151
|
Dong Y, An IS, Ma L, An S. Welcome to a new era of Biomedical Dermatology. BIOMEDICAL DERMATOLOGY 2017. [DOI: 10.1186/s41702-017-0001-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
152
|
Direct conversion from skin fibroblasts to functional dopaminergic neurons for biomedical application. BIOMEDICAL DERMATOLOGY 2017. [DOI: 10.1186/s41702-017-0004-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
153
|
Landeira BS, Araújo JADM, Schroeder T, Müller U, Costa MR. Live Imaging of Primary Cerebral Cortex Cells Using a 2D Culture System. J Vis Exp 2017. [PMID: 28829410 DOI: 10.3791/56063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
During cerebral cortex development, progenitor cells undergo several rounds of symmetric and asymmetric cell divisions to generate new progenitors or postmitotic neurons. Later, some progenitors switch to a gliogenic fate, adding to the astrocyte and oligodendrocyte populations. Using time-lapse video-microscopy of primary cerebral cortex cell cultures, it is possible to study the cellular and molecular mechanisms controlling the mode of cell division and cell cycle parameters of progenitor cells. Similarly, the fate of postmitotic cells can be examined using cell-specific fluorescent reporter proteins or post-imaging immunocytochemistry. More importantly, all these features can be analyzed at the single-cell level, allowing the identification of progenitors committed to the generation of specific cell types. Manipulation of gene expression can also be performed using viral-mediated transfection, allowing the study of cell-autonomous and non-cell-autonomous phenomena. Finally, the use of fusion fluorescent proteins allows the study of symmetric and asymmetric distribution of selected proteins during division and the correlation with daughter cells fate. Here, we describe the time-lapse video-microscopy method to image primary cerebral cortex murine cells for up to several days and analyze the mode of cell division, cell cycle length and fate of newly generated cells. We also describe a simple method to transfect progenitor cells, which can be applied to manipulate genes of interest or simply label cells with reporter proteins.
Collapse
Affiliation(s)
| | | | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zurich
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University
| | - Marcos R Costa
- Brain Institute, Federal University of Rio Grande do Norte;
| |
Collapse
|
154
|
Vermunt MW, Creyghton MP. Transcriptional Dynamics at Brain Enhancers: from Functional Specialization to Neurodegeneration. Curr Neurol Neurosci Rep 2017; 16:94. [PMID: 27628759 PMCID: PMC5023742 DOI: 10.1007/s11910-016-0689-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Over the last decade, the noncoding part of the genome has been shown to harbour thousands of cis-regulatory elements, such as enhancers, that activate well-defined gene expression programs. Driven by the development of numerous techniques, many of these elements are now identified in multiple tissues and cell types, and their characteristics as well as importance in development and disease are becoming increasingly clear. Here, we provide an overview of the insights that were gained from the analysis of noncoding gene regulatory elements in the brain and describe their potential contribution to cell type specialization, brain function and neurodegenerative disease.
Collapse
Affiliation(s)
- Marit W Vermunt
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT, Utrecht, The Netherlands
| | - Menno P Creyghton
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT, Utrecht, The Netherlands.
| |
Collapse
|
155
|
Taguchi J, Yamada Y. In vivo reprogramming for tissue regeneration and organismal rejuvenation. Curr Opin Genet Dev 2017; 46:132-140. [PMID: 28779646 DOI: 10.1016/j.gde.2017.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/22/2017] [Accepted: 07/21/2017] [Indexed: 12/25/2022]
Abstract
Transcription factor-mediated reprogramming has enabled us to induce the fate conversion of somatic cells into other cell types. Although the study of reprogramming mostly occurs at the cellular level in vitro, previous studies have demonstrated that somatic cells are reprogrammable in multicellular organisms too. Recent studies using in vivo reprogramming have provided important insights on regenerative medicine for diseased organs. Moreover, similar studies have revealed unappreciated mechanisms in various biological phenomena, including tissue regeneration, aging, rejuvenation and cancer development in multicellular organisms. Here, we review recent progress and future perspectives of in vivo reprogramming.
Collapse
Affiliation(s)
- Jumpei Taguchi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yasuhiro Yamada
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
156
|
Li H, Chen G. In Vivo Reprogramming for CNS Repair: Regenerating Neurons from Endogenous Glial Cells. Neuron 2017; 91:728-738. [PMID: 27537482 DOI: 10.1016/j.neuron.2016.08.004] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neuroregeneration in the CNS has proven to be difficult despite decades of research. The old dogma that CNS neurons cannot be regenerated in the adult mammalian brain has been overturned; however, endogenous adult neurogenesis appears to be insufficient for brain repair. Stem cell therapy once held promise for generating large quantities of neurons in the CNS, but immunorejection and long-term functional integration remain major hurdles. In this Perspective, we discuss the use of in vivo reprogramming as an emerging technology to regenerate functional neurons from endogenous glial cells inside the brain and spinal cord. Besides the CNS, in vivo reprogramming has been demonstrated successfully in the pancreas, heart, and liver and may be adopted in other organs. Although challenges remain for translating this technology into clinical therapies, we anticipate that in vivo reprogramming may revolutionize regenerative medicine by using a patient's own internal cells for tissue repair.
Collapse
Affiliation(s)
- Hedong Li
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Gong Chen
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
157
|
Pino A, Fumagalli G, Bifari F, Decimo I. New neurons in adult brain: distribution, molecular mechanisms and therapies. Biochem Pharmacol 2017; 141:4-22. [PMID: 28690140 DOI: 10.1016/j.bcp.2017.07.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/05/2017] [Indexed: 12/16/2022]
Abstract
"Are new neurons added in the adult mammalian brain?" "Do neural stem cells activate following CNS diseases?" "How can we modulate their activation to promote recovery?" Recent findings in the field provide novel insights for addressing these questions from a new perspective. In this review, we will summarize the current knowledge about adult neurogenesis and neural stem cell niches in healthy and pathological conditions. We will first overview the milestones that have led to the discovery of the classical ventricular and hippocampal neural stem cell niches. In adult brain, new neurons originate from proliferating neural precursors located in the subventricular zone of the lateral ventricles and in the subgranular zone of the hippocampus. However, recent findings suggest that new neuronal cells can be added to the adult brain by direct differentiation (e.g., without cell proliferation) from either quiescent neural precursors or non-neuronal cells undergoing conversion or reprogramming to neuronal fate. Accordingly, in this review we will also address critical aspects of the newly described mechanisms of quiescence and direct conversion as well as the more canonical activation of the neurogenic niches and neuroblast reservoirs in pathological conditions. Finally, we will outline the critical elements involved in neural progenitor proliferation, neuroblast migration and differentiation and discuss their potential as targets for the development of novel therapeutic drugs for neurodegenerative diseases.
Collapse
Affiliation(s)
- Annachiara Pino
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Italy
| | - Guido Fumagalli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy.
| | - Ilaria Decimo
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Italy.
| |
Collapse
|
158
|
Gascón S, Masserdotti G, Russo GL, Götz M. Direct Neuronal Reprogramming: Achievements, Hurdles, and New Roads to Success. Cell Stem Cell 2017; 21:18-34. [DOI: 10.1016/j.stem.2017.06.011] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
159
|
The doublesex-related Dmrta2 safeguards neural progenitor maintenance involving transcriptional regulation of Hes1. Proc Natl Acad Sci U S A 2017; 114:E5599-E5607. [PMID: 28655839 DOI: 10.1073/pnas.1705186114] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mechanisms that determine whether a neural progenitor cell (NPC) reenters the cell cycle or exits and differentiates are pivotal for generating cells in the correct numbers and diverse types, and thus dictate proper brain development. Combining gain-of-function and loss-of-function approaches in an embryonic stem cell-derived cortical differentiation model, we report that doublesex- and mab-3-related transcription factor a2 (Dmrta2, also known as Dmrt5) plays an important role in maintaining NPCs in the cell cycle. Temporally controlled expression of transgenic Dmrta2 in NPCs suppresses differentiation without affecting their neurogenic competence. In contrast, Dmrta2 knockout accelerates the cell cycle exit and differentiation into postmitotic neurons of NPCs derived from embryonic stem cells and in Emx1-cre conditional mutant mice. Dmrta2 function is linked to the regulation of Hes1 and other proneural genes, as demonstrated by genome-wide RNA-seq and direct binding of Dmrta2 to the Hes1 genomic locus. Moreover, transient Hes1 expression rescues precocious neurogenesis in Dmrta2 knockout NPCs. Our study thus establishes a link between Dmrta2 modulation of Hes1 expression and the maintenance of NPCs during cortical development.
Collapse
|
160
|
Boda E, Nato G, Buffo A. Emerging pharmacological approaches to promote neurogenesis from endogenous glial cells. Biochem Pharmacol 2017. [PMID: 28647491 DOI: 10.1016/j.bcp.2017.06.129] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neurodegenerative disorders are emerging as leading contributors to the global disease burden. While some drug-based approaches have been designed to limit or prevent neuronal loss following acute damage or chronic neurodegeneration, regeneration of functional neurons in the adult Central Nervous System (CNS) still remains an unmet need. In this context, the exploitation of endogenous cell sources has recently gained an unprecedented attention, thanks to the demonstration that, in some CNS regions or under specific circumstances, glial cells can activate spontaneous neurogenesis or can be instructed to produce neurons in the adult mammalian CNS parenchyma. This field of research has greatly advanced in the last years and identified interesting molecular and cellular mechanisms guiding the neurogenic activation/conversion of glia. In this review, we summarize the evolution of the research devoted to understand how resident glia can be directed to produce neurons. We paid particular attention to pharmacologically-relevant approaches exploiting the modulation of niche-associated factors and the application of selected small molecules.
Collapse
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, I-10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, I-10043 Orbassano, Turin, Italy.
| | - Giulia Nato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, I-10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, I-10043 Orbassano, Turin, Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, I-10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, I-10043 Orbassano, Turin, Italy
| |
Collapse
|
161
|
The novel tool of cell reprogramming for applications in molecular medicine. J Mol Med (Berl) 2017; 95:695-703. [PMID: 28597071 PMCID: PMC5487694 DOI: 10.1007/s00109-017-1550-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/15/2017] [Accepted: 05/19/2017] [Indexed: 01/06/2023]
Abstract
Recent discoveries in the field of stem cell biology have enabled scientists to “reprogram” cells from one type to another. For example, it is now possible to place adult skin or blood cells in a dish and convert them into neurons, liver, or heart cells. It is also possible to literally “rejuvenate” adult cells by reprogramming them into embryonic-like stem cells, which in turn can be differentiated into every tissue and cell type of the human body. Our ability to reprogram cell types has four main implications for medicine: (1) scientists can now take skin or blood cells from patients and convert them to other cells to study disease processes. This disease modeling approach has the advantage over animal models because it is directly based on human patient cells. (2) Reprogramming could also be used as a “clinical trial in a dish” to evaluate the general efficacy and safety of newly developed drugs on human patient cells before they would be tested in animal models or people. (3) In addition, many drugs have deleterious side effects like heart arrhythmias in only a small and unpredictable subpopulation of patients. Reprogramming could facilitate precision medicine by testing the safety of already approved drugs first on reprogrammed patient cells in a personalized manner prior to administration. For example, drugs known to sometimes cause arrhythmias could be first tested on reprogrammed heart cells from individual patients. (4) Finally, reprogramming allows the generation of new tissues that could be grafted therapeutically to regenerate lost or damaged cells.
Collapse
|
162
|
Gao C, Wang Q, Chung SK, Shen J. Crosstalk of metabolic factors and neurogenic signaling in adult neurogenesis: Implication of metabolic regulation for mental and neurological diseases. Neurochem Int 2017; 106:24-36. [DOI: 10.1016/j.neuint.2017.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 01/10/2017] [Accepted: 02/03/2017] [Indexed: 12/31/2022]
|
163
|
Induction of functional dopamine neurons from human astrocytes in vitro and mouse astrocytes in a Parkinson's disease model. Nat Biotechnol 2017; 35:444-452. [PMID: 28398344 DOI: 10.1038/nbt.3835] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 02/22/2017] [Indexed: 12/22/2022]
Abstract
Cell replacement therapies for neurodegenerative disease have focused on transplantation of the cell types affected by the pathological process. Here we describe an alternative strategy for Parkinson's disease in which dopamine neurons are generated by direct conversion of astrocytes. Using three transcription factors, NEUROD1, ASCL1 and LMX1A, and the microRNA miR218, collectively designated NeAL218, we reprogram human astrocytes in vitro, and mouse astrocytes in vivo, into induced dopamine neurons (iDANs). Reprogramming efficiency in vitro is improved by small molecules that promote chromatin remodeling and activate the TGFβ, Shh and Wnt signaling pathways. The reprogramming efficiency of human astrocytes reaches up to 16%, resulting in iDANs with appropriate midbrain markers and excitability. In a mouse model of Parkinson's disease, NeAL218 alone reprograms adult striatal astrocytes into iDANs that are excitable and correct some aspects of motor behavior in vivo, including gait impairments. With further optimization, this approach may enable clinical therapies for Parkinson's disease by delivery of genes rather than cells.
Collapse
|
164
|
Metabolic Factors and Adult Neurogenesis: Impacts of Chinese Herbal Medicine on Brain Repair in Neurological Diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 135:117-147. [PMID: 28807156 DOI: 10.1016/bs.irn.2017.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adult neurogenesis plays the important roles in animal cognitive and emotional behaviors. Abnormal proliferation and differentiation of neural stem cells (NSCs) usually associate with the neural dysfunctions induced by different brain disorders. Therefore, targeting neurogenic factors could be a promoting strategy for neural regeneration and brain repair. Importantly, epidemiological studies suggest metabolism disorders like diabetes and obesity significantly increase the risk of neurological and psychiatric diseases. A large number of studies indicate that metabolic factors could serve as the modulators to adult neurogenesis, providing the potentials of metabolic factors to regulate NSCs growth and neural regeneration therapy. This chapter reviews the current studies on the roles of metabolic factors in modulating adult neurogenesis and evaluates the potentials of Chinese Herbal Medicine (CHM) for the treatment of neurological or psychiatric disorders by targeting the metabolic factors. Traditional Chinese Medicine (TCM) including CHM and acupuncture is now widely applied for the treatment of metabolic diseases, and neurological diseases in Asia, because its' therapeutic principles meet the multiple targets and complexity characteristics of most neurological disorders. Different studies indicate that there are many active compounds perform the regulations to metabolic factors and promoting neurogenesis. This chapter systematically summarizes the current progress and understanding of the active compounds and their underlying mechanisms of CHM formulas for promoting neurogenesis. Many CHM formulas and their active ingredients that originally used for metabolic disorders show the promising effects on mediating neurogenesis and brain repair for the treatments of neurodegenerative diseases. Therefore, further investigations about the relationship between neurogenesis and metabolic regulations of CHM will bring new insights into understanding the mechanisms of adult neurogenesis and provide great opportunities to develop new therapeutic strategies for neurological diseases. Those studies will provide scientific guidance to develop the drugs from TCM resource.
Collapse
|
165
|
Fang F, Song R, Ling X, Peng M, Xue Z, Cang J. Multiple sevoflurane anesthesia in pregnant mice inhibits neurogenesis of fetal hippocampus via repressing transcription factor Pax6. Life Sci 2017; 175:16-22. [DOI: 10.1016/j.lfs.2017.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/24/2017] [Accepted: 03/05/2017] [Indexed: 10/20/2022]
|
166
|
Farzanehfar P, Horne MK, Aumann TD. An investigation of gene expression in single cells derived from Nestin-expressing cells in the adult mouse midbrain in vivo. Neurosci Lett 2017; 648:34-40. [PMID: 28363755 DOI: 10.1016/j.neulet.2017.03.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/23/2017] [Accepted: 03/16/2017] [Indexed: 12/21/2022]
Abstract
Generation of new dopamine (DA) neurons in the adult midbrain is a controversial issue in development of better treatments for Parkinson's disease (PD). Previous research suggests Nestin-expressing neural precursor cells (NPCs) have a propensity to differentiate into neurons here, including DA neurons. In the present study we sought confirmation of this by studying gene expression in single Nestin-expressing cells and their progeny/ontogeny within the adult mouse midbrain. Cells were identified by administering a pulse of Tamoxifen to adult Nestin-CreERT2×R26eYFP transgenic mice. Samples of cytoplasm were harvested 4 days to 8 months later from individual eYFP+ cells in acutely prepared midbrain slices and analysed by RT-qPCR for gene expression. Remarkably, most eYFP+ cells co-expressed genes associated with mature (including DA) neurons (i.e. NeuN, Gad1, Gad2, vGlut2, TH and/or D2R) and neurogenesis (i.e. Ki67, Dcx, Ncam, Pax6, Ngn2 and/or Msx1), and this was true at all time-points following Tamoxifen. Indeed, cell proliferation genes (Nestin, Ki67) were exclusively expressed by eYFP+ cells with mature neuronal morphology and gene expression, and only at early time-points after Tamoxifen. Expression of proneuronal genes (Pax6, Msx1, Ngn2) was, however, higher in eYFP+ cells with immature morphology compared with mature morphology. Gene expression bore no relationship to cell location indicating that, in contrast to development, Nestin-expressing cells arise throughout the midbrain parenchyma and do not migrate long distances. On the other hand, gene expression did change with time after Tamoxifen, although not in a way consistent with neurogenesis. Overall, our results suggest that Nestin expression in the adult midbrain occurs in mature neurons, casting doubt on the premise of neurogenesis from Nestin+ NPCs here.
Collapse
Affiliation(s)
- Parisa Farzanehfar
- Florey Institute for Neuroscience & Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Malcolm K Horne
- Florey Institute for Neuroscience & Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tim D Aumann
- Florey Institute for Neuroscience & Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
167
|
The vascular adventitia: An endogenous, omnipresent source of stem cells in the body. Pharmacol Ther 2017; 171:13-29. [DOI: 10.1016/j.pharmthera.2016.07.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/12/2016] [Indexed: 12/22/2022]
|
168
|
Torper O, Götz M. Brain repair from intrinsic cell sources: Turning reactive glia into neurons. PROGRESS IN BRAIN RESEARCH 2017; 230:69-97. [PMID: 28552236 DOI: 10.1016/bs.pbr.2016.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The replacement of lost neurons in the brain due to injury or disease holds great promise for the treatment of neurological disorders. However, logistical and ethical hurdles in obtaining and maintaining viable cells for transplantation have proven difficult to overcome. In vivo reprogramming offers an alternative, to bypass many of the restrictions associated with an exogenous cell source as it relies on a source of cells already present in the brain. Recent studies have demonstrated the possibility to target and reprogram glial cells into functional neurons with high efficiency in the murine brain, using virally delivered transcription factors. In this chapter, we explore the different populations of glial cells, how they react to injury and how they can be exploited for reprogramming purposes. Further, we review the most significant publications and how they have contributed to the understanding of key aspects in direct reprogramming needed to take into consideration, like timing, cell type targeted, and regional differences. Finally, we discuss future challenges and what remains to be explored in order to determine the potential of in vivo reprogramming for future brain repair.
Collapse
Affiliation(s)
- Olof Torper
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, Planegg, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, Munich, Germany; SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig-Maximilians University Munich, Planegg, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, Planegg, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, Munich, Germany; SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig-Maximilians University Munich, Planegg, Germany.
| |
Collapse
|
169
|
Gascón S, Ortega F, Götz M. Transient CREB-mediated transcription is key in direct neuronal reprogramming. NEUROGENESIS 2017; 4:e1285383. [PMID: 28321434 PMCID: PMC5345748 DOI: 10.1080/23262133.2017.1285383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/08/2017] [Accepted: 01/16/2017] [Indexed: 02/08/2023]
Abstract
Combinations of neuronal determinants and/or small-molecules such as Forskolin (Fk) can be used to convert different cell types into neurons. As Fk is known to activate cAMP-dependent pathways including CREB-activity, we aimed here to determine the role of CREB in reprogramming – including its temporal profile. We show that transient expression of the dominant-positive CREB-VP16 followed by its inactivation mediated by the dominant-negative ICER improves neuronal conversion of astrocytes mediated by the neurogenic determinant Ascl1. Contrarily, persistent over-activation by CREB-VP16 or persistent inhibition by ICER interferes with neuronal reprogramming, with the latter enhancing cell death. Taken together our work shows transient CREB activation as a key effector in neuronal reprogramming.
Collapse
Affiliation(s)
- Sergio Gascón
- Ludwig-Maximilians University of Munich, Physiological Genomics, Biomedical Center (BMC), Planegg-Martinsried, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Felipe Ortega
- Biochemistry and Molecular Biology Department IV, Faculty of Veterinary Medicine, Complutense University , Madrid, Spain ; Institute of Neurochemistry (IUIN) , Madrid, Spain ; Health Research Institute of the Hospital Clínico San Carlos (IdISSC) , Madrid, Spain
| | - Magdalena Götz
- Ludwig-Maximilians University of Munich, Physiological Genomics, Biomedical Center (BMC), Planegg-Martinsried, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany; Excellence Cluster of Systems Neurology (SYNERGY), Munich, Germany
| |
Collapse
|
170
|
Pavlakis E, Tonchev AB, Kaprelyan A, Enchev Y, Stoykova A. Interaction between transcription factors PAX6/PAX6-5a and specific members of miR-183-96-182 cluster, may contribute to glioma progression in glioblastoma cell lines. Oncol Rep 2017; 37:1579-1592. [DOI: 10.3892/or.2017.5411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/02/2017] [Indexed: 11/06/2022] Open
|
171
|
Kokaia Z, Tornero D, Lindvall O. Transplantation of reprogrammed neurons for improved recovery after stroke. PROGRESS IN BRAIN RESEARCH 2017; 231:245-263. [PMID: 28554399 DOI: 10.1016/bs.pbr.2016.11.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Somatic cells such as fibroblasts, reprogrammed to induced pluripotent stem cells, can be used to generate neural stem/progenitor cells or neuroblasts for transplantation. In this review, we summarize recent studies demonstrating that when grafted intracerebrally in animal models of stroke, reprogrammed neurons improve function, probably by several different mechanisms, e.g., trophic actions, modulation of inflammation, promotion of angiogenesis, cellular and synaptic plasticity, and neuroprotection. In our own work, we have shown that human skin-derived reprogrammed neurons, fated to cortical progeny, integrate in stroke-injured neuronal network and form functional afferent synapses with host neurons, responding to peripheral sensory stimulation. However, whether neuronal replacement plays a role for the improvement of sensory, motor, and cognitive deficits after transplantation of reprogrammed neurons is still unclear. We conclude that further preclinical studies are needed to understand the therapeutic potential of grafted reprogrammed neurons and to define a road map for their clinical translation in stroke.
Collapse
Affiliation(s)
- Zaal Kokaia
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund, Sweden.
| | - Daniel Tornero
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund, Sweden
| | - Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund, Sweden
| |
Collapse
|
172
|
Engineering cell fate: Spotlight on cell-activation and signaling-directed lineage conversion. Tissue Cell 2016; 48:475-87. [DOI: 10.1016/j.tice.2016.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 06/13/2016] [Accepted: 07/25/2016] [Indexed: 12/23/2022]
|
173
|
Hong S, Jo J, Kim HJ, Lee JE, Shin DH, Lee SG, Baek A, Shim SH, Lee DR. RuvB-Like Protein 2 (Ruvbl2) Has a Role in Directing the Neuroectodermal Differentiation of Mouse Embryonic Stem Cells. Stem Cells Dev 2016; 25:1376-85. [DOI: 10.1089/scd.2016.0076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Soomin Hong
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Junghyun Jo
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Hyung Joon Kim
- Fertility Center, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
| | | | - Dong Hyuk Shin
- Fertility Center, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
| | - Sung-Geum Lee
- CHA Stem Cell Institute, CHA University, Seoul, Korea
| | - Ahmi Baek
- Fertility Center, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
| | - Sung Han Shim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
- Fertility Center, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
| | - Dong Ryul Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
- Fertility Center, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
- CHA Stem Cell Institute, CHA University, Seoul, Korea
| |
Collapse
|
174
|
GABAergic Regulation of Adult Hippocampal Neurogenesis. Mol Neurobiol 2016; 54:5497-5510. [DOI: 10.1007/s12035-016-0072-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/18/2016] [Indexed: 01/17/2023]
|
175
|
Jessberger S. Stem Cell-Mediated Regeneration of the Adult Brain. Transfus Med Hemother 2016; 43:321-326. [PMID: 27781019 DOI: 10.1159/000447646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/10/2016] [Indexed: 12/16/2022] Open
Abstract
Acute or chronic injury of the adult mammalian brain is often associated with persistent functional deficits as its potential for regeneration and capacity to rebuild lost neural structures is limited. However, the discovery that neural stem cells (NSCs) persist throughout life in discrete regions of the brain, novel approaches to induce the formation of neuronal and glial cells, and recently developed strategies to generate tissue for exogenous cell replacement strategies opened novel perspectives how to regenerate the adult brain. Here, we will review recently developed approaches for brain repair and discuss future perspectives that may eventually allow for developing novel treatment strategies in acute and chronic brain injury.
Collapse
Affiliation(s)
- Sebastian Jessberger
- Laboratory of Neural Plasticity, Brain Research Institute, Faculty of Medicine and Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
176
|
McClenahan FK, Sharma H, Shan X, Eyermann C, Colognato H. Dystroglycan Suppresses Notch to Regulate Stem Cell Niche Structure and Function in the Developing Postnatal Subventricular Zone. Dev Cell 2016; 38:548-66. [PMID: 27569418 DOI: 10.1016/j.devcel.2016.07.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 05/09/2016] [Accepted: 07/21/2016] [Indexed: 01/11/2023]
Abstract
While the extracellular matrix (ECM) is known to regulate neural stem cell quiescence in the adult subventricular zone (SVZ), the function of ECM in the developing SVZ remains unknown. Here, we report that the ECM receptor dystroglycan regulates a unique developmental restructuring of ECM in the early postnatal SVZ. Dystroglycan is furthermore required for ependymal cell differentiation and assembly of niche pinwheel structures, at least in part by suppressing Notch activation in radial glial cells, which leads to the increased expression of MCI, Myb, and FoxJ1, transcriptional regulators necessary for acquisition of the multiciliated phenotype. Dystroglycan also regulates perinatal radial glial cell proliferation and transition into intermediate gliogenic progenitors, such that either acute or constitutive loss of function in dystroglycan results in increased oligodendrogenesis. These findings reveal a role for dystroglycan in orchestrating both the assembly and function of the SVZ neural stem cell niche.
Collapse
Affiliation(s)
- Freyja K McClenahan
- Department of Pharmacology, Stony Brook University, Stony Brook, NY 11794-8651, USA
| | - Himanshu Sharma
- Department of Pharmacology, Stony Brook University, Stony Brook, NY 11794-8651, USA
| | - Xiwei Shan
- Department of Pharmacology, Stony Brook University, Stony Brook, NY 11794-8651, USA
| | - Christopher Eyermann
- Department of Pharmacology, Stony Brook University, Stony Brook, NY 11794-8651, USA
| | - Holly Colognato
- Department of Pharmacology, Stony Brook University, Stony Brook, NY 11794-8651, USA.
| |
Collapse
|
177
|
Espinosa Angarica V, del Sol A. Modeling heterogeneity in the pluripotent state: A promising strategy for improving the efficiency and fidelity of stem cell differentiation. Bioessays 2016; 38:758-68. [PMID: 27321053 PMCID: PMC5094535 DOI: 10.1002/bies.201600103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Pluripotency can be considered a functional characteristic of pluripotent stem cells (PSCs) populations and their niches, rather than a property of individual cells. In this view, individual cells within the population independently adopt a variety of different expression states, maintained by different signaling, transcriptional, and epigenetics regulatory networks. In this review, we propose that generation of integrative network models from single cell data will be essential for getting a better understanding of the regulation of self-renewal and differentiation. In particular, we suggest that the identification of network stability determinants in these integrative models will provide important insights into the mechanisms mediating the transduction of signals from the niche, and how these signals can trigger differentiation. In this regard, the differential use of these stability determinants in subpopulation-specific regulatory networks would mediate differentiation into different cell fates. We suggest that this approach could offer a promising avenue for the development of novel strategies for increasing the efficiency and fidelity of differentiation, which could have a strong impact on regenerative medicine.
Collapse
Affiliation(s)
- Vladimir Espinosa Angarica
- Luxembourg Center for Systems Biomedicine (LCSB)University of Luxembourg, Campus BelvalBelvauxLuxembourg
| | - Antonio del Sol
- Luxembourg Center for Systems Biomedicine (LCSB)University of Luxembourg, Campus BelvalBelvauxLuxembourg
| |
Collapse
|
178
|
Chen P, Yan Q, Wang S, Wang C, Zhao P. Transfer of three transcription factors via a lentiviral vector ameliorates spatial learning and memory impairment in a mouse model of Alzheimer's disease. Gene 2016; 587:59-63. [DOI: 10.1016/j.gene.2016.04.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/15/2016] [Indexed: 01/01/2023]
|
179
|
Masserdotti G, Gascón S, Götz M. Direct neuronal reprogramming: learning from and for development. Development 2016; 143:2494-510. [DOI: 10.1242/dev.092163] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The key signalling pathways and transcriptional programmes that instruct neuronal diversity during development have largely been identified. In this Review, we discuss how this knowledge has been used to successfully reprogramme various cell types into an amazing array of distinct types of functional neurons. We further discuss the extent to which direct neuronal reprogramming recapitulates embryonic development, and examine the particular barriers to reprogramming that may exist given a cell's unique developmental history. We conclude with a recently proposed model for cell specification called the ‘Cook Islands’ model, and consider whether it is a fitting model for cell specification based on recent results from the direct reprogramming field.
Collapse
Affiliation(s)
- Giacomo Masserdotti
- Institute of Stem Cell Research, Helmholtz Center Munich, Ingolstädter Landstrasse 1, Neuherberg/Munich D-85764, Germany
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, Großhadernerstrasse 9, Martinsried 82154, Germany
| | - Sergio Gascón
- Institute of Stem Cell Research, Helmholtz Center Munich, Ingolstädter Landstrasse 1, Neuherberg/Munich D-85764, Germany
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, Großhadernerstrasse 9, Martinsried 82154, Germany
| | - Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Center Munich, Ingolstädter Landstrasse 1, Neuherberg/Munich D-85764, Germany
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, Großhadernerstrasse 9, Martinsried 82154, Germany
- Excellence Cluster of Systems Neurology, Großhadernerstrasse 9, Martinsried 82154, Germany
| |
Collapse
|
180
|
Tanabe K, Haag D, Wernig M. Direct somatic lineage conversion. Philos Trans R Soc Lond B Biol Sci 2016; 370:20140368. [PMID: 26416679 DOI: 10.1098/rstb.2014.0368] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The predominant view of embryonic development and cell differentiation has been that rigid and even irreversible epigenetic marks are laid down along the path of cell specialization ensuring the proper silencing of unrelated lineage programmes. This model made the prediction that specialized cell types are stable and cannot be redirected into other lineages. Accordingly, early attempts to change the identity of somatic cells had little success and was limited to conversions between closely related cell types. Nuclear transplantation experiments demonstrated, however, that specialized cells even from adult mammals can be reprogrammed into a totipotent state. The discovery that a small combination of transcription factors can reprogramme cells to pluripotency without the need of oocytes further supported the view that these epigenetic barriers can be overcome much easier than assumed, but the extent of this flexibility was still unclear. When we showed that a differentiated mesodermal cell can be directly converted to a differentiated ectodermal cell without a pluripotent intermediate, it was suggested that in principle any cell type could be converted into any other cell type. Indeed, the work of several groups in recent years has provided many more examples of direct somatic lineage conversions. Today, the question is not anymore whether a specific cell type can be generated by direct reprogramming but how it can be induced.
Collapse
Affiliation(s)
- Koji Tanabe
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daniel Haag
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
181
|
Yang H, Liu CC, Wang CY, Zhang Q, An J, Zhang L, Hao DJ. Therapeutical Strategies for Spinal Cord Injury and a Promising Autologous Astrocyte-Based Therapy Using Efficient Reprogramming Techniques. Mol Neurobiol 2016; 53:2826-2842. [DOI: 10.1007/s12035-015-9157-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/19/2015] [Indexed: 01/01/2023]
|
182
|
Götz M, Nakafuku M, Petrik D. Neurogenesis in the Developing and Adult Brain-Similarities and Key Differences. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a018853. [PMID: 27235475 DOI: 10.1101/cshperspect.a018853] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Adult neurogenesis in the mammalian brain is often viewed as a continuation of neurogenesis at earlier, developmental stages. Here, we will critically review the extent to which this is the case highlighting similarities as well as key differences. Although many transcriptional regulators are shared in neurogenesis at embryonic and adult stages, recent findings on the molecular mechanisms by which these neuronal fate determinants control fate acquisition and maintenance have revealed profound differences between development and adulthood. Importantly, adult neurogenesis occurs in a gliogenic environment, hence requiring adult-specific additional and unique mechanisms of neuronal fate specification and maintenance. Thus, a better understanding of the molecular logic for continuous adult neurogenesis provides important clues to develop strategies to manipulate endogenous stem cells for the purpose of repair.
Collapse
Affiliation(s)
- Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Munich, Germany Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, 80336 Munich, Germany Synergy, Munich Cluster for Systems Neurology, 81377 Munich, Germany
| | - Masato Nakafuku
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45140 Departments of Pediatrics and Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - David Petrik
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Munich, Germany Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, 80336 Munich, Germany
| |
Collapse
|
183
|
Götz M. Glial Cells Generate Neurons—Master Control within CNS Regions: Developmental Perspectives on Neural Stem Cells. Neuroscientist 2016; 9:379-97. [PMID: 14580122 DOI: 10.1177/1073858403257138] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A common problem in neural stem cell research is the poor generation of neuronal or oligodendroglial descendants. The author takes a developmental perspective to propose solutions to this problem. After a general overview of the recent progress in developmental neurobiology, she highlights the necessity of the sequential and hierarchical specification of CNS precursors toward the generation of specific cell types, for example, neurons. In the developing as well as the adult CNS, multipotent stem cells do not directly generate neurons but give rise to precursors that are specified and restricted toward the generation of neurons. Some molecular determinants of this fate restriction have been identified during recent years and reveal that progression via this fate-restricted state is a necessary step of neurogenesis. These discoveries also demonstrate that neuronal fate specification is inseparably linked at the molecular level to regionalization of the developing CNS. These fate determinants and their specific action in distinct region-specific con-texts are essential to direct the progeny of stem cells more efficiently toward the generation of the desired cell types. Recent data are discussed that demonstrate the common identity of precursors and stem cells in the developing and adult nervous system as radial glia, astroglia, or non-myelinating glia. A novel line-age model is proposed that incorporates these new views and explains why the default pathway of stem cells is astroglia. These new insights into the cellular and molecular mechanisms of neurogenesis help to design novel approaches for reconstitutive therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Magdalena Götz
- Max-Planck Institute of Neurobiology, Planegg-Martinsried/Munich, Germany.
| |
Collapse
|
184
|
Rusznák Z, Henskens W, Schofield E, Kim WS, Fu Y. Adult Neurogenesis and Gliogenesis: Possible Mechanisms for Neurorestoration. Exp Neurobiol 2016; 25:103-12. [PMID: 27358578 PMCID: PMC4923354 DOI: 10.5607/en.2016.25.3.103] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/08/2016] [Accepted: 06/08/2016] [Indexed: 12/22/2022] Open
Abstract
The subgranular zone (SGZ) and subventricular zone (SVZ) are developmental remnants of the germinal regions of the brain, hence they retain the ability to generate neuronal progenitor cells in adult life. Neurogenesis in adult brain has an adaptive function because newly produced neurons can integrate into and modify existing neuronal circuits. In contrast to the SGZ and SVZ, other brain regions have a lower capacity to produce new neurons, and this usually occurs via parenchymal and periventricular cell genesis. Compared to neurogenesis, gliogenesis occurs more prevalently in the adult mammalian brain. Under certain circumstances, interaction occurs between neurogenesis and gliogenesis, facilitating glial cells to transform into neuronal lineage. Therefore, modulating the balance between neurogenesis and gliogenesis may present a new perspective for neurorestoration, especially in diseases associated with altered neurogenesis and/or gliogenesis, cell loss, or disturbed homeostasis of cellular constitution. The present review discusses important neuroanatomical features of adult neurogenesis and gliogenesis, aiming to explore how these processes could be modulated toward functional repair of the adult brain.
Collapse
Affiliation(s)
- Zoltán Rusznák
- Neuroscience Research Australia, Sydney, NSW 2031, Australia
| | - Willem Henskens
- Neuroscience Research Australia, Sydney, NSW 2031, Australia.; Prince of Wales Clinical School, UNSW Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Emma Schofield
- Neuroscience Research Australia, Sydney, NSW 2031, Australia
| | - Woojin S Kim
- Neuroscience Research Australia, Sydney, NSW 2031, Australia.; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - YuHong Fu
- Neuroscience Research Australia, Sydney, NSW 2031, Australia.; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
185
|
Jones KS, Connor BJ. The Effect of Pro-Neurogenic Gene Expression on Adult Subventricular Zone Precursor Cell Recruitment and Fate Determination After Excitotoxic Brain Injury. J Stem Cells Regen Med 2016. [PMID: 27397999 PMCID: PMC4929891 DOI: 10.46582/jsrm.1201005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Despite the presence of on-going neurogenesis in the adult mammalian brain, neurons are generally not replaced after injury. Using a rodent model of excitotoxic cell loss and retroviral (RV) lineage tracing, we previously demonstrated transient recruitment of precursor cells from the subventricular zone (SVZ) into the lesioned striatum. In the current study we determined that these cells included migratory neuroblasts and oligodendrocyte precursor cells (OPC), with the predominant response from glial cells. We attempted to override this glial response by ectopic expression of the pro-neurogenic genes Pax6 or Dlx2 in the adult rat SVZ following quinolinic acid lesioning. RV-Dlx2 over-expression stimulated repair at a previously non-neurogenic time point by enhancing neuroblast recruitment and the percentage of cells that retained a neuronal fate within the lesioned area, compared to RV-GFP controls. RV-Pax6 expression was unsuccessful at inhibiting glial fate and intriguingly, increased OPC cell numbers with no change in neuronal recruitment. These findings suggest that gene choice is important when attempting to augment endogenous repair as the lesioned environment can overcome pro-neurogenic gene expression. Dlx2 over-expression however was able to partially overcome an anti-neuronal environment and therefore is a promising candidate for further study of striatal regeneration.
Collapse
Affiliation(s)
- Kathryn S Jones
- Centre for Brain Research, Department of Pharmacology and Clinical Pharmacology, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland
| | - Bronwen J Connor
- Centre for Brain Research, Department of Pharmacology and Clinical Pharmacology, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland
| |
Collapse
|
186
|
Xu A, Cheng L. Chemical transdifferentiation: closer to regenerative medicine. Front Med 2016; 10:152-65. [PMID: 27142989 DOI: 10.1007/s11684-016-0445-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/21/2016] [Indexed: 12/15/2022]
Abstract
Cell transdifferentiation, which directly switches one type of differentiated cells into another cell type, is more advantageous than cell reprogramming to generate pluripotent cells and differentiate them into functional cells. This process is crucial in regenerative medicine. However, the cell-converting strategies, which mainly depend on the virus-mediated expression of exogenous genes, have clinical safety concerns. Small molecules with compelling advantages are a potential alternative in manipulating cell fate conversion. In this review, we briefly retrospect the nature of cell transdifferentiation and summarize the current developments in the research of small molecules in promoting cell conversion. Particularly, we focus on the complete chemical compound-induced cell transdifferentiation, which is closer to the clinical translation in cell therapy. Despite these achievements, the mechanisms underpinning chemical transdifferentiation remain largely unknown. More importantly, identifying drugs that induce resident cell conversion in vivo to repair damaged tissue remains to be the end-goal in current regenerative medicine.
Collapse
Affiliation(s)
- Aining Xu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lin Cheng
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
187
|
Berninger B, Jessberger S. Engineering of Adult Neurogenesis and Gliogenesis. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a018861. [PMID: 27091941 DOI: 10.1101/cshperspect.a018861] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neural stem/progenitor cells (NSPCs) retain their ability to generate newborn neurons throughout life in the mammalian brain. Here, we describe how recently developed virus- and transgenesis-based techniques will help us (1) to understand the functional effects of neurogenesis in health and disease, (2) to design novel approaches to harness the potential for NSPC-associated endogenous repair, and (3) to induce the generation of neurons outside the main neurogenic niches in the adult brain.
Collapse
Affiliation(s)
- Benedikt Berninger
- Adult Neurogenesis and Cellular Reprogramming, Institute of Physiological Chemistry & Focus Program Translational Neuroscience, University Medical Center, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | | |
Collapse
|
188
|
Javaid FZ, Brenton J, Guo L, Cordeiro MF. Visual and Ocular Manifestations of Alzheimer's Disease and Their Use as Biomarkers for Diagnosis and Progression. Front Neurol 2016; 7:55. [PMID: 27148157 PMCID: PMC4836138 DOI: 10.3389/fneur.2016.00055] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/29/2016] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia affecting the growing aging population today, with prevalence expected to rise over the next 35 years. Clinically, patients exhibit a progressive decline in cognition, memory, and social functioning due to deposition of amyloid β (Aβ) protein and intracellular hyperphosphorylated tau protein. These pathological hallmarks of AD are measured either through neuroimaging, cerebrospinal fluid analysis, or diagnosed post-mortem. Importantly, neuropathological progression occurs in the eye as well as the brain, and multiple visual changes have been noted in both human and animal models of AD. The eye offers itself as a transparent medium to cerebral pathology and has thus potentiated the development of ocular biomarkers for AD. The use of non-invasive screening, such as retinal imaging and visual testing, may enable earlier diagnosis in the clinical setting, minimizing invasive and expensive investigations. It also potentially improves disease management and quality of life for AD patients, as an earlier diagnosis allows initiation of medication and treatment. In this review, we explore the evidence surrounding ocular changes in AD and consider the biomarkers currently in development for early diagnosis.
Collapse
Affiliation(s)
- Fatimah Zara Javaid
- Glaucoma and Retinal Degeneration Research Group, Visual Neurosciences, UCL Institute of Ophthalmology, London, UK
| | - Jonathan Brenton
- Glaucoma and Retinal Degeneration Research Group, Visual Neurosciences, UCL Institute of Ophthalmology, London, UK
| | - Li Guo
- Glaucoma and Retinal Degeneration Research Group, Visual Neurosciences, UCL Institute of Ophthalmology, London, UK
| | - Maria F. Cordeiro
- Glaucoma and Retinal Degeneration Research Group, Visual Neurosciences, UCL Institute of Ophthalmology, London, UK
- Western Eye Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
189
|
Liu SJ, Nowakowski TJ, Pollen AA, Lui JH, Horlbeck MA, Attenello FJ, He D, Weissman JS, Kriegstein AR, Diaz AA, Lim DA. Single-cell analysis of long non-coding RNAs in the developing human neocortex. Genome Biol 2016; 17:67. [PMID: 27081004 PMCID: PMC4831157 DOI: 10.1186/s13059-016-0932-1] [Citation(s) in RCA: 257] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/01/2016] [Indexed: 12/22/2022] Open
Abstract
Background Long non-coding RNAs (lncRNAs) comprise a diverse class of transcripts that can regulate molecular and cellular processes in brain development and disease. LncRNAs exhibit cell type- and tissue-specific expression, but little is known about the expression and function of lncRNAs in the developing human brain. Furthermore, it has been unclear whether lncRNAs are highly expressed in subsets of cells within tissues, despite appearing lowly expressed in bulk populations. Results We use strand-specific RNA-seq to deeply profile lncRNAs from polyadenylated and total RNA obtained from human neocortex at different stages of development, and we apply this reference to analyze the transcriptomes of single cells. While lncRNAs are generally detected at low levels in bulk tissues, single-cell transcriptomics of hundreds of neocortex cells reveal that many lncRNAs are abundantly expressed in individual cells and are cell type-specific. Notably, LOC646329 is a lncRNA enriched in single radial glia cells but is detected at low abundance in tissues. CRISPRi knockdown of LOC646329 indicates that this lncRNA regulates cell proliferation. Conclusion The discrete and abundant expression of lncRNAs among individual cells has important implications for both their biological function and utility for distinguishing neural cell types. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-0932-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Siyuan John Liu
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Tomasz J Nowakowski
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,Department of Neurology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Alex A Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,Department of Neurology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Jan H Lui
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,Department of Neurology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA.,Present Address: Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA
| | - Max A Horlbeck
- Department of Cellular and Molecular Pharmacology, San Francisco, CA, 94143, USA.,Howard Hughes Medical Institute, San Francisco, CA, 94143, USA.,California Institute for Quantitative Biomedical Research, San Francisco, CA, 94143, USA.,Center for RNA Systems Biology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Frank J Attenello
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Daniel He
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Jonathan S Weissman
- Department of Cellular and Molecular Pharmacology, San Francisco, CA, 94143, USA.,Howard Hughes Medical Institute, San Francisco, CA, 94143, USA.,California Institute for Quantitative Biomedical Research, San Francisco, CA, 94143, USA.,Center for RNA Systems Biology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Arnold R Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,Department of Neurology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Aaron A Diaz
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA. .,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA. .,University of California, San Francisco, San Francisco, CA, 94143, USA.
| | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA. .,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA. .,University of California, San Francisco, San Francisco, CA, 94143, USA. .,San Francisco Veterans Affairs Medical Center, San Francisco, CA, 94121, USA.
| |
Collapse
|
190
|
Matsumoto S, Banine F, Feistel K, Foster S, Xing R, Struve J, Sherman LS. Brg1 directly regulates Olig2 transcription and is required for oligodendrocyte progenitor cell specification. Dev Biol 2016; 413:173-87. [PMID: 27067865 DOI: 10.1016/j.ydbio.2016.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/28/2016] [Accepted: 04/04/2016] [Indexed: 01/10/2023]
Abstract
The Olig2 basic-helix-loop-helix transcription factor promotes oligodendrocyte specification in early neural progenitor cells (NPCs), including radial glial cells, in part by recruiting SWI/SNF chromatin remodeling complexes to the enhancers of genes involved in oligodendrocyte differentiation. How Olig2 expression is regulated during oligodendrogliogenesis is not clear. Here, we find that the Brg1 subunit of SWI/SNF complexes interacts with a proximal Olig2 promoter and represses Olig2 transcription in the mouse cortex at E14, when oligodendrocyte progenitors (OPCs) are not yet found in this location. Brg1 does not interact with the Olig2 promoter in the E14 ganglionic eminence, where NPCs differentiate into Olig2-positive OPCs. Consistent with these findings, Brg1-null NPCs demonstrate precocious expression of Olig2 in the cortex. However, these cells fail to differentiate into OPCs. We further find that Brg1 is necessary for neuroepithelial-to-radial glial cell transition, but not neuronal differentiation despite a reduction in expression of the pro-neural transcription factor Pax6. Collectively, these and earlier findings support a model whereby Brg1 promotes neurogenic radial glial progenitor cell specification but is dispensable for neuronal differentiation. Concurrently, Brg1 represses Olig2 expression and the specification of OPCs, but is required for OPC differentiation and oligodendrocyte maturation.
Collapse
Affiliation(s)
- Steven Matsumoto
- Integrative Biosciences Department, School of Dentistry, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Fatima Banine
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Kerstin Feistel
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Scott Foster
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Rubing Xing
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Jaime Struve
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA; Department of Cell, Developmental and Cancer Biology, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
191
|
Magnusson JP, Frisén J. Stars from the darkest night: unlocking the neurogenic potential of astrocytes in different brain regions. Development 2016; 143:1075-86. [DOI: 10.1242/dev.133975] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/15/2016] [Indexed: 12/14/2022]
Abstract
In a few regions of the adult brain, specialized astrocytes act as neural stem cells capable of sustaining life-long neurogenesis. In other, typically non-neurogenic regions, some astrocytes have an intrinsic capacity to produce neurons when provoked by particular conditions but do not use this ability to replace neurons completely after injury or disease. Why do astrocytes display regional differences and why do they not use their neurogenic capacity for brain repair to a greater extent? In this Review, we discuss the neurogenic potential of astrocytes in different brain regions and ask what stimulates this potential in some regions but not in others. We discuss the transcriptional networks and environmental cues that govern cell identity, and consider how the activation of neurogenic properties in astrocytes can be understood as the de-repression of a latent neurogenic transcriptional program.
Collapse
Affiliation(s)
- Jens P. Magnusson
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm SE-171 77, Sweden
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm SE-171 77, Sweden
| |
Collapse
|
192
|
White RB, Thomas MG. Developmental transcription factors in age-related CNS disease: a phoenix rising from the ashes? Neural Regen Res 2016; 11:64-5. [PMID: 26981081 PMCID: PMC4774228 DOI: 10.4103/1673-5374.175044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Robert B White
- School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, WA, Australia; Parkinson's Centre, School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia; Experimental and Regenerative Neuroscience, School of Animal Biology, The University of Western Australia, Crawley, WA, Australia
| | - Meghan G Thomas
- Parkinson's Centre, School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia; Experimental and Regenerative Neuroscience, School of Animal Biology, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
193
|
Petersen GF, Strappe PM. Generation of diverse neural cell types through direct conversion. World J Stem Cells 2016; 8:32-46. [PMID: 26981169 PMCID: PMC4766249 DOI: 10.4252/wjsc.v8.i2.32] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/18/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
A characteristic of neurological disorders is the loss of critical populations of cells that the body is unable to replace, thus there has been much interest in identifying methods of generating clinically relevant numbers of cells to replace those that have been damaged or lost. The process of neural direct conversion, in which cells of one lineage are converted into cells of a neural lineage without first inducing pluripotency, shows great potential, with evidence of the generation of a range of functional neural cell types both in vitro and in vivo, through viral and non-viral delivery of exogenous factors, as well as chemical induction methods. Induced neural cells have been proposed as an attractive alternative to neural cells derived from embryonic or induced pluripotent stem cells, with prospective roles in the investigation of neurological disorders, including neurodegenerative disease modelling, drug screening, and cellular replacement for regenerative medicine applications, however further investigations into improving the efficacy and safety of these methods need to be performed before neural direct conversion becomes a clinically viable option. In this review, we describe the generation of diverse neural cell types via direct conversion of somatic cells, with comparison against stem cell-based approaches, as well as discussion of their potential research and clinical applications.
Collapse
|
194
|
Abstract
Acute or chronic injury to the adult brain often results in substantial loss of neural tissue and subsequent permanent functional impairment. Over the last two decades, a number of approaches have been developed to harness the regenerative potential of neural stem cells and the existing fate plasticity of neural cells in the nervous system to prevent tissue loss or to enhance structural and functional regeneration upon injury. Here, we review recent advances of stem cell-associated neural repair in the adult brain, discuss current challenges and limitations, and suggest potential directions to foster the translation of experimental stem cell therapies into the clinic.
Collapse
Affiliation(s)
- Sebastian Jessberger
- Laboratory of Neural Plasticity, Brain Research Institute, Faculty of Medicine and Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
195
|
Abstract
Conversion of one cell type into another cell type by forcibly expressing specific cocktails of transcription factors (TFs) has demonstrated that cell fates are not fixed and that cellular differentiation can be a two-way street with many intersections. These experiments also illustrated the sweeping potential of TFs to “read” genetically hardwired regulatory information even in cells where they are not normally expressed and to access and open up tightly packed chromatin to execute gene expression programs. Cellular reprogramming enables the modeling of diseases in a dish, to test the efficacy and toxicity of drugs in patient-derived cells and ultimately, could enable cell-based therapies to cure degenerative diseases. Yet, producing terminally differentiated cells that fully resemble their in vivo counterparts in sufficient quantities is still an unmet clinical need. While efforts are being made to reprogram cells nongenetically by using drug-like molecules, defined TF cocktails still dominate reprogramming protocols. Therefore, the optimization of TFs by protein engineering has emerged as a strategy to enhance reprogramming to produce functional, stable and safe cells for regenerative biomedicine. Engineering approaches focused on Oct4, MyoD, Sox17, Nanog and Mef2c and range from chimeric TFs with added transactivation domains, designer transcription activator-like effectors to activate endogenous TFs to reprogramming TFs with rationally engineered DNA recognition principles. Possibly, applying the complete toolkit of protein design to cellular reprogramming can help to remove the hurdles that, thus far, impeded the clinical use of cells derived from reprogramming technologies.
Collapse
Affiliation(s)
| | | | - Ralf Jauch
- Genome Regulation Laboratory, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| |
Collapse
|
196
|
Mapping gene regulatory circuitry of Pax6 during neurogenesis. Cell Discov 2016; 2:15045. [PMID: 27462442 PMCID: PMC4860964 DOI: 10.1038/celldisc.2015.45] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/11/2015] [Indexed: 11/19/2022] Open
Abstract
Pax6 is a highly conserved transcription factor among vertebrates and is important in various aspects of the central nervous system development. However, the gene regulatory circuitry of Pax6 underlying these functions remains elusive. We find that Pax6 targets a large number of promoters in neural progenitors cells. Intriguingly, many of these sites are also bound by another progenitor factor, Sox2, which cooperates with Pax6 in gene regulation. A combinatorial analysis of Pax6-binding data set with transcriptome changes in Pax6-deficient neural progenitors reveals a dual role for Pax6, in which it activates the neuronal (ectodermal) genes while concurrently represses the mesodermal and endodermal genes, thereby ensuring the unidirectionality of lineage commitment towards neuronal differentiation. Furthermore, Pax6 is critical for inducing activity of transcription factors that elicit neurogenesis and repress others that promote non-neuronal lineages. In addition to many established downstream effectors, Pax6 directly binds and activates a number of genes that are specifically expressed in neural progenitors but have not been previously implicated in neurogenesis. The in utero knockdown of one such gene, Ift74, during brain development impairs polarity and migration of newborn neurons. These findings demonstrate new aspects of the gene regulatory circuitry of Pax6, revealing how it functions to control neuronal development at multiple levels to ensure unidirectionality and proper execution of the neurogenic program.
Collapse
|
197
|
Bery A, Mérot Y, Rétaux S. Genes expressed in mouse cortical progenitors are enriched in Pax, Lhx, and Sox transcription factor putative binding sites. Brain Res 2015; 1633:37-51. [PMID: 26721689 DOI: 10.1016/j.brainres.2015.12.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 11/25/2015] [Accepted: 12/14/2015] [Indexed: 11/19/2022]
Abstract
Considerable progress has been made in the understanding of molecular and cellular mechanisms controlling the development of the mammalian cortex. The proliferative and neurogenic properties of cortical progenitors located in the ventricular germinal zone start being understood. Little is known however on the cis-regulatory control that finely tunes gene expression in these progenitors. Here, we undertook an in silico-based approach to address this question, followed by some functional validation. Using the Eurexpress database, we established a list of 30 genes specifically expressed in the cortical germinal zone, we selected mouse/human conserved non-coding elements (CNEs) around these genes and we performed motif-enrichment search in these CNEs. We found an over-representation of motifs corresponding to binding sites for Pax, Sox, and Lhx transcription factors, often found as pairs and located within 100bp windows. A small subset of CNEs (n=7) was tested for enhancer activity, by ex-vivo and in utero electroporation assays. Two showed strong enhancer activity in the germinal zone progenitors. Mutagenesis experiments on a selected CNE showed the functional importance of the Pax, Sox, and Lhx TFBS for conferring enhancer activity to the CNE. Overall, from a cis-regulatory viewpoint, our data suggest an input from Pax, Sox and Lhx transcription factors to orchestrate corticogenesis. These results are discussed with regards to the known functional roles of Pax6, Sox2 and Lhx2 in cortical development.
Collapse
Affiliation(s)
- Amandine Bery
- DECA Group, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud, CNRS, UMR 9197, 91198 Gif-sur-Yvette, France.
| | - Yohann Mérot
- DECA Group, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud, CNRS, UMR 9197, 91198 Gif-sur-Yvette, France
| | - Sylvie Rétaux
- DECA Group, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud, CNRS, UMR 9197, 91198 Gif-sur-Yvette, France.
| |
Collapse
|
198
|
Abstract
The direct lineage reprogramming of one specialized cell type into another using defined factors has fundamentally re-shaped traditional concepts regarding the epigenetic stability of differentiated cells. With the rapid increase in cell types generated through direct conversion in recent years, this strategy has become a promising approach for producing functional cells. Here, we review recent advances in lineage reprogramming, including the identification of novel reprogramming factors, underlying molecular mechanisms, strategies for generating functionally mature cells, and assays for characterizing induced cells. We also discuss progress toward the application of lineage reprogramming and the major future challenges for this strategy.
Collapse
|
199
|
Suzuki T, Takayama R, Sato M. eyeless/Pax6 controls the production of glial cells in the visual center of Drosophila melanogaster. Dev Biol 2015; 409:343-53. [PMID: 26670857 DOI: 10.1016/j.ydbio.2015.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 11/11/2015] [Accepted: 12/04/2015] [Indexed: 10/22/2022]
Abstract
Pax6 is known as a neurogenic factor in the development of the central nervous system and regulates proliferation of neuronal progenitor cells and promotes neuronal differentiation. In addition to neurogenesis, Pax6 is also involved in the specification and maturation of glial cells. Here, we show that Eyeless (Ey), Drosophila homolog of Pax6, regulates the production of glial cells in the brain. In the developing fly visual center, the production of neurons and glial cells are controlled by the temporal transcription factors that are sequentially expressed in neuroblasts (NBs). Among them, NBs of the last temporal window produce astrocyte-like glial cells. Ey is strongly expressed in the middle aged NBs, whose temporal window is earlier compared with glia producing older NBs. Weak Ey expression is also detected in the glia producing NBs. Our results suggest that Ey expression in the middle aged NBs indirectly control gliogenesis from the oldest NBs by regulating other temporal transcription factors. Additionally, weak Ey expression in the NBs of last temporal window may directly control gliogenesis. Ey is also expressed in neurons produced from the NBs of Ey-positive temporal window. Interestingly, neuron-specific overexpression of Ey causes significant increase in glial cells suggesting that neuronal expression of Ey may also contribute to gliogenesis. Thus, Pax6-dependent regulation of astrocyte-like glial development is conserved throughout the animal kingdom.
Collapse
Affiliation(s)
- Takumi Suzuki
- Laboratory of Developmental Neurobiology, Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi Kanazawa-shi, Ishikawa 920-8640, Japan
| | - Rie Takayama
- Laboratory of Developmental Neurobiology, Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi Kanazawa-shi, Ishikawa 920-8640, Japan; CREST, JST, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan
| | - Makoto Sato
- Laboratory of Developmental Neurobiology, Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi Kanazawa-shi, Ishikawa 920-8640, Japan; Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi Kanazawa-shi, Ishikawa 920-8640, Japan; CREST, JST, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
200
|
Ghosh M, Lane M, Krizman E, Sattler R, Rothstein JD, Robinson MB. The transcription factor Pax6 contributes to the induction of GLT-1 expression in astrocytes through an interaction with a distal enhancer element. J Neurochem 2015; 136:262-75. [PMID: 26485579 DOI: 10.1111/jnc.13406] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 10/02/2015] [Accepted: 10/06/2015] [Indexed: 12/11/2022]
Abstract
The Na(+) -dependent glutamate transporter GLT-1 (EAAT2) shows selective expression in astrocytes, and neurons induce the expression of GLT-1 in astrocytes. In an unpublished analysis of GLT-1 promoter reporter mice, we identified an evolutionarily conserved domain of 467 nucleotides ~ 8 kb upstream of the GLT-1 translation start site that is required for astrocytic expression. Using in silico approaches, we identified Pax6 as a transcription factor that could contribute to the control of GLT-1 expression by binding within this region. We demonstrated the expression of Pax6 protein in astrocytes in vivo. Lentiviral transduction of astrocytes with exogenous Pax6 increased the expression of enhanced green fluorescent protein (eGFP) in astrocytes prepared from transgenic mice that use a bacterial artificial chromosome containing a large genomic region surrounding the GLT-1 gene to control expression of eGFP. It also increased GLT-1 protein and GLT-1-mediated uptake, whereas there was no effect on the levels of the other astroglial glutamate transporter, glutamate aspartate transporter (GLAST). Transduction of astrocytes with an shRNA directed against Pax6 reduced neuron-dependent induction of GLT-1 or eGFP. Finally, we confirmed Pax6 interaction with the predicted DNA-binding site in electrophoretic mobility assays and chromatin immunoprecipitation (ChIP). Together, these studies show that Pax6 contributes to the regulation of GLT-1 through an interaction with these distal elements and identify a novel role of Pax6 in astrocyte biology. The astroglial glutamate transporter GLT-1 shows selective expression in astrocytes and its expression can be induced by neurons. In this study, we demonstrate that Pax6 is expressed in astrocytes and binds to the GLT-1 promoter in vitro and in vivo. Exogenous expression of Pax6 increases GLT-1 and enhanced green fluorescent protein (eGFP) expression in astrocytes from a transgenic mouse line that uses the GLT-1 gene to drive eGFP expression, and an shRNA directed against Pax6 attenuates neuron-dependent induction of GLT-1/eGFP. We therefore conclude that Pax6 contributes to the neuron-dependent induction of GLT-1.
Collapse
Affiliation(s)
- Mausam Ghosh
- Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meredith Lane
- Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Krizman
- Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rita Sattler
- Department of Neurology and Neuroscience, John Hopkins University, Baltimore, Maryland, USA
| | - Jeffrey D Rothstein
- Department of Neurology and Neuroscience, John Hopkins University, Baltimore, Maryland, USA
| | - Michael B Robinson
- Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|