Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5489, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38942786

ABSTRACT

Lipid droplets (LDs) are dynamic lipid storage organelles. They are tightly linked to metabolism and can exert protective functions, making them important players in health and disease. Most LD studies in vivo rely on staining methods, providing only a snapshot. We therefore developed a LD-reporter mouse by labelling the endogenous LD coat protein perilipin 2 (PLIN2) with tdTomato, enabling staining-free fluorescent LD visualisation in living and fixed tissues and cells. Here we validate this model under standard and high-fat diet conditions and demonstrate that LDs are highly abundant in various cell types in the healthy brain, including neurons, astrocytes, ependymal cells, neural stem/progenitor cells and microglia. Furthermore, we also show that LDs are abundant during brain development and can be visualized using live imaging of embryonic slices. Taken together, our tdTom-Plin2 mouse serves as a novel tool to study LDs and their dynamics under both physiological and diseased conditions in all tissues expressing Plin2.


Subject(s)
Brain , Lipid Droplets , Perilipin-2 , Animals , Perilipin-2/metabolism , Perilipin-2/genetics , Lipid Droplets/metabolism , Brain/metabolism , Mice , Neurons/metabolism , Gene Knock-In Techniques , Mice, Transgenic , Female , Luminescent Proteins/metabolism , Luminescent Proteins/genetics , Male , Astrocytes/metabolism , Diet, High-Fat , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Microglia/metabolism
2.
Trends Endocrinol Metab ; 34(8): 446-461, 2023 08.
Article in English | MEDLINE | ID: mdl-37380501

ABSTRACT

Metabolism has emerged as a key regulator of stem cell behavior. Mitochondria are crucial metabolic organelles that are important for differentiated cells, yet considered less so for stem cells. However, recent studies have shown that mitochondria influence stem cell maintenance and fate decisions, inviting a revised look at this topic. In this review, we cover the current literature addressing the role of mitochondrial metabolism in mouse and human neural stem cells (NSCs) in the embryonic and adult brain. We summarize how mitochondria are implicated in fate regulation and how substrate oxidation affects NSC quiescence. We further explore single-cell RNA sequencing (scRNA-seq) data for metabolic signatures of adult NSCs, highlight emerging technologies reporting on metabolic signatures, and discuss mitochondrial metabolism in other stem cells.


Subject(s)
Adult Stem Cells , Neural Stem Cells , Humans , Mice , Animals , Neural Stem Cells/metabolism , Cell Differentiation/physiology , Mitochondria/metabolism , Adult Stem Cells/metabolism , Oxidation-Reduction
3.
Sci Adv ; 9(9): eadd5220, 2023 03.
Article in English | MEDLINE | ID: mdl-36857455

ABSTRACT

Cellular metabolism is important for adult neural stem/progenitor cell (NSPC) behavior. However, its role in the transition from quiescence to proliferation is not fully understood. We here show that the mitochondrial pyruvate carrier (MPC) plays a crucial and unexpected part in this process. MPC transports pyruvate into mitochondria, linking cytosolic glycolysis to mitochondrial tricarboxylic acid cycle and oxidative phosphorylation. Despite its metabolic key function, the role of MPC in NSPCs has not been addressed. We show that quiescent NSPCs have an active mitochondrial metabolism and express high levels of MPC. Pharmacological MPC inhibition increases aspartate and triggers NSPC activation. Furthermore, genetic Mpc1 ablation in vitro and in vivo also activates NSPCs, which differentiate into mature neurons, leading to overall increased hippocampal neurogenesis in adult and aged mice. These findings highlight the importance of metabolism for NSPC regulation and identify an important pathway through which mitochondrial pyruvate import controls NSPC quiescence and activation.


Subject(s)
Neural Stem Cells , Neurogenesis , Animals , Mice , Neurons , Biological Transport , Mitochondria , Monocarboxylic Acid Transporters
5.
Nat Commun ; 12(1): 7362, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934077

ABSTRACT

Neural stem/progenitor cells (NSPCs) generate new neurons throughout adulthood. However, the underlying regulatory processes are still not fully understood. Lipid metabolism plays an important role in regulating NSPC activity: build-up of lipids is crucial for NSPC proliferation, whereas break-down of lipids has been shown to regulate NSPC quiescence. Despite their central role for cellular lipid metabolism, the role of lipid droplets (LDs), the lipid storing organelles, in NSPCs remains underexplored. Here we show that LDs are highly abundant in adult mouse NSPCs, and that LD accumulation is significantly altered upon fate changes such as quiescence and differentiation. NSPC proliferation is influenced by the number of LDs, inhibition of LD build-up, breakdown or usage, and the asymmetric inheritance of LDs during mitosis. Furthermore, high LD-containing NSPCs have increased metabolic activity and capacity, but do not suffer from increased oxidative damage. Together, these data indicate an instructive role for LDs in driving NSPC behaviour.


Subject(s)
Lipid Droplets/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Inheritance Patterns/genetics , Lipid Peroxidation , Male , Mice, Inbred C57BL , Mitosis , Neurons/cytology , Neurons/metabolism , Perilipin-2/metabolism , Phospholipids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
6.
Immunometabolism ; 3(4): e210034, 2021.
Article in English | MEDLINE | ID: mdl-34754515

ABSTRACT

Research led by Katrin Andreasson suggests that fixing age-induced metabolic defects in myeloid cells would suffice to reverse cognitive impairment and to restore synaptic plasticity to the level of young subjects, at least in mice. This opens up the possibility to develop rejuvenating strategies by targeting immune dysfunction.

7.
Development ; 148(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34042969

ABSTRACT

Cellular metabolism has recently emerged as a key regulator of stem cell behavior. Various studies have suggested that metabolic regulatory mechanisms are conserved in different stem cell niches, suggesting a common level of stem cell regulation across tissues. Although the balance between glycolysis and oxidative phosphorylation has been shown to be distinct in stem cells and their differentiated progeny, much less is known about lipid metabolism in stem cell regulation. In this Review, we focus on how stem cells are affected by two major lipid metabolic pathways: the build-up of lipids, called de novo lipogenesis, and the breakdown of lipids, called fatty acid beta-oxidation. We cover the recent literature on hematopoietic stem cells, intestinal stem cells, neural stem/progenitor cells and cancer stem cells, where these two lipid pathways have been studied in more depth.


Subject(s)
Hematopoietic Stem Cells/metabolism , Lipid Metabolism/physiology , Lipogenesis/physiology , Lipolysis/physiology , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Animals , Energy Metabolism/physiology , Fatty Acids/metabolism , Glycolysis/physiology , Hematopoiesis/physiology , Humans , Neurogenesis/physiology , Oxidative Phosphorylation
8.
Sci Rep ; 10(1): 14642, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32887893

ABSTRACT

The green fluorescent protein (GFP) is a powerful reporter protein that allows labeling of specific proteins or entire cells. However, as GFP is a small soluble protein, it easily crosses membranes if cell integrity is disrupted, and GFP signal is lost or diffuse if the specimen is not fixed beforehand. While pre-fixation is often feasible for histological analyses, many molecular biology procedures and new imaging techniques, such as imaging mass spectrometry, require unfixed specimens. To be able to use GFP labeling in tissues prepared for such applications, we have tested various protocols to minimize the loss of GFP signal. Here we show that, in cryocut sections of snap-frozen brain tissue from two GFP reporter mouse lines, leaking of the GFP signal is prevented by omitting the commonly performed drying of the cryosections, and by direct post-fixation with 4% paraformaldehyde pre-warmed at 30-37 °C. Although the GFP staining does not reach the same quality as obtained with pre-fixed tissue, GFP localization within the cells that express it is preserved with this method. This protocol can thus be used to identify GFP positive cells on sections originating from unfixed, cryosectioned tissue.


Subject(s)
Cryopreservation/methods , Dentate Gyrus/metabolism , Frozen Sections/methods , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/metabolism , Neural Stem Cells/metabolism , Tissue Fixation/methods , Animals , Dentate Gyrus/pathology , Formaldehyde/chemistry , Genes, Reporter , Green Fluorescent Proteins/genetics , Immunohistochemistry/methods , Mice , Mice, Transgenic , Nestin/genetics , Polymers/chemistry , Promoter Regions, Genetic , Staining and Labeling/methods
9.
Stem Cell Reports ; 15(3): 566-576, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32857979

ABSTRACT

Fatty acid ß-oxidation (FAO), the breakdown of lipids, is a metabolic pathway used by various stem cells. FAO levels are generally high during quiescence and downregulated with proliferation. The endogenous metabolite malonyl-CoA modulates lipid metabolism as a reversible FAO inhibitor and as a substrate for de novo lipogenesis. Here we assessed whether malonyl-CoA can be exploited to steer the behavior of hematopoietic stem/progenitor cells (HSPCs), quiescent stem cells of clinical relevance. Treatment of mouse HSPCs in vitro with malonyl-CoA increases HSPC numbers compared with nontreated controls and ameliorates blood reconstitution capacity when transplanted in vivo, mainly through enhanced lymphoid reconstitution. Similarly, human HSPC numbers also increase upon malonyl-CoA treatment in vitro. These data corroborate that lipid metabolism can be targeted to direct cell fate and stem cell proliferation. Physiological modulation of metabolic pathways, rather than genetic or pharmacological inhibition, provides unique perspectives for stem cell manipulations in health and disease.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lipid Metabolism , Lymphocytes/cytology , Metabolome , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation/genetics , Cells, Cultured , Fatty Acids/metabolism , Gene Expression Regulation , Lipid Metabolism/genetics , Lymphocytes/metabolism , Malonyl Coenzyme A/metabolism , Metabolome/genetics , Mice, Inbred C57BL , Oxidation-Reduction
10.
Cell Stem Cell ; 25(6): 729-731, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31809733

ABSTRACT

Metabolism has emerged as a key player for stem cell behavior; however, the role of metabolism in the microenvironment remains poorly understood. In this issue of Cell Stem Cell, Wang et al. (2019) show that brain endothelial cells directly affect adult neural stem cells and maintain a healthy metabolic environment by regulating lactate levels.


Subject(s)
Endothelial Cells , Lactic Acid , Adult , Brain , Hippocampus , Homeostasis , Humans , Neurogenesis
11.
Development ; 146(13)2019 06 21.
Article in English | MEDLINE | ID: mdl-31227641

ABSTRACT

The 2nd SY-Stem Symposium - a symposium for 'the next generation of stem cell researchers' - was held on the 21-23 March 2019 at the Vienna BioCenter in Austria. After the great success of the initial SY-Stem meeting in 2018, this year's event again focused on the work of young scientists. Here, we summarize the impressive amount of new research covering stem cell-related fields that was discussed at the meeting.


Subject(s)
Biomedical Research/trends , Stem Cell Research , Stem Cells/cytology , Systems Biology , Animals , Austria , Biomedical Research/organization & administration , Congresses as Topic/organization & administration , Congresses as Topic/standards , Humans , Regenerative Medicine/organization & administration , Regenerative Medicine/trends , Systems Biology/methods , Systems Biology/trends
13.
Cell Rep ; 20(9): 2144-2155, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28854364

ABSTRACT

Hippocampal neurogenesis is important for certain forms of cognition, and failing neurogenesis has been implicated in neuropsychiatric diseases. The neurogenic capacity of hippocampal neural stem/progenitor cells (NSPCs) depends on a balance between quiescent and proliferative states. Here, we show that the rate of fatty acid oxidation (FAO) regulates the activity of NSPCs. Quiescent NSPCs show high levels of carnitine palmitoyltransferase 1a (Cpt1a)-dependent FAO, which is downregulated in proliferating NSPCs. Pharmacological inhibition and conditional deletion of Cpt1a in vitro and in vivo leads to altered NSPC behavior, showing that Cpt1a-dependent FAO is required for stem cell maintenance and proper neurogenesis. Strikingly, manipulation of malonyl-CoA, the metabolite that regulates levels of FAO, is sufficient to induce exit from quiescence and to enhance NSPC proliferation. Thus, the data presented here identify a shift in FAO metabolism that governs NSPC behavior and suggest an instructive role for fatty acid metabolism in regulating NSPC activity.


Subject(s)
Fatty Acids/metabolism , Neural Stem Cells/metabolism , Animals , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/metabolism , Cell Cycle , Cell Proliferation , Hippocampus/enzymology , Malonyl Coenzyme A/metabolism , Mice, Knockout , Neural Stem Cells/cytology , Neural Stem Cells/enzymology , Neurogenesis , Oxidation-Reduction
14.
Brain Plast ; 3(1): 61-71, 2017 Nov 09.
Article in English | MEDLINE | ID: mdl-29765860

ABSTRACT

Neural stem/progenitor cells (NSPCs) give rise to billions of cells during development and are critical for proper brain formation. The finding that NSPCs persist throughout adulthood has challenged the view that the brain has poor regenerative abilities and raised hope for stem cell-based regenerative therapies. For decades there has been a strong movement towards understanding the requirements of NSPCs and their regulation, resulting in the discovery of many transcription factors and signaling pathways that can influence NSPC behavior and neurogenesis. However, the role of metabolism for NSPC regulation has only gained attention recently. Lipid metabolism in particular has been shown to influence proliferation and neurogenesis, offering exciting new possible mechanisms of NSPC regulation, as lipids are not only the building blocks of membranes, but can also act as alternative energy sources and signaling entities. Here I review the recent literature examining the role of lipid metabolism for NSPC regulation and neurogenesis.

15.
Nature ; 542(7639): 49-54, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28024299

ABSTRACT

Lymphatic vessels are lined by lymphatic endothelial cells (LECs), and are critical for health. However, the role of metabolism in lymphatic development has not yet been elucidated. Here we report that in transgenic mouse models, LEC-specific loss of CPT1A, a rate-controlling enzyme in fatty acid ß-oxidation, impairs lymphatic development. LECs use fatty acid ß-oxidation to proliferate and for epigenetic regulation of lymphatic marker expression during LEC differentiation. Mechanistically, the transcription factor PROX1 upregulates CPT1A expression, which increases acetyl coenzyme A production dependent on fatty acid ß-oxidation. Acetyl coenzyme A is used by the histone acetyltransferase p300 to acetylate histones at lymphangiogenic genes. PROX1-p300 interaction facilitates preferential histone acetylation at PROX1-target genes. Through this metabolism-dependent mechanism, PROX1 mediates epigenetic changes that promote lymphangiogenesis. Notably, blockade of CPT1 enzymes inhibits injury-induced lymphangiogenesis, and replenishing acetyl coenzyme A by supplementing acetate rescues this process in vivo.


Subject(s)
Fatty Acids/chemistry , Fatty Acids/metabolism , Lymphangiogenesis , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Acetates/pharmacology , Acetyl Coenzyme A/metabolism , Acetylation/drug effects , Animals , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epigenesis, Genetic , Female , Histones/metabolism , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lymphangiogenesis/drug effects , Lymphangiogenesis/genetics , Lymphatic Vessels/drug effects , Mice , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Protein Biosynthesis , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Umbilical Arteries/cytology , Up-Regulation
16.
Curr Opin Neurobiol ; 42: 45-52, 2017 02.
Article in English | MEDLINE | ID: mdl-27915086

ABSTRACT

The generation of neurons in the developing and adult mammalian brain by neural stem/progenitor cells (NSPCs) depends on a tight control of NSPC activity and neuronal differentiation that is regulated by a plethora of intrinsic and extrinsic molecular cues. Besides well-studied morphogenic signaling pathways and transcriptional codes that govern the distinct developmental steps from the dividing NSPC to a functional neuron, a critical role of cellular metabolism to determine the functional properties of NSPCs and newborn neurons has been recently identified. Here, we review advances in our understanding of how metabolism affects NSPC behavior and subsequent neuronal differentiation and suggest how metabolism may serve as a common signal integrator to ensure life-long addition of new neurons in the mammalian brain.


Subject(s)
Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Animals , Brain/cytology , Brain/embryology , Brain/metabolism , Humans , Neurons/cytology , Neurons/metabolism , Signal Transduction
17.
Stem Cell Reports ; 3(5): 735-42, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25418721

ABSTRACT

Proliferation of neural stem/progenitor cells (NSPCs) in the adult brain is tightly controlled to prevent exhaustion and to ensure proper neurogenesis. Several extrinsic stimuli affect NSPC regulation. However, the lack of unique markers led to controversial results regarding the in vivo behavior of NSPCs to different stimuli. We recently identified SPOT14, which controls NSPC proliferation through regulation of de novo lipogenesis, selectively in low-proliferating NSPCs. Whether SPOT14-expressing (SPOT14+) NSPCs react in vivo to neurogenic regulators is not known. We show that aging is accompanied by a marked disappearance of SPOT14+ NSPCs, whereas running, a positive neurogenic stimulus, increases proliferation of SPOT14+ NSPCs. Furthermore, transient depletion of highly proliferative cells recruits SPOT14+ NSPCs into the proliferative pool. Additionally, we have established endogenous SPOT14 protein staining, reflecting transgenic SPOT14-GFP expression. Thus, our data identify SPOT14 as a potent marker for adult NSPCs that react dynamically to positive and negative neurogenic regulators.


Subject(s)
Hippocampus/metabolism , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Age Factors , Animals , Antineoplastic Agents, Alkylating/pharmacology , Biomarkers/metabolism , Cell Proliferation/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Hippocampus/growth & development , Immunohistochemistry , Mice, Transgenic , Microscopy, Fluorescence , Neurogenesis/drug effects , Nuclear Proteins/genetics , Temozolomide , Transcription Factors/genetics
18.
Development ; 140(2): 459-70, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23193167

ABSTRACT

Stem cell self-renewal, commitment and reprogramming rely on a poorly understood coordination of cell cycle progression and execution of cell fate choices. Using existing experimental paradigms, it has not been possible to probe this relationship systematically in live stem cells in vitro or in vivo. Alterations in stem cell cycle kinetics probably occur long before changes in phenotypic markers are apparent and could be used as predictive parameters to reveal changes in stem cell fate. To explore this intriguing concept, we developed a single-cell tracking approach that enables automatic detection of cell cycle phases in live (stem) cells expressing fluorescent ubiquitylation-based cell-cycle indicator (FUCCI) probes. Using this tool, we have identified distinctive changes in lengths and fluorescence intensities of G1 (red fluorescence) and S/G2-M (green) that are associated with self-renewal and differentiation of single murine neural stem/progenitor cells (NSCs) and embryonic stem cells (ESCs). We further exploited these distinctive features using fluorescence-activated cell sorting to select for desired stem cell fates in two challenging cell culture settings. First, as G1 length was found to nearly double during NSC differentiation, resulting in progressively increasing red fluorescence intensity, we successfully purified stem cells from heterogeneous cell populations by their lower fluorescence. Second, as ESCs are almost exclusively marked by the green (S/G2-M) FUCCI probe due to their very short G1, we substantially augmented the proportion of reprogramming cells by sorting green cells early on during reprogramming from a NSC to an induced pluripotent stem cell state. Taken together, our studies begin to shed light on the crucial relationship between cell cycle progression and fate choice, and we are convinced that the presented approach can be exploited to predict and manipulate cell fate in a wealth of other mammalian cell systems.


Subject(s)
Cell Lineage , Embryonic Stem Cells/cytology , Stem Cells/cytology , Animals , Cell Cycle , Cell Differentiation , Cell Division , Cell Separation , Crosses, Genetic , Developmental Biology/methods , Flow Cytometry , Heterozygote , Kinetics , Mice , Mice, Inbred C57BL , Microscopy/methods , Neurons/metabolism
19.
Nature ; 493(7431): 226-30, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23201681

ABSTRACT

Mechanisms controlling the proliferative activity of neural stem and progenitor cells (NSPCs) have a pivotal role to ensure life-long neurogenesis in the mammalian brain. How metabolic programs are coupled with NSPC activity remains unknown. Here we show that fatty acid synthase (Fasn), the key enzyme of de novo lipogenesis, is highly active in adult NSPCs and that conditional deletion of Fasn in mouse NSPCs impairs adult neurogenesis. The rate of de novo lipid synthesis and subsequent proliferation of NSPCs is regulated by Spot14, a gene previously implicated in lipid metabolism, that we found to be selectively expressed in low proliferating adult NSPCs. Spot14 reduces the availability of malonyl-CoA, which is an essential substrate for Fasn to fuel lipogenesis. Thus, we identify here a functional coupling between the regulation of lipid metabolism and adult NSPC proliferation.


Subject(s)
Adult Stem Cells/metabolism , Fatty Acid Synthases/metabolism , Lipogenesis , Neural Stem Cells/metabolism , Adult Stem Cells/cytology , Animals , Cell Proliferation , Dentate Gyrus/metabolism , Fatty Acid Synthases/deficiency , Fatty Acid Synthases/genetics , Gene Expression Profiling , Hippocampus/cytology , Hippocampus/metabolism , Malonyl Coenzyme A/metabolism , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neurogenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
20.
J Neurosci ; 32(10): 3376-87, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22399759

ABSTRACT

Neural stem cells (NSCs) generate neurons throughout life in the hippocampal dentate gyrus (DG). How gene expression signatures differ among NSCs and immature neurons remains largely unknown. We isolated NSCs and their progeny in the adult DG using transgenic mice expressing a GFP reporter under the control of the Sox2 promoter (labeling NSCs) and transgenic mice expressing a DsRed reporter under the control of the doublecortin (DCX) promoter (labeling immature neurons). Transcriptome analyses revealed distinct gene expression profiles between NSCs and immature neurons. Among the genes that were expressed at significantly higher levels in DG NSCs than in immature neurons was the growth factor insulin-like growth factor 2 (IGF2). We show that IGF2 selectively controls proliferation of DG NSCs in vitro and in vivo through AKT-dependent signaling. Thus, by gene expression profiling of NSCs and their progeny, we have identified IGF2 as a novel regulator of adult neurogenesis.


Subject(s)
Adult Stem Cells/physiology , Cell Differentiation/genetics , Gene Expression Profiling/methods , Hippocampus/physiology , Insulin-Like Growth Factor II/physiology , Neural Stem Cells/physiology , Neurogenesis/genetics , Adult Stem Cells/cytology , Animals , Cells, Cultured , Doublecortin Protein , Female , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/cytology , Neurons/cytology , Neurons/physiology , Transcriptome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...