiotis Papasaikas, Juan Valcárcel, Bernhard Payer, Clara Berenguer Balaguer, Jose Luis Sardina, Serena Francesca Generoso, Claudia Vivori, and Anna Mallol
- Subjects
TIA1 ,RNA splicing ,Alternative splicing ,RNA-binding protein ,Epigenetics ,Biology ,Induced pluripotent stem cell ,Reprogramming ,Embryonic stem cell ,Cell biology - Abstract
In contrast to the extensively studied rewiring of epigenetic and transcriptional programs required for cell reprogramming, the dynamics of post-transcriptional changes and their associated regulatory mechanisms remain poorly understood. Here we have studied the dynamics of alternative splicing (AS) changes occurring during efficient reprogramming of mouse B cells into induced pluripotent stem (iPS) cells. These changes, generally uncoupled from transcriptional regulation, significantly overlapped with splicing programs reported during reprogramming of mouse embryonic fibroblasts (MEFs). Correlation between gene expression of potential regulators and specific clusters of AS changes enabled the identification and subsequent validation of CPSF3 and hnRNP UL1 as facilitators, and TIA1 as repressor of MEFs reprogramming. These RNA-binding proteins control partially overlapping programs of splicing regulation affecting genes involved in developmental and morphogenetic processes. Our results reveal common programs of splicing regulation during reprogramming of different cell types and identify three novel regulators of this process.
- Published
- 2020
- Full Text
- View/download PDF
14. Reduced MEK inhibition preserves genomic stability in naive human embryonic stem cells
- Author
-
Anna Sahakyan, Mai Ueda, Keiko Shioda, Yasuhiro Takashima, Justin Brumbaugh, Aaron J. Huebner, Kendell Clement, Shan Sabri, Steven P. Gygi, Alison R. Erickson, Toshi Shioda, Hongcang Gu, Alexander Meissner, Katie J. Clowers, Kathrin Plath, Konrad Hochedlinger, and Bruno Di Stefano
- Subjects
0301 basic medicine ,Genome instability ,MAPK/ERK pathway ,Technology ,Proteome ,DNA damage ,1.1 Normal biological development and functioning ,Biology ,Regenerative Medicine ,Medical and Health Sciences ,Biochemistry ,Genomic Instability ,03 medical and health sciences ,Underpinning research ,Humans ,Stem Cell Research - Embryonic - Human ,Protein Kinase Inhibitors ,Molecular Biology ,Embryonic Stem Cells ,Cell Biology ,DNA Methylation ,Biological Sciences ,Cell cycle ,MAP Kinase Kinase Kinases ,Stem Cell Research ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,Epiblast ,embryonic structures ,DNA methylation ,Generic health relevance ,Transcriptome ,Ex vivo ,Developmental Biology ,Biotechnology - Abstract
Human embryonic stem cells (hESCs) can be captured in a primed state in which they resemble the postimplantation epiblast, or in a naive state where they resemble the preimplantation epiblast. Naive-cell-specific culture conditions allow the study of preimplantation development ex vivo but reportedly lead to chromosomal abnormalities, which compromises their utility in research and potential therapeutic applications. Although MEK inhibition is essential for the naive state, here we show that reduced MEK inhibition facilitated the establishment and maintenance of naive hESCs that retained naive-cell-specific features, including global DNA hypomethylation, HERVK expression, and two active X chromosomes. We further show that hESCs cultured under these modified conditions proliferated more rapidly; accrued fewer chromosomal abnormalities; and displayed changes in the phosphorylation levels of MAPK components, regulators of DNA damage/repair, and cell cycle. We thus provide a simple modification to current methods that can enable robust growth and reduced genomic instability in naive hESCs.
- Published
- 2018
- Full Text
- View/download PDF
15. An ES-like pluripotent state in FGF-dependent murine iPS cells.
- Author
-
Bruno Di Stefano, Christa Buecker, Federica Ungaro, Alessandro Prigione, Hsu-Hsin Chen, Maaike Welling, Maureen Eijpe, Gustavo Mostoslavsky, Paul Tesar, James Adjaye, Niels Geijsen, and Vania Broccoli
- Subjects
Medicine ,Science - Abstract
Recent data demonstrates that stem cells can exist in two morphologically, molecularly and functionally distinct pluripotent states; a naïve LIF-dependent pluripotent state which is represented by murine embryonic stem cells (mESCs) and an FGF-dependent primed pluripotent state represented by murine and rat epiblast stem cells (EpiSCs). We find that derivation of induced pluripotent stem cells (iPSCs) under EpiSC culture conditions yields FGF-dependent iPSCs from hereon called FGF-iPSCs) which, unexpectedly, display naïve ES-like/ICM properties. FGF-iPSCs display X-chromosome activation, multi-lineage differentiation, teratoma competence and chimera contribution in vivo. Our findings suggest that in 129 and Bl6 mouse strains, iPSCs can dominantly adopt a naive pluripotent state regardless of culture growth factor conditions. Characterization of the key molecular signalling pathways revealed FGF-iPSCs to depend on the Activin/Nodal and FGF pathways, while signalling through the JAK-STAT pathway is not required for FGF-iPS cell maintenance. Our findings suggest that in 129 and Bl6 mouse strains, iPSCs can dominantly adopt a naive pluripotent state regardless of culture growth factor conditions.
- Published
- 2010
- Full Text
- View/download PDF
16. Prolonged Mek1/2 suppression impairs the developmental potential of embryonic stem cells
- Author
-
Arman W. Mohammad, Konrad Hochedlinger, Jiho Choi, Bruno Di Stefano, Steven P. Gygi, Sang Yong Kim, Ruslan I. Sadreyev, Kaixuan Lin, Ryan M. Walsh, Haruhiko Koseki, Andrew Xiao, Kendell Clement, Andrej J. Savol, Junko Odajima, Justin Brumbaugh, Toshihiro Shioda, Jean Charron, Hongcang Gu, Alexander Meissner, Christopher M. Rose, Aaron J. Huebner, Andreas Gnirke, and Jafar Sharif
- Subjects
0301 basic medicine ,Genetics ,Multidisciplinary ,Methyltransferase ,urogenital system ,Wnt signaling pathway ,Biology ,Embryonic stem cell ,Article ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Downregulation and upregulation ,embryonic structures ,DNA methylation ,Inner cell mass ,Epigenetics ,biological phenomena, cell phenomena, and immunity ,Genomic imprinting ,reproductive and urinary physiology - Abstract
Concomitant activation of the Wnt pathway and suppression of Mapk signalling by two small molecule inhibitors (2i) in the presence of leukaemia inhibitory factor (LIF) (hereafter termed 2i/L) induces a naive state in mouse embryonic stem (ES) cells that resembles the inner cell mass (ICM) of the pre-implantation embryo. Since the ICM exists only transiently in vivo, it remains unclear how sustained propagation of naive ES cells in vitro affects their stability and functionality. Here we show that prolonged culture of male mouse ES cells in 2i/L results in irreversible epigenetic and genomic changes that impair their developmental potential. Furthermore, we find that female ES cells cultured in conventional serum plus LIF medium phenocopy male ES cells cultured in 2i/L. Mechanistically, we demonstrate that the inhibition of Mek1/2 is predominantly responsible for these effects, in part through the downregulation of DNA methyltransferases and their cofactors. Finally, we show that replacement of the Mek1/2 inhibitor with a Src inhibitor preserves the epigenetic and genomic integrity as well as the developmental potential of ES cells. Taken together, our data suggest that, although short-term suppression of Mek1/2 in ES cells helps to maintain an ICM-like epigenetic state, prolonged suppression results in irreversible changes that compromise their developmental potential.
- Published
- 2017
- Full Text
- View/download PDF
17. Reprogramming: identifying the mechanisms that safeguard cell identity
- Author
-
Justin Brumbaugh, Bruno Di Stefano, and Konrad Hochedlinger
- Subjects
Cell type ,Lineage (genetic) ,Context (language use) ,Computational biology ,Review ,Biology ,Cell fate determination ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Cellular Reprogramming ,Cell identity ,Chromatin ,Neoplasm Proteins ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Reprogramming ,Function (biology) ,Developmental Biology ,Transcription Factors - Abstract
Development and homeostasis rely upon concerted regulatory pathways to establish the specialized cell types needed for tissue function. Once a cell type is specified, the processes that restrict and maintain cell fate are equally important in ensuring tissue integrity. Over the past decade, several approaches to experimentally reprogram cell fate have emerged. Importantly, efforts to improve and understand these approaches have uncovered novel molecular determinants that reinforce lineage commitment and help resist cell fate changes. In this Review, we summarize recent studies that have provided insights into the various chromatin factors, post-transcriptional processes and features of genomic organization that safeguard cell identity in the context of reprogramming to pluripotency. We also highlight how these factors function in other experimental, physiological and pathological cell fate transitions, including direct lineage conversion, pluripotency-to-totipotency reversion and cancer.
- Published
- 2019
18. The RNA Helicase DDX6 Controls Cellular Plasticity by Modulating P-Body Homeostasis
- Author
-
Kendell Clement, Chuck Haggerty, Stefan Aigner, Hongcang Gu, Alexander Meissner, Inna Lipchina, Anthony Anselmo, Steven P. Gygi, Justin Brumbaugh, Fei Ji, John L. Pulice, Mattia F. M. Gerli, Katie J. Clowers, Aaron J. Huebner, Inés Rabano Jiménez, En-Ching Luo, Ruslan I. Sadreyev, Bruno Di Stefano, Konrad Hochedlinger, Jocelyn Charlton, Marit A.C. de Kort, and Gene W. Yeo
- Subjects
Jumonji Domain-Containing Histone Demethylases ,Messenger ,RNA helicase DDX6 ,Cell Plasticity ,adult progenitor cells ,P-body ,Inbred C57BL ,self-renewal ,Regenerative Medicine ,Medical and Health Sciences ,DEAD-box RNA Helicases ,Mice ,0302 clinical medicine ,Gene expression ,Homeostasis ,primed pluripotency ,RNA-Seq ,0303 health sciences ,Cell Differentiation ,differentiation ,Nanog Homeobox Protein ,Biological Sciences ,RNA Helicase A ,Cell biology ,Chromatin ,Organoids ,Ribonucleoproteins ,DNA methylation ,Molecular Medicine ,Stem Cell Research - Nonembryonic - Non-Human ,Stem cell ,Pluripotent Stem Cells ,exit from pluripotency ,1.1 Normal biological development and functioning ,Induced Pluripotent Stem Cells ,Cell fate determination ,Biology ,Article ,Cell Line ,03 medical and health sciences ,Underpinning research ,Proto-Oncogene Proteins ,Genetics ,Animals ,Humans ,RNA, Messenger ,Post-transcriptional regulation ,Embryonic Stem Cells ,030304 developmental biology ,naive pluripotency ,Proteins ,Cell Biology ,DNA Methylation ,Stem Cell Research ,Chromatin Assembly and Disassembly ,Embryonic stem cell ,Mice, Inbred C57BL ,Gene Ontology ,Gene Expression Regulation ,Protein Biosynthesis ,RNA ,chromatin ,Generic health relevance ,Ribosomes ,030217 neurology & neurosurgery ,post-transcriptional regulation ,Developmental Biology - Abstract
Post-transcriptional mechanisms have the potential to influence complex changes in gene expression, yet their role in cell fate transitions remains largely unexplored. Here, we show that suppression of the RNA helicase DDX6 endows human and mouse primed embryonic stem cells (ESCs) with a differentiation-resistant, "hyper-pluripotent" state, which readily reprograms to a naive state resembling the preimplantation embryo. We further demonstrate that DDX6 plays a key role in adult progenitors where it controls the balance between self-renewal and differentiation in a context-dependent manner. Mechanistically, DDX6 mediates the translational suppression of target mRNAs in P-bodies. Upon loss of DDX6 activity, P-bodies dissolve and release mRNAs encoding fate-instructive transcription and chromatin factors that re-enter the ribosome pool. Increased translation of these targets impacts cell fate by rewiring the enhancer, heterochromatin, and DNA methylation landscapes of undifferentiated cell types. Collectively, our data establish a link between P-body homeostasis, chromatin organization, and stem cell potency.
- Published
- 2019
19. Single cell RNA-seq identifies the origins of heterogeneity in efficient cell transdifferentiation and reprogramming
- Author
-
Marcos Plana-Carmona, Marta Gut, Bruno Di Stefano, Thomas Graf, Maria Mendez Lago, Amy Guillaumet-Adkins, Luisa de Andrés-Aguayo, Gustavo Rodriguez-Esteban, Clara Berenguer, Holger Heyn, Mirko Francesconi, Ivo Gut, and Ben Lehner
- Subjects
0301 basic medicine ,Mouse ,Somatic cell ,transdifferentiation ,Stem cells ,Computational biology ,Mice ,0302 clinical medicine ,RNA-Seq ,Biology (General) ,Induced pluripotent stem cell ,Mice, Knockout ,education.field_of_study ,General Neuroscience ,Transdifferentiation ,General Medicine ,Cellular Reprogramming ,Stem Cells and Regenerative Medicine ,Cell biology ,single cell ,Cell Transdifferentiation ,Regenerative medicine ,Medicine ,Stem cell ,Single-Cell Analysis ,Systems biology ,Reprogramming ,Research Article ,Computational and Systems Biology ,Signal Transduction ,Cell type ,QH301-705.5 ,Science ,Population ,Induced Pluripotent Stem Cells ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Animals ,Cell Lineage ,education ,General Immunology and Microbiology ,Precursor Cells, B-Lymphoid ,reprogramming ,Mice, Inbred C57BL ,030104 developmental biology ,Transcriptome ,030217 neurology & neurosurgery - Abstract
Forced transcription factor expression can transdifferentiate somatic cells into other specialised cell types or reprogram them into induced pluripotent stem cells (iPSCs) with variable efficiency. To better understand the heterogeneity of these processes, we used single-cell RNA sequencing to follow the transdifferentation of murine pre-B cells into macrophages as well as their reprogramming into iPSCs. Even in these highly efficient systems, there was substantial variation in the speed and path of fate conversion. We predicted and validated that these differences are inversely coupled and arise in the starting cell population, with Mychigh large pre-BII cells transdifferentiating slowly but reprogramming efficiently and Myclow small pre-BII cells transdifferentiating rapidly but failing to reprogram. Strikingly, differences in Myc activity predict the efficiency of reprogramming across a wide range of somatic cell types. These results illustrate how single cell expression and computational analyses can identify the origins of heterogeneity in cell fate conversion processes. We would like to thank Ido Amit and Diego Adhemar Jaitin for help with the MARS-Seq technique and the CRG/UPF FACS Unit for help with the cell sorting. Work in the lab of TG was supported by the European Research Council (ERC) Synergy Grant (4D-Genome) and by AGAUR (SGR-1136). Research in the lab of BL was supported by an ERC Consolidator grant (616434), the Spanish Ministry of Economy and Competitiveness (BFU2011-26206), the AXA Research Fund, the Bettencourt Schueller Foundation and AGAUR (SGR-831). We acknowledge support of the Spanish Ministry of Economy and Competitiveness, Centro de Excelencia Severo Ochoa 2013–2017 (SEV-2012–0208) and of the CERCA Programme/Generalitat de Catalunya.
- Published
- 2019
20. Cell-of-Origin-Specific 3D Genome Structure Acquired during Somatic Cell Reprogramming
- Author
-
Wouter de Laat, Francesco Limone, Peter H.L. Krijger, Thomas Graf, Elzo de Wit, Chris van Oevelen, Bruno Di Stefano, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
0301 basic medicine ,Genetics ,Regulation of gene expression ,Cèl·lules somàtiques ,Somatic cell ,Chromosome ,Computational biology ,Cell Biology ,Biology ,Genome ,Embryonic stem cell ,3. Good health ,Chromatin ,03 medical and health sciences ,030104 developmental biology ,Journal Article ,Molecular Medicine ,Induced pluripotent stem cell ,Reprogramming - Abstract
Forced expression of reprogramming factors can convert somatic cells into induced pluripotent stem cells (iPSCs). Here we studied genome topology dynamics during reprogramming of different somatic cell types with highly distinct genome conformations. We find large-scale topologically associated domain (TAD) repositioning and alterations of tissue-restricted genomic neighborhoods and chromatin loops, effectively erasing the somatic-cell-specific genome structures while establishing an embryonic stem-cell-like 3D genome. Yet, early passage iPSCs carry topological hallmarks that enable recognition of their cell of origin. These hallmarks are not remnants of somatic chromosome topologies. Instead, the distinguishing topological features are acquired during reprogramming, as we also find for cell-of-origin-dependent gene expression patterns. This work was supported by an NWO/CW TOP grant (714.012.002), an NWO VICI grant 724.012.003, a NanoNextNL grant, and a European Research Council Starting Grant (209700, ‘‘4C’’) to W.d.L.; a Ministerio de Educacion y Ciencia, SAF.2012-37167, Fundació La Marató TV3 120410, AGAUR SGR 1136, and European Research Council Synergy Grant (‘‘4D-Genome) to T.G.; and an ERC Stg (637587, ‘‘HAP-PHEN’’) to E.d.W
- Published
- 2016
21. Author response: Single cell RNA-seq identifies the origins of heterogeneity in efficient cell transdifferentiation and reprogramming
- Author
-
Mirko Francesconi, Ivo Gut, Amy Guillaumet-Adkins, Maria Mendez-Lago, Thomas Graf, Ben Lehner, Gustavo Rodriguez-Esteban, Holger Heyn, Bruno Di Stefano, Clara Berenguer, Marcos Plana-Carmona, Luisa de Andrés-Aguayo, and Marta Gut
- Subjects
medicine.anatomical_structure ,Cell Transdifferentiation ,Cell ,medicine ,RNA-Seq ,Computational biology ,Biology ,Reprogramming - Published
- 2019
- Full Text
- View/download PDF
22. Whsc1 links pluripotency exit with mesendoderm specification
- Author
-
Bruno Di Stefano, Thomas Graf, Alessandro Dasti, Tian V. Tian, Luisa de Andrés-Aguayo, Carolina Segura-Morales, Johanna Goldmann, Antonio Gomez, Gregoire Stik, Enrique Vidal, Jose Luis Sardina, Rudolf Jaenisch, and Maria Vila-Casadesús
- Subjects
Pluripotent Stem Cells ,education ,Library science ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Blueprint ,Political science ,Animals ,Cell Lineage ,Embryonic Stem Cells ,030304 developmental biology ,0303 health sciences ,Neural Plate ,Endoderm ,Nuclear Proteins ,Cell Differentiation ,Cell Biology ,Histone-Lysine N-Methyltransferase ,Cell biology ,030220 oncology & carcinogenesis ,General partnership ,embryonic structures ,Christian ministry ,Biologia del desenvolupament ,Enzims ,Proteïnes ,Genètica ,Germ Layers ,Transcription Factors - Abstract
How pluripotent stem cells differentiate into the main germ layers is a key question of developmental biology. Here, we show that the chromatin-related factor Whsc1 (also known as Nsd2 and MMSET) has a dual role in pluripotency exit and germ layer specification of embryonic stem cells. On induction of differentiation, a proportion of Whsc1-depleted embryonic stem cells remain entrapped in a pluripotent state and fail to form mesendoderm, although they are still capable of generating neuroectoderm. These functions of Whsc1 are independent of its methyltransferase activity. Whsc1 binds to enhancers of the mesendodermal regulators Gata4, T (Brachyury), Gata6 and Foxa2, together with Brd4, and activates the expression of these genes. Depleting each of these regulators also delays pluripotency exit, suggesting that they mediate the effects observed with Whsc1. Our data indicate that Whsc1 links silencing of the pluripotency regulatory network with activation of mesendoderm lineages. J.L.S. were supported by Juan de la Cierva postdoctoral fellowships (MINECO; FJCI-2014-22946 and IJCI-2014-21872), B.D.S. by an EMBO long-term fellowship (number ALTF 1143-2015), G.S. by a Marie Sklodowska-Curie fellowship (H2020-MSCA-IF-2016, miRStem), A.D. by a Severo Ochoa fellowship and J.G. by a Boehringer Ingelheim Graduate Student Fellowship. R.J. was supported by NIH grants R01 NS088538-01 and 2R01MH104610-15. This work was supported by the EU-FP7 project BLUEPRINT, the Spanish Ministry of Economy, Industry and Competitiveness to the EMBL partnership, Centro de Excelencia Severo Ochoa 2013–2017 and the CERCA Program Generalitat de Catalunya. T.V.T., J.L.S. and B.D.S. were supported by a CRG award for junior collaborative projects
- Published
- 2019
- Full Text
- View/download PDF
23. Novel roles for SUMOylation in cellular plasticity
- Author
-
Konrad Hochedlinger and Bruno Di Stefano
- Subjects
0301 basic medicine ,Heterochromatin ,Cellular differentiation ,Cell Plasticity ,SUMO protein ,Biology ,Cell fate determination ,Article ,03 medical and health sciences ,Kruppel-Like Factor 4 ,Mice ,Animals ,Enhancer ,Cells, Cultured ,Sumoylation ,Cell Differentiation ,Mouse Embryonic Stem Cells ,Cell Biology ,Cellular Reprogramming ,Chromatin ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Small Ubiquitin-Related Modifier Proteins ,Protein Processing, Post-Translational ,Reprogramming ,Transcription Factors - Abstract
Understanding general principles that safeguard cellular identity should reveal critical insights into common mechanisms underlying specification of varied cell types. Here, we show that SUMO modification acts to stabilize cell fate in a variety of contexts. Hyposumoylation enhances pluripotency reprogramming in vitro and in vivo, increases lineage transdifferentiation, and facilitates leukemic cell differentiation. Suppressing sumoylation in embryonic stem cells (ESCs) promotes their conversion into 2-cell-embryo-like (2C-like) cells. During reprogramming to pluripotency, SUMO functions on fibroblastic enhancers to retain somatic transcription factors together with Oct4, Sox2, and Klf4, thus impeding somatic enhancer inactivation. In contrast, in ESCs, SUMO functions on heterochromatin to silence the 2C program, maintaining both proper H3K9me3 levels genome-wide and repression of the Dux locus by triggering recruitment of the sumoylated PRC1.6 and Kap/Setdb1 repressive complexes. Together, these studies show that SUMO acts on chromatin as a glue to stabilize key determinants of somatic and pluripotent states.
- Published
- 2018
24. Reduced MEK inhibition preserves genomic stability in naïve human ES cells
- Author
-
Bruno Di Stefano and Konrad Hochedlinger
- Subjects
0301 basic medicine ,Genome instability ,MAPK/ERK pathway ,DNA damage ,Cell cycle ,Biology ,Embryonic stem cell ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Epiblast ,embryonic structures ,General Earth and Planetary Sciences ,Ex vivo ,General Environmental Science ,DNA hypomethylation - Abstract
Human embryonic stem cells (hESCs) can be captured in a primed state resembling the postimplantation epiblast or in a naive state resembling the preimplantation epiblast. Naive conditions allow the study of preimplantation development ex vivo but reportedly lead to chromosomal abnormalities, compromising their utility in research and potential therapeutic applications. Although MEK inhibition is essential for the naive state, here we show that reduced MEK inhibition facilitates the establishment and maintenance of naive hESCs that retain naive-specific features, including global DNA hypomethylation, HERVK expression and X chromosome reactivation. We further show that hESCs cultured under these modified conditions proliferate more rapidly, accrue fewer chromosomal abnormalities and display changes in the phosphorylation levels of MAPK components, regulators of DNA damage/repair, and cell cycle. We thus provide a simple modification to current methods to enable robust growth and reduced genomic instability in naive hESCs.
- Published
- 2018
- Full Text
- View/download PDF
25. Single cell expression analysis uncouples transdifferentiation and reprogramming
- Author
-
Ben Lehner, Amy Guillaumet-Adkins, Clara Berenguer, Marta Gut, Bruno Di Stefano, Luisa de Andrés-Aguayo, Gustavo Rodriguez-Esteban, Thomas Graf, Holger Heyn, Maria Mendez Lago, Mirko Francesconi, and Ivo Gut
- Subjects
0303 health sciences ,Cell type ,Somatic cell ,Chemistry ,Cellular differentiation ,Cell ,Transdifferentiation ,Cell fate determination ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Induced pluripotent stem cell ,Reprogramming ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Many somatic cell types are plastic, having the capacity to convert into other specialized cells (transdifferentiation)(1) or into induced pluripotent stem cells (iPSCs, reprogramming)(2) in response to transcription factor over-expression. To explore what makes a cell plastic and whether these different cell conversion processes are coupled, we exposed bone marrow derived pre-B cells to two different transcription factor overexpression protocols that efficiently convert them either into macrophages or iPSCs and monitored the two processes over time using single cell gene expression analysis. We found that even in these highly efficient cell fate conversion systems, cells differ in both their speed and path of transdifferentiation and reprogramming. This heterogeneity originatesin two starting pre-B cell subpopulations,large pre-BII and the small pre-BII cells they normally differentiate into. The large cells transdifferentiate slowly but exhibit a high efficiency of iPSC reprogramming. In contrast, the small cells transdifferentiate rapidly but are highly resistant to reprogramming. Moreover, the large B cells induce a stronger transient granulocyte/macrophage progenitor (GMP)-like state, while the small B cells undergo a more direct conversion to the macrophage fate. The large cells are cycling and exhibit high Myc activity whereas the small cells are Myc low and mostly quiescent. The observed heterogeneity of the two cell conversion processes can therefore be traced to two closely related cell types in the starting population that exhibit different types of plasticity. These data show that a somatic cell’s propensity for either transdifferentiation and reprogramming can be uncoupled.One sentence summarySingle cell transcriptomics of cell conversions
- Published
- 2018
- Full Text
- View/download PDF
26. IEEE 1855™: The First IEEE Standard Sponsored by IEEE Computational Intelligence Society [Society Briefs]
- Author
-
Bruno Di Stefano, Autilia Vitiello, and Giovanni Acampora
- Subjects
Artificial Intelligence ,Computer science ,business.industry ,0202 electrical engineering, electronic engineering, information engineering ,020206 networking & telecommunications ,020201 artificial intelligence & image processing ,Computational intelligence ,02 engineering and technology ,Telecommunications ,business ,Theoretical Computer Science - Published
- 2016
- Full Text
- View/download PDF
27. TBR2 antagonizes retinoic acid dependent neuronal differentiation by repressing ZFP423 during corticogenesis
- Author
-
Bruno Di Stefano, Lisbeth Flores-Garcia, Mattia Zaghi, Luca Massimino, Gaia Colasante, Bruce A. Hamilton, Cecilia Icoresi-Mazzeo, and Alessandro Sessa
- Subjects
0301 basic medicine ,Cellular differentiation ,Organogenesis ,Retinoic acid ,Tretinoin ,Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Cell Line, Tumor ,medicine ,Animals ,Progenitor cell ,Molecular Biology ,Transcription factor ,Cerebral Cortex ,Cell Differentiation ,Cell Biology ,Cell cycle ,Neural stem cell ,Cell biology ,DNA-Binding Proteins ,Corticogenesis ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Cerebral cortex ,T-Box Domain Proteins ,030217 neurology & neurosurgery ,Developmental Biology ,Signal Transduction ,Transcription Factors - Abstract
During cerebral cortex development, neural progenitors are required to elaborate a variety of cell differentiation signals to which they are continuously exposed. RA acid is a potent inducer of neuronal differentiation as it was found to influence cortical development. We report herein that TBR2, a transcription factor specific to Intermediate (Basal) Neural Progenitors (INPs), represses activation of the RA responsive element and expression of RA target genes in cell lines. This repressive action on RA signaling was functionally confirmed by the decrease of RA-mediated neuronal differentiation in neural stem cells stably overexpressing TBR2. In vivo mapping of RA activity in the developing cortex indicated that RA activity is detected in radial glial cells and subsequently downregulated in INPs, revealing a fine cell-type specific regulation of its signaling. Thus, TBR2 might be a molecular player in opposing RA signaling in INPs. Interestingly, this negative regulation is achieved at least in part by directly repressing the critical nuclear RA co-factor ZFP423. Indeed, we found ZFP423 to be expressed in the developing cortex and promote RA-dependent neuronal differentiation. These data indicate that TBR2 contributes to suppressing RA signaling in INPs, thereby enabling them to re-enter the cell cycle and delay neuronal differentiation.
- Published
- 2018
28. Direct Reprogramming of Mouse Fibroblasts into Functional Skeletal Muscle Progenitors
- Author
-
Konrad Hochedlinger, Priscilla Cheung, Amy Coffey, A. Almada, Sylvain Paisant, Duygu Payzin-Dogru, Mattia F. M. Gerli, Harald C. Ott, Aaron J. Huebner, Amy Galvin, Anthony Anselmo, Ori Bar-Nur, Amy J. Wagers, Bruno Di Stefano, Shahragim Tajbakhsh, Michael A. Rudnicki, Peter Feige, Cassandra Verheul, and Ruslan I. Sadreyev
- Subjects
0301 basic medicine ,Skeletal muscle ,Satellite cells ,Direct lineage reprogramming ,Induced muscle progenitor cells ,MyoD ,Pax7 ,Small molecules ,Transplantation ,Muscular dystrophy ,Muscle Fibers, Skeletal ,Muscle Development ,Biochemistry ,Mice ,Myocyte ,Transgenes ,Cell Self Renewal ,Stem Cell Niche ,lcsh:QH301-705.5 ,lcsh:R5-920 ,Stem Cells ,PAX7 Transcription Factor ,Cell Differentiation ,musculoskeletal system ,Cellular Reprogramming ,Cell biology ,medicine.anatomical_structure ,MYF5 ,Stem cell ,lcsh:Medicine (General) ,tissues ,Reprogramming ,Cell type ,Satellite Cells, Skeletal Muscle ,Biology ,Article ,Small Molecule Libraries ,03 medical and health sciences ,Genetics ,medicine ,Animals ,Regeneration ,Progenitor cell ,Muscle, Skeletal ,MyoD Protein ,Cell Biology ,Fibroblasts ,Muscular Dystrophy, Animal ,030104 developmental biology ,lcsh:Biology (General) ,Biomarkers ,Developmental Biology ,Stem Cell Transplantation - Abstract
Summary Skeletal muscle harbors quiescent stem cells termed satellite cells and proliferative progenitors termed myoblasts, which play pivotal roles during muscle regeneration. However, current technology does not allow permanent capture of these cell populations in vitro. Here, we show that ectopic expression of the myogenic transcription factor MyoD, combined with exposure to small molecules, reprograms mouse fibroblasts into expandable induced myogenic progenitor cells (iMPCs). iMPCs express key skeletal muscle stem and progenitor cell markers including Pax7 and Myf5 and give rise to dystrophin-expressing myofibers upon transplantation in vivo. Notably, a subset of transplanted iMPCs maintain Pax7 expression and sustain serial regenerative responses. Similar to satellite cells, iMPCs originate from Pax7+ cells and require Pax7 itself for maintenance. Finally, we show that myogenic progenitor cell lines can be established from muscle tissue following small-molecule exposure alone. This study thus reports on a robust approach to derive expandable myogenic stem/progenitor-like cells from multiple cell types., Graphical Abstract, Highlights • MyoD and small molecules reprogram fibroblasts to myogenic progenitors termed iMPCs • iMPCs self-renew and express key satellite cell and myoblast markers • iMPC growth is driven by Pax7+ cells and requires Pax7 gene function • Transplanted iMPCs engraft and sustain muscle regeneration in vivo, In this article, Hochedlinger and colleagues reprogrammed mouse fibroblasts into induced myogenic progenitors (iMPCs) by transient expression of MyoD and treatment with small molecules. iMPCs can be extensively propagated in vitro and exhibit skeletal muscle stem/progenitor cell characteristics, including the requirement for Pax7 function as well as the ability to sustain muscle regeneration upon repeated injury.
- Published
- 2018
- Full Text
- View/download PDF
29. Logical modeling of lymphoid and myeloid cell specification and transdifferentiation
- Author
-
Wassim Abou-Jaoudé, Morgane Thomas-Chollier, Samuel Collombet, Bruno Di Stefano, Jose Luis Sardina Ortega, Thomas Graf, Chris van Oevelen, Denis Thieffry, Institut de biologie de l'ENS Paris (IBENS), Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Département de Biologie - ENS Paris, École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut de biologie de l'ENS Paris (UMR 8197/1024) (IBENS), Département de Biologie - ENS Paris, and Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Myeloid ,Cellular differentiation ,Gene regulatory network ,Biology ,Sackler Colloquium on Gene Regulatory Networks and Network Models in Development and Evolution ,Models, Biological ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Gene Regulatory Networks ,Myeloid Cells ,Lymphocytes ,Transcription factor ,Genetics ,B-Lymphocytes ,Multidisciplinary ,Macrophages ,Transdifferentiation ,Cell Differentiation ,[SDV.BIBS]Life Sciences [q-bio]/Quantitative Methods [q-bio.QM] ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Cell Transdifferentiation ,Stem cell ,Reprogramming ,030217 neurology & neurosurgery - Abstract
International audience; Blood cells are derived from a common set of hematopoietic stem cells, which differentiate into more specific progenitors of the myeloid and lymphoid lineages, ultimately leading to differentiated cells. This developmental process is controlled by a complex regulatory network involving cytokines and their receptors, transcription factors, and chromatin remodelers. Using public data and data from our own molecular genetic experiments (quantitative PCR, Western blot, EMSA) or genome-wide assays (RNA-sequencing, ChIP-sequencing), we have assembled a comprehensive regulatory network encompassing the main transcription factors and signaling components involved in myeloid and lymphoid development. Focusing on B-cell and macrophage development, we defined a qualitative dynamical model recapitulating cytokine-induced differentiation of common progenitors, the effect of various reported gene knockdowns, and the reprogramming of pre-B cells into macrophages induced by the ectopic expression of specific transcription factors. The resulting network model can be used as a template for the integration of new hematopoietic differentiation and transdifferentiation data to foster our understanding of lymphoid/myeloid cell-fate decisions.
- Published
- 2017
- Full Text
- View/download PDF
30. IEEE 1849: The XES Standard: The Second IEEE Standard Sponsored by IEEE Computational Intelligence Society [Society Briefs]
- Author
-
Eric Verbeek, CW Christian Günther, Giovanni Acampora, Wil M. P. van der Aalst, Bruno Di Stefano, Autilia Vitiello, Acampora, Giovanni, Vitiello, Autilia, Di Stefano, Bruno, van der Aalst, Wil, Gunther, Christian, and Verbeek, Eric
- Subjects
IEEE 802.1AE ,IEEE 802.11u ,IEEE 802.11w-2009 ,Computer science ,business.industry ,DySPAN ,Computational intelligence ,02 engineering and technology ,010501 environmental sciences ,IEEE 802.6 ,01 natural sciences ,IEEE floating point ,Theoretical Computer Science ,Artificial Intelligence ,0202 electrical engineering, electronic engineering, information engineering ,020201 artificial intelligence & image processing ,Telecommunications ,business ,0105 earth and related environmental sciences - Published
- 2017
31. Cell reprogramming: Brain versus brawn
- Author
-
Bruno, Di Stefano and Konrad, Hochedlinger
- Subjects
Neurons ,Sequence Analysis, RNA ,Animals ,Fibroblasts ,Single-Cell Analysis ,Cellular Reprogramming ,Article - Abstract
Direct lineage reprogramming represents a remarkable conversion of cellular and transcriptome states1–3. However, the intermediates through which individual cells progress are largely undefined. Here we used single-cell RNA-seq4–7 at multiple time points to dissect direct reprogramming from mouse embryonic fibroblasts (MEFs) to induced neuronal (iN) cells. By deconstructing heterogeneity at each time point and ordering cells by transcriptome similarity, we find that the molecular reprogramming path is remarkably continuous. Overexpression of the proneural pioneer factor Ascl1 results in a well-defined initialization, causing cells to exit the cell cycle and re-focus gene expression through distinct neural transcription factors. The initial transcriptional response is relatively homogeneous among fibroblasts suggesting the early steps are not limiting for productive reprogramming. Instead, the later emergence of a competing myogenic program and variable transgene dynamics over time appear to be the major efficiency limits of direct reprogramming. Moreover, a transcriptional state, distinct from donor and target cell programs, is transiently induced in cells undergoing productive reprogramming. Our data provide a high-resolution approach for understanding transcriptome states during lineage differentiation.
- Published
- 2016
32. Rapid Generation of Induced Pluripotent Stem Cells from Mouse Pre-B Cells
- Author
-
Bruno Di Stefano and Thomas Graf
- Subjects
General Earth and Planetary Sciences ,General Environmental Science - Published
- 2016
- Full Text
- View/download PDF
33. Importance of Shank3 Protein in Regulating Metabotropic Glutamate Receptor 5 (mGluR5) Expression and Signaling at Synapses
- Author
-
Renato Mantegazza, Michael Schoen, Chiara Verpelli, Bruno Di Stefano, Alexander Dityatev, Michela Chiappalone, Vania Broccoli, Elena Dvoretskova, Tobias M. Böckers, Carlo Sala, Cinzia Vicidomini, and Francesca Rossi
- Subjects
Dendritic spine ,Dendritic Spines ,Receptor, Metabotropic Glutamate 5 ,Nerve Tissue Proteins ,Receptor Regulation ,Glutamate Receptors Ionotropic (AMPA, NMDA) ,Neurotransmission ,Biology ,Receptors, Metabotropic Glutamate ,Hippocampus ,Models, Biological ,Receptors, N-Methyl-D-Aspartate ,Biochemistry ,Mice ,Neurobiology ,Mental Retardation ,shRNA ,Synaptic augmentation ,mental disorders ,Animals ,Receptors, AMPA ,Phosphorylation ,Long-term depression ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Neurons ,Metabotropic glutamate receptor 5 ,Cell Biology ,Rats ,Cell biology ,Mice, Inbred C57BL ,Postsynaptic Density ,nervous system ,Metabotropic glutamate receptor ,Synapses ,Synaptic plasticity ,Postsynaptic density ,Glutamate Receptors Metabotropic ,Signal Transduction - Abstract
Shank3/PROSAP2 gene mutations are associated with cognitive impairment ranging from mental retardation to autism. Shank3 is a large scaffold postsynaptic density protein implicated in dendritic spines and synapse formation; however, its specific functions have not been clearly demonstrated. We have used RNAi to knockdown Shank3 expression in neuronal cultures and showed that this treatment specifically reduced the synaptic expression of the metabotropic glutamate receptor 5 (mGluR5), but did not affect the expression of other major synaptic proteins. The functional consequence of Shank3 RNAi knockdown was impaired signaling via mGluR5, as shown by reduction in ERK1/2 and CREB phosphorylation induced by stimulation with (S)-3,5-dihydroxyphenylglycine (DHPG) as the agonist of mGluR5 receptors, impaired mGluR5-dependent synaptic plasticity (DHPG-induced long-term depression), and impaired mGluR5-dependent modulation of neural network activity. We also found morphological abnormalities in the structure of synapses (spine number, width, and length) and impaired glutamatergic synaptic transmission, as shown by reduction in the frequency of miniature excitatory postsynaptic currents (mEPSC). Notably, pharmacological augmentation of mGluR5 activity using 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)-benzamide as the positive allosteric modulator of these receptors restored mGluR5-dependent signaling (DHPG-induced phosphorylation of ERK1/2) and normalized the frequency of mEPSCs in Shank3-knocked down neurons. These data demonstrate that a deficit in mGluR5-mediated intracellular signaling in Shank3 knockdown neurons can be compensated by 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)-benzamide; this raises the possibility that pharmacological augmentation of mGluR5 activity represents a possible new therapeutic approach for patients with Shank3 mutations.
- Published
- 2011
- Full Text
- View/download PDF
34. Transcription Factors Drive Tet2-Mediated Enhancer Demethylation to Reprogram Cell Fate
- Author
-
Marta Gut, Clara Berenguer, Antonio Gomez, Tian V. Tian, Luciano Di Croce, Denis Thieffry, Bruno Di Stefano, Carolina Segura-Morales, Thomas Graf, Ralph Stadhouders, Jose Luis Sardina, Konrad Hochedlinger, Sergi Aranda, Ivo Gut, Justin Brumbaugh, Samuel Collombet, Simon Heath, and Pulmonary Medicine
- Subjects
Male ,0301 basic medicine ,DNA Hydroxymethylation ,Induced Pluripotent Stem Cells ,Biology ,Article ,Dioxygenases ,Kruppel-Like Factor 4 ,Mice ,03 medical and health sciences ,Proto-Oncogene Proteins ,Genetics ,Animals ,Epigenetics ,Cells, Cultured ,Mice, Knockout ,Pioneer factor ,Cell Biology ,Methylation ,DNA Methylation ,Fibroblasts ,Cellular Reprogramming ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Enhancer Elements, Genetic ,030104 developmental biology ,DNA methylation ,biology.protein ,Molecular Medicine ,Demethylase ,Female ,Reprogramming ,Transcription Factors - Abstract
Here, we report DNA methylation and hydroxymethylation dynamics at nucleotide resolution using C/EBPα-enhanced reprogramming of B cells into induced pluripotent cells (iPSCs). We observed successive waves of hydroxymethylation at enhancers, concomitant with a decrease in DNA methylation, suggesting active demethylation. Consistent with this finding, ablation of the DNA demethylase Tet2 almost completely abolishes reprogramming. C/EBPα, Klf4, and Tfcp2l1 each interact with Tet2 and recruit the enzyme to specific DNA sites. During reprogramming, some of these sites maintain high levels of 5hmC, and enhancers and promoters of key pluripotency factors become demethylated as early as 1 day after Yamanaka factor induction. Surprisingly, methylation changes precede chromatin opening in distinct chromatin regions, including Klf4 bound sites, revealing a pioneer factor activity associated with alteration in DNA methylation. Rapid changes in hydroxymethylation similar to those in B cells were also observed during compound-accelerated reprogramming of fibroblasts into iPSCs, highlighting the generality of our observations. Using a highly efficient reprogramming system, Sardina et al. examined the dynamics of DNA methylation and hydroxymethylation. They found that throughout the process several transcription factors can recruit Tet2 to specific sites, leading to demethylation. Some of these sites became demethylated before chromatin opening.
- Published
- 2018
- Full Text
- View/download PDF
35. Constitutively Active SMAD2/3 Are Broad-Scope Potentiators of Transcription-Factor-Mediated Cellular Reprogramming
- Author
-
Tyson, Ruetz, Ulrich, Pfisterer, Bruno, Di Stefano, James, Ashmore, Meryam, Beniazza, Tian V, Tian, Daniel F, Kaemena, Luca, Tosti, Wenfang, Tan, Jonathan R, Manning, Eleni, Chantzoura, Daniella Rylander, Ottosson, Samuel, Collombet, Anna, Johnsson, Erez, Cohen, Kosuke, Yusa, Sten, Linnarsson, Thomas, Graf, Malin, Parmar, and Keisuke, Kaji
- Subjects
Induced Pluripotent Stem Cells ,Humans ,Smad2 Protein ,Smad3 Protein ,Cellular Reprogramming ,Cell Line ,Transcription Factors - Abstract
Reprogramming of cellular identity using exogenous expression of transcription factors (TFs) is a powerful and exciting tool for tissue engineering, disease modeling, and regenerative medicine. However, generation of desired cell types using this approach is often plagued by inefficiency, slow conversion, and an inability to produce mature functional cells. Here, we show that expression of constitutively active SMAD2/3 significantly improves the efficiency of induced pluripotent stem cell (iPSC) generation by the Yamanaka factors. Mechanistically, SMAD3 interacts with reprogramming factors and co-activators and co-occupies OCT4 target loci during reprogramming. Unexpectedly, active SMAD2/3 also markedly enhances three other TF-mediated direct reprogramming conversions, from B cells to macrophages, myoblasts to adipocytes, and human fibroblasts to neurons, highlighting broad and general roles for SMAD2/3 as cell-reprogramming potentiators. Our results suggest that co-expression of active SMAD2/3 could enhance multiple types of TF-based cell identity conversion and therefore be a powerful tool for cellular engineering.
- Published
- 2016
36. C/EBPα creates elite cells for iPSC reprogramming by upregulating Klf4 and increasing the levels of Lsd1 and Brd4
- Author
-
Janus S. Jakobsen, Francesco Limone, Bruno Di Stefano, Bo T. Porse, Mirko Francesconi, Matthias Mann, Samuel Collombet, Denis Thieffry, Carolina Segura-Morales, Michael Wierer, Jose Luis Sardina, Thomas Graf, Ralph Stadhouders, and Andreas Lackner
- Subjects
0301 basic medicine ,Male ,Proteomics ,Blotting, Western ,Induced Pluripotent Stem Cells ,Kruppel-Like Transcription Factors ,Biology ,Cell Line ,03 medical and health sciences ,Kruppel-Like Factor 4 ,Mice ,Downregulation and upregulation ,CCAAT-Enhancer-Binding Protein-alpha ,Animals ,Humans ,Induced pluripotent stem cell ,Cells, Cultured ,Histone Demethylases ,Induced stem cells ,B-Lymphocytes ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,HEK 293 cells ,Nuclear Proteins ,Mouse Embryonic Stem Cells ,Cell Biology ,Cellular Reprogramming ,Cell biology ,Up-Regulation ,Mice, Inbred C57BL ,030104 developmental biology ,Gene Ontology ,HEK293 Cells ,Cell culture ,KLF4 ,Cancer research ,Female ,Signal transduction ,Reprogramming ,Transcription Factors - Abstract
Reprogramming somatic cells into induced pluripotent stem cells (iPSCs) is typically inefficient and has been explained by elite-cell and stochastic models. We recently reported that B cells exposed to a pulse of C/EBPα (Bα' cells) behave as elite cells, in that they can be rapidly and efficiently reprogrammed into iPSCs by the Yamanaka factors OSKM. Here we show that C/EBPα post-transcriptionally increases the abundance of several hundred proteins, including Lsd1, Hdac1, Brd4, Med1 and Cdk9, components of chromatin-modifying complexes present at super-enhancers. Lsd1 was found to be required for B cell gene silencing and Brd4 for the activation of the pluripotency program. C/EBPα also promotes chromatin accessibility in pluripotent cells and upregulates Klf4 by binding to two haematopoietic enhancers. Bα' cells share many properties with granulocyte/macrophage progenitors, naturally occurring elite cells that are obligate targets for leukaemic transformation, whose formation strictly requires C/EBPα.
- Published
- 2015
37. C/EBPα creates elite cells for iPSC reprogramming by upregulating Klf4 and increasing the levels of Lsd1 and Brd4
- Author
-
Jose Luis Sardina, Bruno Di Stefano, and Thomas Graf
- Subjects
0301 basic medicine ,Induced stem cells ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Transdifferentiation ,Cell Differentiation ,Cell Biology ,Editorials: Cell Cycle Features ,Biology ,Cellular Reprogramming ,Embryonic stem cell ,Cell Line ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,Immunology ,CCAAT-Enhancer-Binding Protein-alpha ,Stem cell ,Induced pluripotent stem cell ,Molecular Biology ,Reprogramming ,Developmental Biology - Abstract
Much of what is known about how mammalian cells decide what to become derives from the study of transcription factor (TF)-induced transdifferentiation and reprogramming into induced pluripotent stem (iPS) cells. Among the plethora of transdifferentiation systems described1 the C/EBPα-induced conversion of lymphoid progenitors as well as of mature B cells into macrophages is probably the most efficient, reaching 100%.2 This lineage switch does not involve an overt retrodifferentiation into embryonic stem cells (ESCs) or haematopoietic stem cells, but the transient activation of genes specific for granulocyte/macrophage precursors (GMPs)2 (and see below).
- Published
- 2016
- Full Text
- View/download PDF
38. Brain versus brawn
- Author
-
Konrad Hochedlinger and Bruno Di Stefano
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,Multidisciplinary ,medicine.anatomical_structure ,Single-cell analysis ,Cell ,medicine ,Stem cell ,Biology ,Reprogramming ,Mature cell ,Cell biology - Abstract
The mechanisms that underlie enforced transitions between mature cell lineages are poorly understood. Profiling single skin cells that are induced to become neurons reveals that, unexpectedly, they often become muscle. See Letter p.391
- Published
- 2016
- Full Text
- View/download PDF
39. Nudt21 Controls Cell Fate by Connecting Alternative Polyadenylation to Chromatin Signaling
- Author
-
Fei Ji, Ruslan I. Sadreyev, Benjamin A. Schwarz, Bruno Di Stefano, Katie J. Clowers, Steven P. Gygi, Yongsheng Shi, Elmira Forouzmand, Justin Brumbaugh, Stephen J. Elledge, Konrad Hochedlinger, Marian Kalocsay, Marti Borkent, Guang Hu, Xiuye Wang, and Yue Chen
- Subjects
0301 basic medicine ,Biology ,Cell fate determination ,Polyadenylation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,Mice ,03 medical and health sciences ,Animals ,Humans ,Induced pluripotent stem cell ,Cells, Cultured ,Embryonic Stem Cells ,Cleavage And Polyadenylation Specificity Factor ,Transdifferentiation ,Cellular Reprogramming ,Chromatin Assembly and Disassembly ,Embryonic stem cell ,Chromatin ,Cell biology ,HEK293 Cells ,030104 developmental biology ,Stem cell ,Reprogramming ,Signal Transduction - Abstract
Cell fate transitions involve rapid gene expression changes and global chromatin remodeling, yet the underlying regulatory pathways remain incompletely understood. Here, we identified the RNA-processing factor Nudt21 as a novel regulator of cell fate change using transcription-factor-induced reprogramming as a screening assay. Suppression of Nudt21 enhanced the generation of induced pluripotent stem cells, facilitated transdifferentiation into trophoblast stem cells, and impaired differentiation of myeloid precursors and embryonic stem cells, suggesting a broader role for Nudt21 in cell fate change. We show that Nudt21 directs differential polyadenylation of over 1,500 transcripts in cells acquiring pluripotency, although only a fraction changed protein levels. Remarkably, these proteins were strongly enriched for chromatin regulators, and their suppression neutralized the effect of Nudt21 during reprogramming. Collectively, our data uncover Nudt21 as a novel post-transcriptional regulator of cell fate and establish a direct, previously unappreciated link between alternative polyadenylation and chromatin signaling.
- Published
- 2018
- Full Text
- View/download PDF
40. Transcription factor induced reprogramming of B cells
- Author
-
Ralph Stadhouders, Bruno Di Stefano, Ben Lehner, Johanna Goldmann, Jose Luis Sardina, Thomas Graf, Gregoire Stik, Mirko Francesconi, and Samuel Collombet
- Subjects
Cancer Research ,Genetics ,Cell Biology ,Hematology ,Biology ,Molecular Biology ,Reprogramming ,Transcription factor ,Cell biology - Published
- 2017
- Full Text
- View/download PDF
41. Very Rapid and Efficient Generation of Induced Pluripotent Stem Cells from Mouse Pre-B Cells
- Author
-
Bruno, Di Stefano and Thomas, Graf
- Subjects
Precursor Cells, B-Lymphoid ,SOXB1 Transcription Factors ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Genes, myc ,Kruppel-Like Transcription Factors ,Gene Expression ,Cell Differentiation ,Fibroblasts ,Cellular Reprogramming ,Antigens, Differentiation ,Culture Media ,Proto-Oncogene Proteins c-myc ,Kruppel-Like Factor 4 ,Mice ,Doxycycline ,CCAAT-Enhancer-Binding Proteins ,Animals ,Cellular Reprogramming Techniques ,Stromal Cells ,Octamer Transcription Factor-3 ,Cells, Cultured - Abstract
One of the major obstacles in generating induced pluripotent stem (iPS) cells suitable for therapeutic application is the low efficiency of the process and the long time required, with many iPS lines acquiring genomic aberrations. In this chapter we describe a highly efficient iPS reprogramming system based on the transient expression in pre-B cells of the transcription factor C/EBPα, followed by the induction of the four Yamanaka factors (OSKM). In addition, the process is very rapid, yielding Oct4 positive cells within 2 days and Nanog-positive iPS cell colonies within a week.
- Published
- 2014
42. Time-resolved gene expression profiling during reprogramming of C/EBPα-pulsed B cells into iPS cells
- Author
-
Samuel Collombet, Thomas Graf, and Bruno Di Stefano
- Subjects
Statistics and Probability ,Data Descriptor ,Somatic cell ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Gene Expression ,Biology ,Library and Information Sciences ,Education ,Mice ,Animals ,Induced pluripotent stem cell ,B-Lymphocytes ,Induced stem cells ,Ccaat-enhancer-binding proteins ,Microarray analysis techniques ,Gene Expression Profiling ,Cell Differentiation ,Cellular Reprogramming ,Microarray Analysis ,Molecular biology ,Cell biology ,Computer Science Applications ,Gene expression profiling ,CCAAT-Enhancer-Binding Proteins ,Statistics, Probability and Uncertainty ,Reprogramming ,Information Systems - Abstract
The reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) is lengthy and inefficient. The development of a reprogramming system that allows rapid and synchronous reprogramming to pluripotency is imperative for understanding the mechanism of iPSC formation and for future therapeutic applications. We have recently reported that a short expression in mouse primary B cells of the transcription factor C/EBPα before the induction of pluripotency factors increases the iPSC reprogramming efficiency >100-fold, involving 95% of the cells within a week. Here we present a dataset containing the time course of gene expression during this process as determined by microarray and RNA-seq techniques.
- Published
- 2014
- Full Text
- View/download PDF
43. Very Rapid and Efficient Generation of Induced Pluripotent Stem Cells from Mouse Pre-B Cells
- Author
-
Bruno Di Stefano and Thomas Graf
- Subjects
SOX2 ,KLF4 ,Biology ,Pre-B-Cells ,Induced pluripotent stem cell ,Reprogramming ,Transcription factor ,Embryonic stem cell ,Cell biology - Abstract
One of the major obstacles in generating induced pluripotent stem (iPS) cells suitable for therapeutic application is the low efficiency of the process and the long time required, with many iPS lines acquiring genomic aberrations. In this chapter we describe a highly efficient iPS reprogramming system based on the transient expression in pre-B cells of the transcription factor C/EBPα, followed by the induction of the four Yamanaka factors (OSKM). In addition, the process is very rapid, yielding Oct4 positive cells within 2 days and Nanog-positive iPS cell colonies within a week.
- Published
- 2014
- Full Text
- View/download PDF
44. Polycomb complexes in stem cells and embryonic development
- Author
-
Luigi Aloia, Luciano Di Croce, and Bruno Di Stefano
- Subjects
Cellular differentiation ,Embryonic Development ,macromolecular substances ,Embryoid body ,Biology ,Histones ,Mice ,Animals ,Drosophila Proteins ,Induced pluripotent stem cell ,Promoter Regions, Genetic ,Molecular Biology ,Embryonic Stem Cells ,Genetics ,Polycomb Repressive Complex 1 ,Induced stem cells ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,Cellular Reprogramming ,Embryonic stem cell ,Cell biology ,Multipotent Stem Cell ,CpG Islands ,Drosophila ,Stem cell ,Developmental Biology ,Adult stem cell - Abstract
Polycomb group (PcG) proteins are epigenetic modifiers involved in controlling gene repression. Organized within multiprotein complexes, they regulate developmental genes in multiple cell types and tissue contexts, including embryonic and adult stem cells, and are essential for cell fate transitions and proper development. Here, we summarize recent breakthroughs that have revealed the diversity of PcG complexes acting in different cell types and genomic contexts. Intriguingly, it appears that particular PcG proteins have specific functions in embryonic development, in pluripotent stem cells and in reprogramming somatic cells into a pluripotent-like state. Finally, we highlight recent results from analyzing PcG protein functions in multipotent stem cells, such as neural, hematopoietic and epidermal stem cells.
- Published
- 2013
45. C/EBPα poises B cells for rapid reprogramming into induced pluripotent stem cells
- Author
-
Chris van Oevelen, Jun Lu, Guillermo P. Vicent, Jose Luis Sardina, Bruno Di Stefano, Denis Thieffry, Miguel Beato, Thomas Graf, Eric M Kallin, and Samuel Collombet
- Subjects
Epithelial-Mesenchymal Transition ,Induced Pluripotent Stem Cells ,Kruppel-Like Transcription Factors ,Biology ,Dioxygenases ,Proto-Oncogene Proteins c-myc ,Cytosine ,Kruppel-Like Factor 4 ,Mice ,SOX2 ,Enhancer binding ,Proto-Oncogene Proteins ,CCAAT-Enhancer-Binding Protein-alpha ,Animals ,Deoxyribonuclease I ,Induced pluripotent stem cell ,Transcription factor ,Cells, Cultured ,B-Lymphocytes ,Multidisciplinary ,SOXB1 Transcription Factors ,Transdifferentiation ,DNA Methylation ,Cellular Reprogramming ,Molecular biology ,Chromatin ,Up-Regulation ,DNA-Binding Proteins ,KLF4 ,Cell Transdifferentiation ,Reprogramming ,Octamer Transcription Factor-3 - Abstract
A pulse of C/EBPα followed by overexpression of the transcription factors Oct4, Sox2, Klf4 and Myc leads to fast and very efficient reprogramming of B cell precursors to induced pluripotent stem cells; C/EBPα facilitates transient chromatin accessibility and accelerates expression of pluripotency genes through a mechanism that involves activation of the Tet2 enzyme. A new study by Thomas Graf and colleagues describes how a pulse of C/EBPα (the transcription factor CCAAT/enhancer binding protein-α) followed by overexpression of the Yamanaka 'OSKM' reprogramming factors leads to fast and very efficient reprogramming of B-cell precursors to induced pluripotent stem (iPS) cells. The authors found that C/EBPα facilitates chromatin accessibility and accelerates expression of pluripotency genes through a mechanism that involves activation of the Tet2 enzyme. This demonstration of highly efficient and fast reprogramming of B cells into iPS cells provides a model for the study of the reprogramming process and may also have clinical relevance. CCAAT/enhancer binding protein-α (C/EBPα) induces transdifferentiation of B cells into macrophages at high efficiencies and enhances reprogramming into induced pluripotent stem (iPS) cells when co-expressed with the transcription factors Oct4 (Pou5f1), Sox2, Klf4 and Myc (hereafter called OSKM)1,2. However, how C/EBPα accomplishes these effects is unclear. Here we find that in mouse primary B cells transient C/EBPα expression followed by OSKM activation induces a 100-fold increase in iPS cell reprogramming efficiency, involving 95% of the population. During this conversion, pluripotency and epithelial–mesenchymal transition genes become markedly upregulated, and 60% of the cells express Oct4 within 2 days. C/EBPα acts as a ‘path-breaker’ as it transiently makes the chromatin of pluripotency genes more accessible to DNase I. C/EBPα also induces the expression of the dioxygenase Tet2 and promotes its translocation to the nucleus where it binds to regulatory regions of pluripotency genes that become demethylated after OSKM induction. In line with these findings, overexpression of Tet2 enhances OSKM-induced B-cell reprogramming. Because the enzyme is also required for efficient C/EBPα-induced immune cell conversion3, our data indicate that Tet2 provides a mechanistic link between iPS cell reprogramming and B-cell transdifferentiation. The rapid iPS reprogramming approach described here should help to fully elucidate the process and has potential clinical applications.
- Published
- 2013
46. Hi-TEC reprogramming for organ regeneration
- Author
-
Bruno Di Stefano and Thomas Graf
- Subjects
Ectopic thymus ,Transdifferentiation ,Cell ,FOXN1 ,Cell Biology ,Biology ,medicine.disease ,Regenerative medicine ,Cell biology ,Transplantation ,medicine.anatomical_structure ,medicine ,Transcription factor ,Reprogramming - Abstract
The induction of cell reprogramming by transcription factors into alternative cell fates opens new avenues for regenerative medicine. Thymic epithelial cells that were obtained from fibroblasts by Foxn1 overexpression support the formation of an ectopic thymus following transplantation.
- Published
- 2014
- Full Text
- View/download PDF
47. An ES-like pluripotent state in FGF-dependent murine iPS cells
- Author
-
Niels Geijsen, Federica Ungaro, Hsu-Hsin Chen, Christa Buecker, Bruno Di Stefano, Vania Broccoli, Alessandro Prigione, James Adjaye, Maureen Eijpe, Gustavo Mostoslavsky, Maaike Welling, and Paul J. Tesar
- Subjects
KOSR ,X Chromosome ,Cellular differentiation ,Green Fluorescent Proteins ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,lcsh:Medicine ,Mice, Transgenic ,Biology ,Fibroblast growth factor ,Embryo Culture Techniques ,Mice ,Species Specificity ,Animals ,lcsh:Science ,Induced pluripotent stem cell ,Embryonic Stem Cells ,Multidisciplinary ,Stem Cells ,lcsh:R ,Teratoma ,Fibroblasts ,Embryonic stem cell ,Molecular biology ,Cell biology ,Fibroblast Growth Factors ,Retroviridae ,Cell culture ,Epiblast ,Intercellular Signaling Peptides and Proteins ,lcsh:Q ,Stem cell ,Function and Dysfunction of the Nervous System ,Research Article ,Developmental Biology - Abstract
Recent data demonstrates that stem cells can exist in two morphologically, molecularly and functionally distinct pluripotent states; a naive LIF-dependent pluripotent state which is represented by murine embryonic stem cells (mESCs) and an FGF-dependent primed pluripotent state represented by murine and rat epiblast stem cells (EpiSCs). We find that derivation of induced pluripotent stem cells (iPSCs) under EpiSC culture conditions yields FGF-dependent iPSCs from hereon called FGF-iPSCs) which, unexpectedly, display naive ES-like/ICM properties. FGF-iPSCs display X-chromosome activation, multi-lineage differentiation, teratoma competence and chimera contribution in vivo. Our findings suggest that in 129 and Bl6 mouse strains, iPSCs can dominantly adopt a naive pluripotent state regardless of culture growth factor conditions. Characterization of the key molecular signalling pathways revealed FGF-iPSCs to depend on the Activin/Nodal and FGF pathways, while signalling through the JAK-STAT pathway is not required for FGF-iPS cell maintenance. Our findings suggest that in 129 and Bl6 mouse strains, iPSCs can dominantly adopt a naive pluripotent state regardless of culture growth factor conditions.
- Published
- 2010
48. Efficient genetic reprogramming of unmodified somatic neural progenitors uncovers the essential requirement of Oct4 and Klf4
- Author
-
Bruno Di Stefano, Alessandro Prigione, and Vania Broccoli
- Subjects
Pluripotent Stem Cells ,Somatic cell ,Cellular differentiation ,Virus Integration ,Kruppel-Like Transcription Factors ,Biology ,Kruppel-Like Factor 4 ,Mice ,SOX2 ,Animals ,Genetics ,Neurons ,Stem Cells ,Teratoma ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell Biology ,Hematology ,Cellular Reprogramming ,Embryonic stem cell ,Cell biology ,KLF4 ,Somatic cell nuclear transfer ,biological phenomena, cell phenomena, and immunity ,Reprogramming ,Octamer Transcription Factor-3 ,Developmental Biology ,Adult stem cell - Abstract
Significant breakthroughs have been recently achieved in reprogramming somatic cells to a pluripotent embryonic state by the ectopic expression of specific transcription factors. One of the major drawbacks of reprogramming strategies lays in the low efficiency of the process. It is likely that the required complex epigenetic-remodeling events could be cell-type specific and more rational approaches to cell source selection might help to improve the outcome of the procedure. Because the use of somatic stem cells, and specifically neural stem cells (NSCs), as nuclear donors significantly increased the efficiency of somatic cell nuclear transfer, we aimed to determine whether genetically unmodified somatic NSCs could be more easily reprogrammed to pluripotency than unmodified mouse embryonic fibroblasts. Retroviral transduction of the factors Oct4, Sox2, Klf4, and c-Myc successfully reverted NSCs to a pluripotent embryonic stem cell-like state with a 2-fold efficiency increase, faster kinetic, and with a lower number of viral integrations. Quantification analysis of reprogramming-associated genes revealed that NSCs endogenously expressed high levels of Sox2 and c-Myc. Accordingly, NSCs could be successfully induced to pluripotency through the ectopic viral expression of the other two factors (Oct4 and Klf4). These findings suggest that endogenous expression of reprogramming genes could help the reprogramming process and somatic stem cells might be more prone to reprogramming due to their specific genetic background. Genetic-based somatic cell screening might provide essential information for the selection of alternative cell sources more suitable to direct reprogramming.
- Published
- 2008
49. Leptin/HER2 crosstalk in breast cancer: in vitro study and preliminary in vivo analysis
- Author
-
Terrasi M, M. Franco Bonetti, Elena Fiorio, Veronica Parolin, R. Micciolo, Alessandra Auriemma, Gian Luigi Cetto, Antonio Giordano, Bruno Di Stefano, Annamaria Molino, Andrea Remo, Anna Mercanti, and Eva Surmacz
- Subjects
Leptin ,Transcriptional Activation ,medicine.medical_specialty ,Cancer Research ,Receptor, ErbB-2 ,Breast Neoplasms ,lcsh:RC254-282 ,Breast cancer ,Surgical oncology ,Risk Factors ,Internal medicine ,Cell Line, Tumor ,medicine ,Genetics ,Humans ,Obesity ,Receptor ,skin and connective tissue diseases ,neoplasms ,Leptin receptor ,business.industry ,Carcinoma, Ductal, Breast ,Receptor Cross-Talk ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Postmenopause ,Endocrinology ,Oncology ,Receptors, Leptin ,Female ,Signal transduction ,business ,Immunostaining ,Protein Binding ,Research Article - Abstract
Background Obesity in postmenopausal women is associated with increased breast cancer risk, development of more aggressive tumors and resistance to certain anti-breast cancer treatments. Some of these effects might be mediated by obesity hormone leptin, acting independently or modulating other signaling pathways. Here we focused on the link between leptin and HER2. We tested if HER2 and the leptin receptor (ObR) can be coexpressed in breast cancer cell models, whether these two receptors can physically interact, and whether leptin can transactivate HER2. Next, we studied if leptin/ObR can coexist with HER2 in breast cancer tissues, and if presence of these two systems correlates with specific clinicopathological features. Methods Expression of ObR, HER2, phospo-HER2 was assessed by immonoblotting. Physical interactions between ObR and HER2 were probed by immunoprecipitation and fluorescent immunostaining. Expression of leptin and ObR in breast cancer tissues was detected by immunohistochemistry (IHC). Associations among markers studied by IHC were evaluated using Fisher's exact test for count data. Results HER2 and ObR were coexpressed in all studied breast cancer cell lines. In MCF-7 cells, HER2 physically interacted with ObR and leptin treatment increased HER2 phosphorylation on Tyr 1248. In 59 breast cancers, the presence of leptin was correlated with ObR (the overall association was about 93%). This result was confirmed both in HER2-positive and in HER2-negative subgroups. The expression of leptin or ObR was numerically more frequent in larger (> 10 mm) tumors. Conclusion Coexpression of HER2 and the leptin/ObR system might contribute to enhanced HER2 activity and reduced sensitivity to anti-HER2 treatments.
- Published
- 2008
50. AllProbes_AllReplicates.xls
- Author
-
Bruno Di Stefano, Samuel Collombet, Thomas Graf, Bruno Di Stefano, Samuel Collombet, and Thomas Graf
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.