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










Publication year range
1.
Cell Mol Life Sci ; 80(1): 36, 2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36627412

ABSTRACT

Cell differentiation involves profound changes in global gene expression that often has to occur in coordination with cell cycle exit. Because cyclin-dependent kinase inhibitor p27 reportedly regulates proliferation of neural progenitor cells in the subependymal neurogenic niche of the adult mouse brain, but can also have effects on gene expression, we decided to molecularly analyze its role in adult neurogenesis and oligodendrogenesis. At the cell level, we show that p27 restricts residual cyclin-dependent kinase activity after mitogen withdrawal to antagonize cycling, but it is not essential for cell cycle exit. By integrating genome-wide gene expression and chromatin accessibility data, we find that p27 is coincidentally necessary to repress many genes involved in the transit from multipotentiality to differentiation, including those coding for neural progenitor transcription factors SOX2, OLIG2 and ASCL1. Our data reveal both a direct association of p27 with regulatory sequences in the three genes and an additional hierarchical relationship where p27 repression of Sox2 leads to reduced levels of its downstream targets Olig2 and Ascl1. In vivo, p27 is also required for the regulation of the proper level of SOX2 necessary for neuroblasts and oligodendroglial progenitor cells to timely exit cell cycle in a lineage-dependent manner.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27 , Neurogenesis , SOXB1 Transcription Factors , Animals , Mice , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Division , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression , Neurogenesis/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
2.
Nat Commun ; 14(1): 373, 2023 01 23.
Article in English | MEDLINE | ID: mdl-36690670

ABSTRACT

Neural stem cells (NSCs) in the adult murine subependymal zone balance their self-renewal capacity and glial identity with the potential to generate neurons during the lifetime. Adult NSCs exhibit lineage priming via pro-neurogenic fate determinants. However, the protein levels of the neural fate determinants are not sufficient to drive direct differentiation of adult NSCs, which raises the question of how cells along the neurogenic lineage avoid different conflicting fate choices, such as self-renewal and differentiation. Here, we identify RNA-binding protein MEX3A as a post-transcriptional regulator of a set of stemness associated transcripts at critical transitions in the subependymal neurogenic lineage. MEX3A regulates a quiescence-related RNA signature in activated NSCs that is needed for their return to quiescence, playing a role in the long-term maintenance of the NSC pool. Furthermore, it is required for the repression of the same program at the onset of neuronal differentiation. Our data indicate that MEX3A is a pivotal regulator of adult murine neurogenesis acting as a translational remodeller.


Subject(s)
Neural Stem Cells , Neurogenesis , Mice , Animals , Neurogenesis/genetics , Neurons/physiology , Neural Stem Cells/metabolism , Cell Differentiation/genetics , RNA-Binding Proteins/metabolism
4.
Nat Commun ; 13(1): 334, 2022 01 17.
Article in English | MEDLINE | ID: mdl-35039505

ABSTRACT

RNF43/ZNRF3 negatively regulate WNT signalling. Both genes are mutated in several types of cancers, however, their contribution to liver disease is unknown. Here we describe that hepatocyte-specific loss of Rnf43/Znrf3 results in steatohepatitis and in increase in unsaturated lipids, in the absence of dietary fat supplementation. Upon injury, Rnf43/Znrf3 deletion results in defective hepatocyte regeneration and liver cancer, caused by an imbalance between differentiation/proliferation. Using hepatocyte-, hepatoblast- and ductal cell-derived organoids we demonstrate that the differentiation defects and lipid alterations are, in part, cell-autonomous. Interestingly, ZNRF3 mutant liver cancer patients present poorer prognosis, altered hepatic lipid metabolism and steatohepatitis/NASH signatures. Our results imply that RNF43/ZNRF3 predispose to liver cancer by controlling the proliferative/differentiation and lipid metabolic state of hepatocytes. Both mechanisms combined facilitate the progression towards malignancy. Our findings might aid on the management of those RNF43/ZNRF3 mutated individuals at risk of developing fatty liver and/or liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Liver Regeneration , Liver/metabolism , Ubiquitin-Protein Ligases/metabolism , Adult , Animals , Carcinoma, Hepatocellular/pathology , Cell Differentiation , Cell Proliferation , Fatty Liver/pathology , Gene Deletion , Gene Expression Regulation , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatomegaly/pathology , Humans , Hyperplasia , Lipid Droplets/metabolism , Lipid Metabolism/genetics , Lipidomics , Liver/pathology , Liver Neoplasms/pathology , Mice , Prognosis
5.
Cell Stem Cell ; 28(11): 1907-1921.e8, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34343491

ABSTRACT

In the liver, ductal cells rarely proliferate during homeostasis but do so transiently after tissue injury. These cells can be expanded as organoids that recapitulate several of the cell-autonomous mechanisms of regeneration but lack the stromal interactions of the native tissue. Here, using organoid co-cultures that recapitulate the ductal-to-mesenchymal cell architecture of the portal tract, we demonstrate that a subpopulation of mouse periportal mesenchymal cells exerts dual control on proliferation of the epithelium. Ductal cell proliferation is either induced and sustained or, conversely, completely abolished, depending on the number of direct mesenchymal cell contacts, through a mechanism mediated, at least in part, by Notch signaling. Our findings expand the concept of the cellular niche in epithelial tissues, whereby not only soluble factors but also cell-cell contacts are the key regulatory cues involved in the control of cellular behaviors, suggesting a critical role for cell-cell contacts during regeneration.


Subject(s)
Epithelial Cells , Mesoderm , Animals , Cell Proliferation , Epithelium , Liver , Mice
6.
STAR Protoc ; 2(2): 100425, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33899012

ABSTRACT

This protocol provides a flow-cytometry-based procedure to classify and isolate all cells of the adult rodent subependymal zone (SEZ) neurogenic lineage, without the need for reporter mice, into different cell populations, including three neural stem cell (NSC) fractions with molecular signatures that are coherent with single-cell transcriptomics. Additionally, their cycling behavior can be assessed by means of 5-ethynyl-2'-deoxyuridine (EdU) incorporation. Our method allows the isolation of different NSC fractions and the functional assay of their cycling heterogeneity and quiescence-activation transitions. For complete details on the use, execution, and outcomes of this protocol, please refer to Belenguer et al. (2021).


Subject(s)
Ependyma/cytology , Flow Cytometry/methods , Neural Stem Cells/cytology , Single-Cell Analysis/methods , Transcriptome/genetics , Animals , Cell Culture Techniques , Cell Line , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL
7.
Cell Stem Cell ; 28(2): 285-299.e9, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33207218

ABSTRACT

Adult stem cells (SCs) transit between the cell cycle and a poorly defined quiescent state. Single neural SCs (NSCs) with quiescent, primed-for-activation, and activated cell transcriptomes have been obtained from the subependymal zone (SEZ), but the functional regulation of these states under homeostasis is not understood. Here, we develop a multilevel strategy to analyze these NSC states with the aim to uncover signals that regulate their level of quiescence/activation. We show that transitions between states occur in vivo and that activated and primed, but not quiescent, states can be captured and studied in culture. We also show that peripherally induced inflammation promotes a transient activation of primed NSCs (pNSCs) mediated by tumor necrosis factor α (TNF-α) acting through its receptor, TNF receptor 2 (TNFR2), and a return to quiescence in a TNF receptor 1 (TNFR1)-dependent manner. Our data identify a signaling pathway promoting NSC alertness and add to the emerging concept that SCs can respond to the systemic milieu.


Subject(s)
Adult Stem Cells , Neural Stem Cells , Humans , Inflammation , Lateral Ventricles , Neurogenesis , Receptors, Tumor Necrosis Factor , Receptors, Tumor Necrosis Factor, Type I , Signal Transduction , Tumor Necrosis Factor-alpha
8.
Proc Natl Acad Sci U S A ; 117(49): 31448-31458, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229571

ABSTRACT

Adult neural stem cells (NSC) serve as a reservoir for brain plasticity and origin for certain gliomas. Lineage tracing and genomic approaches have portrayed complex underlying heterogeneity within the major anatomical location for NSC, the subventricular zone (SVZ). To gain a comprehensive profile of NSC heterogeneity, we utilized a well-validated stem/progenitor-specific reporter transgene in concert with single-cell RNA sequencing to achieve unbiased analysis of SVZ cells from infancy to advanced age. The magnitude and high specificity of the resulting transcriptional datasets allow precise identification of the varied cell types embedded in the SVZ including specialized parenchymal cells (neurons, glia, microglia) and noncentral nervous system cells (endothelial, immune). Initial mining of the data delineates four quiescent NSC and three progenitor-cell subpopulations formed in a linear progression. Further evidence indicates that distinct stem and progenitor populations reside in different regions of the SVZ. As stem/progenitor populations progress from neonatal to advanced age, they acquire a deficiency in transition from quiescence to proliferation. Further data mining identifies stage-specific biological processes, transcription factor networks, and cell-surface markers for investigation of cellular identities, lineage relationships, and key regulatory pathways in adult NSC maintenance and neurogenesis.


Subject(s)
Aging/genetics , Cell Lineage , Lateral Ventricles/anatomy & histology , Lateral Ventricles/cytology , Stem Cell Niche/genetics , Transcriptome/genetics , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Lineage/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Transgenes
9.
Cells ; 8(12)2019 11 30.
Article in English | MEDLINE | ID: mdl-31801296

ABSTRACT

The renin-angiotensin system (RAS), and particularly its angiotensin type-2 receptors (AT2), have been classically involved in processes of cell proliferation and maturation during development. However, the potential role of RAS in adult neurogenesis in the ventricular-subventricular zone (V-SVZ) and its aging-related alterations have not been investigated. In the present study, we analyzed the role of major RAS receptors on neurogenesis in the V-SVZ of adult mice and rats. In mice, we showed that the increase in proliferation of cells in this neurogenic niche was induced by activation of AT2 receptors but depended partially on the AT2-dependent antagonism of AT1 receptor expression, which restricted proliferation. Furthermore, we observed a functional dependence of AT2 receptor actions on Mas receptors. In rats, where the levels of the AT1 relative to those of AT2 receptor are much lower, pharmacological inhibition of the AT1 receptor alone was sufficient in increasing AT2 receptor levels and proliferation in the V-SVZ. Our data revealed that interactions between RAS receptors play a major role in the regulation of V-SVZ neurogenesis, particularly in proliferation, generation of neuroblasts, and migration to the olfactory bulb, both in young and aged brains, and suggest potential beneficial effects of RAS modulators on neurogenesis.


Subject(s)
Lateral Ventricles/metabolism , Neurogenesis , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Age Factors , Angiotensin II/metabolism , Animals , Immunohistochemistry , Male , Mice , Mice, Knockout , Models, Biological , Neural Stem Cells/metabolism , Neurogenesis/genetics , Protein Binding , Rats , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/genetics
10.
Nat Cell Biol ; 21(11): 1321-1333, 2019 11.
Article in English | MEDLINE | ID: mdl-31685987

ABSTRACT

Following severe or chronic liver injury, adult ductal cells (cholangiocytes) contribute to regeneration by restoring both hepatocytes and cholangiocytes. We recently showed that ductal cells clonally expand as self-renewing liver organoids that retain their differentiation capacity into both hepatocytes and ductal cells. However, the molecular mechanisms by which adult ductal-committed cells acquire cellular plasticity, initiate organoids and regenerate the damaged tissue remain largely unknown. Here, we describe that ductal cells undergo a transient, genome-wide, remodelling of their transcriptome and epigenome during organoid initiation and in vivo following tissue damage. TET1-mediated hydroxymethylation licences differentiated ductal cells to initiate organoids and activate the regenerative programme through the transcriptional regulation of stem-cell genes and regenerative pathways including the YAP-Hippo signalling. Our results argue in favour of the remodelling of genomic methylome/hydroxymethylome landscapes as a general mechanism by which differentiated cells exit a committed state in response to tissue damage.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Epigenome , Liver Regeneration/genetics , Liver/metabolism , Organoids/metabolism , Proto-Oncogene Proteins/genetics , Transcriptome , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bile Ducts/cytology , Bile Ducts/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Hippo Signaling Pathway , Liver/cytology , Male , Mice, Transgenic , Organoids/cytology , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , YAP-Signaling Proteins
11.
Neuroscience ; 405: 77-91, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30677487

ABSTRACT

Microglia are the prototypical innate immune cells of the central nervous system. They constitute a unique type of tissue-resident mononuclear phagocytes which act as glial cells. Elegant experiments in the last few years have revealed the origin, extraordinary molecular diversity, and phenotypic plasticity of these cells and how their potential relates to both immune and non-immune actions in the normal and diseased brain. Microglial cells originate in the yolk sac and colonize the brain during embryogenesis, playing a role in neural development and later in adult brain function. Neurogenesis continues after birth in discrete areas of the mammalian brain sustained by the postnatal persistence of neural stem cells in specific neurogenic niches. Recent data indicate that microglial cells are distinct cellular elements of these neurogenic niches where they regulate different aspects of stem cell biology. Interestingly, microglial and neural stem cells are specified very early in fetal development and persist as self-renewing populations throughout life, suggesting potential life-long interactions between them. We aim at reviewing these interactions in one neurogenic niche, the subependymal zone.


Subject(s)
Cell Communication/physiology , Microglia/physiology , Neural Stem Cells/physiology , Adult , Adult Stem Cells/cytology , Adult Stem Cells/physiology , Animals , Brain/cytology , Brain/physiology , Humans , Microglia/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Neurons/physiology , Stem Cell Niche/physiology
12.
J Neurosci ; 38(4): 814-825, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29217686

ABSTRACT

Synaptic protein α-synuclein (α-SYN) modulates neurotransmission in a complex and poorly understood manner and aggregates in the cytoplasm of degenerating neurons in Parkinson's disease. Here, we report that α-SYN present in dopaminergic nigral afferents is essential for the normal cycling and maintenance of neural stem cells (NSCs) in the brain subependymal zone of adult male and female mice. We also show that premature senescence of adult NSCs into non-neurogenic astrocytes in mice lacking α-SYN resembles the effects of dopaminergic fiber degeneration resulting from chronic exposure to 1-methyl-4-phenyl-1,2,3,6-tetra-hydropyridine or intranigral inoculation of aggregated toxic α-SYN. Interestingly, NSC loss in α-SYN-deficient mice can be prevented by viral delivery of human α-SYN into their sustantia nigra or by treatment with l-DOPA, suggesting that α-SYN regulates dopamine availability to NSCs. Our data indicate that α-SYN, present in dopaminergic nerve terminals supplying the subependymal zone, acts as a niche component to sustain the neurogenic potential of adult NSCs and identify α-SYN and DA as potential targets to ameliorate neurogenic defects in the aging and diseased brain.SIGNIFICANCE STATEMENT We report an essential role for the protein α-synuclein present in dopaminergic nigral afferents in the regulation of adult neural stem cell maintenance, identifying the first synaptic regulator with an implication in stem cell niche biology. Although the exact role of α-synuclein in neural transmission is not completely clear, our results indicate that it is required for stemness and the preservation of neurogenic potential in concert with dopamine.


Subject(s)
Brain/metabolism , Dopaminergic Neurons/metabolism , Neural Stem Cells/metabolism , Stem Cell Niche/physiology , alpha-Synuclein/metabolism , Animals , Brain/cytology , Cellular Senescence/physiology , Dopamine/metabolism , Dopaminergic Neurons/cytology , Female , Humans , Male , Mice , Mice, Mutant Strains , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons, Afferent/cytology , Neurons, Afferent/metabolism
13.
Differentiation ; 91(4-5): 28-41, 2016.
Article in English | MEDLINE | ID: mdl-27016251

ABSTRACT

Individual cells dissected from the subependymal neurogenic niche of the adult mouse brain proliferate in medium containing basic fibroblast growth factor (bFGF) and/or epidermal growth factor (EGF) as mitogens, to produce multipotent clonal aggregates called neurospheres. These cultures constitute a powerful tool for the study of neural stem cells (NSCs) provided that they allow the analysis of their features and potential capacity in a controlled environment that can be modulated and monitored more accurately than in vivo. Clonogenic and population analyses under mitogen addition or withdrawal allow the quantification of the self-renewing and multilineage potency of these cells and the identification of the mechanisms involved in these properties. Here, we describe a set of procedures developed and/or modified by our group including several experimental options that can be used either independently or in combination for the ex vivo assessment of cell properties of NSCs obtained from the adult subependymal niche.


Subject(s)
Cell Culture Techniques , Ependyma/growth & development , Neural Stem Cells/cytology , Neurogenesis/genetics , Adult Stem Cells , Animals , Cell Differentiation/genetics , Ependyma/cytology , Humans , Mice , Neurons
SELECTION OF CITATIONS
SEARCH DETAIL
...