Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(20)2022 Oct 16.
Article in English | MEDLINE | ID: mdl-36293245

ABSTRACT

Clonal cell analysis outlines the ontogenic potential of single progenitor cells, allowing the elucidation of the neural heterogeneity among different cell types and their lineages. In this work, we analyze the potency of retinal stem/progenitor cells through development using the chick embryo as a model. We implemented in ovo the clonal genetic tracing strategy UbC-StarTrack for tracking retinal cell lineages derived from individual progenitors of the ciliary margin at E3.5 (HH21-22). The clonal assignment of the derived-cell progeny was performed in the neural retina at E11.5-12 (HH38) through the identification of sibling cells as cells expressing the same combination of fluorophores. Moreover, cell types were assessed based on their cellular morphology and laminar location. Ciliary margin derived-cell progenies are organized in columnar associations distributed along the peripheral retina with a limited tangential dispersion. The analysis revealed that, at the early stages of development, this region harbors multipotent and committed progenitor cells.


Subject(s)
Retina , Stem Cells , Animals , Chick Embryo , Stem Cells/metabolism , Cell Differentiation , Retina/metabolism , Cell Lineage , Cells, Cultured
2.
Sci Adv ; 7(15)2021 04.
Article in English | MEDLINE | ID: mdl-33827819

ABSTRACT

Neural cell diversity is essential to endow distinct brain regions with specific functions. During development, progenitors within these regions are characterized by specific gene expression programs, contributing to the generation of diversity in postmitotic neurons and astrocytes. While the region-specific molecular diversity of neurons and astrocytes is increasingly understood, whether these cells share region-specific programs remains unknown. Here, we show that in the neocortex and thalamus, neurons and astrocytes express shared region-specific transcriptional and epigenetic signatures. These signatures not only distinguish cells across these two brain regions but are also detected across substructures within regions, such as distinct thalamic nuclei, where clonal analysis reveals the existence of common nucleus-specific progenitors for neurons and astrocytes. Consistent with their shared molecular signature, regional specificity is maintained following astrocyte-to-neuron reprogramming. A detailed understanding of these regional-specific signatures may thus inform strategies for future cell-based brain repair.


Subject(s)
Astrocytes , Neocortex , Astrocytes/metabolism , Epigenomics , Neurons/physiology , Thalamus
3.
Cell Mol Life Sci ; 78(5): 1971-1982, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33151389

ABSTRACT

Understanding how an adult brain reaches an appropriate size and cell composition from a pool of progenitors that proliferates and differentiates is a key question in Developmental Neurobiology. Not only the control of final size but also, the proper arrangement of cells of different embryonic origins is fundamental in this process. Each neural progenitor has to produce a precise number of sibling cells that establish clones, and all these clones will come together to form the functional adult nervous system. Lineage cell tracing is a complex and challenging process that aims to reconstruct the offspring that arise from a single progenitor cell. This tracing can be achieved through strategies based on genetically modified organisms, using either genetic tracers, transfected viral vectors or DNA constructs, and even single-cell sequencing. Combining different reporter proteins and the use of transgenic mice revolutionized clonal analysis more than a decade ago and now, the availability of novel genome editing tools and single-cell sequencing techniques has vastly improved the capacity of lineage tracing to decipher progenitor potential. This review brings together the strategies used to study cell lineages in the brain and the role they have played in our understanding of the functional clonal relationships among neural cells. In addition, future perspectives regarding the study of cell heterogeneity and the ontogeny of different cell lineages will also be addressed.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Gene Expression Profiling/methods , Nervous System/metabolism , Neural Stem Cells/metabolism , Animals , CRISPR-Cas Systems/genetics , Gene Editing/methods , High-Throughput Nucleotide Sequencing/methods , Nervous System/cytology , Neural Stem Cells/cytology , Single-Cell Analysis/methods
4.
Genes (Basel) ; 11(3)2020 03 13.
Article in English | MEDLINE | ID: mdl-32183100

ABSTRACT

The large phenotypic variation in the olfactory bulb may be related to heterogeneity in the progenitor cells. Accordingly, the progeny of subventricular zone (SVZ) progenitor cells that are destined for the olfactory bulb is of particular interest, specifically as there are many facets of these progenitors and their molecular profiles remain unknown. Using modified StarTrack genetic tracing strategies, specific SVZ progenitor cells were targeted in E12 mice embryos, and the cell fate of these neural progenitors was determined in the adult olfactory bulb. This study defined the distribution and the phenotypic diversity of olfactory bulb interneurons from specific SVZ-progenitor cells, focusing on their spatial pallial origin, heterogeneity, and genetic profile.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Olfactory Bulb/growth & development , Stem Cells/metabolism , Animals , Cell Movement/genetics , Interneurons/cytology , Interneurons/metabolism , Lateral Ventricles , Mice , Olfactory Bulb/cytology , Stem Cells/classification
5.
Stem Cell Reports ; 13(4): 700-712, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31543472

ABSTRACT

Understanding the contribution of adult neural progenitor cells (NPCs) and their lineage potential is a great challenge in neuroscience. To reveal progenitor diversity and cell-lineage relationships of postnatal NPCs in the subventricular zone (SVZ), we performed in vivo lineage-tracing genetic analysis using the UbC-StarTrack. We determined the progeny of single SVZ-NPCs, the number of cells per clone, the dispersion of sibling cells, and the cell types within clones. Long-term analysis revealed that both the cell-dispersion pattern and number of cells comprising clones varied depending on the glial/neuronal nature of sibling cells. Sibling-olfactory interneurons were primarily located within the same layer, while sibling-glial cells populated SVZ-adjacent areas. Sibling astrocytes and interneurons did not form big clones, whereas oligodendroglial-lineage clones comprised the largest clones originated in adult brains. These results demonstrate the existence of SVZ postnatal bipotential progenitors that give rise to clones widely dispersed across the olfactory bulb and SVZ-adjacent areas.


Subject(s)
Action Potentials , Cell Lineage , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Cells, Cultured , Clone Cells , Fluorescent Antibody Technique , Interneurons/cytology , Interneurons/metabolism , Mice , Neuroglia/cytology , Olfactory Bulb/cytology , Oligodendroglia/cytology
6.
Cell Rep ; 27(2): 429-441.e3, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30970247

ABSTRACT

The adult mouse brain contains an extensive neurogenic niche in the lateral walls of the lateral ventricles. This epithelium, which has a unique pinwheel organization, contains multiciliated ependymal (E1) cells and neural stem cells (B1). This postnatal germinal epithelium develops from the embryonic ventricular zone, but the lineage relationship between E1 and B1 cells remains unknown. Distinct subpopulations of radial glia (RG) cells in late embryonic and early postnatal development either expand their apical domain >11-fold to form E1 cells or retain small apical domains that coalesce into the centers of pinwheels to form B1 cells. Using independent methods of lineage tracing, we show that individual RG cells can give rise to clones containing E1 and B1 cells. This study reveals key developmental steps in the formation of the postnatal germinal niche and the shared cellular origin of E1 and B1 cells.


Subject(s)
Ependyma/embryology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Animals , Humans , Mice
7.
Cell Stem Cell ; 23(4): 557-571.e8, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30290178

ABSTRACT

A broad molecular framework of how neural stem cells are specified toward astrocyte fate during brain development has proven elusive. Here we perform comprehensive and integrated transcriptomic and epigenomic analyses to delineate gene regulatory programs that drive the developmental trajectory from mouse embryonic stem cells to astrocytes. We report molecularly distinct phases of astrogliogenesis that exhibit stage- and lineage-specific transcriptomic and epigenetic signatures with unique primed and active chromatin regions, thereby revealing regulatory elements and transcriptional programs underlying astrocyte generation and maturation. By searching for transcription factors that function at these elements, we identified NFIA and ATF3 as drivers of astrocyte differentiation from neural precursor cells while RUNX2 promotes astrocyte maturation. These transcription factors facilitate stage-specific gene expression programs by switching the chromatin state of their target regulatory elements from primed to active. Altogether, these findings provide integrated insights into the genetic and epigenetic mechanisms steering the trajectory of astrogliogenesis.


Subject(s)
Activating Transcription Factor 3/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation/genetics , NFI Transcription Factors/metabolism , Neurogenesis/genetics , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL
8.
PLoS Biol ; 16(9): e2005513, 2018 09.
Article in English | MEDLINE | ID: mdl-30260948

ABSTRACT

The morphological, molecular, and functional heterogeneity of astrocytes is under intense scrutiny, but how this diversity is ontogenetically achieved remains largely unknown. Here, by quantitative in vivo clonal analyses and proliferation studies, we demonstrate that the major cerebellar astrocyte types emerge according to an unprecedented and remarkably orderly developmental program comprising (i) a time-dependent decline in both clone size and progenitor multipotency, associated with clone allocation first to the hemispheres and then to the vermis(ii) distinctive clonal relationships among astrocyte types, revealing diverse lineage potentials of embryonic and postnatal progenitors; and (iii) stereotyped clone architectures and recurrent modularities that correlate to layer-specific dynamics of postnatal proliferation/differentiation. In silico simulations indicate that the sole presence of a unique multipotent progenitor at the source of the whole astrogliogenic program is unlikely and rather suggest the involvement of additional committed components.


Subject(s)
Astrocytes/cytology , Cerebellum/cytology , Animals , Animals, Newborn , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Proliferation , Cell Size , Cerebellum/embryology , Clone Cells , Computer Simulation , Female , Humans , Mice, Inbred C57BL , Models, Biological , White Matter/cytology
9.
Sci Rep ; 6: 33896, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27654510

ABSTRACT

Clonal cell analysis defines the potential of single cells and the diversity they can produce. To achieve this, we have developed a novel adaptation of the genetic tracing strategy, UbC-StarTrack, which attributes a specific and unique color-code to single neural precursors, allowing all their progeny to be tracked. We used integrable fluorescent reporters driven by a ubiquitous promoter in PiggyBac-based vectors to achieve inheritable and stable clonal cell labeling. In addition, coupling this to an inducible Cre-LoxP system avoids the expression of non-integrated reporters. To assess the utility of this system, we first analyzed images of combinatorial expression of fluorescent reporters in transfected cells and their progeny. We also validated the efficiency of the UbC-StarTrack to trace cell lineages through in vivo, in vitro and ex vivo strategies. Finally, progenitors located in the lateral ventricles were targeted at embryonic or postnatal stages to determine the diversity of neurons and glia they produce, and their clonal relationships. In this way we demonstrate that UbC-StarTrack can be used to identify all the progeny of a single cell and that it can be employed in a wide range of contexts.

10.
Front Neurosci ; 10: 194, 2016.
Article in English | MEDLINE | ID: mdl-27242400

ABSTRACT

Neurons are generated during embryonic development and in adulthood, although adult neurogenesis is restricted to two main brain regions, the hippocampus and olfactory bulb. The subventricular zone (SVZ) of the lateral ventricles generates neural stem/progenitor cells that continually provide the olfactory bulb (OB) with new granule or periglomerular neurons, cells that arrive from the SVZ via the rostral migratory stream. The continued neurogenesis and the adequate integration of these newly generated interneurons is essential to maintain homeostasis in the olfactory bulb, where the differentiation of these cells into specific neural cell types is strongly influenced by temporal cues. Therefore, identifying the critical features that control the generation of adult OB interneurons at either pre- or post-natal stages is important to understand the dynamic contribution of neural stem cells. Here, we used in utero and neonatal SVZ electroporation along with a transposase-mediated stable integration plasmid, in order to track interneurons and glial lineages in the OB. These plasmids are valuable tools to study the development of OB interneurons from embryonic and post-natal SVZ progenitors. Accordingly, we examined the location and identity of the adult progeny of embryonic and post-natally transfected progenitors by examining neurochemical markers in the adult OB. These data reveal the different cell types in the olfactory bulb that are generated in function of age and different electroporation conditions.

11.
J Neurosci ; 35(19): 7388-402, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25972168

ABSTRACT

Cerebellar GABAergic interneurons in mouse comprise multiple subsets of morphologically and neurochemically distinct phenotypes located at strategic nodes of cerebellar local circuits. These cells are produced by common progenitors deriving from the ventricular epithelium during embryogenesis and from the prospective white matter (PWM) during postnatal development. However, it is not clear whether these progenitors are also shared by other cerebellar lineages and whether germinative sites different from the PWM originate inhibitory interneurons. Indeed, the postnatal cerebellum hosts another germinal site along the Purkinje cell layer (PCL), in which Bergmann glia are generated up to first the postnatal weeks, which was proposed to be neurogenic. Both PCL and PWM comprise precursors displaying traits of juvenile astroglia and neural stem cell markers. First, we examine the proliferative and fate potential of these niches, showing that different proliferative dynamics regulate progenitor amplification at these sites. In addition, PCL and PWM differ in the generated progeny. GABAergic interneurons are produced exclusively by PWM astroglial-like progenitors, whereas PCL precursors produce only astrocytes. Finally, through in vitro, ex vivo, and in vivo clonal analyses we provide evidence that the postnatal PWM hosts a bipotent progenitor that gives rise to both interneurons and white matter astrocytes.


Subject(s)
Cell Proliferation/physiology , Cerebellum/cytology , Gene Expression Regulation, Developmental/physiology , Interneurons/physiology , Neuroglia/physiology , Stem Cells/physiology , Actins/genetics , Actins/metabolism , Animals , Animals, Newborn , CD2 Antigens/genetics , CD2 Antigens/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Cerebellum/embryology , Cerebellum/growth & development , Embryo, Mammalian , Estrogen Antagonists/pharmacology , Excitatory Amino Acid Transporter 1/genetics , Female , GABAergic Neurons/physiology , Gene Expression Regulation, Developmental/drug effects , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tamoxifen/pharmacology , White Matter/cytology , White Matter/metabolism
12.
Front Neurosci ; 9: 87, 2015.
Article in English | MEDLINE | ID: mdl-25852461

ABSTRACT

Genetic lineage tracing with electroporation is one of the most powerful techniques to target neural progenitor cells and their progeny. However, the spatiotemporal relationship between neural progenitors and their final phenotype remain poorly understood. One critical factor to analyze the cell fate of progeny is reporter integration into the genome of transfected cells. To address this issue, we performed postnatal and in utero co-electroporations of different fluorescent reporters to label, in both cerebral cortex and olfactory bulb, the progeny of subventricular zone neural progenitors. By comparing fluorescent reporter expression in the adult cell progeny, we show a differential expression pattern within the same cell lineage, depending on electroporation stage and cell identity. Further, while neuronal lineages arise from many progenitors in proliferative zones after few divisions, glial lineages come from fewer progenitors that accomplish many cell divisions. Together, these data provide a useful guide to select a strategy to track the cell fate of a specific cell population and to address whether a different proliferative origin might be correlated with functional heterogeneity.

13.
Front Neuroanat ; 8: 55, 2014.
Article in English | MEDLINE | ID: mdl-25071462

ABSTRACT

The olfactory system has a highly regular organization of interconnected synaptic circuits from the periphery. It is therefore an excellent model for understanding general principles about how the brain processes information. Cajal revealed the basic cell types and their interconnections at the end of the XIX century. Since his original descriptions, the observation and analysis of the olfactory system and its components represents a major topic in neuroscience studies, providing important insights into the neural mechanisms. In this review, we will highlight the importance of Cajal contributions and his legacy to the actual knowledge of the olfactory system.

SELECTION OF CITATIONS
SEARCH DETAIL
...